1
|
Xu H, Yao Q, Hu X, Zheng D, Ren C, Ren Z, Gao Y. On-Membrane Supramolecular Assemblies Serving as Bioorthogonal Gating for Melphalan. Angew Chem Int Ed Engl 2025:e202502922. [PMID: 40272883 DOI: 10.1002/anie.202502922] [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: 02/05/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025]
Abstract
Covalent drugs have experienced a revival in recent decades due to their advantageous pharmacodynamic profiles and targeting of "undruggable" proteins. However, balancing selectivity, reactivity, and potency is essential for safe and effective drugs. Here, we employ a cell-selective bioorthogonal prodrug design to enhance the selectivity for covalent inhibitors without compromising the reactivity and potency. The upregulation of phosphatase and integrin facilitates the formation of enzyme-instructed supramolecular assemblies (EISA) on the cancer cell membrane. These assemblies localize bioorthogonal reaction handles tetrazine (Tz), which liberate Melphalan from its bioorthogonal prodrug TCO-Mel. The TCO modification disrupts the LAT1-mediated transportation, reducing cellular permeability of TCO-Mel and the corresponding cytotoxicity to normal cells. Although the cell-selective on-membrane assemblies directed prodrug activation restores Melphalan influx to inhibit cancer cell growth. This prodrug activation strategy further demonstrates potent tumor suppression with satisfactory biocompatibility in vivo. Overall, we extend the scope of bioorthogonal prodrug design for covalent drugs via regulating cellular influx of active pharmaceutical ingredients (APIs).
Collapse
Affiliation(s)
- Hanlin Xu
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qingxin Yao
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoqian Hu
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Debin Zheng
- Medicine Medical Innovation Research Division of the Chinese PLA General Hospital, Beijing, 100853, China
| | - Chao Ren
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhibin Ren
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yuan Gao
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
2
|
Yamashita K, Ito A, Ishida M, Shintani Y, Ikeda M, Hadano S, Izumi M, Ochi R. Stereoisomerism-dependent gelation and crystal structures of glycosylated N-methylbromomaleimide-based supramolecular hydrogels. SOFT MATTER 2025. [PMID: 39981688 DOI: 10.1039/d4sm01325e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
In this study, we developed glycosylated N-methylbromomaleimide-based supramolecular hydrogels exhibiting colour change along with gel-sol transition and found that the stereoisomerism of the saccharide residue affects their gelation ability. Single-crystal X-ray diffraction analysis revealed the molecular packing and hydrogen-bonding networks contributed by the saccharide residues. Interestingly, it was found that water molecules were incorporated into the hydrogen-bonding network in the crystals of the compounds that showed gelation ability.
Collapse
Affiliation(s)
- Kotoyo Yamashita
- Graduate School of Integrated Arts and Sciences, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan.
| | - Akitaka Ito
- School of Engineering Science, Kochi University of Technology, Kami, Kochi 782-8502, Japan
- Research Center for Molecular Design, Kochi University of Technology, Kami, Kochi 782-8502, Japan
| | - Masashi Ishida
- Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| | - Yuki Shintani
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Masato Ikeda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Innovation Research Center for Quantum Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Shingo Hadano
- Graduate School of Integrated Arts and Sciences, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan.
- Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
- Research and Education Faculty, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| | - Masayuki Izumi
- Graduate School of Integrated Arts and Sciences, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan.
- Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
- Research and Education Faculty, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| | - Rika Ochi
- Graduate School of Integrated Arts and Sciences, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan.
- Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
- Research and Education Faculty, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| |
Collapse
|
3
|
Bigo-Simon A, Estrozi LF, Chaumont A, Schurhammer R, Schoehn G, Combet J, Schmutz M, Schaaf P, Jierry L. 3D Cryo-Electron Microscopy Reveals the Structure of a 3-Fluorenylmethyloxycarbonyl Zipper Motif Ensuring the Self-Assembly of Tripeptide Nanofibers. ACS NANO 2024; 18:30448-30462. [PMID: 39441741 DOI: 10.1021/acsnano.4c08043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Short peptide-based supramolecular hydrogels appeared as highly interesting materials for applications in many fields. The optimization of their properties relies mainly on the design of a suitable hydrogelator through an empirical trial-and-error strategy based on the synthesis of various types of peptides. This approach is in part due to the lack of prior structural knowledge of the molecular architecture of the various families of nanofibers. The 3D structure of the nanofibers determines their ability to interact with entities present in their surrounding environment. Thus, it is important to resolve the internal structural organization of the material. Herein, using Fmoc-FFY tripeptide as a model amphiphilic hydrogelator and cryo-EM reconstruction approach, we succeeded to obtain a 3.8 Å resolution 3D structure of a self-assembled nanofiber with a diameter of approximately 4.1 nm and with apparently "infinite" length. The elucidation of the spatial organization of such nano-objects addresses fundamental questions about the way short amphiphilic N-Fmoc peptides lacking secondary structure can self-assemble and ensure the cohesion of such a lengthy nanostructure. This nanofiber is organized into a triple-stranded helix with an asymmetric unit composed of two Fmoc-FFY peptides per strand. The three identical amphiphilic strands are maintained together by strong lateral interactions coming from a 3-Fmoc zipper motif. This hydrophobic core of the nanofiber is surrounded by 12 phenyl groups from phenylalanine residues, nonplanar with the six Fmoc groups. Polar tyrosine residues at the C-term position constitute the hydrophilic shell and are exposed all around the external part of the assembly. This fiber has a highly hydrophobic central core with an internal diameter of only 2.4 Å. Molecular dynamics simulations highlight van der Waals and hydrogen bonds between peptides placed on top of each other. We demonstrate that the self-assembly of Fmoc-FFY, whether induced by annealing or by the action of a phosphatase on the phosphorylated precursor Fmoc-FFpY, results in two nanostructures with minor differences that we are unable to distinguish.
Collapse
Affiliation(s)
- Alexis Bigo-Simon
- CNRS, Institut Charles Sadron (UPR22), Université de Strasbourg, 23 rue du Loess, BP 84047,Strasbourg Cedex 2 67034, France
- Faculté de Chimie, Université de Strasbourg, UMR7140, 1 rue Blaise Pascal, Strasbourg Cedex 67008, France
| | - Leandro F Estrozi
- CNRS, CEA, IBSUniversité de Grenoble Alpes, Grenoble F-38000, France
| | - Alain Chaumont
- Faculté de Chimie, Université de Strasbourg, UMR7140, 1 rue Blaise Pascal, Strasbourg Cedex 67008, France
| | - Rachel Schurhammer
- Faculté de Chimie, Université de Strasbourg, UMR7140, 1 rue Blaise Pascal, Strasbourg Cedex 67008, France
| | - Guy Schoehn
- CNRS, CEA, IBSUniversité de Grenoble Alpes, Grenoble F-38000, France
| | - Jérôme Combet
- CNRS, Institut Charles Sadron (UPR22), Université de Strasbourg, 23 rue du Loess, BP 84047,Strasbourg Cedex 2 67034, France
| | - Marc Schmutz
- CNRS, Institut Charles Sadron (UPR22), Université de Strasbourg, 23 rue du Loess, BP 84047,Strasbourg Cedex 2 67034, France
| | - Pierre Schaaf
- CNRS, Institut Charles Sadron (UPR22), Université de Strasbourg, 23 rue du Loess, BP 84047,Strasbourg Cedex 2 67034, France
- INSERM Unite 1121, CRBSInstitut National de la Santé et de la Recherche Médicale, 1 rue Eugène Boeckel, Strasbourg 67000, France
- Faculté de Chirurgie Dentaire, Université de Strasbourg, 8 rue Sainte Elisabeth, Strasbourg 67000, France
| | - Loïc Jierry
- CNRS, Institut Charles Sadron (UPR22), Université de Strasbourg, 23 rue du Loess, BP 84047,Strasbourg Cedex 2 67034, France
| |
Collapse
|
4
|
Chabatake Y, Tanigawa T, Hirayama Y, Taniguchi R, Ito A, Takahashi K, Noro SI, Akutagawa T, Nakamura T, Izumi M, Ochi R. A 15-crown-5-ether-based supramolecular hydrogel with selection ability for potassium cations via gelation and colour change. SOFT MATTER 2024; 20:8170-8173. [PMID: 39291588 DOI: 10.1039/d4sm00906a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
We developed a novel supramolecular hydrogelator possessing a benzo-15-crown-5 (B15C5) moiety. The hydrogelator can detect colourless potassium cations (K+) via easily readable gelation and colour change arising from a change in the molecular assembling ability through host-guest interactions between B15C5 and K+, which afford a B15C5/K+/B15C5 sandwich complex.
Collapse
Affiliation(s)
- Yuta Chabatake
- Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan.
| | - Tomoki Tanigawa
- Graduate School of Integrated Arts and Sciences, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| | - Yuto Hirayama
- Graduate School of Integrated Arts and Sciences, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| | - Ryo Taniguchi
- Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan.
| | - Akitaka Ito
- School of Engineering Science, Kochi University of Technology, Kami, Kochi 782-8502, Japan
- Research Center for Molecular Design, Kochi University of Technology, Kami, Kochi 782-8502, Japan
| | - Kiyonori Takahashi
- Research Institute for Electronic Science (RIES), Hokkaido University, N20W10, Kita-ku, Sapporo 001-0020, Japan
| | - Shin-Ichiro Noro
- Faculty of Environmental Earth Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Tomoyuki Akutagawa
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - Takayoshi Nakamura
- Research Institute for Electronic Science (RIES), Hokkaido University, N20W10, Kita-ku, Sapporo 001-0020, Japan
- Department of Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-3-1, Kagamiyama, Higashi-hiroshima 739-8526, Japan
| | - Masayuki Izumi
- Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan.
