1
|
Zhang H, Wang Y, Jiang M, Wang K, Yan J, Li G, Zheng Z. Inherently anti-metastatic peptide hydrogels for sonodynamic-amplified ferroptosis in cancer therapy. Mater Today Bio 2025; 32:101688. [PMID: 40206142 PMCID: PMC11980000 DOI: 10.1016/j.mtbio.2025.101688] [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: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer metastasis remains a significant challenge in oncology, prompting the exploration of innovative biomaterials to enhance treatment efficacy. While many hydrogels only serve as passive carriers, this study presents two novel self-assembling peptides, CWEWTWY and NapFFSGP, which form supramolecular hydrogels with intrinsic anti-metastatic properties. We demonstrate a correlation between the nanofibrous morphology of these peptides and their enhanced anti-metastatic activity, mediated by disruption of F-actin organization and impacting pathways related to cancer cell adhesion and actin filament dynamics. In vivo studies confirm a significant reduction in lung metastasis using a 4T1 pulmonary metastasis model. We also demonstrate their potential as a simple, synergistic platform integrating sonodynamic therapy (SDT) and ferroptosis. Ironporphyrin (FP), incorporated into Gel@FP, acts as both a sonosensitizer and ferroptosis inducer. Upon ultrasound irradiation, FP generates localized reactive oxygen species, further amplifying ferroptosis through enhanced lipid peroxidation. Gel@FP combined with ultrasound demonstrates potent antitumor efficacy in vitro and in vivo, promoting apoptosis, ferroptosis, and immunogenic cell death, leading to enhanced tumor regression and robust immune activation. Our findings highlight the potential of anti-metastatic hydrogels as a promising multifunctional platform to address the challenges of metastasis while enhancing antitumor immunity.
Collapse
Affiliation(s)
- Hongxia Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Yamei Wang
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Mengmeng Jiang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Kunyu Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Jingru Yan
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Gongyu Li
- Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Science, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhen Zheng
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
2
|
Ma Y, Jiang Q, Liu X, Sun X, Liang G. In situ peptide assembly for cell membrane rewiring in tumor therapy. J Control Release 2025; 381:113637. [PMID: 40107514 DOI: 10.1016/j.jconrel.2025.113637] [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: 01/14/2025] [Revised: 02/14/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Peptide assembly on the cell membrane is capable of endowing cells with novel biological properties that are distinct from their original states, thereby playing a pivotal role in the regulation of diverse cellular biological events. In practical biomedical scenarios, in order to make peptide assembly more precisely meet the requirements of cells at different physiological stages and conditions to achieve desired effects of cell function regulation, it becomes particularly crucial to conduct precise in situ spatiotemporal control of peptide assembly on the cell membrane, thus attracting great attentions. Particularly for tumor treatment, this artificially manipulated cell surface engineering can achieve excellent anti-tumor effects by altering the cell membrane structure, influencing receptor clustering or interfering with relevant signal pathways. Of note, membrane-anchoring peptides play a key role in these processes. In this review, we focus on three main types of membrane-anchoring peptides, elaborating in detail on how their assembly regulation mechanisms influence the cell membrane remodeling effect, and further exert therapeutic effects on tumors. On this basis, we further introduce a variety of tumor treatment strategies combined with in situ peptide assembly on the cell membrane, and discuss the current opportunities and challenges in this field, aiming to present the overall research panorama and trend of in situ peptide self-assembly on the cell membrane for efficient tumor treatment.
Collapse
Affiliation(s)
- Yu Ma
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China
| | - Qiaochu Jiang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China
| | - Xiaoyang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China
| | - Xianbao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China.
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing 211189, China.
| |
Collapse
|
3
|
Zhou Z, Zhang P, Chen D, Kong N, Liu H, Liang J, Huang K, Wang H. Cecropin A-Derived Peptide for the Treatment of Osteomyelitis by Inhibiting the Growth of Multidrug-Resistant Bacteria and Eliminating Inflammation. ACS NANO 2025; 19:15733-15750. [PMID: 40231707 DOI: 10.1021/acsnano.4c18858] [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: 04/16/2025]
Abstract
Osteomyelitis poses substantial therapeutic challenges due to the prevalence of multidrug-resistant bacterial infections and associated inflammation. Current treatment regimens often rely on a combination of corticosteroids and antibiotics, which can lead to complications and impede effective bacterial clearance. In this study, we present CADP-10, a Cecropin A-derived peptide, designed to target methicillin-resistant Staphylococcus aureus (MRSA) and multidrug-resistant Escherichia coli (MRE), while simultaneously addressing inflammatory responses. CADP-10 self-assembles into nanobacterial net (NBacN) that selectively identify and bind to bacterial endotoxins (LPS and LTA), disrupting membrane integrity and depolarizing membrane potential, which culminates in bacterial death. Importantly, these NBacN are bound to LPS and LTA from dead bacteria, preventing their engagement with TLR receptors and effectively blocking downstream inflammatory pathways. Our assessments of CADP-10 demonstrate good biosafety in both in vitro and in vivo models. Notably, in a rabbit osteomyelitis model, CADP-10 eliminated MRSA-induced bone infections, mitigated inflammation, and promoted bone tissue regeneration. This research highlights the potential of CADP-10 as a multifunctional antimicrobial agent for the management of infectious inflammatory diseases.
Collapse
Affiliation(s)
- Ziao Zhou
- Department of Chemistry, Zhejiang University, Hangzhou 310027, Zhejiang Province, China
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Peng Zhang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Dinghao Chen
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Nan Kong
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Huayang Liu
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Juan Liang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Kai Huang
- Department of Orthopedics, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Hangzhou 310012, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| |
Collapse
|
4
|
Li Q, Mu Z, Dong Y, Ouyang Z, Zuo J, Wu Y, Yang Y, Sun S, Liang H, Bai L. In Situ Self-Assembly of Artificial Topological Nanostructures Enhances In Vivo Efficacy of PCSK9 Inhibitory Peptides. Angew Chem Int Ed Engl 2025; 64:e202502559. [PMID: 39945488 DOI: 10.1002/anie.202502559] [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: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
Optimizing the stability and affinity of peptides in vivo is critical for their development as alternatives to approved monoclonal antibodies. In recent years, efforts in academia and industry have focused on modifying Pep2-8, a classical antagonistic peptide targeting proprotein convertase subtilisin/kexin type 9 (PCSK9), to enhance its specificity and affinity. However, developing effective PCSK9 inhibitory peptides remains challenging, especially given the limited examples of their successful in vivo applications. Here, we designed transformable inhibitory peptide (TIP) against PCSK9 based on the modular structure of Pep2-8. Upon encountering PCSK9, TIP undergoes in situ self-assembly at the epidermal growth factor-like domain A (EGF-A) binding domain of PCSK9 to form artificial topological nanostructures (ATNs). The ATNs not only enhance peptide stability and prolong in vivo retention time but also strengthen PCSK9 binding through multivalent synergistic effects. We demonstrate that compared to Pep2-8, TIP forms ATNs which increasing its binding affinity for PCSK9 by approximately 18.7-fold in vitro. In high-fat diet mouse models, TIP significantly increase hepatic LDLR levels (2.0-fold) and reduced LDL-C and TC levels. We envision that the in situ formation of ATNs by peptides enhances in vivo stability and affinity, which offering an approach for development as antibody alternatives in clinical.
