1
|
Zhang W, Wang F, Wang H, Xu T, Su H, Cui H. Balancing Chemical and Supramolecular Stability in OEGylated Supramolecular Polymers for Systemic Drug Delivery. J Am Chem Soc 2025. [PMID: 40375654 DOI: 10.1021/jacs.5c03253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
The chemical conjugation of poly(ethylene glycol) (PEG) to therapeutic agents, known as PEGylation, is a well-established strategy for enhancing drug solubility, chemical stability, and pharmacokinetics. Here, we report on a class of supramolecular polymeric prodrugs by utilizing oligo(ethylene glycol) (OEG) to modify the hydrophobic anticancer drug camptothecin (CPT). These OEGylated prodrugs, despite their low molecular weight, spontaneously self-assemble into therapeutic supramolecular polymers (SPs) with a tubular morphology, featuring a dense OEG coating on the surface. By designing biodegradable linkers with varying chemical stabilities, we investigated how the release kinetics of CPT influence the in vitro and in vivo performance of these SPs. Our findings demonstrate that self-assembling prodrugs (SAPDs) with a self-immolative disulfanyl-ethyl carbonate (etcSS) linker exhibit a faster drug release rate than those with a reducible disulfanyl butyrate (buSS) linker, leading to higher potency and significantly improved antitumor efficacy. Notably, two stable tubular SPs, Tubustecan (TT) 1E and TT 7E, outperformed irinotecan─a clinically approved CPT prodrug─in a colon cancer model, achieving enhanced tumor growth inhibition and prolonged animal survival. These results highlight the potential of supramolecular OEGylation as an important strategy for engineering drug-based supramolecular polymers and underscore the critical role of chemical stability vs supramolecular stability in optimizing supramolecular prodrug design.
Collapse
Affiliation(s)
- Weijie Zhang
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Feihu Wang
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Han Wang
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Tian Xu
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Hao Su
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
2
|
Bai L, Yi M, Xu B. Self-Assembly of Noncanonical Peptides: A New Frontier in Cancer Therapeutics and Beyond. Macromol Biosci 2025:e2500153. [PMID: 40260674 DOI: 10.1002/mabi.202500153] [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: 02/28/2025] [Revised: 04/03/2025] [Indexed: 04/24/2025]
Abstract
In addition to the 20 standard amino acids that form the building blocks of proteins, nature employs alternative amino acids to create specialized "noncanonical peptides." These unique peptides, found in organisms from bacteria to humans, often exhibit unconventional structures and functionalities, playing critical roles in modulating cellular processes, particularly as antibiotics. Their potential has attracted significant interest for designing novel functional materials based on noncanonical peptides. This review highlights recent advances in the generation and application of noncanonical peptide assemblies. It begins with a definition of noncanonical peptides, including classic examples that showcase their distinct structures and useful biological activities. Then the applications of noncanonical peptide assemblies in developing anticancer therapeutics are discussed, focusing on recent and representative studies that demonstrate their efficacy and versatility in targeting tumor cells. Beyond oncology, it is explored how noncanonical peptide assemblies have been utilized in biomaterials, regenerative medicine, molecular imaging and catalysis. Finally, perspectives are offered on future directions in this rapidly evolving field, emphasizing exciting opportunities and remaining challenges that will drive continued innovation in designing and applying noncanonical peptide-based assemblies.
Collapse
Affiliation(s)
- Lin Bai
- School of Chemistry, Brandeis University, 415 South St, Waltham, MA, 02453, USA
| | - Meihui Yi
- School of Chemistry, Brandeis University, 415 South St, Waltham, MA, 02453, USA
| | - Bing Xu
- School of Chemistry, Brandeis University, 415 South St, Waltham, MA, 02453, USA
| |
Collapse
|
3
|
Liu X, Hu B, Yu Z. Noncanonical Amino Acids Dictate Peptide Assembly in Living Cells. Acc Chem Res 2025; 58:1081-1093. [PMID: 40105513 DOI: 10.1021/acs.accounts.4c00796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
ConspectusEmulating the structural features or functions of natural systems has been demonstrated as a state-of-the-art strategy to create artificial functional materials. Inspired by the assembly and bioactivity of proteins, the self-assembly of peptides into nanostructures represents a promising approach for creating biomaterials. Conventional assembled peptide biomaterials are typically formulated in solution and delivered to pathological sites for implementing theranostic objectives. However, this translocation entails a switch from formulation conditions to the physiological environment and raises concerns about material performance. In addition, the precise and efficient accumulation of administered biomaterials at target sites remains a significant challenge, leading to potential biosafety issues associated with off-target effects. These limitations significantly hinder the progress of advanced biomaterials. To address these concerns, the past few years have witnessed the development of in situ assembly of peptides in living systems as a new endeavor for optimizing biomaterial performance benefiting from the advances of stimuli-responsive reactions regulating noncovalent interactions. In situ assembly of peptides refers to the processes of regulating assembly via stimuli-responsive reactions at target sites. Due to the advantages of precisely forming well-defined nanostructures at pathological lesions, in situ-formed assemblies with integrated bioactivity are interesting for the development of next-generation biomedical agents.Despite the great potential of in situ assembly of peptides for developing biomedical agents, this research area still suffers from a limited toolkit for operating peptide assembly under complicated physiological conditions. Considering the advantages of amino acids in being incorporated into peptide backbones and modified with stimuli-responsive units, development of an amino acid toolkit is promising to address this concern. Therefore, our laboratory has been intensively engaged in designing and discovering