- Graduate School of Integrated Arts and Sciences, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
- Research and Education Faculty, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| | - Rika Ochi
- Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan.
- Graduate School of Integrated Arts and Sciences, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
- Research and Education Faculty, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| |
Collapse
|
5
|
Xiao X, Huang J. Enzyme-Responsive Supramolecular Self-Assembly in Small Amphiphiles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024. [PMID: 39018035 DOI: 10.1021/acs.langmuir.4c01762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Enzyme-responsive molecular assemblies have recently made remarkable progress, owing to their widespread applications. As a class of catalysts with high specificity and efficiency, enzymes play a critical role in producing new molecules and maintaining metabolic stability in living organisms. Therefore, the study of enzyme-responsive assembly aids in understanding the origin of life and the physiological processes occurring within living bodies, contributing to further advancements across various disciplines. In this Review, we summarize three kinds of enzyme-responsive assembly systems in amphiphiles: enzyme-triggered assembly, disassembly, and structural transformation. Furthermore, motivated by the fact that biological macromolecules and complex structures all originated with small molecules, our focus lies on the small amphiphiles (e.g., peptides, surfactants, fluorescent molecules, and drug molecules). We also provide an outlook on the potential of enzyme-responsive assembly systems for biomimetic development and hope this Review will attract more attention to this emerging research branch at the intersection of assembly chemistry and biological science.
Collapse
Affiliation(s)
- Xiao Xiao
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, Nanjing 211816, PR China
| | - Jianbin Huang
- Beijing National Laboratory for Molecular Sciences (BNLMS), State Key Laboratory for Structural Chemistry of Unstable and Stable Species, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, PR China
| |
Collapse
|
6
|
Zhang W, Zeng Y, Xiao Q, Wu Y, Liu J, Wang H, Luo Y, Zhan J, Liao N, Cai Y. An in-situ peptide-antibody self-assembly to block CD47 and CD24 signaling enhances macrophage-mediated phagocytosis and anti-tumor immune responses. Nat Commun 2024; 15:5670. [PMID: 38971872 PMCID: PMC11227529 DOI: 10.1038/s41467-024-49825-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 06/20/2024] [Indexed: 07/08/2024] Open
Abstract
Targeted immunomodulation for reactivating innate cells, especially macrophages, holds great promise to complement current adaptive immunotherapy. Nevertheless, there is still a lack of high-performance therapeutics for blocking macrophage phagocytosis checkpoint inhibitors in solid tumors. Herein, a peptide-antibody combo-supramolecular in situ assembled CD47 and CD24 bi-target inhibitor (PAC-SABI) is described, which undergoes biomimetic surface propagation on cancer cell membranes through ligand-receptor binding and enzyme-triggered reactions. By simultaneously blocking CD47 and CD24 signaling, PAC-SABI enhances the phagocytic ability of macrophages in vitro and in vivo, promoting anti-tumor responses in breast and pancreatic cancer mouse models. Moreover, building on the foundation of PAC-SABI-induced macrophage repolarization and increased CD8+ T cell tumor infiltration, sequential anti-PD-1 therapy further suppresses 4T1 tumor progression, prolonging survival rate. The in vivo construction of PAC-SABI-based nano-architectonics provides an efficient platform for bridging innate and adaptive immunity to maximize therapeutic potency.
Collapse
Affiliation(s)
- Weiqi Zhang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Breast Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yinghua Zeng
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qiuqun Xiao
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuanyuan Wu
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiale Liu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haocheng Wang
- Department of Gastrointestinal Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuting Luo
- Department of Breast Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jie Zhan
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Ning Liao
- Department of Breast Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Yanbin Cai
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Ding Y, Zhang S, Li W, Chen X, Li J, Zhang X, Zhang Z, Hu Y, Yang Z, Hu ZW, Shen X. Enzyme-Instructed Photoactivatable Supramolecular Antigens on Cancer Cell Membranes for Precision-Controlled T-Cell-Based Cancer Immunotherapy. NANO LETTERS 2024. [PMID: 38838340 DOI: 10.1021/acs.nanolett.4c01587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Cancer immunotherapies based on cytotoxic CD8+ T lymphocytes (CTLs) are highly promising for cancer treatment. The specific interaction between T-cell receptors and peptide-MHC-I complexes (pMHC-I) on cancer cell membranes critically determines their therapeutic outcomes. However, the lack of appropriate endogenous antigens for MHC-I presentation disables tumor recognition by CTLs. By devising three antigen-loaded self-assembling peptides of pY-K(Ag)-ERGD, pY-K(Ag)-E, and Y-K(Ag)-ERGD to noncovalently generate light-activatable supramolecular antigens at tumor sites in different manners, we report pY-K(Ag)-ERGD as a promising candidate to endow tumor cells with pMHC-I targets on demand. Specifically, pY-K(Ag)-ERGD first generates low-antigenic supramolecular antigens on cancer cell membranes, and a successive light pulse allows antigen payloads to efficiently release from the supramolecular scaffold, directly producing antigenic pMHC-I. Intravenous administration of pY-K(Ag)-ERGD enables light-controlled tumor inhibition when combined with adoptively transferred antigen-specific CTLs. Our strategy is feasible for broadening tumor antigen repertoires for T-cell immunotherapies and advancing precision-controlled T-cell immunotherapies.
Collapse
Affiliation(s)
- Yinghao Ding
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, P. R. China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Shengyi Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Wei Li
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Xiaodong Chen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, P. R. China
| | - Jun Li
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Xiangyang Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Zhenghao Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Yuanbo Hu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, P. R. China
| | - Zhimou Yang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, P. R. China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Zhi-Wen Hu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, P. R. China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Xian Shen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, P. R. China
| |
Collapse
|
8
|
Zeng Y, Liao X, Guo Y, Liu F, Bu F, Zhan J, Zhang J, Cai Y, Shen M. Baicalin-peptide supramolecular self-assembled nanofibers effectively inhibit ferroptosis and attenuate doxorubicin-induced cardiotoxicity. J Control Release 2024; 366:838-848. [PMID: 38145663 DOI: 10.1016/j.jconrel.2023.12.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
Doxorubicin, an anthracycline chemotherapeutic agent, elicits a deleterious cardiotoxicity known as doxorubicin-induced cardiomyopathy (DIC) that circumscribes its chemotherapy utility for malignancies. Recent empirical evidence implicates ferroptosis, an iron-dependent form of regulated cell death, as playing a pivotal role in the pathogenesis of DIC. We postulated that anti-ferroptosis agents may constitute a novel therapeutic strategy for mitigating DIC. To test this hypothesis, we engineered baicalin-peptide supramolecular self-assembled nanofibers designed to selectively target the angiotensin II type I receptor (AT1R), which is upregulated in doxorubicin-damaged cardiomyocytes. This enabled targeted delivery of baicalin, a natural antioxidant compound, to inhibit ferroptosis in the afflicted myocardium. In vitro, the nanofibers ameliorated cardiomyocyte death by attenuating peroxide accumulation and suppressing ferroptosis. In a murine model of DIC, AT1R-targeted baicalin delivery resulted in efficacious cardiac accumulation and superior therapeutic effects compared to systemic administration. This investigation delineates a promising framework for developing targeted therapies that alleviate doxorubicin-induced cardiotoxicity by inhibiting the ferroptosis pathway in cardiomyocytes.
Collapse
Affiliation(s)
- Yinghua Zeng
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xu Liao
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuting Guo
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Cardiology, Hainan Hospital of Chinese PLA General Hosptial, Hainan Geriatric Disease Clinical Medical Research Center, Hainan Branch of China Geriatric Disease Clinical Research Center, Sanya, China
| | - Fengjiao Liu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Bu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Cardiology, Hainan Hospital of Chinese PLA General Hosptial, Hainan Geriatric Disease Clinical Medical Research Center, Hainan Branch of China Geriatric Disease Clinical Research Center, Sanya, China
| | - Jie Zhan
- Department of Laboratory Medicine, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianwu Zhang
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China; Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yanbin Cai
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China.
| | - Mingzhi Shen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of General Practice, Hainan Hospital of Chinese PLA General Hosptial, Sanya, China.
| |
Collapse
|
9
|
Kim K, Park MH. Role of Functionalized Peptides in Nanomedicine for Effective Cancer Therapy. Biomedicines 2024; 12:202. [PMID: 38255307 PMCID: PMC10813321 DOI: 10.3390/biomedicines12010202] [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: 12/27/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Peptide-functionalized nanomedicine, which addresses the challenges of specificity and efficacy in drug delivery, is emerging as a pivotal approach for cancer therapy. Globally, cancer remains a leading cause of mortality, and conventional treatments, such as chemotherapy, often lack precision and cause adverse effects. The integration of peptides into nanomedicine offers a promising solution for enhancing the targeting and delivery of therapeutic agents. This review focuses on the three primary applications of peptides: cancer cell-targeting ligands, building blocks for self-assembling nanostructures, and elements of stimuli-responsive systems. Nanoparticles modified with peptides improved targeting of cancer cells, minimized damage to healthy tissues, and optimized drug delivery. The versatility of self-assembled peptide structures makes them an innovative vehicle for drug delivery by leveraging their biocompatibility and diverse nanoarchitectures. In particular, the mechanism of cell death induced by self-assembled structures offers a novel approach to cancer therapy. In addition, peptides in stimuli-responsive systems enable precise drug release in response to specific conditions in the tumor microenvironment. The use of peptides in nanomedicine not only augments the efficacy and safety of cancer treatments but also suggests new research directions. In this review, we introduce systems and functionalization methods using peptides or peptide-modified nanoparticles to overcome challenges in the treatment of specific cancers, including breast cancer, lung cancer, colon cancer, prostate cancer, pancreatic cancer, liver cancer, skin cancer, glioma, osteosarcoma, and cervical cancer.