Collapse
Affiliation(s)
- Qin Li
- College of Pharmacy, Chongqing Medical University, Chongqing Research Center for Pharmaceutical Engineering, 1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Zhaode Mu
- College of Pharmacy, Chongqing Medical University, Chongqing Research Center for Pharmaceutical Engineering, 1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Yuhan Dong
- College of Pharmacy, Chongqing Medical University, Chongqing Research Center for Pharmaceutical Engineering, 1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Zubin Ouyang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Jianli Zuo
- College of Pharmacy, Chongqing Medical University, Chongqing Research Center for Pharmaceutical Engineering, 1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Yijie Wu
- College of Pharmacy, Chongqing Medical University, Chongqing Research Center for Pharmaceutical Engineering, 1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Yuxuan Yang
- College of Pharmacy, Chongqing Medical University, Chongqing Research Center for Pharmaceutical Engineering, 1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Si Sun
- National Engineering Research Center of Immunological Products, Third Military Medical University, 30 Gaotanyan Zheng Street, Shapingba District, Chongqing, China
| | - Hongwen Liang
- College of Pharmacy, Chongqing Medical University, Chongqing Research Center for Pharmaceutical Engineering, 1 Yixueyuan Road, Yuzhong District, Chongqing, China
| | - Lijuan Bai
- College of Pharmacy, Chongqing Medical University, Chongqing Research Center for Pharmaceutical Engineering, 1 Yixueyuan Road, Yuzhong District, Chongqing, China
| |
Collapse
|
5
|
Pei S, Liu Z, Jiao Q, Jin Q, Luo X, Liu Y, Zhou S, Pang S, Wu X, Xu K, Zhong W. Self-Reporting Ratiometric AIEgen-Peptide Nanoprobes for Activatable Chemotherapy and Noninvasive Imaging of Therapeutic Outcomes. J Med Chem 2025; 68:7767-7779. [PMID: 40170557 DOI: 10.1021/acs.jmedchem.5c00372] [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: 04/03/2025]
Abstract
Efficacious chemotherapy and real-time therapeutic monitoring remain major challenges in cancer treatment. Traditional systems often lack tumor specificity, limiting efficacy, and hindering therapy optimization. Moreover, the absence of real-time monitoring can lead to missed opportunities and increased risks of side effects. Herein, we designed a self-reporting ratiometric AIEgen-peptide nanoprobe (TPE-1(Hyd-DOX)-DEVD) for activatable chemotherapy and noninvasive imaging of therapeutic outcomes. When doxorubicin (DOX) in the nanoprobe is selectively activated in the acidic tumor microenvironment, the ensuing caspase-3 cascade triggers a morphological transformation of the nanoprobe that amplifies the TPE fluorescence. This enhancement allows the TPE/DOX fluorescence ratio to serve as an indicator for monitoring DOX activation and for providing therapeutic feedback. Both in vitro and in vivo studies demonstrated that TPE-1(Hyd-DOX)-DEVD exhibited an impressive tumor suppression efficacy and excellent biocompatibility. This study highlights the strong potential of this nanoprobe as a valuable tool for cancer theranostics, offering hope for more effective and personalized treatment strategies.
Collapse
Affiliation(s)
- Shicheng Pei
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Zhaohan Liu
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Qishu Jiao
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Qiling Jin
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Xuan Luo
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Yuanyuan Liu
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Shuyao Zhou
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Shuqin Pang
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoxing Wu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China
| | - Keming Xu
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 210009, China
| | - Wenying Zhong
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
6
|
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
|
7
|
Liu W, Wang B, Wei M, Hai Z. In situ self-assembled peptide nanoparticles improve the anti-hepatic fibrosis effect. J Mater Chem B 2025; 13:4080-4084. [PMID: 40079763 DOI: 10.1039/d4tb02819h] [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: 03/15/2025]
Abstract
Antagonistic peptide Leu-Ser-Lys-Leu (LSKL) is capable of blocking the transforming growth factor-β1 (TGF-β1) signaling pathway and exhibits anti-fibrotic effects. Herein, we constructed LSKL nanoparticles (NPs) in situ based on an alkaline phosphatase (ALP)-instructed self-assembly strategy for improving its specific therapeutic effect against liver fibrosis.
Collapse
Affiliation(s)
- Wenbin Liu
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China.
| | - Beibei Wang
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China.
| | - Mengxing Wei
- Department of Radiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
- Medical Imagine Research Center, Anhui Medical University, Hefei, Anhui 230601, China
| | - Zijuan Hai
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China.
| |
Collapse
|
8
|
Chung MC, Mendez‐Gomez HR, Soni D, McGinley R, Zacharia A, Ashbrook J, Stover B, Grippin AJ, Sayour EJ, Guan J. Multi-Step Assembly of an RNA-Liposome Nanoparticle Formulation Revealed by Real-Time, Single-Particle Quantitative Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414305. [PMID: 39887619 PMCID: PMC11948016 DOI: 10.1002/advs.202414305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Indexed: 02/01/2025]
Abstract
Self-assembly plays a critical role in nanoparticle-based applications. However, it remains challenging to monitor the self-assembly of multi-component nanomaterials at a single-particle level, in real-time, with high throughput, and in a model-independent manner. Here, multi-color fluorescence microscopy is applied to track the assembly of both liposomes and mRNA simultaneously in clinical mRNA-based cancer immunotherapy. Imaging reveals that the assembly occurs in discrete steps: initially, RNA adsorbs onto the liposomes; then, the RNA-coated liposomes cluster into heterogeneous structures ranging from sub-micrometer to tens of micrometers. The clustering process is consistent with a Smoluchowski model with a Brownian diffusion kernel. The transition between the two steps of assembly is determined by the orientation of RNA-mediated interactions. Given the facile application of this approach and the ubiquity of the components studied, the imaging and analysis in this work are readily applied to monitor multi-component assembly of diverse nanomaterials.
Collapse
Affiliation(s)
- Michael C. Chung
- Division of Chemical Biology and Medicinal ChemistryCollege of PharmacyUniversity of Texas at AustinAustinTX78712USA
- Department of PhysicsUniversity of FloridaGainesvilleFL32611USA
| | - Hector R. Mendez‐Gomez
- Department of NeurosurgeryPreston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida Lillian S. WellsGainesvilleFL32610USA
| | - Dhruvkumar Soni
- Department of NeurosurgeryPreston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida Lillian S. WellsGainesvilleFL32610USA
| | - Reagan McGinley
- Department of Microbiology and Cell ScienceUniversity of FloridaGainesvilleFL32603USA
| | - Alen Zacharia
- Department of PhysicsUniversity of FloridaGainesvilleFL32611USA
| | - Jewel Ashbrook
- Middlebury College Department of PhysicsMcCardell Bicentennial HallMiddleburyVT05753USA
| | - Brian Stover
- Department of PediatricsDivision of Pediatric Hematology OncologyUniversity of FloridaGainesvilleFL32610USA
| | - Adam J. Grippin
- MD Anderson Cancer CenterDivision of Radiation OncologyUniversity of TexasHoustonTX77030USA
| | - Elias J. Sayour
- Department of NeurosurgeryPreston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida Lillian S. WellsGainesvilleFL32610USA
- Department of PediatricsDivision of Pediatric Hematology OncologyUniversity of FloridaGainesvilleFL32610USA
| | - Juan Guan
- Division of Chemical Biology and Medicinal ChemistryCollege of PharmacyUniversity of Texas at AustinAustinTX78712USA
| |
Collapse
|
9
|
Liao X, Tang M, Li J, Guo R, Zhong C, Chen X, Zhang X, Mo H, Que D, Yu W, Song X, Li H, Cai Y, Yang P. Acid-Triggered Cascaded Responsive Supramolecular Peptide Alleviates Myocardial Ischemia‒Reperfusion Injury by Restoring Redox Homeostasis and Protecting Mitochondrial Function. Adv Healthc Mater 2025; 14:e2404319. [PMID: 39831810 DOI: 10.1002/adhm.202404319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Redox imbalance, including excessive production of reactive oxygen species (ROS) caused by mitochondrial dysfunction and insufficient endogenous antioxidant capacity, is the primary cause of myocardial ischemia‒reperfusion (I/R) injury. In the exploration of reducing myocardial I/R injury, it is found that protecting myocardial mitochondrial function after reperfusion not only reduces ROS bursts but also inhibits cell apoptosis triggered by the release of cytochrome c. Additionally, nuclear factor erythroid 2-related factor 2 (Nrf2) is considered a potential therapeutic target for treating myocardial I/R injury by enhancing the cellular antioxidant capacity through the induction of endogenous antioxidant enzymes. In this study, a peptide‒drug conjugate OI-FFG-ss-SS31(ISP) is developed by integrating the Nrf2 activator 4-octyl itaconate (OI) and the mitochondria-targeting protective peptide elamipretide (SS31), and its therapeutic potential for myocardial I/R injury is explored. The results showed that ISP could self-assemble into nanofibers in response to the acidic microenvironment and bind to Keap-1 with high affinity, thereby activating Nrf2 and enhancing antioxidant capacity. Simultaneously, the release of SS31 could improve mitochondrial function and reduce ROS, ultimately providing a restoration of redox homeostasis to effectively alleviate myocardial I/R injury. This study presents a promising acid-triggered peptide-drug conjugate for treating myocardial I/R injury.