stimuli-responsive noncanonical amino acids (ncAAs) to expand the toolkit for manipulating peptide assembly under various biological conditions. Thus far, we have synthesized peptides containing ncAAs 4-aminoproline, 2-nitroimidazole alanine, Se-methionine, sulfated tyrosine, and glycosylated serine, which allow us to develop acid-responsive, redox-responsive, and enzyme-responsive assembly systems. Based on these stimuli-responsive ncAAs, we have established complex self-sorting assembly, self-amplified assembly, and dissipative assembly systems in living cells to optimize the bioactivity of peptides. The resulting in situ assembly systems exhibit morphological adaptability to the biological microenvironment, which contributes to overcoming delivery barriers and improvement of targeting accumulation. Therefore, by utilizing the developed toolkit, we have further created supramolecular PROTACs, supramolecular antagonists, and supramolecular probes for cancer treatment and diagnosis to highlight the implications of ncAAs for biomedical usage. In this Account, we summarize our journey of in situ self-assembly of peptides in living cells utilizing stimuli-responsive ncAAs, with an emphasis on the mechanism for regulating peptide assembly and optimizing the bioactivity of peptides. Eventually, we also provide our forward conceiving prospects on the challenges for the further development of in situ assembly of peptides in living systems and the clinical translation of in situ-formulated biomaterials.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Binbin Hu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zhilin Yu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
4
|
Wang H, Zheng YT, Zhang J, Gao Y, Chen J, Cai P, Wang J, van Esch JH, Guo X, Li H, Wang Y. Synthesis of Abiotic Supramolecular Polymers Inside Living Cells via Organocatalysis-Mediated Self-Assembly. Angew Chem Int Ed Engl 2025:e202500998. [PMID: 40059797 DOI: 10.1002/anie.202500998] [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: 01/13/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/18/2025]
Abstract
Cells execute mesmerizing functions using supramolecular polymers (SPs) formed through the self-assembly of biological precursors. Integration of the vast array of synthetic SPs with living cells would offer a powerful way to remold cellular functions and bridge the gap between synthetic materials and the biological realm, yet remains a challenge because of the lack of robust abiotic SP systems that can be triggered to self-assemble inside cells. Here, we report how fully abiotic SPs can be synthesized inside living cells via an organocatalysis-responsive self-assembly strategy, and how the in situ-generated SPs are capable of interfering and can interfere with cellular functions. The incorporation of a nucleophilic organocatalyst (CAT) into living cells accelerates the intracellular conversion of hydrazide (H) and aldehyde-derived precursors (A) to hydrazone-based monomers (HA3) that locally self-assemble into SPs. Interestingly, the in situ-generated SPs possess ignorable effects on cell viability and proliferation but remarkably hinder cell migration. Furthermore, the presence of SPs is found to retard intracellular diffusion and alter the organization of the actin cytoskeleton, both of which are suggested to be responsible for the hindered cellular migration. In considering the vastly wide range of synthetic SPs, tremendous non-natural cellular functionalities can be obtained by in situ-synthesizing SPs.
Collapse
Affiliation(s)
- Hucheng Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Ya-Ting Zheng
- School of Systems Science and Institute of Nonequilibrium Systems, Beijing Normal University, Beijing, 100875, P.R. China
| | - Jiahao Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Yuliang Gao
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Jingjing Chen
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Peiwen Cai
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Junyou Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Jan H van Esch
- Department of Chemical Engineering, Delft University of Technology, Delft, 2629 HZ, The Netherlands
| | - Xuhong Guo
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Hui Li
- School of Systems Science and Institute of Nonequilibrium Systems, Beijing Normal University, Beijing, 100875, P.R. China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, 100875, P.R. China
| | - Yiming Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
- Shanghai Key Laboratory for Intelligent Sensing and Detection Technology, East China University of Science and Technology, Shanghai, 200237, P.R. China
| |
Collapse
|
5
|
Wu C, Zhang XW, Wang M, Sun J, Chen J, Guan Y, Pang X. Trypsin-instructed bioactive peptide nanodrugs with cascading transformations to improve chemotherapy against colon cancer. J Nanobiotechnology 2025; 23:66. [PMID: 39891144 PMCID: PMC11784115 DOI: 10.1186/s12951-025-03143-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025] Open
Abstract
Chemotherapy remains an effective treatment for colon cancer but is hampered by its limited response rate. Bioactive peptides, marked with intracellular transformations, have been reported as an effective approach to boosting chemotherapeutic activity. Herein, a promising trypsin-responsive bioactive peptide-based nanodrug is constructed, which could significantly prolong the drug retention time in vivo by cascading transformations and improve chemotherapeutic efficacy. Initially, 1-Pept co-assembles with Dox into a few nanofibers called 1-Pept/Dox NFs, inducing an enhanced cellular uptake via caveolae-mediated endocytosis by avoiding lysosomal degradation and further promoting perinuclear transportation, thus enlarging the drug efficacy in target areas. After nanofiber disassembly, the released 1-Pept converts into Pept under the catalysis of intracellular overexpressed trypsin, which then reassembles into denser Pept NFs, inducing a cascade of effects including disruption of the cytoskeleton, mitochondrial dysfunction, and activation of caspase-3. By the synergism of Pept NFs and Dox, caspase-3 can be further activated, and cause greater damage to nuclear, thereby leading to tumor ablation. As the first example of employing trypsin-mediated nanodrugs with cascading transformations to promote chemotherapeutic activity, this work promises a strategy for novel therapies for efficiently combating colon cancer.