Collapse
Affiliation(s)
- Kibeom Kim
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University, Seoul 01795, Republic of Korea;
- Department of Chemistry and Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| | - Myoung-Hwan Park
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University, Seoul 01795, Republic of Korea;
- Department of Chemistry and Life Science, Sahmyook University, Seoul 01795, Republic of Korea
- Department of Convergence Science, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
10
|
Chen J, Wang H, Long F, Bai S, Wang Y. Dynamic supramolecular hydrogels mediated by chemical reactions. Chem Commun (Camb) 2023; 59:14236-14248. [PMID: 37964743 DOI: 10.1039/d3cc04353c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Supramolecular self-assembly in a biological system is usually dominated by sophisticated metabolic processes (chemical reactions) such as catalysis of enzymes and consumption of high energy chemicals, leading to groups of biomolecules with unique dynamics and functions in an aqueous environment. In recent years, increasing efforts have been made to couple chemical reactions to molecular self-assembly, with the aim of creating supramolecular materials with lifelike properties and functions. In this feature article, after summarising the work of chemical reaction mediated supramolecular hydrogels, we first focus on a typical example where dynamic self-assembly of molecular hydrogels is activated by in situ formation of a hydrazone bond in water. We discuss how the formation of the hydrazone-based supramolecular hydrogels can be controlled in time and space. After that, we describe transient assembly of supramolecular hydrogels powered by out-of-equilibrium chemical reaction networks regulated by chemical fuels, which show unique properties such as finite lifetime, dynamic structures, and regenerative capabilities. Finally, we provide a perspective on the future investigations that need to be done urgently, which range from fundamental research to real-life applications of dynamic supramolecular hydrogels.
Collapse
Affiliation(s)
- Jingjing Chen
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China.
| | - Hucheng Wang
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China.
| | - Feng Long
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China.
| | - Shengyu Bai
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China.
| | - Yiming Wang
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China.
| |
Collapse
|
11
|
Xie L, Ding Y, Zhang X, Zhang Z, Zeng S, Wang L, Yang Z, Liu Q, Hu ZW. A Cascade-Targeted Enzyme-Instructed Peptide Self-Assembly Strategy for Cancer Immunotherapy through Boosting Immunogenic Cell Death. SMALL METHODS 2023; 7:e2201416. [PMID: 36965100 DOI: 10.1002/smtd.202201416] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/04/2023] [Indexed: 05/17/2023]
Abstract
Immunogenic cell death (ICD) approaches by encumbering mitochondrial functions provide great promise for the treatment of malignant tumors, but these kinds of ICD strategies are still in their infancy. Here, one multifunctional drug-loaded, cascade-targeted, and enzyme-instructed self-assembling peptide nanomedicine (Comp. 4) for ICD-based cancer therapy is constructed. Comp. 4 consists of 1) lonidamine (LND) that specifically interferes with mitochondrial functions; 2) a programmed death ligand 1 (PD-L1) binding peptide sequence (NTYYEDQG) and a mitochondria-specific motif (triphenylphosphonium, TPP) that can sequentially control the cell membrane and mitochondria targeting capacities, respectively; and 3) a -GD FD FpD Y- assembly core to in situ organize peptide assemblies responsive to alkaline phosphatase (ALP). Comp. 4 demonstrates noticeable structural and morphological transformations in the presence of ALP and produces peptide assemblies in mouse colon cancer cells (CT26) with high expressions of both ALP and PD-L1. Moreover, the presence of PD-L1- and mitochondria-specific motifs can assist Comp. 4 for effective endocytosis and endosomal escape, forming peptide assemblies and delivering LND into mitochondria. Consequently, Comp. 4 shows superior capacities to in vivo induce abundant mitochondrial oxidative stress, provoke robust ICD responses, and produce an immunogenic tumor microenvironment, successfully inhibiting CT26 tumor growth by eliciting a systemic ICD-based antitumor immunity.
Collapse
Affiliation(s)
- Limin Xie
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300071, P. R. China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Yinghao Ding
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Xiangyang Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Zhenghao Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Sheng Zeng
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300192, P. R. China
- School of Medicine, Nankai University, Tianjin, 300071, P. R. China
| | - Ling Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, 300071, P. R. China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300192, P. R. China
- School of Medicine, Nankai University, Tianjin, 300071, P. R. China
| | - Zhi-Wen Hu
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
12
|
Kaur H, Sharma P, Pal VK, Sen S, Roy S. Exploring Supramolecular Interactions between the Extracellular-Matrix-Derived Minimalist Bioactive Peptide and Nanofibrillar Cellulose for the Development of an Advanced Biomolecular Scaffold. ACS Biomater Sci Eng 2023; 9:1422-1436. [PMID: 36826412 DOI: 10.1021/acsbiomaterials.3c00014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
It has been increasingly evident over the last few years that bioactive peptide hydrogels in conjugation with polymer hydrogels are emerging as a new class of supramolecular materials suitable for various biomedical applications owing to their specificity, tunability, and nontoxicity toward the biological system. Despite their unique biocompatible features, both polymer- and peptide-based scaffolds suffer from certain limitations, which restrict their use toward developing efficient matrices for controlling cellular behavior. The peptide hydrogels usually form soft matrices with low mechanical strength, whereas most of the polymer hydrogels lack biofunctionality. In this direction, combining polymers with peptides to develop a conjugate hydrogel can be explored as an emergent approach to overcome the limitations of the individual components. The polymer will provide high mechanical strength, whereas the biofunctionality of the material can be induced by the bioactive peptide sequence. In this study, we utilized TEMPO-oxidized nanofibrillar cellulose as the polymer counterpart, which was co-assembled with a short N-cadherin mimetic bioactive peptide sequence, Nap-HAVDI, to fabricate an NFC-peptide conjugate hydrogel. Interestingly, the mechanical strength of the peptide hydrogel was found to be significantly improved by combining the peptide with the NFC in the conjugate hydrogel. The addition of the peptide into the NFC also reduced the pore size within NFC matrices, which further helped in improving cellular adhesion, survival, and proliferation. Furthermore, the cells grown on the NFC and NFC-peptide hybrid hydrogel demonstrated normal expression of cytoskeleton proteins, i.e., β-tubulin in C6 cells and actin in L929 cells, respectively. The selective response of neuronal cells toward the specific bioactive peptide was further observed through a protein expression study. Thus, our study demonstrated the collective role of the cellulose-peptide composite material that revealed superior physical properties and biological response of this composite scaffold, which may open up a new platform for biomedical applications.
Collapse
Affiliation(s)
- Harsimran Kaur
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab 140306, India
| | - Pooja Sharma
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab 140306, India
| | - Vijay K Pal
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab 140306, India
| | - Sourav Sen
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab 140306, India
| | - Sangita Roy
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab 140306, India
| |
Collapse
|
13
|
Hamley IW. Self-Assembly, Bioactivity, and Nanomaterials Applications of Peptide Conjugates with Bulky Aromatic Terminal Groups. ACS APPLIED BIO MATERIALS 2023; 6:384-409. [PMID: 36735801 PMCID: PMC9945136 DOI: 10.1021/acsabm.2c01041] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The self-assembly and structural and functional properties of peptide conjugates containing bulky terminal aromatic substituents are reviewed with a particular focus on bioactivity. Terminal moieties include Fmoc [fluorenylmethyloxycarbonyl], naphthalene, pyrene, naproxen, diimides of naphthalene or pyrene, and others. These provide a driving force for self-assembly due to π-stacking and hydrophobic interactions, in addition to the hydrogen bonding, electrostatic, and other forces between short peptides. The balance of these interactions leads to a propensity to self-assembly, even for conjugates to single amino acids. The hybrid molecules often form hydrogels built from a network of β-sheet fibrils. The properties of these as biomaterials to support cell culture, or in the development of molecules that can assemble in cells (in response to cellular enzymes, or otherwise) with a range of fascinating bioactivities such as anticancer or antimicrobial activity, are highlighted. In addition, applications of hydrogels as slow-release drug delivery systems and in catalysis and other applications are discussed. The aromatic nature of the substituents also provides a diversity of interesting optoelectronic properties that have been demonstrated in the literature, and an overview of this is also provided. Also discussed are coassembly and enzyme-instructed self-assembly which enable precise tuning and (stimulus-responsive) functionalization of peptide nanostructures.