Collapse
Affiliation(s)
- Xu Liao
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Min Tang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Department of Cardiology of Zhuzhou Central Hospital, NO.116 Changjiang South Road Tianyuan District, Zhuzhou, 412000, P. R. China
| | - Jiejing Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Runze Guo
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Chongbin Zhong
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Xiangzhou Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Xuwei Zhang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Hongwei Mo
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Dongdong Que
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Wenjie Yu
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Xudong Song
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Hekai Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Yanbin Cai
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Pingzhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| |
Collapse
|
10
|
Zhang Z, Liu X, Tian F, Wang Y, Hu B, Yu Z. Aspartate Isomerization Regulates in Situ Assembly of Peptides into Supramolecular Probes for Detection of Protein Carboxyl Methyltransferase in Bladder Cancer. NANO LETTERS 2025. [PMID: 40016095 DOI: 10.1021/acs.nanolett.4c06452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Protein carboxyl methyltransferase (PCMT) restores aspartate isomers in proteins and plays a critical role in cancer prognosis. However, in vivo detection of PCMT remains challenging. Here, we report the aspartate isomerization-regulated in situ assembly of peptides into supramolecular probes within living cells for PCMT detection in bladder cancer. The peptide consists of alternating hydrophobic and hydrophilic residues and contains an isoAsp residue as a kinked site to prevent the facial amphiphilicity of the peptide. Exposure to PCMT converts isoAsp to Asp within the peptide, thereby promoting its assembly into nanofibers. Incorporation of 7-nitro-2,1,3-benzoxadiazole (NBD) into the nanofibers enables PCMT detection based on hydrophobicity-dependent fluorescence of NBD units. Both cellular and animal studies confirm the capability of supramolecular probes for efficient detection of PCMT. Our finding demonstrates an efficient strategy for regulating peptide assembly in living systems and thus provides a new tool for creation of biomedical agents in the future.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Yushi 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
| | - 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
| |
Collapse
|
11
|
Guan T, Chen Z, Wang X, Gao S, Lu X, Li Y, Wang Z, Zhang S, Guo Y, Guo M, Cui Y, Wang Y, Chen C. Harnessing Mn 2+ Ions and Antitumor Peptides: A Robust Hydrogel for Enhanced Tumor Immunotherapy. J Am Chem Soc 2025; 147:6523-6535. [PMID: 39950218 DOI: 10.1021/jacs.4c14700] [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: 02/27/2025]
Abstract
Immunotherapy has attracted widespread attention because of its durable and effective antitumor properties. However, systemic delivery strategies often result in immune-related off-target toxicity effects and inadequate drug retention at the tumor site, which limits its broader application. In this research, we designed a dual-functional antitumor peptide (N-Pep) that serves as both a therapeutic agent and metal ions (Mn2+) immunomodulator carrier. The rational designed antitumor peptide self-assembles into a hydrogel through coordination with Mn2+ ions (referred to as N-Pep-Mn gel). The multiporous hydrogel network allows for efficient loading of antiprogrammed death-1 antibody (αPD-1). The hydrogel served as a depot for the sustained release of Mn2+ ions and encapsulated αPD-1, effectively activating dendritic cells, polarizing tumor-associated macrophages and enhancing effector T cell infiltration, thereby leading to the effective inhibition of tumor growth through intratumoral and systemic immune responses. Additionally, the hydrogel induces robust immune memory, providing substantial protection against tumor recurrence. These findings underscore the potential of Mn2+ ion-coordinated antitumor peptide hydrogel as an advanced platform for enhancing antitumor immunotherapy.
Collapse
Affiliation(s)
- Tong Guan
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ziwei Chen
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Susu Gao
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyi Lu
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Li
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhichao Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuhan Zhang
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuecong Guo
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengyu Guo
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanyan Cui
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China
| | - Yaling Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunying Chen
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
12
|
Feng R, Xu W, Ning J, Ma Q, Wang H, Li L, Xu S, Wang L. Design of Fluorinated Peptides as Biotransformed Urinalysis Biomarkers for Non-Invasive Diagnosis and Treatment of Liver Injury through Enzyme Directed Kinetics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413571. [PMID: 39817848 DOI: 10.1002/adma.202413571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/29/2024] [Indexed: 01/18/2025]
Abstract
Urinalysis, as a non-invasive and efficient diagnostic method, is very important but faces great challenges due to the complex compositions of urine and limited naturally occurring biomarkers for diseases. Herein, by leveraging the intrinsic absence of endogenous fluorinated interference, a strategy with the enzymatically activated assembly of synthetic fluorinated peptide for cholestatic liver injury (CLI) diagnosis and treatment through 19F nuclear magnetic resonance (NMR) urinalysis and efficient drug retention is developed. Specifically, alkaline phosphatase (ALP), overexpressed in the liver of CLI mice, triggers the assembly of fluorinated peptide, thus, directing the traffic and dynamic distribution of the synthetic biomarkers after administration, whereas CLI mice display much slower clearance of peptides through urine as compared with healthy counterparts. As such, it enables to transform pathophysiological information into exogenous signals via noninvasive urinary monitoring. Moreover, as a proof-of-concept, by grafting different functional groups to peptides, the theranostic platforms can be established to provide a new paradigm for the design of multifunctional peptides.
Collapse
Affiliation(s)
- Ruxin Feng
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Weilu Xu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jinhui Ning
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qian Ma
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Hui Wang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Liangyu Li
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Suying Xu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Leyu Wang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
13
|
Yang Y, Zheng Y, Tong T, Dong J, Zhao M, Zhang G, Yu Z, Dong L, Jiang J, Yuan Y. Size-Dependent Cascade Enhancement of T 1-T 2 Dual-Modal MRI in Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414201. [PMID: 39828516 DOI: 10.1002/adma.202414201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Currently, there is no conclusive evidence indicating that in situ self-assembled Gd nanostructures of varying sizes demonstrate distinct T1 and T2 signal enhancement capabilities. Furthermore, it remains uncertain whether size adjustment can effectively achieve enhanced T1-T2 dual-modal MRI. To address these uncertainties, a two-step in situ self-assembly strategy is developed. This approach began with a small-sized nanoprobe, Gd-TCO-P, with a hydrodynamic diameter (dH) of 16 ± 3 nm. This nanoprobe underwent alkaline phosphatase (ALP) cleavage and self-assembled intracellularly into short nanofibers termed Gd-NFs (dH: 200 ± 51 nm). The subsequent introduction of tetrazine-tetrazine crosslinked these Gd-NFs, leading to the formation of larger two-stage dendritic nanofibers known as Gd-TS-NFs (dH: 4371 ± 236 nm). This process achieves size-dependent enhancement of both T1 and T2 signals, which is validated through both in vitro and in vivo experiments, enabling precise long-term imaging of ALP-overexpressing tumors. This study not only provides valuable insights into the relationship between the size of in situ formed Gd nanostructures and T1/T2 MRI contrast enhancement, but also suggests a promising strategy for clinical applications of T1-T2 dual-modal MRI.
Collapse
Affiliation(s)
- Yanyun Yang
- State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yifan Zheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Tong Tong
- State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Jiajing Dong
- State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Miaoxin Zhao
- State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Guangtao Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Zian Yu
- State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Ling Dong
- Department of Chemistry and Chemical Engineering, Hefei Normal University, Hefei, Anhui, 230061, China
| | - Jinhui Jiang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Yue Yuan
- State Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
14
|
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
|
15
|
Zhu W, Yu M, Wang M, Zhang M, Hai Z. Sequential self-assembly and release of a camptothecin prodrug for tumor-targeting therapy. NANOSCALE 2025; 17:2061-2067. [PMID: 39648922 DOI: 10.1039/d4nr03519d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Chemotherapy is the most commonly used method to treat malignant tumors with a wide range of drugs. However, chemotherapeutic drugs are characterized by poor solubility, low stability and specificity, as well as drug resistance, which led to their limited bioavailability and severe adverse effects. Therefore, most researches focus on one or two strategies while a few researches focus on three strategies to improve the efficacy of drugs. Herein, we combined three strategies (targeted therapy, prodrug design and drug delivery) to exploit a self-assembled camptothecin (CPT) prodrug (CPT-SS-FFEYp-Biotin) for enhancing therapeutic efficacy and reducing side effects of CPT. CPT-SS-FFEYp-Biotin enters into tumor cells following the recognition between biotin and biotin receptors. Moreover, the over-expressed alkaline phosphatase (ALP) on cell membranes specifically dephosphorylates CPT-SS-FFEYp-Biotin to CPT-SS-FFEY-Biotin, which self-assembles into a CPT hydrogel with the local enrichment of CPT. Subsequently, excess glutathione (GSH) in tumor cells can reduce the disulfide bond of CPT-SS-FFEY-Biotin to slowly release CPT for sustained tumor therapy. Cell experiments demonstrated that CPT-SS-FFEYp-Biotin enhances therapeutic efficacy of CPT on tumor cells while being safer to normal cells than CPT. Moreover, CPT-SS-FFEYp-Biotin effectively improved anti-tumor treatment of CPT in vivo. We envision that the integration of these three strategies is helpful to exploit a variety of prodrugs for effective anti-tumor treatment in the future.