Collapse
Affiliation(s)
- Can Wu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China
| | - Xiao Wei Zhang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Manman Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jinpan Sun
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jianfei Chen
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yanbin Guan
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xin Pang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
6
|
Zhang J, Jin K, Feng Y, Lu D, Chen M, Wang H, Jin C, Wang D, Li Z, Wang Y. Injectable Self-Healing and Anti-Dissolving Low-Molecular-Weight Hydrogels Enabled by Ionic Cross-Linking for Cell Encapsulation. ACS Macro Lett 2025; 14:20-25. [PMID: 39690898 DOI: 10.1021/acsmacrolett.4c00725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Injectable behavior is often observed in polymer-based hydrogels yet is rarely achieved in low-molecular-weight hydrogels (LMWHs), the realization of which may boost the development of new soft materials for biomedical applications. Here, we report on injectable self-healing and antidissolving LMWHs that are formed through a simple ionic cross-linking strategy, showing a fundamental application for the encapsulation of living cells. The LMWHs are formed by simply mixing Ca2+ with negatively charged supramolecular polymers. Surprisingly, the resultant hydrogels are capable of rapidly self-healing within seconds after damage, showing an unexpected injectable function. When the hydrogel is injected into an aqueous medium, continuous macroscopic hydrogel fibers can be produced. Interestingly, the hydrogel can remain intact in the aqueous medium, showing impressive antidissolving behavior which is less observed in other LMWHs. Furthermore, the hydrogel is demonstrated to be nontoxic and can be used as a cytocompatible scaffold for living cells. This work may open an avenue toward injectable and antidissolving LMWHs for the ever-expanding list of applications in biotherapy and bioprinting.
Collapse
Affiliation(s)
- Jiahao Zhang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Kaiyu Jin
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yifei Feng
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Da Lu
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Mai Chen
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Hucheng Wang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Cheng Jin
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Dengyu Wang
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zhiling Li
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yiming Wang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory for Intelligent Sensing and Detection Technology, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
7
|
Cui H, Tirrell M. Self-Assembling Peptides, Conjugates, and Mimics: A Versatile Platform for Materials and Beyond. Acc Chem Res 2025; 58:163-164. [PMID: 39834233 DOI: 10.1021/acs.accounts.4c00805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Affiliation(s)
- Honggang Cui
- Department of Chemical and Biomolecular Engineering and Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Matthew Tirrell
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois 60637, United States
- Argonne National Laboratory, Lemont, Illinois 60439, United States
| |
Collapse
|
8
|
Choi HJ, Jin S, Park J, Lee D, Jeong HJ, Kim OH, Ryu JH, Kim SJ. Assessing the Efficacy of Mitochondria-Accumulating Self-Assembly Peptides in Pancreatic Cancer: An Animal Study. Int J Mol Sci 2025; 26:784. [PMID: 39859498 PMCID: PMC11766353 DOI: 10.3390/ijms26020784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Although pancreatic cancer presents with one of the most unfavorable prognoses, its treatment options are very limited. Mitochondria-targeting moieties, considered a new and prominent treatment modality, are expected to demonstrate synergistic anticancer effects due to their distinct mechanism compared to conventional chemotherapeutic approaches. This study evaluated the therapeutic potential of mitochondria-accumulating self-assembly peptides, referred to as Mito-FFs, utilizing both in vitro and in vivo pancreatic cancer models. Cellular viability assays revealed a concentration-dependent decrease in the survival of MIA-PACA2 pancreatic cancer cells upon exposure to Mito-FF treatment (p < 0.05). Subsequent in vitro Mito-FF treatments prompted the use of several molecular analyses, including Real-time PCR, Western blot analysis, and MitoSOX staining, which collectively indicated an upsurge in apoptosis, a concurrent reduction in the antioxidant enzyme expression, and an elevation in mitochondrial ROS levels (p < 0.05). In a murine xenograft model of pancreatic cancer, the intravenous administration of Mito-FF yielded a notable reduction in the tumor volume. Moreover, it upregulated the expression of pro-apoptotic markers, such as cleaved PARP and c-caspase 3, while concurrently downregulating the expression of an anti-apoptotic marker, MCL-1, as evidenced by both Western blot analysis and immunohistochemical staining (p < 0.05). It also resulted in the reduced expression of antioxidant enzymes like HO-1, catalase, and SOD2 within excised tumor tissues, as confirmed using Western blot analysis (p < 0.05). Cumulatively, the findings underscore the significant anticancer efficacy of Mito-FF against pancreatic cancer cells, predominantly mediated through the induction of apoptosis, suppression of antioxidant enzyme expression, and enhancement of mitochondrial ROS levels within the tumor microenvironment.
Collapse
Affiliation(s)
- Ho Joong Choi
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.J.C.); (D.L.)
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.P.); (H.J.J.); (O.-H.K.)
| | - Seongeon Jin
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea; (S.J.); (J.-H.R.)
| | - Junghyun Park
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.P.); (H.J.J.); (O.-H.K.)
- Department of Surgery, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, Republic of Korea
| | - Dosang Lee
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.J.C.); (D.L.)
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.P.); (H.J.J.); (O.-H.K.)
| | - Hee Jeong Jeong
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.P.); (H.J.J.); (O.-H.K.)
| | - Ok-Hee Kim
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.P.); (H.J.J.); (O.-H.K.)
- Translational Research Team, Surginex Co., Ltd., Seoul 06591, Republic of Korea
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea; (S.J.); (J.-H.R.)
| | - Say-June Kim
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.J.C.); (D.L.)
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.P.); (H.J.J.); (O.-H.K.)
| |
Collapse
|
9
|
Dai SY, Xiao Z, Shen F, Lim I, Rao J. Light-Controlled Intracellular Synthesis of Poly(luciferin) Polymers Induces Cell Paraptosis. J Am Chem Soc 2025; 147:2037-2048. [PMID: 39757486 DOI: 10.1021/jacs.4c15644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Accumulation of misfolded proteins challenges cellular proteostasis and is implicated in aging and chronic disorders. Cancer cells, moreover, face an elevated level of basal proteotoxic stress; hence, exacerbating endoplasmic reticulum (ER) stress has been shown to induce programmed cell death while enhancing anticancer immunogenicity. We hypothesize that hydrophobic abiotic macromolecules can trigger a similar stress response. Most polymers and nanoparticles, however, are sequestered in endo/lysosomes after endocytosis, which prevents their interaction with the proteostasis machinery. We adopted an in situ polymerization approach to synthesize polymers in cells with cell-permeable monomers. Specifically, we developed a biocompatible polycondensation between l-cysteine and 2-cyanobenzothiazole (CBT) with photochemical control to form insoluble poly(luciferin) aggregates. We identified that in situ polymerization activates the BiP-PERK-CHOP pathway of the unfolded protein response and that the unresolved ER stress initiates a form of regulated cell death consistent with paraptosis. In addition, the dying cells emit damage-associated molecular patterns (DAMPs), indicating an immunogenic cell death that could potentiate antitumor immunity. Our results show that in situ polymerization mimics misfolded protein aggregates to induce proteotoxic stress and cancer cell death, offering a novel therapeutic strategy to exploit cancer vulnerability.