Collapse
|
14
|
Chen K, Wu X, Wang Q, Wang Y, Zhang H, Zhao S, Li C, Hu Z, Yang Z, Li L. The protective effects of a d-tetra-peptide hydrogel adjuvant vaccine against H7N9 influenza virus in mice. CHINESE CHEM LETT 2023; 34:107446. [DOI: 10.1016/j.cclet.2022.04.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Jeena MT, Jin S, Jana B, Ryu JH. Enzyme-instructed morphology transformation of mitochondria-targeting peptide for the selective eradication of osteosarcoma. RSC Chem Biol 2022; 3:1416-1421. [PMID: 36544576 PMCID: PMC9709777 DOI: 10.1039/d2cb00166g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022] Open
Abstract
The treatment of osteosarcoma involves an adjuvant therapy that combines surgery and chemotherapy. However, considering that children are the main victims of osteosarcoma, replacing such a harsh treatment with a soft but powerful method that ensures a complete cure while having no adverse effects is highly desirable. To achieve this aim, we have developed a supramolecular therapeutic strategy based on morphology-transformable mitochondria-targeting peptides for the eradication of osteosarcoma with enhanced selectivity and reduced side effects. A newly designed micelle-forming amphiphilic peptide, l-Mito-FFYp, consisting of a phosphate substrate for the biomarker enzyme of osteosarcoma alkaline phosphatase (ALP), disassembles in response to the ALP enzyme in the cell membrane to generate positively charged l-Mito-FFY molecules, which diffuse inside the targeted cell and self-assemble to form nanostructures specifically inside the mitochondria to induce cell apoptosis.
Collapse
Affiliation(s)
- M. T. Jeena
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST)Ulsan 44919Republic of Korea
| | - Seongeon Jin
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST)Ulsan 44919Republic of Korea
| | - Batakrishna Jana
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST)Ulsan 44919Republic of Korea
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST)Ulsan 44919Republic of Korea
| |
Collapse
|
16
|
Sun S, Liang HW, Wang H, Zou Q. Light-Triggered Self-Assembly of Peptide Nanoparticles into Nanofibers in Living Cells through Molecular Conformation Changes and H-Bond Interactions. ACS NANO 2022; 16:18978-18989. [PMID: 36354757 DOI: 10.1021/acsnano.2c07895] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Controlled self-assembly has attracted extensive interest in biological and nanotechnological applications. Enzymatic or biocatalytic triggered self-assembly is widely used for the diagnostic and prognostic marker in different pathologies because of their nanostructures and biological effects. However, it remains a great challenge to control the self-assembly of peptides in living cells with a high degree of spatial and temporal precision. Here we demonstrate a light-triggered platform that enables spatiotemporal control of self-assembly from nanoparticles into nanofibers in living cells through subtle molecular conformational changes and internal H-bonding interactions. The platform contained 3-methylene-2-(quinolin-8-yl) isoindolin-1-one, which acts as the light-controlled unit to disrupt the hydrophilic/lipophilic balance through the change of molecular conformation, and a peptide that can be a faster recombinant to assemble via H-bonding interactions. The process has good biocompatibility because it does not involve waste generation or oxygen consumption; moreover, the assembly rate constant was fast and up to 0.17 min-1. It is applied to the regulation of molecular assembly in living cells. As such, our findings demonstrate that light-triggered controllable assembly can be applied for initiative regulating cellular behaviors in living systems.
Collapse
Affiliation(s)
- Si Sun
- National Engineering Research Center of Immunological Products, Third Military Medical University, Gaotanyan No. 30, Shapingba District, Chongqing, 400038, China
| | - Hong-Wen Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing100190, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing100190, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Third Military Medical University, Gaotanyan No. 30, Shapingba District, Chongqing, 400038, China
| |
Collapse
|
17
|
Lin F, Jia C, Wu FG. Intracellular Enzyme-Instructed Self-Assembly of Peptides (IEISAP) for Biomedical Applications. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196557. [PMID: 36235094 PMCID: PMC9571778 DOI: 10.3390/molecules27196557] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/13/2022]
Abstract
Despite the remarkable significance and encouraging breakthroughs of intracellular enzyme-instructed self-assembly of peptides (IEISAP) in disease diagnosis and treatment, a comprehensive review that focuses on this topic is still desirable. In this article, we carefully review the advances in the applications of IEISAP, including the development of various bioimaging techniques, such as fluorescence imaging, photoacoustic imaging, magnetic resonance imaging, positron-emission tomography imaging, radiation imaging, and multimodal imaging, which are successfully leveraged in visualizing cancer tissues and cells, bacteria, and enzyme activity. We also summarize the utilization of IEISAP in disease treatments, including anticancer, antibacterial, and antiinflammation applications, among others. We present the design, action modes, structures, properties, functions, and performance of IEISAP materials, such as nanofibers, nanoparticles, nanoaggregates, and hydrogels. Finally, we conclude with an outlook towards future developments of IEISAP materials for biomedical applications. It is believed that this review may foster the future development of IEISAP with better performance in the biomedical field.
Collapse
|
18
|
Zheng J, Song X, Yang Z, Yin C, Luo W, Yin C, Ni Y, Wang Y, Zhang Y. Self-assembly hydrogels of therapeutic agents for local drug delivery. J Control Release 2022; 350:898-921. [PMID: 36089171 DOI: 10.1016/j.jconrel.2022.09.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 10/14/2022]
Abstract
Advanced drug delivery systems are of vital importance to enhance therapeutic efficacy. Among various recently developed formulations, self-assembling hydrogels composed of therapeutic agents have shown promising potential for local drug delivery owing to their excellent biocompatibility, high drug-loading efficiency, low systemic toxicity, and sustained drug release behavior. In particular, therapeutic agents self-assembling hydrogels with well-defined nanostructures are beneficial for direct delivery to the target site via injection, not only improving drug availability, but also extending their retention time and promoting cellular uptake. In brief, the self-assembly approach offers better opportunities to improve the precision of pharmaceutical treatment and achieve superior treatment efficacies. In this review, we intend to cover the recent developments in therapeutic agent self-assembling hydrogels. First, the molecular structures, self-assembly mechanisms, and application of self-assembling hydrogels are systematically outlined. Then, we summarize the various self-assembly strategies, including the single therapeutic agent, metal-coordination, enzyme-instruction, and co-assembly of multiple therapeutic agents. Finally, the potential challenges and future perspectives are discussed. We hope that this review will provide useful insights into the design and preparation of therapeutic agent self-assembling hydrogels.
Collapse
Affiliation(s)
- Jun Zheng
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Xianwen Song
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Zhaoyu Yang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chao Yin
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Weikang Luo
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chunyang Yin
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yaqiong Ni
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yang Wang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Yi Zhang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China.
| |
Collapse
|
19
|
Mo Y, Danovich D, Shaik S. The roles of charge transfer and polarization in non-covalent interactions: a perspective from ab initio valence bond methods. J Mol Model 2022; 28:274. [PMID: 36006511 DOI: 10.1007/s00894-022-05187-8] [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: 08/14/2021] [Accepted: 12/03/2021] [Indexed: 11/28/2022]
Abstract
Noncovalent interactions are ubiquitous and have been well recognized in chemistry, biology and material science. Yet, there are still recurring controversies over their natures, due to the wide range of noncovalent interaction terms. In this Essay, we employed the Valence Bond (VB) methods to address two types of interactions which recently have drawn intensive attention, i.e., the halogen bonding and the CH‧‧‧HC dihydrogen bonding. The VB methods have the advantage of interpreting molecular structures and properties in the term of electron-localized Lewis (resonance) states (structures), which thereby shed specific light on the alteration of the bonding patterns. Due to the electron localization nature of Lewis states, it is possible to define individually and measure both polarization and charge transfer effects which have different physical origins. We demonstrated that both the ab initio VB method and the block-localized wavefunction (BLW) method can provide consistent pictures for halogen bonding systems, where strong Lewis bases NH3, H2O and NMe3 partake as the halogen bond acceptors, and the halogen bond donors include dihalogen molecules and XNO2 (X = Cl, Br, I). Based on the structural, spectral, and energetic changes, we confirm the remarkable roles of charge transfer in these halogen bonding complexes. Although the weak C-H∙∙∙H-C interactions in alkane dimers and graphene sheets are thought to involve dispersion only, we show that this term embeds delicate yet important charge transfer, bond reorganization and polarization interactions.