Collapse
Affiliation(s)
- Wujuan Zhu
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China.
| | - Minghui Yu
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China.
| | - Minghui Wang
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China.
| | - Miaomiao Zhang
- College of Chemistry and Center for Advanced Analysis & Gene Sequencing, Zhengzhou University, Zhengzhou 450001, China.
| | - Zijuan Hai
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China.
| |
Collapse
|
16
|
Ji S, Pan T, Wang K, Zai W, Jia R, Wang N, Jia S, Ding D, Shi Y. A Membrane-Anchoring Self-Assembling Peptide Allows Bioorthogonal Coupling of Type-I AIEgens for Pyroptosis-Induced Cancer Therapy. Angew Chem Int Ed Engl 2025; 64:e202415735. [PMID: 39223092 DOI: 10.1002/anie.202415735] [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: 08/17/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/04/2024]
Abstract
Enrichment of photosensitizers (PSs) on cancer cell membranes via bioorthogonal reactions is considered to be a very promising therapeutic modality. However, azide-modified sugars-based metabolic labeling processes usually lack targeting and the labeling speed is relatively slow. Moreover, it has been rarely reported that membrane-anchoring pure type-I PSs can induce cancer cell pyroptosis. Here, we report an alkaline phosphatase (ALP) and cholecystokinin-2 receptor (CCK2R) dual-targeting peptide named DBCO-pYCCK6, which can selectively and rapidly self-assemble on cancer cell membrane, and then bioorthogonal enrich type-I aggregation-induced emission luminogens (AIEgen) PSs (SAIE-N3) on the cell membrane. Upon light irradiation, the membrane-anchoring SAIE-N3 could effectively generate type-I reactive oxygen species (ROS) to induce gasdermin E (GSDME)-mediated pyroptosis. In vivo experiments demonstrated that the bioorthogonal combination strategy of peptide and AIEgen PSs could significantly inhibit tumor growth, which is accompanied by CD8+ cytotoxic T cell infiltration. This work provides a novel self-assembly peptide-mediated bioorthogonal reaction strategy to bridge the supramolecular self-assembly and AIE field through strain-promoted azide-alkyne cycloaddition (SPAAC) and elucidates that pure type-I membrane-anchoring PSs can be used for cancer therapy via GSDME-mediated pyroptosis.
Collapse
Affiliation(s)
- Shenglu Ji
- The Key Laboratory of Biomedical Materials, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tengwu Pan
- Frontiers Science Center for New Organic Matter, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Kaiyuan Wang
- The Key Laboratory of Biomedical Materials, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Weiqi Zai
- The Key Laboratory of Biomedical Materials, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Ruikang Jia
- Frontiers Science Center for New Organic Matter, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Nannan Wang
- The Key Laboratory of Biomedical Materials, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shaorui Jia
- Frontiers Science Center for New Organic Matter, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Dan Ding
- The Key Laboratory of Biomedical Materials, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Frontiers Science Center for New Organic Matter, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yang Shi
- Frontiers Science Center for New Organic Matter, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| |
Collapse
|
17
|
Zhang Z, Hu Y, Ding Y, Zhang X, Dong X, Xie L, Yang Z, Hu ZW. Dual-Enzyme-Instructed Peptide Self-Assembly to Boost Immunogenic Cell Death by Coordinating Intracellular Calcium Overload and Chemotherapy. ACS NANO 2025; 19:488-503. [PMID: 39754594 DOI: 10.1021/acsnano.4c10119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The concept of immunogenic cell death (ICD) induced by chemotherapy as a potential synergistic modality for cancer immunotherapy has been widely discussed. Unfortunately, most chemotherapeutic agents failed to dictate effective ICD responses due to their defects in inducing potent ICD signaling. Here, we report a dual-enzyme-instructed peptide self-assembly platform of CPMC (CPT-GFFpY-PLGVRK-Caps) that cooperatively utilizes camptothecin (CPT) and capsaicin (Caps) to promote ICD and engage systemic adaptive immunity for tumor rejection. Although CPT and Caps respectively prevent tumor progression by inhibiting type-I DNA topoisomerase and activating transient receptor potential cation channel subfamily V member 1 (TRPV1) for intracellular calcium overload, neither alone effectively stimulates sufficient ICD signaling to meet immunotherapeutic needs. CPMC, sequentially allowing an active Caps derivative of VRK-Caps and CPT to release extracellularly and intracellularly, can synergize two distinct apoptosis pathways stimulated by Caps and CPT to increase tumor immunogenicity and elicit systemic T-cell-based immunity. Consequently, CPMC facilitates the generation of improved tumor-specific cytotoxic T-cell responses and sustained immunological memory, successfully suppressing both primary and distant tumors. Moreover, CPMC can render tumors susceptible to PD-L1 blockade and synergize with an antiprogrammed cell death-ligand 1 (aPDL1) antibody for tumor inhibition. Combining two cancer chemotherapeutic drugs with low ICD-stimulating capacity using a peptide self-assembly strategy was demonstrated to boost ICD responses and potentiate cancer immunotherapy.
Collapse
Affiliation(s)
- Zhenghao Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, 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
| | - Yuhan 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
| | - Yinghao Ding
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, 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
| | - 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
| | - Xiao Dong
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| | - Limin Xie
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, 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
| | - 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
- 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, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, 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
| |
Collapse
|
18
|
Guo P, Jiang YB, Jiang T. Peptide Designs for Cell-Interfacing Assemblies. Chemistry 2025; 31:e202402880. [PMID: 39648953 DOI: 10.1002/chem.202402880] [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: 07/30/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 12/10/2024]
Abstract
Constructing synthetic materials to interact with cells offers significant promise for understanding natural cellular processes and manipulating cell behaviors beyond nature's capabilities. Peptide assemblies are particular promising in this regard, as they have demonstrated efficacy in promoting cell differentiation, repair, and regeneration as supportive scaffolds. However, distinct gaps persist between natural and synthetic assembly systems in various aspects. This Concept review explores representative studies focusing on developing chemical strategies to design peptide assemblies with precise control over displayed molecules, adjustable molecular dynamics to modulate cell behaviors, and responsiveness to biological stimuli. This paper would inspire more fundamental studies on molecular assemblies and foster inter-disciplinary interests in material applications, advancing the interplay between natural cells and synthetic materials.
Collapse
Affiliation(s)
- Pan Guo
- Department of Chemistry, Collage of Chemistry and Chemical Engineering, Xiamen University, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, China
| | - Yun-Bao Jiang
- Department of Chemistry, Collage of Chemistry and Chemical Engineering, Xiamen University, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, China
| | - Tao Jiang
- Department of Chemistry, Collage of Chemistry and Chemical Engineering, Xiamen University, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
19
|
Jia F, Luo T, Zhuang JY, Guo P, Fang N, Jiang YB, Jiang T. Noncovalently Bridging Cell-Surface Proteins Using Synthetic Peptides to Modulate Cell Apoptosis. NANO LETTERS 2025; 25:268-275. [PMID: 39680928 DOI: 10.1021/acs.nanolett.4c04959] [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: 12/18/2024]
Abstract
Controlled high-order clustering of cell-surface proteins is an essential but unmatched regulatory mechanism in living systems for the modulation of cell behavior. Here, we present a strategy for generating extended and tunable one-dimensional clusters of death receptors on live cell surfaces by employing synthetic peptides to noncovalently bridging the proteins. The on-cell assembly process is validated through super-resolution fluorescence imaging and fluorescence lifetime imaging analyses. By adjusting the number of spacing peptides between the receptors before and even after the cluster formation, receptor separation can be precisely varied at nanoscale to drive cells into apoptotic or antiapoptotic states. Remarkably, this approach results in higher levels of cell apoptosis compared to the conventional practice of using preformed ligand-appended peptide coassemblies. These results demonstrate that in situ fabrication of cell-interfacing materials with compositional control permits robust and effective manipulation of high-order clustering of cell-surface proteins, advancing our ability to regulate cell behavior.
Collapse
Affiliation(s)
- Fan Jia
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen 361005, China
| | - Tian Luo
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen 361005, China
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Xiamen 361005, China
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen 361005, China
| | - Jin-Yan Zhuang
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen 361005, China
| | - Pan Guo
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen 361005, China
| | - Ning Fang
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen 361005, China
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Xiamen 361005, China
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen 361005, China
| | - Yun-Bao Jiang
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen 361005, China
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen 361005, China
| | - Tao Jiang
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen 361005, China
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen 361005, China
| |
Collapse
|
20
|
Wen Z, Song ZZ, Cai MZ, Zhang NY, Li HZ, Yang Y, Wang QT, Ghafoor MH, An HW, Wang H. Biomimetic Nanomaterials Based on Peptide In Situ Self-Assembly for Immunotherapy Applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70005. [PMID: 39895019 DOI: 10.1002/wnan.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/04/2024] [Accepted: 01/01/2025] [Indexed: 02/04/2025]
Abstract
Cancer remains the leading cause of patient death worldwide and its incidence continues to rise. Immunotherapy is rapidly developing due to its significant differences in the mechanism of action from conventional radiotherapy and targeted antitumor drugs. In the past decades, many biomaterials have been designed and prepared to construct therapeutic platforms that modulate the immune system against cancer. Immunotherapeutic platforms utilizing biomaterials can markedly enhance therapeutic efficacy by optimizing the delivery of therapeutic agents, minimizing drug loss during circulation, and amplifying immunomodulatory effects. The intricate physiological barriers of tumors, coupled with adverse immune environments such as inadequate infiltration, off-target effects, and immunosuppression, have emerged as significant obstacles impeding the effectiveness of oncology drug therapy. However, most of the current studies are devoted to the development of complex immunomodulators that exert immunomodulatory functions by loading drugs or adjuvants, ignoring the complex physiological barriers and adverse immune environments of tumors. Compared with conventional biomaterials, biomimetic nanomaterials based on peptide in situ self-assembly with excellent functional characteristics of biocompatibility, biodegradability, and bioactivity have emerged as a novel and effective tool for cancer immunotherapy. This article presents a comprehensive review of the latest research findings on biomimetic nanomaterials based on peptide in situ self-assembly in tumor immunotherapy. Initially, we categorize the structural types of biomimetic peptide nanomaterials and elucidate their intrinsic driving forces. Subsequently, we delve into the in situ self-assembly strategies of these peptide biomimetic nanomaterials, highlighting their advantages in immunotherapy. Furthermore, we detail the applications of these biomimetic nanomaterials in antigen presentation and modulation of the immune microenvironment. In conclusion, we encapsulate the challenges and prospective developments of biomimetic nanomaterials based on peptide in situ self-assembly for clinical translation in immunotherapy.