Collapse
Affiliation(s)
- Sheng-Yao Dai
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Zhen Xiao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Fangfang Shen
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Irene Lim
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jianghong Rao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
10
|
Bharathidasan D, Maity C. Organelle-Specific Smart Supramolecular Materials for Bioimaging and Theranostics Application. Top Curr Chem (Cham) 2024; 383:1. [PMID: 39607460 DOI: 10.1007/s41061-024-00483-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
In cellular environments, certain synthetic molecules can form nanostructures via self-assembly, impacting molecular imaging, and biomedical applications. Control over the formation of these self-assembled nanostructures in subcellular organelle is challenging. By the action of stimuli, either present in the cellular environment or applied externally, in situ generation of molecular precursors can lead to accumulation and supramolecular nanostructure formation, resulting in efficient bioimaging. Here, we summarize smart fluorophore-based ordered nanostructure preparation at specific organelles for efficient bioimaging and therapeutic application towards cancer theranostics. We also present challenges and an outlook regarding intercellular self-assembly for theranostics application. Altogether, smart nanostructured materials with fluorescence read-outs at specific subcellular compartments would be beneficial in synthetic biology and precision therapeutics.
Collapse
Affiliation(s)
- Dineshkumar Bharathidasan
- (Organic)Material Science and Engineering Laboratory, Centre for Nanobiotechnology (CNBT), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamilnadu, 632014, India
| | - Chandan Maity
- (Organic)Material Science and Engineering Laboratory, Centre for Nanobiotechnology (CNBT), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamilnadu, 632014, India.
| |
Collapse
|
11
|
Shang Y, Wang J, Feng Y, Liu Z, Lu J, Deng S, Li L, Zhang H, Li L, Wang Z, Yang Z. A dual-targeted trinity of antibody-peptide-drug delivery consortium to combat HER2+ tumor. Chem Commun (Camb) 2024; 60:13742-13745. [PMID: 39494544 DOI: 10.1039/d4cc05115g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
We pioneered a dual-targeted trinity of antibody-peptide-drug delivery consortium to combat HER2+ tumors. This innovative approach leverages the self-assembly of peptides with high affinity to antibodies to create nanofibers for antibody encapsulation, offering a novel strategy in antibody drug delivery.
Collapse
Affiliation(s)
- Yuna Shang
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, P. R. China
| | - Jiayu Wang
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, P. R. China
| | - Yinyin Feng
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, P. R. China
| | - Zhengyu Liu
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, P. R. China
| | - Jiayi Lu
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, P. R. China
| | - Sijia Deng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P. R. China.
| | - Li Li
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, P. R. China
| | - Hongxu Zhang
- Academy of Interdisciplinary Studies on Intelligent Molecules, Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, P. R. China
| | - Liantao Li
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P. R. China.
| | - Zhongyan Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, P. R. China.
| | - Zhimou Yang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China.
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P. R. China.
| |
Collapse
|
12
|
Guo Y, Li P, Guo X, Yao C, Yang D. Synthetic Nanoassemblies for Regulating Organelles: From Molecular Design to Precision Therapeutics. ACS NANO 2024; 18:30224-30246. [PMID: 39441007 DOI: 10.1021/acsnano.4c10194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Each organelle referring to a complex multiorder architecture executes respective biological processes via its distinct spatial organization and internal microenvironment. As the assembly of biomolecules is the structural basis of living cells, creating synthetic nanoassemblies with specific physicochemical and morphological properties in living cells to interfere or couple with the natural organelle architectures has attracted great attention in precision therapeutics of cancers. In this review, we give an overview of the latest advances in the synthetic nanoassemblies for precise organelle regulation, including the formation mechanisms, triggering strategies, and biomedical applications in precision therapeutics. We summarize the emerging material systems, including polymers, peptides, and deoxyribonucleic acids (DNAs), and their respective intermolecular interactions for intercellular synthetic nanoassemblies, and highlight their design principles in constructing precursors that assemble into synthetic nanoassemblies targeting specific organelles in the complex cellular environment. We further showcase the developed intracellular synthetic nanoassemblies targeting specific organelles including mitochondria, the endoplasmic reticulum, lysosome, Golgi apparatus, and nucleus and describe their underlying mechanisms for organelle regulation and precision therapeutics for cancer. Last, the essential challenges in this field and prospects for future precision therapeutics of synthetic nanoassemblies are discussed. This review should facilitate the rational design of organelle-targeting synthetic nanoassemblies and the comprehensive recognition of organelles by materials and contribute to the deep understanding and application of the synthetic nanoassemblies for precision therapeutics.