Collapse
Affiliation(s)
- Yirong Mo
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, 27401, USA.
| | - David Danovich
- Institute of Chemistry, The Hebrew University of Jerusalem, 9190407, Jerusalem, Israel
| | - Sason Shaik
- Institute of Chemistry, The Hebrew University of Jerusalem, 9190407, Jerusalem, Israel.
| |
Collapse
|
20
|
Zhou Z, Maxeiner K, Moscariello P, Xiang S, Wu Y, Ren Y, Whitfield CJ, Xu L, Kaltbeitzel A, Han S, Mücke D, Qi H, Wagner M, Kaiser U, Landfester K, Lieberwirth I, Ng DYW, Weil T. In Situ Assembly of Platinum(II)-Metallopeptide Nanostructures Disrupts Energy Homeostasis and Cellular Metabolism. J Am Chem Soc 2022; 144:12219-12228. [PMID: 35729777 PMCID: PMC9284552 DOI: 10.1021/jacs.2c03215] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Nanostructure-based functions are omnipresent in nature and essential for the diversity of life. Unlike small molecules, which are often inhibitors of enzymes or biomimetics with established methods of elucidation, we show that functions of nanoscale structures in cells are complex and can implicate system-level effects such as the regulation of energy and redox homeostasis. Herein, we design a platinum(II)-containing tripeptide that assembles into intracellular fibrillar nanostructures upon molecular rearrangement in the presence of endogenous H2O2. The formed nanostructures blocked metabolic functions, including aerobic glycolysis and oxidative phosphorylation, thereby shutting down ATP production. As a consequence, ATP-dependent actin formation and glucose metabolite-dependent histone deacetylase activity are downregulated. We demonstrate that assembly-driven nanomaterials offer a rich avenue to achieve broad-spectrum bioactivities that could provide new opportunities in drug discovery.
Collapse
Affiliation(s)
- Zhixuan Zhou
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Konrad Maxeiner
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | | | - Siyuan Xiang
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Yingke Wu
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Yong Ren
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | | | - Lujuan Xu
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | | | - Shen Han
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - David Mücke
- Central Facility of Materials Science Electron Microscopy, Universität Ulm, 89081 Ulm, Germany
| | - Haoyuan Qi
- Central Facility of Materials Science Electron Microscopy, Universität Ulm, 89081 Ulm, Germany.,Faculty of Chemistry and Food Chemistry & Center for Advancing Electronics Dresden (cfaed), Technische Universität Dresden, 01062 Dresden, Germany
| | - Manfred Wagner
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Ute Kaiser
- Central Facility of Materials Science Electron Microscopy, Universität Ulm, 89081 Ulm, Germany
| | | | | | - David Y W Ng
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Tanja Weil
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| |
Collapse
|
21
|
Mañas‐Torres MC, Illescas‐Lopez S, Gavira JA, de Cienfuegos LÁ, Marchesan S. Interactions Between Peptide Assemblies and Proteins for Medicine. Isr J Chem 2022. [DOI: 10.1002/ijch.202200018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Mari C. Mañas‐Torres
- Departamento de Química Orgánica, Facultad de Ciencias Unidad de Excelencia Química Aplicada a Biomedicina y Medioambiente (UEQ) Universidad de Granada, (UGR) C. U. Fuentenueva Avda. Severo Ochoa s/n E-18071 Granada
| | - Sara Illescas‐Lopez
- Departamento de Química Orgánica, Facultad de Ciencias Unidad de Excelencia Química Aplicada a Biomedicina y Medioambiente (UEQ) Universidad de Granada, (UGR) C. U. Fuentenueva Avda. Severo Ochoa s/n E-18071 Granada
| | - José A. Gavira
- Laboratorio de Estudios Cristalográficos Instituto Andaluz de Ciencias de la Tierra (Consejo Superior de Investigaciones Científicas-UGR) Avenida de las Palmeras 4 18100 Armilla, UEQ Granada Spain
| | - Luis Álvarez de Cienfuegos
- Departamento de Química Orgánica, Facultad de Ciencias Unidad de Excelencia Química Aplicada a Biomedicina y Medioambiente (UEQ) Universidad de Granada, (UGR) C. U. Fuentenueva Avda. Severo Ochoa s/n E-18071 Granada
- Instituto de Investigación Biosanitaria ibs Granada Spain
| | - Silvia Marchesan
- Chemical and Pharmaceutical Sciences Department University of Trieste Via L. Giorgieri 1 Trieste 34127 Italy
| |
Collapse
|
22
|
Localized Enzyme-Assisted Self-Assembly of low molecular weight hydrogelators. Mechanism, applications and perspectives. Adv Colloid Interface Sci 2022; 304:102660. [PMID: 35462266 DOI: 10.1016/j.cis.2022.102660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/28/2022] [Accepted: 03/31/2022] [Indexed: 01/31/2023]
Abstract
Nature uses systems of high complexity coordinated by the precise spatial and temporal control of associated processes, working from the molecular to the macroscopic scale. This living organization is mainly ensured by enzymatic actions. Herein, we review the concept of Localized Enzyme-Assisted Self-Assembly (LEASA). It is defined and presented as a straightforward and insightful strategy to achieve high levels of control in artificial systems. Indeed, the use of immobilized enzymes to drive self-assembly events leads not only to the local formation of supramolecular structures but also to tune their kinetics and their morphologies. The possibility to design tailored complex systems taking advantage of self-assembled networks through their inherent and emergent properties offers new perspectives for the design of novel, more adaptable materials. As a result, some applications have already been developed and are gathered in this review. Finally, challenges and perspectives of LEASA are introduced and discussed.
Collapse
|
23
|
Zhang Y, Tian X, Li X. Supramolecular assemblies of histidine-containing peptides with switchable hydrolase and peroxidase activities through Cu(II) binding and co-assembling. J Mater Chem B 2022; 10:3716-3722. [PMID: 35451448 DOI: 10.1039/d2tb00375a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Modulating enzyme activities or functionalities is one of the primary features of biological systems, which is, however, a great challenge for artificial enzyme systems. In this work, we designed and synthesized a series of self-assembling peptides from histidine and other amino acids (Asp, Ser, Lys or Arg), which exist in the active site of natural enzymes. These peptides could undergo a conformational transition from random coils to β-sheet structures under physiological conditions and formed self-assembled nanotubes with obvious hydrolase activities. After incorporation of transition metal ions such as Cu2+, these peptides could coordinate with Cu2+ ions, switch molecular conformations, and self-assemble into hybrid nanomaterials with altered morphologies and peroxidase-like activities. This work illustrates a facile approach for constructing artificial enzymes from self-assembling peptides with histidine residues whose catalytic functions could be modulated by incorporation of Cu2+ ions.
Collapse
Affiliation(s)
- Yue Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China.
| | - Xin Tian
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, 215123, China.
| | - Xinming Li
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China.
| |
Collapse
|
24
|
Guan T, Li J, Chen C, Liu Y. Self-Assembling Peptide-Based Hydrogels for Wound Tissue Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104165. [PMID: 35142093 PMCID: PMC8981472 DOI: 10.1002/advs.202104165] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/23/2021] [Indexed: 05/02/2023]
Abstract
Wound healing is a long-term, multistage biological process that includes hemostasis, inflammation, proliferation, and tissue remodeling and requires intelligent designs to provide comprehensive and convenient treatment. The complexity of wounds has led to a lack of adequate wound treatment materials, which must systematically regulate unique wound microenvironments. Hydrogels have significant advantages in wound treatment due to their ability to provide spatiotemporal control over the wound healing process. Self-assembling peptide-based hydrogels are particularly attractive due to their innate biocompatibility and biodegradability along with additional advantages including ligand-receptor recognition, stimulus-responsive self-assembly, and the ability to mimic the extracellular matrix. The ability of peptide-based materials to self-assemble in response to the physiological environment, resulting in functionalized microscopic structures, makes them conducive to wound treatment. This review introduces several self-assembling peptide-based systems with various advantages and emphasizes recent advances in self-assembling peptide-based hydrogels that allow for precise control during different stages of wound healing. Moreover, the development of multifunctional self-assembling peptide-based hydrogels that can regulate and remodel the wound immune microenvironment in wound therapy with spatiotemporal control has also been summarized. Overall, this review sheds light on the future clinical and practical applications of self-assembling peptide-based hydrogels.
Collapse
Affiliation(s)
- Tong Guan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Jiayang Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
- GBA National Institute for Nanotechnology InnovationGuangdong510700P. R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
- GBA National Institute for Nanotechnology InnovationGuangdong510700P. R. China
| |
Collapse
|
25
|
Kim BJ. Enzyme-Instructed Self-Assembly of Peptides: From Concept to Representative Applications. Chem Asian J 2022; 17:e202200094. [PMID: 35213091 DOI: 10.1002/asia.202200094] [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: 01/30/2022] [Revised: 02/23/2022] [Indexed: 11/11/2022]
Abstract
Enzyme-instructed self-assembly, integrating enzymatic reaction and molecular self-assembly, has drawn noticeable attention over the last decade with the intension of being used in valuable applications. Recent advances in the field allow it possible to spatiotemporally control peptide self-assembly in cellular milieu, broadening the potential applications of peptide assemblies to cancer therapy and subcellular delivery. In this minireview, the concept of enzyme-instructed self-assembly of peptide, containing enzymatic trigger and spatiotemporal control, is described. Representative applications in cells are also discussed, followed by outlook on the field of enzyme-instructed self-assembly.