Collapse
Affiliation(s)
- Zhuan Wen
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
| | - Zhang-Zhi Song
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
| | - Ming-Ze Cai
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
| | - Ni-Yuan Zhang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Hao-Ze Li
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
| | - Yang Yang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
| | - Qian-Ting Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
| | - Muhammad Hamza Ghafoor
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
| | - Hong-Wei An
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Hao Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
21
|
Ma J, Yang H, Tian X, Meng F, Zhai X, Li A, Li C, Wang M, Wang G, Lu C, Bai J. Matrix metalloproteinase 2-responsive dual-drug-loaded self-assembling peptides suppress tumor growth and enhance breast cancer therapy. Bioeng Transl Med 2024; 9:e10702. [PMID: 39545088 PMCID: PMC11558207 DOI: 10.1002/btm2.10702] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/07/2024] [Accepted: 07/04/2024] [Indexed: 11/17/2024] Open
Abstract
Conventional chemotherapeutic agents are limited by their lack of targeting and penetration and their short retention time, and chemotherapy might induce an immune suppressive environment. Peptide self-assembly can result in a specific morphology, and the resulting morphological changes are stimuli responsive to the external environment, which is important for drug permeation and retention of encapsulated chemotherapeutic agents. In this study, a polypeptide (Pep1) containing the peptide sequences PLGLAG and RGD that is responsive to matrix metalloproteinase 2 (MMP-2) was successfully developed. Pep1 underwent a morphological transformation from a spherical structure to aggregates with a high aspect ratio in response to MMP-2 induction. This drug delivery system (DI/Pep1) can transport doxorubicin (DOX) and indomethacin (IND) simultaneously to target tumor cells for subsequent drug release while extending drug retention within tumor cells, which increases immunogenic cell death and facilitates the immunotherapeutic effect of CD4+ T cells. Ultimately, DI/Pep1 attenuated tumor-associated inflammation, enhanced the body's immune response, and inhibited breast cancer growth by combining the actions of DOX and IND. Our research offers an approach to hopefully enhance the effectiveness of cancer treatment.
Collapse
Affiliation(s)
- Jihong Ma
- School of Clinical MedicineShandong Second Medical UniversityWeifangChina
| | - Haiyan Yang
- Emergency DepartmentYantaishan HospitalYantaiChina
| | - Xue Tian
- School of Basic Medical SciencesShandong Second Medical UniversityWeifangChina
| | - Fanhu Meng
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Xiaoqing Zhai
- School of Clinical MedicineShandong Second Medical UniversityWeifangChina
| | - Aimei Li
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Chuntao Li
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Min Wang
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Guohui Wang
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Chunbo Lu
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Jingkun Bai
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| |
Collapse
|
22
|
Shi K, Fu W, Farhadi Sabet Z, Ye J, Liang S, Liu T, Liu Q, Guo M, You M, Wu J, Bai R, Liu Y, Hu B, Cui X, Li J, Chen C. Hydrogel-Mediated Jamming of Exosome Communications That Counter Tumor Adaption in the Tumor Immune Microenvironment. ACS NANO 2024. [PMID: 39441690 DOI: 10.1021/acsnano.4c07603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Hypoxia, a common occurrence within solid tumors, can stimulate the dissemination of deceptive tumor exosomes, which function as communicative bridges and orchestrate the recruitment of various supportive cell types for enhanced tumor adaptability in a tumor immune microenvironment. Current nanotechnology provides us intelligent strategies to combat the hypoxic tumor microenvironment. However, once exposed to external stimuli, such as chemotherapy, tumor cells simultaneously release malignant signals to develop tumor migration and immunosuppression, posing challenges to clinical practice. Taking advantage of the membrane-targeting therapeutic strategy, the application of a self-assembled short peptide (PepABS-py), affording hydrogels on tumor cell surfaces, can block exosome dissemination with fiber-like nanostructures and effectively limit the systemic adverse effects of traditional therapeutics. Moreover, PepABS-py can attenuate the hypoxic tumor microenvironment in vivo by carrying an inhibitor of the hypoxic tumor-overexpressed CA IX enzyme, where hypoxia is also a crucial regulator to induce tumor exosomes and mediate intercellular communications within the immune system. Herein, its application on jamming exosome communications can target the T cell-related signaling pathway by regulating microRNAs in exosome cargoes and ultimately enhances CD8+ T cell infiltration and alleviates inflammatory monocytes at metastasis sites. Collectively, with the capability of blocking exosome dissemination, PepABS-py can be applied as a promising tumor membrane-targeting therapeutic tool to counter tumor adaption within an immune microenvironment and further advance traditional chemotherapy.
Collapse
Affiliation(s)
- Kejian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Wenjiao Fu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Zeinab Farhadi Sabet
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Jinmin Ye
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Shijian Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Tao Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Qiaolin Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Min You
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Junguang Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Ru Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Bin Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Xuejing Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Jiayang Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100021, People's Republic of China
| |
Collapse
|
23
|
Tang R, Zhang Z, Liu X, Zhu L, Xu Y, Chai R, Zhan W, Shen S, Liang G. Fibroblast Growth Factor Receptor 1-Specific Dehydrogelation to Release Its Inhibitor for Enhanced Lung Tumor Therapy. ACS NANO 2024; 18:29223-29232. [PMID: 39392940 DOI: 10.1021/acsnano.4c11548] [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: 10/13/2024]
Abstract
Fibroblast growth factor receptor 1 (FGFR1) is emerging as a promising molecular target of lung cancer, and various FGFR1 inhibitors have exhibited significant therapeutic effects on lung cancer in preclinical research. Due to their low targeting ability or bioavailability, direct administration of these inhibitors may cause side effects. Herein, a hydrogelator, Nap-Phe-Phe-Phe-Glu-Thr-Glu-Leu-Tyr-OH (Nap-Y), was rationally designed to coassemble with an FGFR1 inhibitor nintedanib (Nin) to form a peptide hydrogel Gel Y/Nin for localized administration and FGFR1-triggered release of Nin. Upon specific phosphorylation by FGFR1 overexpressed on lung cancer cells, Nap-Y in Gel Y/Nin is converted to the hydrophilic product Nap-Phe-Phe-Phe-Glu-Thr-Glu-Leu-Tyr(H2PO3)-OH (Nap-Yp), leading to dehydrogelation of the gel and subsequent Nin release. In vitro experiments demonstrate that the release of Nin in a sustained manner from Gel Y/Nin significantly suppresses the survival, migration, and invasion of A549 cells by inhibiting FGFR1 expression and its phosphorylation function on downstream signaling molecules. Nude mouse studies show that Gel Y/Nin exhibits enhanced therapeutic efficacy on lung tumor than free Nin. We anticipate that Gel Y/Nin will be utilized for lung cancer treatment in clinical settings in the near future.