Collapse
Affiliation(s)
- Yanfei Guo
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, P.R. China
| | - Peiran Li
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Xiaocui Guo
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Chi Yao
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Dayong Yang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, P.R. China
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| |
Collapse
|
13
|
Yu S, Webber MJ. Engineering disease analyte response in peptide self-assembly. J Mater Chem B 2024; 12:10757-10769. [PMID: 39382032 DOI: 10.1039/d4tb01860e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
A need to enhance the precision and specificity of therapeutic nanocarriers inspires the development of advanced nanomaterials capable of sensing and responding to disease-related cues. Self-assembled peptides offer a promising nanocarrier platform with versatile use to create precisely defined nanoscale materials. Disease-relevant cues can range from large biomolecules, such as enzymes, to ubiquitous small molecules with varying concentrations in healthy versus diseased states. Notably, pH changes (i.e., H+ concentration), redox species (e.g., H2O2), and glucose levels are significant spatial and/or temporal indicators of therapeutic need. Self-assembled peptides respond to these cues by altering their solubility, modulating electrostatic interactions, or facilitating chemical transformations through dynamic or labile bonds. This review explores the design and construction of therapeutic nanocarriers using self-assembled peptides, focusing on how peptide sequence engineering along with the inclusion of non-peptidic components can link the assembly state of these nanocarriers to the presence of disease-relevant small molecules.
Collapse
Affiliation(s)
- Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
14
|
Guo J, Zia A, Qiu Q, Norton M, Qiu K, Usuba J, Liu Z, Yi M, Rich-New ST, Hagan M, Fraden S, Han GD, Diao J, Wang F, Xu B. Cell-Free Nonequilibrium Assembly for Hierarchical Protein/Peptide Nanopillars. J Am Chem Soc 2024; 146:26102-26112. [PMID: 39255453 PMCID: PMC11669155 DOI: 10.1021/jacs.4c06775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Cells contain intricate protein nanostructures, but replicating them outside of cells presents challenges. One such example is the vertical fibronectin pillars observed in embryos. Here, we demonstrate the creation of cell-free vertical fibronectin pillar mimics using nonequilibrium self-assembly. Our approach utilizes enzyme-responsive phosphopeptides that assemble into nanotubes. Enzyme action triggers shape changes in peptide assemblies, driving the vertical growth of protein nanopillars into bundles. These bundles, with peptide nanotubes serving as a template to remodel fibronectin, can then recruit collagen, which forms aggregates or bundles depending on their types. Nanopillar formation relies on enzyme-catalyzed nonequilibrium self-assembly and is governed by the concentrations of enzyme, protein, peptide, the structure of the peptide, and peptide assembly morphologies. Cryo-EM reveals unexpected nanotube thinning and packing after dephosphorylation, indicating a complex sculpting process during assembly. Our study demonstrates a cell-free method for constructing intricate, multiprotein nanostructures with directionality and composition.
Collapse
Affiliation(s)
- Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Ayisha Zia
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Qianfeng Qiu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Michael Norton
- Department of Physics, Brandeis University, Waltham, MA, 02453, USA
| | - Kangqiang Qiu
- Department of Cancer Biology, Center for Chemical Imaging in Biomedicine, Advanced Cell Analysis Service Center, University of Cincinnati College of Medicine, Cincinnati OH, 45267, USA
| | - Junichi Usuba
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Zhiyu Liu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Meihui Yi
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Shane T. Rich-New
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Michael Hagan
- Department of Physics, Brandeis University, Waltham, MA, 02453, USA
| | - Seth Fraden
- Department of Physics, Brandeis University, Waltham, MA, 02453, USA
| | - Grace D. Han
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Jiajie Diao
- Department of Cancer Biology, Center for Chemical Imaging in Biomedicine, Advanced Cell Analysis Service Center, University of Cincinnati College of Medicine, Cincinnati OH, 45267, USA
| | - Fengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- O’Neal Comprehensive Cancer Center University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| |
Collapse
|
15
|
Xu D, Bi S, Li J, Ma S, Yu ZA, Wang Y, Chen H, Zhan J, Song X, Cai Y. Legumain-Guided Ferulate-Peptide Self-Assembly Enhances Macrophage-Endotheliocyte Partnership to Promote Therapeutic Angiogenesis After Myocardial Infarction. Adv Healthc Mater 2024:e2402056. [PMID: 39252665 DOI: 10.1002/adhm.202402056] [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: 06/24/2024] [Revised: 08/31/2024] [Indexed: 09/11/2024]
Abstract
Promoting angiogenesis and modulating the inflammatory microenvironment are promising strategies for treating acute myocardial infarction (MI). Macrophages are crucial in regulating inflammation and influencing angiogenesis through interactions with endothelial cells. However, current therapies lack a comprehensive assessment of pathological and physiological subtleties, resulting in limited myocardial recovery. In this study, legumain-guided ferulate-peptide nanofibers (LFPN) are developed to facilitate the interaction between macrophages and endothelial cells in the MI lesion and modulate their functions. LFPN exhibits enhanced ferulic acid (FA) aggregation and release, promoting angiogenesis and alleviating inflammation. The multifunctional role of LFPN is validated in cells and an MI mouse model, where it modulated macrophage polarization, attenuated inflammatory responses, and induces endothelial cell neovascularization compare to FA alone. LFPN supports the preservation of border zone cardiomyocytes by regulating inflammatory infiltration in the ischemic core, leading to significant functional recovery of the left ventricle. These findings suggest that synergistic therapy exploiting multicellular interaction and enzyme guidance may enhance the clinical translation potential of smart-responsive drug delivery systems to treat MI. This work emphasizes macrophage-endothelial cell partnerships as a novel paradigm to enhance cell interactions, control inflammation, and promote therapeutic angiogenesis.