Collapse
Affiliation(s)
- Beom Jin Kim
- University of Ulsan, Chemistry, 12, Techno Industrial Complex-ro, 55 beon-gil, 4776, Ulsan, KOREA, REPUBLIC OF
| |
Collapse
|
26
|
Wang BX, Xu W, Yang Z, Wu Y, Pi F. An Overview on Recent Progress of the Hydrogels: From Material Resources, Properties to Functional Applications. Macromol Rapid Commun 2022; 43:e2100785. [PMID: 35075726 DOI: 10.1002/marc.202100785] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/04/2022] [Indexed: 11/06/2022]
Abstract
Hydrogels, as the most typical elastomer materials with three-dimensional network structures, have attracted wide attention owing to their outstanding features in fields of sensitive stimulus response, low surface friction coefficient, good flexibility and bio-compatibility. Because of numerous fresh polymer materials (or polymerization monomers), hydrogels with various structure diversities and excellent properties are emerging, and the development of hydrogels is very vigorous over the past decade. This review focuses on state-of-the-art advances, systematically reviews the recent progress on construction of novel hydrogels utilized several kinds of typical polymerization monomers, and explores the main chemical and physical cross-linking methods to develop the diversity of hydrogels. Following the aspects mentioned above, the classification and emerging applications of hydrogels, such as pH response, ionic response, electrical response, thermal response, biomolecular response, and gas response, are extensively summarized. Finally, we have done this review with the promises and challenges for the future evolution of hydrogels and their biological applications. cross-linking methods; functional applications; hydrogels; material resources This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ben-Xin Wang
- School of Science, Jiangnan University, Wuxi, 214122, China
| | - Wei Xu
- School of Science, Jiangnan University, Wuxi, 214122, China
| | - Zhuchuang Yang
- School of Science, Jiangnan University, Wuxi, 214122, China
| | - Yangkuan Wu
- School of Science, Jiangnan University, Wuxi, 214122, China
| | - Fuwei Pi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
27
|
Huang F, Liu J, Liu Y. Engineering living cells with cucurbit[7]uril-based supramolecular polymer chemistry: from cell surface engineering to manipulation of subcellular organelles. Chem Sci 2022; 13:8885-8894. [PMID: 35975152 PMCID: PMC9350592 DOI: 10.1039/d2sc02797f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/04/2022] [Indexed: 12/20/2022] Open
Abstract
Supramolecular polymer chemistry, which closely integrates noncovalent interactions with polymeric structures, is a promising toolbox for living cell engineering. Here, we report our recent progress in exploring the applications of cucurbit[7]uril (CB[7])-based supramolecular polymer chemistry for engineering living cells. First, a modular polymer-analogous approach was established to prepare multifunctional polymers that contain CB[7]-based supramolecular recognition motifs. The supramolecular polymeric systems were successfully applied to cell surface engineering and subcellular organelle manipulation. By anchoring polymers on the cell membranes, cell–cell interactions were established by CB[7]-based host–guest recognition, which further facilitated heterogeneous cell fusion. In addition to cell surface engineering, placing the multifunctional polymers on specific subcellular organelles, including the mitochondria and endoplasmic reticulum, has led to enhanced physical contact between subcellular organelles. It is highly anticipated that the CB[7]-based supramolecular polymer chemistry will provide a new strategy for living cell engineering to advance the development of cell-based therapeutic materials. Cucurbit[7]uril-based supramolecular polymer chemistry, which closely integrates host–guest recognition with multifunctional polymeric structures, is a promising toolbox for living cell engineering.![]()
Collapse
Affiliation(s)
- Fang Huang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Jiaxiong Liu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Yiliu Liu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
28
|
Tong KC, Wan PK, Lok CN, Che CM. Dynamic supramolecular self-assembly of platinum(ii) complexes perturbs an autophagy-lysosomal system and triggers cancer cell death. Chem Sci 2021; 12:15229-15238. [PMID: 34976343 PMCID: PMC8635173 DOI: 10.1039/d1sc02841c] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/17/2021] [Indexed: 12/27/2022] Open
Abstract
Self-assembly of platinum(ii) complexes to form supramolecular structures/nanostructures due to intermolecular ligand π-π stacking and metal-ligand dispersive interactions is widely used to develop functional molecular materials, but the application of such non-covalent molecular interactions has scarcely been explored in medical science. Herein is described the unprecedented biological properties of platinum(ii) complexes relevant to induction of cancer cell death via manifesting such intermolecular interactions. With conjugation of a glucose moiety to the planar platinum(ii) terpyridyl scaffold, the water-soluble complex [Pt(tpy)(C[triple bond, length as m-dash]CArOGlu)](CF3SO3) (1a, tpy = 2,2':6',2''-terpyridine, Glu = glucose) is able to self-assemble into about 100 nm nanoparticles in physiological medium, be taken up by lung cancer cells via energy-dependent endocytosis, and eventually transform into other superstructures distributed in endosomal/lysosomal and mitochondrial compartments apparently following cleavage of the glycosidic linkage. Accompanying the formation of platinum-containing superstructures are increased autophagic vacuole formation, lysosomal membrane permeabilization, and mitochondrial membrane depolarization, as well as anti-tumor activity of 1a in a mouse xenograft model. These findings highlight the dynamic, multi-stage extracellular and intracellular supramolecular self-assembly of planar platinum(ii) complexes driven by modular intermolecular interactions with potential anti-cancer application.
Collapse
Affiliation(s)
- Ka-Chung Tong
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China .,Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong China
| | - Pui-Ki Wan
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China .,Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong China
| | - Chun-Nam Lok
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China .,Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong China
| | - Chi-Ming Che
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China .,Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong China
| |
Collapse
|
29
|
Panja S, Adams DJ. Urea-Urease Reaction in Controlling Properties of Supramolecular Hydrogels: Pros and Cons. Chemistry 2021; 27:8928-8939. [PMID: 33861488 PMCID: PMC8360084 DOI: 10.1002/chem.202100490] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Indexed: 12/18/2022]
Abstract
Supramolecular hydrogels are useful in many areas such as cell culturing, catalysis, sensing, tissue engineering, drug delivery, environmental remediation and optoelectronics. The gels need specific properties for each application. The properties arise from a fibrous network that forms the matrix. A common method to prepare hydrogels is to use a pH change. Most methods result in a sudden pH jump and often lead to gels that are hard to reproduce and control. The urease-urea reaction can be used to control hydrogel properties by a uniform and controlled pH increase as well as to set up pH cycles. The reaction involves hydrolysis of urea by urease and production of ammonia which increases the pH. The rate of ammonia production can be controlled which can be used to prepare gels with differing properties. Herein, we show how the urease-urea reaction can be used for the construction of next generation functional materials.
Collapse
Affiliation(s)
- Santanu Panja
- School of ChemistryUniversity of GlasgowGlasgowG12 8QQUK
| | - Dave J. Adams
- School of ChemistryUniversity of GlasgowGlasgowG12 8QQUK
| |
Collapse
|
30
|
|
31
|
Hamley IW. Biocatalysts Based on Peptide and Peptide Conjugate Nanostructures. Biomacromolecules 2021; 22:1835-1855. [PMID: 33843196 PMCID: PMC8154259 DOI: 10.1021/acs.biomac.1c00240] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/31/2021] [Indexed: 12/15/2022]
Abstract
Peptides and their conjugates (to lipids, bulky N-terminals, or other groups) can self-assemble into nanostructures such as fibrils, nanotubes, coiled coil bundles, and micelles, and these can be used as platforms to present functional residues in order to catalyze a diversity of reactions. Peptide structures can be used to template catalytic sites inspired by those present in natural enzymes as well as simpler constructs using individual catalytic amino acids, especially proline and histidine. The literature on the use of peptide (and peptide conjugate) α-helical and β-sheet structures as well as turn or disordered peptides in the biocatalysis of a range of organic reactions including hydrolysis and a variety of coupling reactions (e.g., aldol reactions) is reviewed. The simpler design rules for peptide structures compared to those of folded proteins permit ready ab initio design (minimalist approach) of effective catalytic structures that mimic the binding pockets of natural enzymes or which simply present catalytic motifs at high density on nanostructure scaffolds. Research on these topics is summarized, along with a discussion of metal nanoparticle catalysts templated by peptide nanostructures, especially fibrils. Research showing the high activities of different classes of peptides in catalyzing many reactions is highlighted. Advances in peptide design and synthesis methods mean they hold great potential for future developments of effective bioinspired and biocompatible catalysts.