Collapse
Affiliation(s)
- Runqun Tang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Ziyi Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Xiaoyang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Liangxi Zhu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Yuting Xu
- Breast Surgery, Wenzhou Central Hospital, Wenzhou 325000, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China
| | - Wenjun Zhan
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Shurong Shen
- Breast Surgery, Wenzhou Central Hospital, Wenzhou 325000, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
24
|
Chen Y, Tan BSN, Cheng Y, Zhao Y. Artificial Polymerizations in Living Organisms for Biomedical Applications. Angew Chem Int Ed Engl 2024; 63:e202410579. [PMID: 39086115 DOI: 10.1002/anie.202410579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/16/2024] [Accepted: 07/31/2024] [Indexed: 08/02/2024]
Abstract
Within living organisms, numerous nanomachines are constantly involved in complex polymerization processes, generating a diverse array of biomacromolecules for maintaining biological activities. Transporting artificial polymerizations from lab settings into biological contexts has expanded opportunities for understanding and managing biological events, creating novel cellular compartments, and introducing new functionalities. This review summarizes the recent advancements in artificial polymerizations, including those responding to external stimuli, internal environmental factors, and those that polymerize spontaneously. More importantly, the cutting-edge biomedical application scenarios of artificial polymerization, notably in safeguarding cells, modulating biological events, improving diagnostic performance, and facilitating therapeutic efficacy are highlighted. Finally, this review outlines the key challenges and technological obstacles that remain for polymerizations in biological organisms, as well as offers insights into potential directions for advancing their practical applications and clinical trials.
Collapse
Affiliation(s)
- Yun Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Brynne Shu Ni Tan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Yu Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| |
Collapse
|
25
|
Zhang H, Mou J, Ding J, Qin W. Peptide-Driven Assembly of Magnetic Beads for Potentiometric Sensing of Bacterial Enzyme at a Subcellular Level. ACS Sens 2024; 9:4947-4955. [PMID: 39180154 DOI: 10.1021/acssensors.4c01632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
Bacterial enzymes with different subcellular localizations play a critical ecological role in biogeochemical processing. However, precisely quantifying enzymes localized at certain subcellular levels, such as extracellular enzymes, has not yet been fully realized due to the complexity and dynamism of the bacterial outer membrane. Here we present a magneto-controlled potentiometric sensing platform for the specific detection of extracellular enzymatic activity. Alkaline phosphatase (ALP), which is one of the crucial hydrolytic enzymes in the ocean, was selected as the target enzyme. Magnetic beads functionalized with an ALP-responsive self-assembled peptide (GGGGGFFFpYpYEEE, MBs-peptides) prevent negatively charged peptides from entering the bacterial outer membrane, thereby enabling direct potentiometric sensing of extracellular ALP both attached to the bacterial cell surface and released into the surrounding environment. The dephosphorylation-triggered assembly of peptide-coupled magnetic beads can be directly and sensitively measured by using a magneto-controlled sensor. In this study, extracellular ALP activity of Pseudomonas aeruginosa at concentrations ranging from 10 to 1.0 × 105 CFU mL-1 was specifically and sensitively monitored. Moreover, this magneto-controlled potentiometric method enabled a simple and accurate assay of ALP activity across different subcellular localizations.
Collapse
Affiliation(s)
- Han Zhang
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai, Shandong 264003, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Junsong Mou
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai, Shandong 264003, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Jiawang Ding
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai, Shandong 264003, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Wei Qin
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai, Shandong 264003, P.R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, P. R. China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, Shandong 266071, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
26
|
Wu C, Jiang P, Su W, Yan Y. Alkaline Phosphatase-Instructed Peptide Assemblies for Imaging and Therapeutic Applications. Biomacromolecules 2024; 25:5609-5629. [PMID: 39185628 DOI: 10.1021/acs.biomac.4c00795] [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: 08/27/2024]
Abstract
Self-assembly, a powerful strategy for constructing highly stable and well-ordered supramolecular structures, widely exists in nature and in living systems. Peptides are frequently used as building blocks in the self-assembly process due to their advantageous characteristics, such as ease of synthesis, tunable mechanical stability, good biosafety, and biodegradability. Among the initiators for peptide self-assembly, enzymes are excellent candidates for guiding this process under mild reaction conditions. As a crucial and commonly used biomarker, alkaline phosphatase (ALP) cleaves phosphate groups, triggering a hydrophilicity-to-hydrophobicity transformation that induces peptide self-assembly. In recent years, ALP-instructed peptide self-assembly has made breakthroughs in biological imaging and therapy, inspiring the development of self-assembly biomaterials for diagnosis and therapeutics. In this review, we highlight the most recent advancements in ALP-instructed peptide assemblies and provide perspectives on their potential impact. Finally, we briefly discuss the ongoing challenges for future research in this field.
Collapse
Affiliation(s)
- Chengfan Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Pingge Jiang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Wen Su
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| |
Collapse
|
27
|
Li Y, Hu L, Wang J, Wang H. Clustering of the Membrane Protein by Molecular Self-Assembly Downregulates the Signaling Pathway for Cancer Cell Inhibition. NANO LETTERS 2024; 24:10681-10690. [PMID: 39158180 DOI: 10.1021/acs.nanolett.4c03217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
This work reports a cyclic peptide appended self-assembled scaffold that recognizes the membrane protein EGFR and arrests the EGFR signaling through multivalent interactions by assembly-induced aggregation. When incubated with cells, the oligomers of PAD-1 first recognize the overexpressed EGFR on cancer cell membranes for arresting EGFR, which then initiates cellular uptake through endocytosis. The accumulation of PAD-1 and EGFR in the lysosome results in the formation of nanofibers, leading to the lysosomal membrane permeabilization (LMP). These processes disrupt the homeostasis of EGFR and inhibit the downstream signaling transduction of EGFR for cancer cell survival. Moreover, LMP induced the release of protein aggregates that could generate endoplasmic reticulum (ER) stress, resulting in cancer cell death selectively. In vivo studies indicate the efficient antitumor efficiency of PAD-1 in tumor-bearing mice. As a first example, this work provides an alternative strategy for controlling protein behavior for tuning cellular events in living cells.
Collapse
Affiliation(s)
- Ying Li
- Department of Chemistry, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
- Department of Chemistry, School of Science, Westlake University; Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| | - Liangbo Hu
- Department of Chemistry, School of Science, Westlake University; Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| | - Jing Wang
- Department of Chemistry, School of Science, Westlake University; Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Westlake University; Institute of Natural Sciences, Westlake Institute for Advanced Study, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| |
Collapse
|
28
|
Zhang X, Zhang B, Zhang Y, Ding Y, Zhang Z, Liu Q, Yang Z, Wang L, Gao J. Copper-Induced Supramolecular Peptide Assemblies for Multi-Pathway Cell Death and Tumor Inhibition. Angew Chem Int Ed Engl 2024; 63:e202406602. [PMID: 38837577 DOI: 10.1002/anie.202406602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024]
Abstract
Although self-assembly has emerged as an effective tool for fabricating biomaterials, achieving precise control over the morphologies and functionalities of the resultant assemblies remains an ongoing challenge. Inspired by the copper peptide naturally present in human plasma, in this study, we designed a synthetic precursor, FcGH. FcGH can self-assemble via two distinct pathways: spontaneous and Cu2+-induced. These two assembly pathways enabled the formation of assemblies with tunable morphologies by adjusting the amount of added Cu2+. We found that the nanoparticles formed by Cu2+-induced self-assembly exhibited a significantly higher cellular uptake efficiency than the wormlike fibers formed spontaneously. Moreover, this Cu2+-induced assembly process occurred spontaneously at a 1 : 1 molar ratio of Cu2+ to FcGH, avoiding the excessive use of Cu2+ and a tedious preparation procedure. By co-assembling with 10-hydroxycamptothecin (HCPT)-conjugated FcGH, Cu2+-induced supramolecular nanodrugs elicited multiple cell death modalities in cancer cells with elevated immunogenicity, enhancing the therapeutic effect compared to free HCPT. This study highlights Cu2+-induced self-assembly as an efficient tool for directing the assembly of nanodrugs and for synergistic tumor therapy.
Collapse
Affiliation(s)
- Xiangyang Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Buyue Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Ying Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Yinghao Ding
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Zhenghao Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University Xuzhou, Jiangsu, 221002, China
| | - Ling Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| |
Collapse
|
29
|
Pu G, Liang Z, Shi J, Tao Y, Lu P, Qing H, Zhang J. Enhancing the Inhibition of Corneal Neovascularization Efficacy by Self-Assembled into Supramolecular Hydrogel of Anti-Angiogenic Peptide. Int J Nanomedicine 2024; 19:7605-7616. [PMID: 39081898 PMCID: PMC11287374 DOI: 10.2147/ijn.s465965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Background Corneal neovascularization (CNV) is a common eye disease that leads to blindness. New treatment strategies are urgently needed due to the limitations of current treatment methods. Methods We report the synthesis of peptide Nap-FFEEPCAIWF ( Comp.3 ) via chemical conjugation of Nap-FFEE ( Comp.2 ) to antiangiogenic peptide PCAIWF (Comp.1). Comp.3 self-assembled into a hydrogel ( gel of 3 ) composed of nanofibers, which enhanced the antiangiogenic function of the epitope. Results We developed a novel peptide with an amphiphilic framework, Comp.3 , which could self-assemble into a supramolecular hydrogel with a well-ordered nanofiber structure. The nanofibers exhibited good biocompatibility with corneal epithelial cells, presenting a promising strategy to enhance the efficacy of free peptide-based drugs in the treatment of ocular vascular diseases, such as CNV and other angiogenesis-related diseases. Conclusion Nap-FFEEPCAIWF nanofibers provide an alternative approach to enhancing the therapeutic efficiency of free peptide-based drugs against ocular vascular diseases.