Collapse
Affiliation(s)
- Delong Xu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Shenghui Bi
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jiejing Li
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Shaodan Ma
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Ze-An Yu
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yenan Wang
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Huiming Chen
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jie Zhan
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xudong Song
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, 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, 510280, China
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
16
|
Liu X, Tian F, Zhang Z, Liu J, Wang S, Guo RC, Hu B, Wang H, Zhu H, Liu AA, Shi L, Yu Z. In Vivo Self-Sorting of Peptides via In Situ Assembly Evolution. J Am Chem Soc 2024; 146:24177-24187. [PMID: 39140408 DOI: 10.1021/jacs.4c10309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Despite significant progress achieved in artificial self-sorting in solution, operating self-sorting in the body remains a considerable challenge. Here, we report an in vivo self-sorting peptide system via an in situ assembly evolution for combined cancer therapy. The peptide E3C16-SS-EIY consists of two disulfide-connected segments, E3C16SH and SHEIY, capable of independent assembly into twisted or flat nanoribbons. While E3C16-SS-EIY assembles into nanorods, exposure to glutathione (GSH) leads to the conversion of the peptide into E3C16SH and SHEIY, thus promoting in situ evolution from the nanorods into self-sorted nanoribbons. Furthermore, incorporation of two ligand moieties targeting antiapoptotic protein XIAP and organellar endoplasmic reticulum (ER) into the self-sorted nanoribbons allows for simultaneous inhibition of XIAP and accumulation surrounding ER. This leads to the cytotoxicity toward the cancer cells with elevated GSH levels, through activating caspase-dependent apoptosis and inducing ER dysfunction. In vivo self-sorting of E3C16-SS-EIY decorated with ligand moieties is thoroughly validated by tissue studies. Tumor-bearing mouse experiments confirm the therapeutic efficacy of the self-sorted assemblies for inhibiting tumor growth, with excellent biosafety. Our findings demonstrate an efficient approach to develop in vivo self-sorting systems and thereby facilitating in situ formulation of biomedical agents.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Feng Tian
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zeyu Zhang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Juanzu Liu
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shuya Wang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Ruo-Chen Guo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Binbin Hu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Hao Wang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Han Zhu
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - An-An Liu
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zhilin Yu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
17
|
Yang A, Song J, Li J, Li Y, Bai S, Zhou C, Wang M, Zhou Y, Wen K, Luo M, Chen P, Liu B, Yang H, Bai Y, Wong WL, Cai Q, Pu H, Qian Y, Hu W, Huang W, Wan M, Zhang C, Feng X. Ligand-Receptor Interaction-Induced Intracellular Phase Separation: A Global Disruption Strategy for Resistance-Free Lethality of Pathogenic Bacteria. J Am Chem Soc 2024; 146:23121-23137. [PMID: 38980064 DOI: 10.1021/jacs.4c04749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Addressing the global challenge of bacterial resistance demands innovative approaches, among which multitargeting is a widely used strategy. Current strategies of multitargeting, typically achieved through drug combinations or single agents inherently aiming at multiple targets, face challenges such as stringent pharmacokinetic and pharmacodynamic requirements and cytotoxicity concerns. In this report, we propose a bacterial-specific global disruption approach as a vastly expanded multitargeting strategy that effectively disrupts bacterial subcellular organization. This effect is achieved through a pioneering chemical design of ligand-receptor interaction-induced aggregation of small molecules, i.e., DNA-induced aggregation of a diarginine peptidomimetic within bacterial cells. These intracellular aggregates display affinity toward various proteins and thus substantially interfere with essential bacterial functions and rupture bacterial cell membranes in an "inside-out" manner, leading to robust antibacterial activities and suppression of drug resistance. Additionally, biochemical analysis of macromolecule binding affinity, cytoplasmic localization patterns, and bacterial stress responses suggests that this bacterial-specific intracellular aggregation mechanism is fundamentally different from nonselective classic DNA or membrane binding mechanisms. These mechanistic distinctions, along with the peptidomimetic's selective permeation of bacterial membranes, contribute to its favorable biocompatibility and pharmacokinetic properties, enabling its in vivo antimicrobial efficacy in several animal models, including mice-based superficial wound models, subcutaneous abscess models, and septicemia infection models. These results highlight the great promise of ligand-receptor interaction-induced intracellular aggregation in achieving a globally disruptive multitargeting effect, thereby offering potential applications in the treatment of malignant cells, including pathogens, tumor cells, and infected tissues.
Collapse
Affiliation(s)
- Anming Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Junfeng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Jiaqi Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Youzhi Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Silei Bai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Cailing Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Min Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yu Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Kang Wen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Miaomiao Luo
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Peiren Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Bo Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No.555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Huan Yang
- School of Medical Technology, Xuzhou Medical University, Xuzhou 221004, China
| | - Yugang Bai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR 999077, China
| | - Qingyun Cai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Huangsheng Pu
- College of Advanced Interdisciplinary Studies & Hunan Provincial Key Laboratory of Novel NanoOptoelectronic Information Materials and Devices, National University of Defense Technology, Changsha, Hunan 410073, China
- Nanhu Laser Laboratory, National University of Defense Technology, Changsha 410073, China
| | - Yu Qian
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wenhao Hu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wei Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No.555 Zuchongzhi Rd, Pudong, Shanghai 201203, China
| | - Muyang Wan
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Chunhui Zhang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Xinxin Feng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
18
|
Tang Z, Zhang J, Li W, Wen K, Gu Z, Zhou D, Su H. Supramolecular assembly of isomeric SN-38 prodrugs regulated by conjugation sites. J Mater Chem B 2024; 12:6146-6154. [PMID: 38842181 DOI: 10.1039/d4tb00717d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Supramolecular polymers (SPs) are an emerging class of drug transporters employed to improve drug therapy. Through the rational design of self-assembling monomers, one can optimize the properties of the resulting supramolecular nanostructures, such as size, shape, surface chemistry, release, and, therefore, biological fates. This study highlights the design of isomeric SN38 prodrugs through the conjugation of hydrophilic oligo(ethylene glycol) (OEG) with hydroxyls at positions 10 and 20 on hydrophobic SN-38. Self-assembling prodrug (SAPD) isomers 10-OEG-SN38 and 20-OEG-SN38 can self-assemble into giant nanotubes and filamentous assemblies, respectively, via aromatic associations that dominate self-assembly. Our study reveales the influence of modification sites on the assembly behavior and ability of the SN38 SAPDs, as well as drug release and subsequent in vitro and in vivo antitumor effects. The SAPD modified at position 20 exhibits stronger π-π interactions among SN38 units, leading to more compact packing and enhanced assembly capability, whereas OEG at position 10 poses steric hindrance for aromatic associations. Importantly, owing to its higher chemical and supramolecular stability, 20-OEG-SN38 outperforms 10-OEG-SN38 and irinotecan, a clinically used prodrug of SN38, in a CT26 tumor model, demonstrating enhanced tumor growth inhibition and prolonged animal survival. This study presents a new strategy of using interactions among drug molecules as dominating features to create supramolecular assemblies. It also brings some insights into creating effective supramolecular drug assemblies via the engineering of self-assembling building blocks, which could contribute to the optimization of design principles for supramolecular drug delivery systems.