Collapse
Affiliation(s)
- Ian W. Hamley
- Department of Chemistry, University of Reading, RG6 6AD Reading, United Kingdom
| |
Collapse
|
32
|
Evaluation of a Model Photo-Caged Dehydropeptide as a Stimuli-Responsive Supramolecular Hydrogel. NANOMATERIALS 2021; 11:nano11030704. [PMID: 33799670 PMCID: PMC8001155 DOI: 10.3390/nano11030704] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 01/17/2023]
Abstract
Short peptides capped on the N-terminus with aromatic groups are often able to form supramolecular hydrogels, via self-assembly, in aqueous media. The rheological properties of these readily tunable hydrogels resemble those of the extracellular matrix (ECM) and therefore have potential for various biological applications, such as tissue engineering, biosensors, 3D bioprinting, drug delivery systems and wound dressings. We herein report a new photo-responsive supramolecular hydrogel based on a "caged" dehydropeptide (CNB-Phe-ΔPhe-OH 2), containing a photo-cleavable carboxy-2-nitrobenzyl (CNB) group. We have characterized this hydrogel using a range of techniques. Irradiation with UV light cleaves the pendant aromatic capping group, to liberate the corresponding uncaged model dehydropeptide (H-Phe-ΔPhe-OH 3), a process which was investigated by 1H NMR and HPLC studies. Crucially, this cleavage of the capping group is accompanied by dissolution of the hydrogel (studied visually and by fluorescence spectroscopy), as the delicate balance of intramolecular interactions within the hydrogel structure is disrupted. Hydrogels which can be disassembled non-invasively with temporal and spatial control have great potential for specialized on-demand drug release systems, wound dressing materials and various topical treatments. Both 2 and 3 were found to be non-cytotoxic to the human keratinocyte cell line, HaCaT. The UV-responsive hydrogel system reported here is complementary to previously reported related UV-responsive systems, which are generally composed of peptides formed from canonical amino acids, which are susceptible to enzymatic proteolysis in vivo. This system is based on a dehydrodipeptide structure which is known to confer proteolytic resistance. We have investigated the ability of the photo-activated system to accelerate the release of the antibiotic, ciprofloxacin, as well as some other small model drug compounds. We have also conducted some initial studies towards skin-related applications. Moreover, this model system could potentially be adapted for on-demand "self-delivery", through the uncaging of known biologically active dehydrodipeptides.
Collapse
|
33
|
Chen J, Zhao Y, Yao Q, Gao Y. Pathological environment directed in situ peptidic supramolecular assemblies for nanomedicines. Biomed Mater 2021; 16:022011. [PMID: 33630754 DOI: 10.1088/1748-605x/abc2e9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peptidic self-assembly provides a powerful method to build biomedical materials with integrated functions. In particular, pathological environment instructed peptidic supramolecular have gained great progress in treating various diseases. Typically, certain pathology related factors convert hydrophilic precursors to corresponding more hydrophobic motifs to assemble into supramolecular structures. Herein, we would like to review the recent progress of nanomedicines based on the development of instructed self-assembly against several specific disease models. Firstly we introduce the cancer instructed self-assembly. These assemblies have exhibited great inhibition efficacy, as well as enhanced imaging contrast, against cancer models both in vitro and in vivo. Then we discuss the infection instructed peptidic self-assembly. A number of different molecular designs have demonstrated the potential antibacterial application with satisfied efficiency for peptidic supramolecular assemblies. Further, we discuss the application of instructed peptidic self-assembly for other diseases including neurodegenerative disease and vaccine. The assemblies have succeeded in down-regulating abnormal Aβ aggregates and immunotherapy. In summary, the self-assembly precursors are typical two-component molecules with (1) a self-assembling motif and (2) a cleavable trigger responsive to the pathological environment. Upon cleavage, the self-assembly occurs selectively in pathological loci whose targeting capability is independent from active targeting. Bearing the novel targeting regime, we envision that the pathological conditions instructed peptidic self-assembly will lead a paradigm shift on biomedical materials.
Collapse
Affiliation(s)
- Jiali Chen
- CAS Center of Excellence for Nanoscience, Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yan Zhao
- CAS Center of Excellence for Nanoscience, Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Qingxin Yao
- CAS Center of Excellence for Nanoscience, Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yuan Gao
- CAS Center of Excellence for Nanoscience, Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
34
|
Karcher J, Kirchner S, Leistner AL, Hald C, Geng P, Bantle T, Gödtel P, Pfeifer J, Pianowski ZL. Selective release of a potent anticancer agent from a supramolecular hydrogel using green light. RSC Adv 2021. [DOI: 10.1039/d0ra08893e] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Selective green-light triggered release of an anticancer agent under physiological conditions from a supramolecular hydrogel.
Collapse
Affiliation(s)
- Johannes Karcher
- Institut für Organische Chemie
- Karlsruher Institut für Technologie
- 76131 Karlsruhe
- Germany
| | - Susanne Kirchner
- Institut für Organische Chemie
- Karlsruher Institut für Technologie
- 76131 Karlsruhe
- Germany
| | - Anna-Lena Leistner
- Institut für Organische Chemie
- Karlsruher Institut für Technologie
- 76131 Karlsruhe
- Germany
| | - Christian Hald
- Institut für Organische Chemie
- Karlsruher Institut für Technologie
- 76131 Karlsruhe
- Germany
| | - Philipp Geng
- Institut für Organische Chemie
- Karlsruher Institut für Technologie
- 76131 Karlsruhe
- Germany
| | - Tobias Bantle
- Institut für Organische Chemie
- Karlsruher Institut für Technologie
- 76131 Karlsruhe
- Germany
| | - Peter Gödtel
- Institut für Organische Chemie
- Karlsruher Institut für Technologie
- 76131 Karlsruhe
- Germany
| | - Juliana Pfeifer
- Institut für Funktionelle Grenzflächen IFG
- Karlsruher Institut für Technologie
- Germany
| | - Zbigniew L. Pianowski
- Institut für Organische Chemie
- Karlsruher Institut für Technologie
- 76131 Karlsruhe
- Germany
- Institute of Biological and Chemical Systems – FMS
| |
Collapse
|
35
|
Zhou Q, Guo M, Ni K, Kerton FM. Construction of supramolecular laccase enzymes and understanding of catalytic dye degradation using multispectral and molecular docking approaches. REACT CHEM ENG 2021. [DOI: 10.1039/d1re00111f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A non-covalent supramolecular enzyme system which was successfully constructed by non-covalent interaction of enzyme with substrates analogs can effectively recognize and degrade 13 kinds of dyes.
Collapse
Affiliation(s)
- Qingteng Zhou
- College of Engineering, Zhejiang A&F University, Hangzhou, Zhejiang 311300, China
| | - Ming Guo
- College of Engineering, Zhejiang A&F University, Hangzhou, Zhejiang 311300, China
- College of Science, Zhejiang A&F University, Hangzhou, Zhejiang 311300, China
| | - Kaijie Ni
- College of Science, Zhejiang A&F University, Hangzhou, Zhejiang 311300, China
| | - Francesca M. Kerton
- Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, Canada A1B 3X7
| |
Collapse
|
36
|
Shy AN, Wang H, Feng Z, Xu B. Heterotypic Supramolecular Hydrogels Formed by Noncovalent Interactions in Inflammasomes. Molecules 2020; 26:E77. [PMID: 33375296 PMCID: PMC7795891 DOI: 10.3390/molecules26010077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 01/04/2023] Open
Abstract
The advance of structural biology has revealed numerous noncovalent interactions between peptide sequences in protein structures, but such information is less explored for developing peptide materials. Here we report the formation of heterotypic peptide hydrogels by the two binding motifs revealed by the structures of an inflammasome. Specifically, conjugating a self-assembling motif to the positively or negatively charged peptide sequence from the ASCPYD filaments of inflammasome produces the solutions of the peptides. The addition of the peptides of the oppositely charged and complementary peptides to the corresponding peptide solution produces the heterotypic hydrogels. Rheology measurement shows that ratios of the complementary peptides affect the viscoelasticity of the resulted hydrogel. Circular dichroism indicates that the addition of the complementary peptides results in electrostatic interactions that modulate self-assembly. Transmission electron microscopy reveals that the ratio of the complementary peptides controls the morphology of the heterotypic peptide assemblies. This work illustrates a rational, biomimetic approach that uses the structural information from the protein data base (PDB) for developing heterotypic peptide materials via self-assembly.
Collapse
Affiliation(s)
| | | | | | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA; (A.N.S.); (H.W.); (Z.F.)
| |
Collapse
|
37
|
Yang HH, Liu PP, Hu JP, Fang H, Lin Q, Hong Y, Zhang YM, Qu WJ, Wei TB. A fluorescent supramolecular gel and its application in the ultrasensitive detection of CN - by anion-π interactions. SOFT MATTER 2020; 16:9876-9881. [PMID: 33006593 DOI: 10.1039/d0sm01392g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Supramolecular gels have been widely reported on account of their unique superiority and application prospects. In this work, we constructed a novel supramolecular gel (HD-G) by using hydroxy-naphthaldehyde decorated with naphthalimide in DMSO solution, which exhibited excellent selectivity and ultrasensitive sensing properties toward CN- (the lowest detection limit is 1.82 × 10-10 M). The sensing mechanism of this supramolecular gel takes advantage of π-π stacking interactions and anion-π interactions, which is different from the other familiar methods.
Collapse
Affiliation(s)
- Hao-Hang Yang
- Key Laboratory of Polymer Materials of Gansu Province, Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, College of Chemistry and Chemical Engineering, Northwest Normal University, Anning East Road 967, Lanzhou, Gansu 730070, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Yang D, He H, Xu B. Enzyme-instructed morphological transition of the supramolecular assemblies of branched peptides. Beilstein J Org Chem 2020; 16:2709-2718. [PMID: 33214796 PMCID: PMC7653338 DOI: 10.3762/bjoc.16.221] [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: 07/25/2020] [Accepted: 10/16/2020] [Indexed: 11/26/2022] Open
Abstract
Here, we report the use of an enzymatic reaction to cleave the branch off branched peptides for inducing the morphological transition of the assemblies of the peptides. The attachment of DEDDDLLI sequences to the ε-amine of the lysine residue of a tetrapeptide produces branched peptides that form micelles. Upon the proteolytic cleavage of the branch, catalyzed by proteinase K, the micelles turn into nanofibers. We also found that the acetylation of the N-terminal of the branch increased the stability of the branched peptides. Moreover, these branched peptides facilitate the delivery of the proteins into cells. This work contributes insights for the development of peptide supramolecular assemblies via enzymatic noncovalent synthesis in cellular environment.