Collapse
Affiliation(s)
- Guojuan Pu
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Zhen Liang
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Jieran Shi
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Yuan Tao
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Ping Lu
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Huiling Qing
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Junjie Zhang
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| |
Collapse
|
30
|
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
|
31
|
Li S, Wang H. Lysosomal Peptide Self-Assembly to Control Cell Behavior. Chembiochem 2024; 25:e202400232. [PMID: 38660742 DOI: 10.1002/cbic.202400232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/14/2024] [Accepted: 04/23/2024] [Indexed: 04/26/2024]
Abstract
Lysosomes are membrane-enclosed organelles that play key roles in degrading and recycling cellular debris, cellular signaling, and energy metabolism processes. Confinement of amphiphilic peptides in the lysosome to construct functional nanostructures through noncovalent interactions is an emerging approach to tune the homeostasis of lysosome. After briefly introducing the importance of lysosome and its functions, we discuss the advantages of lysosomal nanostructure formation for disease therapy. We next discuss the strategy for triggering the self-assembly of peptides in the lysosome, followed by a concise outlook of the future perspective about this emerging research direction.
Collapse
Affiliation(s)
- Sangshuang Li
- Department of Chemistry, School of Science, Westlake University, No. 600 Yungu Road, 310030, Hangzhou, Zhejiang Province, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Westlake University, No. 600 Yungu Road, 310030, Hangzhou, Zhejiang Province, China
| |
Collapse
|
32
|
Hua Y, Shen Y. Applications of self-assembled peptide hydrogels in anti-tumor therapy. NANOSCALE ADVANCES 2024; 6:2993-3008. [PMID: 38868817 PMCID: PMC11166105 DOI: 10.1039/d4na00172a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/29/2024] [Indexed: 06/14/2024]
Abstract
Peptides are a class of active substances composed of a variety of amino acids with special physiological functions. The rational design of peptide sequences at the molecular level enables their folding into diverse secondary structures. This property has garnered significant attention in the biomedical sphere owing to their favorable biocompatibility, adaptable mechanical traits, and exceptional loading capabilities. Concurrently with advancements in modern medicine, the diagnosis and treatment of tumors have increasingly embraced targeted and personalized approaches. This review explores recent applications of self-assembled peptides derived from natural amino acids in chemical therapy, immunotherapy, and other adjunctive treatments. We highlighted the utilization of peptide hydrogels as delivery systems for chemotherapeutic drugs and other bioactive molecules and then discussed the challenges and prospects for their future application.
Collapse
Affiliation(s)
- Yue Hua
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University Nanjing Jiangsu 210009 China
| | - Yang Shen
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University Nanjing Jiangsu 210009 China
| |
Collapse
|
33
|
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
|
34
|
Liu H, Wang H. From cells to subcellular organelles: Next-generation cancer therapy based on peptide self-assembly. Adv Drug Deliv Rev 2024; 209:115327. [PMID: 38703895 DOI: 10.1016/j.addr.2024.115327] [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: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 05/06/2024]
Abstract
Due to the editability, functionality, and excellent biocompatibility of peptides, in situ self-assembly of peptides in cells is a powerful strategy for biomedical applications. Subcellular organelle targeting of peptides assemblies enables more precise drug delivery, enhances selectivity to disease cells, and mitigates drug resistance, providing an effective strategy for disease diagnosis and therapy. This reviewer first introduces the triggering conditions, morphological changes, and intracellular locations of self-assembling peptides. Then, the functions of peptide assemblies are summarized, followed by a comprehensive understanding of the interactions between peptide assemblies and subcellular organelles. Finally, we provide a brief outlook and the remaining challenges in this field.
Collapse
Affiliation(s)
- Huayang Liu
- Department of Chemistry, School of Science, Westlake University, No. 600 Dunyu Road, Sandun Town, Hangzhou 310024, Zhejiang Province, China; Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Westlake University, No. 600 Dunyu Road, Sandun Town, Hangzhou 310024, Zhejiang Province, China; Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| |
Collapse
|
35
|
Mo X, Zhang Z, Song J, Wang Y, Yu Z. Self-assembly of peptides in living cells for disease theranostics. J Mater Chem B 2024; 12:4289-4306. [PMID: 38595070 DOI: 10.1039/d4tb00365a] [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: 04/11/2024]
Abstract
The past few decades have witnessed substantial progress in biomedical materials for addressing health concerns and improving disease therapeutic and diagnostic efficacy. Conventional biomedical materials are typically created through an ex vivo approach and are usually utilized under physiological environments via transfer from preparative media. This transfer potentially gives rise to challenges for the efficient preservation of the bioactivity and implementation of theranostic goals on site. To overcome these issues, the in situ synthesis of biomedical materials on site has attracted great attention in the past few years. Peptides, which exhibit remarkable biocompability and reliable noncovalent interactions, can be tailored via tunable assembly to precisely create biomedical materials. In this review, we summarize the progress in the self-assembly of peptides in living cells for disease diagnosis and therapy. After a brief introduction to the basic design principles of peptide assembly systems in living cells, the applications of peptide assemblies for bioimaging and disease treatment are highlighted. The challenges in the field of peptide self-assembly in living cells and the prospects for novel peptide assembly systems towards next-generation biomaterials are also discussed, which will hopefully help elucidate the great potential of peptide assembly in living cells for future healthcare applications.
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.
| | - 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.
| | - 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.
| | - Yushi 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.
| | - 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
|
36
|
Gong Z, Zhao H, Bai J. pH-responsive drug-loaded peptides enhance drug accumulation and promote apoptosis in tumor cells. Colloids Surf B Biointerfaces 2024; 239:113954. [PMID: 38744076 DOI: 10.1016/j.colsurfb.2024.113954] [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: 03/25/2024] [Revised: 04/26/2024] [Accepted: 05/04/2024] [Indexed: 05/16/2024]
Abstract
The efficacy of chemotherapeutic drugs in tumor treatment is limited by their toxicity and side effects due to their inability to selectively accumulate in tumor tissue. In addition, chemotherapeutic agents are easily pumped out of tumor cells, resulting in their inadequate accumulation. To overcome these challenges, a drug delivery system utilizing the amphiphilic peptide Pep1 was designed. Pep1 can self-assemble into spherical nanoparticles (PL/Pep1) and encapsulate paclitaxel (PTX) and lapatinib (LAP). PL/Pep1 transformed into nanofibers in an acidic environment, resulting in longer drug retention and higher drug concentrations within tumor cells. Ultimately, PL/Pep1 inhibited tumor angiogenesis and enhanced tumor cell apoptosis. The use of shape-changing peptides as drug carriers to enhance cancer cell apoptosis is promising.
Collapse
Affiliation(s)
- Zhongying Gong
- College of Economics and Management, Qingdao University of Science and Technology, Qingdao, China
| | - Hongxia Zhao
- College of Economics and Management, Qingdao University of Science and Technology, Qingdao, China.
| | - Jingkun Bai
- School of Bioscience and Technology, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
37
|
Yi Y, An HW, Wang H. Intelligent Biomaterialomics: Molecular Design, Manufacturing, and Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305099. [PMID: 37490938 DOI: 10.1002/adma.202305099] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/14/2023] [Indexed: 07/27/2023]
Abstract
Materialomics integrates experiment, theory, and computation in a high-throughput manner, and has changed the paradigm for the research and development of new functional materials. Recently, with the rapid development of high-throughput characterization and machine-learning technologies, the establishment of biomaterialomics that tackles complex physiological behaviors has become accessible. Breakthroughs in the clinical translation of nanoparticle-based therapeutics and vaccines have been observed. Herein, recent advances in biomaterials, including polymers, lipid-like materials, and peptides/proteins, discovered through high-throughput screening or machine learning-assisted methods, are summarized. The molecular design of structure-diversified libraries; high-throughput characterization, screening, and preparation; and, their applications in drug delivery and clinical translation are discussed in detail. Furthermore, the prospects and main challenges in future biomaterialomics and high-throughput screening development are highlighted.