Collapse
Affiliation(s)
- Zhenhai Tang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Jianhua Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Wenting Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Kaiying Wen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Zhipeng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Dongdong Zhou
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Hao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
19
|
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
|
20
|
Yi M, Ashton-Rickardt G, Tan W, Liu Z, He H, Hsieh JT, Xu B. Accelerating Cellular Uptake with Unnatural Amino Acid for Inhibiting Immunosuppressive Cancer Cells. Chemistry 2024; 30:e202400691. [PMID: 38527252 PMCID: PMC11132931 DOI: 10.1002/chem.202400691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 03/27/2024]
Abstract
Targeting immunosuppressive metastatic cancer cells is a key challenge in therapy. We recently have shown that a rigid-rod aromatic, pBP-NBD, that responds to enzymes and kill immunosuppressive metastatic osteosarcoma (mOS) and castration resistant prostate cancer (CRPC) cells in mimetic bone microenvironment. However, pBP-NBD demonstrated moderate efficacy against CRPC cells. To enhance activity, we incorporated the unnatural amino acid L- or D-4,4'-biphenylalanine (L- or D-BiP) into pBP-NBD, drastically increasing cellular uptake and CRPC inhibition. Specifically, we inserted BiP into pBP-NBD to target mOS (Saos2 and SJSA1) and CRPC (VCaP and PC3) cells with overexpressed phosphatases. Our results show that the D-peptide backbone with an aspartate methyl diester at the C-terminal offers the highest activity against these immunosuppressive mOS and CRPC cells. Importantly, imaging shows that the peptide assemblies almost instantly enter the cells and accumulate primarily within the endoplasmic reticulum of Saos2, SJSA1, and PC3 cells and at the lysosomes of VCaP cells. By using BiP to boost cellular uptake and self-assembly within cancer cells, this work illustrates an unnatural hydrophobic amino acid as a versatile and effective residue to boost endocytosis of synthetic peptides for intracellular self-assembly.
Collapse
Affiliation(s)
- Meihui Yi
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | | | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Zhiyu Liu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Jer-Tsong Hsieh
- Department of Urology, Southwestern Medical Center, University of Texas, Dallas, TX 75235, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| |
Collapse
|
21
|
Ren Y, Zhou Z, Maxeiner K, Kaltbeitzel A, Harley I, Xing J, Wu Y, Wagner M, Landfester K, Lieberwirth I, Weil T, Ng DYW. Supramolecular Assembly in Live Cells Mapped by Real-Time Phasor-Fluorescence Lifetime Imaging. J Am Chem Soc 2024; 146:11991-11999. [PMID: 38639465 PMCID: PMC11066860 DOI: 10.1021/jacs.4c01279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024]
Abstract
The complex dynamics and transience of assembly pathways in living systems complicate the understanding of these molecular to nanoscale processes. Current technologies are unable to track the molecular events leading to the onset of assembly, where real-time information is imperative to correlate their rich biology. Using a chemically designed pro-assembling molecule, we map its transformation into nanofibers and their fusion with endosomes to form hollow fiber clusters. Tracked by phasor-fluorescence lifetime imaging (phasor-FLIM) in epithelial cells (L929, A549, MDA-MB 231) and correlative light-electron microscopy and tomography (CLEM), spatiotemporal splicing of the assembly events shows time-correlated metabolic dysfunction. The biological impact begins with assembly-induced endosomal disruption that reduces glucose transport into the cells, which, in turn, stymies mitochondrial respiration.
Collapse
Affiliation(s)
- Yong Ren
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Zhixuan Zhou
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Konrad Maxeiner
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | | | - Iain Harley
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Jiaqi Xing
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Yingke Wu
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Manfred Wagner
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | | | | | - Tanja Weil
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - David Y. W. Ng
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| |
Collapse
|
22
|
Mo X, Song J, Liu X, Guo RC, Hu B, Yu Z. Redox-Regulated In Situ Seed-Induced Assembly of Peptides. Biomacromolecules 2024; 25:2497-2508. [PMID: 38478850 DOI: 10.1021/acs.biomac.3c01453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Morphology-transformational self-assembly of peptides allows for manipulation of the performance of nanostructures and thereby advancing the development of biomaterials. Acceleration of the morphological transformation process under a biological microenvironment is important to efficiently implement the tailored functions in living systems. Herein, we report redox-regulated in situ seed-induced assembly of peptides via design of two co-assembled bola-amphiphiles serving as a redox-resistant seed and a redox-responsive assembly monomer, respectively. Both of the peptides are able to independently assemble into nanoribbons, while the seed monomer exhibits stronger assembling propensity. The redox-responsive monomer undergoes morphological transformation from well-defined nanoribbons to nanoparticles. Kinetics studies validate the role of the assembled inert monomer as the seeds in accelerating the assembly of the redox-responsive monomer. Alternative addition of oxidants and reductants into the co-assembled monomers promotes the redox-regulated assembly of the peptides facilitated by the in situ-formed seeds. The reduction-induced assembly of the peptide could also be accelerated by in situ-formed seeds in cancer cells with a high level of reductants. Our findings demonstrate that through precisely manipulating the assembling propensity of co-assembled monomers, the in situ seed-induced assembly of peptides could be achieved. Combining the rapid assembly kinetics of the seed-induced assembly with the common presence of redox agents in a biological microenvironment, this strategy potentially offers a new method for developing biomedical materials in living systems.