Collapse
Affiliation(s)
- Dongsik Yang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| |
Collapse
|
39
|
Guo J, Tian C, Xu B. Biomaterials based on noncovalent interactions of small molecules. EXCLI JOURNAL 2020; 19:1124-1140. [PMID: 33088250 PMCID: PMC7573174 DOI: 10.17179/excli2020-2656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 07/27/2020] [Indexed: 11/10/2022]
Abstract
Unlike conventional materials that covalent bonds connecting atoms as the major force to hold the materials together, supramolecular biomaterials rely on noncovalent intermolecular interactions to assemble. The reversibility and biocompatibility of supramolecular biomaterials render them with diverse range of functions and lead to rapid development in the past two decades. This review focuses on the noncovalent and enzymatic control of supramolecular biomaterials, with the introduction to various triggering mechanism to initiate self-assembly. Representative applications of supramolecular biomaterials are highlighted in four categories: tissue engineering, cancer therapy, drug delivery, and molecular imaging. By introducing various applications, we intend to show enzymatic control and noncovalent interactions as a powerful tool for achieving spatiotemporal control of biomaterials both invitro and in vivo for biomedicine.
Collapse
Affiliation(s)
- Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA
| | - Changhao Tian
- Department of Physics, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, China
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA
| |
Collapse
|
40
|
Abstract
Enzymatic reactions and noncovalent (i.e., supramolecular) interactions are two fundamental nongenetic attributes of life. Enzymatic noncovalent synthesis (ENS) refers to a process where enzymatic reactions control intermolecular noncovalent interactions for spatial organization of higher-order molecular assemblies that exhibit emergent properties and functions. Like enzymatic covalent synthesis (ECS), in which an enzyme catalyzes the formation of covalent bonds to generate individual molecules, ENS is a unifying theme for understanding the functions, morphologies, and locations of molecular ensembles in cellular environments. This review intends to provide a summary of the works of ENS within the past decade and emphasize ENS for functions. After comparing ECS and ENS, we describe a few representative examples where nature uses ENS, as a rule of life, to create the ensembles of biomacromolecules for emergent properties/functions in a myriad of cellular processes. Then, we focus on ENS of man-made (synthetic) molecules in cell-free conditions, classified by the types of enzymes. After that, we introduce the exploration of ENS of man-made molecules in the context of cells by discussing intercellular, peri/intracellular, and subcellular ENS for cell morphogenesis, molecular imaging, cancer therapy, and other applications. Finally, we provide a perspective on the promises of ENS for developing molecular assemblies/processes for functions. This review aims to be an updated introduction for researchers who are interested in exploring noncovalent synthesis for developing molecular science and technologies to address societal needs.
Collapse
Affiliation(s)
- Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Meihui Yi
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Adrianna N Shy
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| |
Collapse
|
41
|
Jain R, Pal VK, Roy S. Triggering Supramolecular Hydrogelation Using a Protein–Peptide Coassembly Approach. Biomacromolecules 2020; 21:4180-4193. [DOI: 10.1021/acs.biomac.0c00984] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Rashmi Jain
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Vijay Kumar Pal
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Sangita Roy
- Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| |
Collapse
|
42
|
Shy AN, Li J, Shi J, Zhou N, Xu B. Enzyme-instructed self-assembly of the stereoisomers of pentapeptides to form biocompatible supramolecular hydrogels. J Drug Target 2020; 28:760-765. [PMID: 32668995 PMCID: PMC7729926 DOI: 10.1080/1061186x.2020.1797048] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/19/2020] [Accepted: 07/08/2020] [Indexed: 10/23/2022]
Abstract
This article reports enzyme-instructed self-assembly (EISA) of stereoisomers of pentapeptides as a simple approach for generating biocompatible supramolecular hydrogels as potential soft bionanomaterials. Peptide-based supramolecular hydrogels are emerging as a new type of biomaterials. The use of tyrosine phosphate offers a trigger for enzymatic hydrogelation, and the incorporation of D-amino acids can increase the proteolytic stability of peptides. This work compared four phosphorpeptides that are stereoisomers in terms of rate of dephosphorylation, proteolytic stability, and cell compatibility. The results show that the naphthyl (Nap)-capped pentapeptides, containing the amino acid sequence of Phe-Phe-Gly-Glu-pTyr, are able to undergo EISA to form the hydrogels consisting the nanofibres of the dephosphorylated pentapeptides. The naphthyl-capped D-phosphopentpeptides, contrasting to a naphthyl-capped D-phosphotripeptide (Nap-D-Phe-D-Phe-D-pTyr), are largely cell compatible. This result, suggesting that the sequence of phophopeptides also dedicates the cell compatibility of the peptide assemblies resulted from EISA, provides useful insights for developing supramolecular hydrogels as potential biomaterials with tailored properties.
Collapse
Affiliation(s)
- Adrianna N. Shy
- Department of Chemistry, Brandeis University, Waltham, MA USA
| | - Jie Li
- Department of Chemistry, Brandeis University, Waltham, MA USA
| | - Junfeng Shi
- Department of Chemistry, Brandeis University, Waltham, MA USA
| | - Ning Zhou
- Department of Chemistry, Brandeis University, Waltham, MA USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, Waltham, MA USA
| |
Collapse
|
43
|
Gao J, Wu P, Fernandez A, Zhuang J, Thayumanavan S. Cellular AND Gates: Synergistic Recognition to Boost Selective Uptake of Polymeric Nanoassemblies. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202002748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Jingjing Gao
- Department of Chemistry University of Massachusetts Amherst Amherst MA 01003 USA
| | - Peidong Wu
- Department of Chemistry University of Massachusetts Amherst Amherst MA 01003 USA
| | - Ann Fernandez
- Department of Chemistry University of Massachusetts Amherst Amherst MA 01003 USA
| | - Jiaming Zhuang
- Department of Chemistry University of Massachusetts Amherst Amherst MA 01003 USA
| | - S. Thayumanavan
- Department of Chemistry University of Massachusetts Amherst Amherst MA 01003 USA
- Molecular and Cellular Biology Program University of Massachusetts Amherst Amherst USA
- Center for Bioactive Delivery University of Massachusetts Amherst Amherst USA
| |
Collapse
|
44
|
Gao J, Wu P, Fernandez A, Zhuang J, Thayumanavan S. Cellular AND Gates: Synergistic Recognition to Boost Selective Uptake of Polymeric Nanoassemblies. Angew Chem Int Ed Engl 2020; 59:10456-10460. [DOI: 10.1002/anie.202002748] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Jingjing Gao
- Department of Chemistry University of Massachusetts Amherst Amherst MA 01003 USA
| | - Peidong Wu
- Department of Chemistry University of Massachusetts Amherst Amherst MA 01003 USA
| | - Ann Fernandez
- Department of Chemistry University of Massachusetts Amherst Amherst MA 01003 USA
| | - Jiaming Zhuang
- Department of Chemistry University of Massachusetts Amherst Amherst MA 01003 USA
| | - S. Thayumanavan
- Department of Chemistry University of Massachusetts Amherst Amherst MA 01003 USA
- Molecular and Cellular Biology Program University of Massachusetts Amherst Amherst USA
- Center for Bioactive Delivery University of Massachusetts Amherst Amherst USA
| |
Collapse
|
45
|
Abstract
Enzymatic reactions and self-assembly are two fundamental attributes of cells. It is not surprising that one can use enzyme-instructed self-assembly (EISA)-the integration of enzymatic transformation and molecular self-assembly-to modulate the emergent properties of supramolecular assemblies for controlling cell behaviors. The exploration of EISA for developing cancer therapy and imaging has made considerable progress over the last five years. In this Topical Review, we discuss these exciting results and the future promise of EISA. After describing several key studies to illustrate the progress of EISA in developing cancer therapy, we discuss the use of EISA for molecular imaging. Then, we give the outlook of EISA for developing supramolecular anticancer medicine that inhibits multiple hallmark capabilities of cancer.
Collapse
Affiliation(s)
- Beom Jin Kim
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| |
Collapse
|
46
|
Chi X, Liu K, Luo X, Yin Z, Lin H, Gao J. Recent advances of nanomedicines for liver cancer therapy. J Mater Chem B 2020; 8:3747-3771. [DOI: 10.1039/c9tb02871d] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This review highlights recent advancements in nanomedicines for liver cancer therapy.
Collapse
Affiliation(s)
- Xiaoqin Chi
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma
- Zhongshan Hospital
- Xiamen University
- Xiamen 361004
- China
| | - Kun Liu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
- The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- Xiamen University
- Xiamen 361005
| | - Xiangjie Luo
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
- The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- Xiamen University
- Xiamen 361005
| | - Zhenyu Yin
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma
- Zhongshan Hospital
- Xiamen University
- Xiamen 361004
- China
| | - Hongyu Lin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
- The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- Xiamen University
- Xiamen 361005
| | - Jinhao Gao
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
- The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- Xiamen University
- Xiamen 361005
| |
Collapse
|