Collapse
Affiliation(s)
- Xiaowei Mo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jinyan Song
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xin Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Ruo-Chen Guo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Binbin Hu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zhilin Yu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
- Haihe Laboratory of Synthetic Biology, 21 West 15th Avenue, Tianjin 300308, China
| |
Collapse
|
23
|
Zhou Y, Xu L, Sun X, Zhan W, Liang G. In situ peptide assemblies for bacterial infection imaging and treatment. NANOSCALE 2024; 16:3211-3225. [PMID: 38288668 DOI: 10.1039/d3nr05557d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Bacterial infections, especially antibiotic-resistant ones, remain a major threat to human health. Advances in nanotechnology have led to the development of numerous antimicrobial nanomaterials. Among them, in situ peptide assemblies, formed by biomarker-triggered self-assembly of peptide-based building blocks, have received increasing attention due to their unique merits of good spatiotemporal controllability and excellent disease accumulation and retention. In recent years, a variety of "turn on" imaging probes and activatable antibacterial agents based on in situ peptide assemblies have been developed, providing promising alternatives for the treatment and diagnosis of bacterial infections. In this review, we introduce representative design strategies for in situ peptide assemblies and highlight the bacterial infection imaging and treatment applications of these supramolecular materials. Besides, current challenges in this field are proposed.
Collapse
Affiliation(s)
- Yanyan Zhou
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing 210093, China
- Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Lingling Xu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, Jiangsu 210096, China.
| | - Xianbao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, Jiangsu 210096, China.
| | - Wenjun Zhan
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, Jiangsu 210096, China.
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, Jiangsu 210096, China.
| |
Collapse
|
24
|
Qiao Y, Wu G, Liu Z, He H, Tan W, Xu B. Assessment of the Enzymatic Dephosphorylation Kinetics in the Assemblies of a Phosphopentapeptide that Forms Intranuclear Nanoribbons. Biomacromolecules 2024; 25:1310-1318. [PMID: 38265878 PMCID: PMC11071069 DOI: 10.1021/acs.biomac.3c01288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Although the formation of peptide assemblies catalyzed by alkaline phosphatase (ALP) has received increasing attention in inhibiting cancer cells, the detailed enzyme kinetics of the dephosphorylation of the corresponding phosphopeptide assemblies have yet to be determined. We recently discovered that assemblies from a phosphopentapeptide can form intracellular nanoribbons that kill induced pluripotent stem cells or osteosarcoma cells, but the kinetics of enzymatic dephosphorylation remain unknown. Thus, we chose to examine the enzyme kinetics of the dephosphorylation of the phosphopentapeptide [NBD-LLLLpY (1)] from concentrations below to above its critical micelle concentration (CMC). Our results show that the phosphopeptide exhibits a CMC of 75 μM in phosphate saline buffer, and the apparent Vmax and Km values of alkaline phosphatase catalyzed dephosphorylation are approximately 0.24 μM/s and 5.67 mM, respectively. Despite dephosphorylation remaining incomplete at 60 min in all the concentrations tested, dephosphorylation of the phosphopeptide at concentrations of 200 μM or above mainly results in nanoribbons, dephosphorylation at concentrations of CMC largely produces nanofibers, and dephosphorylation below the CMC largely generates nanoparticles. Moreover, the formation of nanoribbons correlates with the intranuclear accumulation of the pentapeptide. By providing the first examination of the enzymatic kinetics of phosphopeptide assemblies, this work further supports the notion that the assemblies of phosphopentapeptides can act as a new functional entity for controlling cell fates.
Collapse
Affiliation(s)
- Yuchen Qiao
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Grace Wu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Zhiyu Liu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - 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
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| |
Collapse
|
25
|
Lu H, Yang X, Wang H. Tuning Phase Transition of Molecular Self-Assembly by Artificial Chaperones through Aromatic-Aromatic Interactions. Biomacromolecules 2024; 25:466-473. [PMID: 38147794 DOI: 10.1021/acs.biomac.3c01082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
The molecular chaperones are essential and play significant roles in controlling the protein phase transition and maintaining physiological homeostasis. However, manipulating phase transformation in biomimetic peptide self-assembly is still challenging. This work shows that an artificial chaperone modulates the energy landscape of supramolecular polymerization, thus controlling the phase transition of amyloid-like assemblies from crystals to hydrogels to solution. The absence of a chaperone allows the NapP to form crystals, while the presence of the chaperone biases the pathway to form nanofibrous hydrogels to soluble oligomers by adjusting the chaperone ratios. Mechanistic studies reveal that the aromatic-aromatic interaction is the key to trapping the molecules in a higher energy fold. Adding the chaperone relieves this restriction, lowers the energy barrier, and transforms the crystal into a hydrogel. This phase transformation can also be achieved in the macromolecular crowding environment, thus providing new insights into understanding molecular self-assembly in multiple component systems.
Collapse
Affiliation(s)
- Honglei Lu
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province 310027, China
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou, Zhejiang Province 310024, China
| | - Xuejiao Yang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou, Zhejiang Province 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, 18 Shilongshan Road, Hangzhou, Zhejiang Province 310024, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou, Zhejiang Province 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, 18 Shilongshan Road, Hangzhou, Zhejiang Province 310024, China
| |
Collapse
|