Collapse
Affiliation(s)
- Yu Yi
- 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, Beijing, 100190, China
| | - Hong-Wei An
- 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, Beijing, 100190, 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, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
38
|
Wang H, Jiao D, Feng D, Liu Q, Huang Y, Hou J, Ding D, Zhang W. Transformable Supramolecular Self-Assembled Peptides for Cascade Self-Enhanced Ferroptosis Primed Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311733. [PMID: 38339920 DOI: 10.1002/adma.202311733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/02/2024] [Indexed: 02/12/2024]
Abstract
Immunotherapy has received widespread attention for its effective and long-term tumor-eliminating ability. However, for immunogenic "cold" tumors, such as prostate cancer (PCa), the low immunogenicity of the tumor itself is a serious obstacle to efficacy. Here, this work reports a strategy to enhance PCa immunogenicity by triggering cascade self-enhanced ferroptosis in tumor cells, turning the tumor from "cold" to "hot". This work develops a transformable self-assembled peptide TEP-FFG-CRApY with alkaline phosphatase (ALP) responsiveness and glutathione peroxidase 4 (GPX4) protein targeting. TEP-FFG-CRApY self-assembles into nanoparticles under aqueous conditions and transforms into nanofibers in response to ALP during endosome/lysosome uptake into tumor cells, promoting lysosomal membrane permeabilization (LMP). On the one hand, the released TEP-FFG-CRAY nanofibers target GPX4 and selectively degrade the GPX4 protein under the light irradiation, inducing ferroptosis; on the other hand, the large amount of leaked Fe2+ further cascade to amplify the ferroptosis through the Fenton reaction. TEP-FFG-CRApY-induced immunogenic ferroptosis improves tumor cell immunogenicity by promoting the maturation of dendritic cells (DCs) and increasing intratumor T-cell infiltration. More importantly, recovered T cells further enhance ferroptosis by secreting large amounts of interferon-gamma (IFN-γ). This work provides a novel strategy for the molecular design of synergistic molecularly targeted therapy for immunogenic "cold" tumors.
Collapse
Affiliation(s)
- He Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Di Jiao
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Dexiang Feng
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Dan Ding
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education and College of Life Sciences, Nankai University, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Weijie Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| |
Collapse
|
39
|
Yin H, Hua Y, Feng S, Xu Y, Ding Y, Liu S, Chen D, Du F, Liang G, Zhan W, Shen Y. In Situ Nanofiber Formation Blocks AXL and GAS6 Binding to Suppress Ovarian Cancer Development. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308504. [PMID: 38546279 DOI: 10.1002/adma.202308504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/25/2024] [Indexed: 04/05/2024]
Abstract
Anexelekto (AXL) is an attractive molecular target for ovarian cancer therapy because of its important role in ovarian cancer initiation and progression. To date, several AXL inhibitors have entered clinical trials for the treatment of ovarian cancer. However, the disadvantages of low AXL affinity and severe off-target toxicity of these inhibitors limit their further clinical applications. Herein, by rational design of a nonapeptide derivative Nap-Phe-Phe-Glu-Ile-Arg-Leu-Arg-Phe-Lys (Nap-IR), a strategy of in situ nanofiber formation is proposed to suppress ovarian cancer growth. After administration, Nap-IR specifically targets overexpressed AXL on ovarian cancer cell membranes and undergoes a receptor-instructed nanoparticle-to-nanofiber transition. In vivo and in vitro experiments demonstrate that in situ formed Nap-IR nanofibers efficiently induce apoptosis of ovarian cancer cells by blocking AXL activation and disrupting subsequent downstream signaling events. Remarkably, Nap-IR can synergistically enhance the anticancer effect of cisplatin against HO8910 ovarian tumors. It is anticipated that the Nap-IR can be applied in clinical ovarian cancer therapy in the near future.
Collapse
Affiliation(s)
- Han Yin
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu, 210009, China
| | - Yue Hua
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu, 210009, China
| | - Songwei Feng
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu, 210009, China
| | - Yi Xu
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu, 210009, China
| | - Yue Ding
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu, 210009, China
| | - Sicong Liu
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu, 210009, China
| | - Dongsheng Chen
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., 699-18 Xuanwu Avenue, Nanjing, Jiangsu, 210042, China
| | - Furong Du
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., 699-18 Xuanwu Avenue, Nanjing, Jiangsu, 210042, 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
| | - Wenjun Zhan
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, Jiangsu, 210096, China
| | - Yang Shen
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu, 210009, China
| |
Collapse
|
40
|
Liu S, Zhang Q, Peng X, Hu C, Wang S, Sun Y. Intranuclear assembly of leucine-rich peptides for selective death of osteosarcoma cells. Biomater Sci 2024; 12:1274-1280. [PMID: 38251092 DOI: 10.1039/d3bm02054a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Herein, we show a pair of leucine-rich L- and D-phosphopeptides which self-assemble into twisting nanofibers, whose secondary structures contain a strong β-sheet component after being dephosphorylated by alkaline phosphatase (ALP). While being incubated with ALP overexpressing osteosarcoma cells, both of the peptides self-assemble in the nuclei and induce cell death. The cell death involves multiple cell death modalities and occurs along with the disruption of cell membranes. Enzyme-instructed self-assembly (EISA) inhibits osteosarcoma cells and shows no side effect to other cells. In addition, the cancer cells hardly gain drug resistance after repeated treatment. This work reports a pair of EISA-based nanofibers to target cell nuclei, and also provides a novel chemotherapeutic agent to inhibit osteosarcoma cells without side effects and drug resistance.
Collapse
Affiliation(s)
- Shuang Liu
- School of Materials Science and Engineering, Wuhan University of Technology, 122 Luoshi Road, Wuhan, Hubei, 430070, China.
- State Key Laboratory of Infrared Physics, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, 420 Zhongshan Road, Shanghai, 200434, China.
| | - Qiuxin Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| | - Xingrao Peng
- School of Materials Science and Engineering, Wuhan University of Technology, 122 Luoshi Road, Wuhan, Hubei, 430070, China.
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| | - Cong Hu
- Guangxi Key Laboratory of Automatic Detecting Technology and Instruments, Guilin University of Electronic Technology, Guilin 541004, China
| | - Shaowei Wang
- State Key Laboratory of Infrared Physics, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, 420 Zhongshan Road, Shanghai, 200434, China.
| | - Yao Sun
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, 430079, China.
| |
Collapse
|
41
|
Guo XY, Yi L, Yang J, An HW, Yang ZX, Wang H. Self-assembly of peptide nanomaterials at biointerfaces: molecular design and biomedical applications. Chem Commun (Camb) 2024; 60:2009-2021. [PMID: 38275083 DOI: 10.1039/d3cc05811e] [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: 01/27/2024]
Abstract
Self-assembly is an important strategy for constructing ordered structures and complex functions in nature. Based on this, people can imitate nature and artificially construct functional materials with novel structures through the supermolecular self-assembly pathway of biological interfaces. Among the many assembly units, peptide molecular self-assembly has received widespread attention in recent years. In this review, we introduce the interactions (hydrophobic interaction, hydrogen bond, and electrostatic interaction) between peptide nanomaterials and biological interfaces, summarizing the latest advancements in multifunctional self-assembling peptide materials. We systematically demonstrate the assembly mechanisms of peptides at biological interfaces, such as proteins and cell membranes, while highlighting their application potential and challenges in fields like drug delivery, antibacterial strategies, and cancer therapy.
Collapse
Affiliation(s)
- Xin-Yuan Guo
- College of Chemistry, Huazhong Agricultural University, Shizishan 1, Hongshan District, Wuhan, 430070, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China.
| | - Li Yi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China.
| | - Jia Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China.
| | - Hong-Wei An
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China.
| | - Zi-Xin Yang
- College of Chemistry, Huazhong Agricultural University, Shizishan 1, Hongshan District, Wuhan, 430070, China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China.
| |
Collapse
|
42
|
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
|
43
|
Mondal T, Chatterjee A, Hansda B, Mondal B, Sen P, Banerjee A. Cationic and amphiphilic peptide-based hydrogels with dual activities as anticancer and antibacterial agents. SOFT MATTER 2024; 20:1236-1244. [PMID: 38230549 DOI: 10.1039/d3sm01291c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
The emergence of peptide-based functional biomaterials is on the rise. To fulfil this purpose, a series of amphiphilic peptides, such as H2N-X-Met-Phe-C12H25, where X = L-lysine (CP1), X = L-histidine (CP2), and X = L-leucine (CP3), have been designed, synthesised, purified and fully characterised. Herein, we reported peptide-based supramolecular hydrogels with antibacterial and anticancer activities. An attempt has been made to investigate the antibacterial properties of these peptide-based hydrogels against Gram-positive (S. aureus and B. subtilis) and Gram-negative (E. coli and P. aeruginosa) bacteria. Investigations show that the L-lysine containing gelator, CP1, is active against both Gram-positive and Gram-negative bacteria and the L-histidine containing gelator, CP2, selectively inhibits the growth of Gram-negative bacteria. Interestingly, the L-leucine containing gelator, CP3, does not show any antibacterial properties. Moreover, the L-lysine containing gelator exhibits the best potency. Generation of reactive oxygen species (ROS) is a probable way to damage the bacterial membrane. To explore the cytotoxic properties and to determine the efficacy of the synthesized compounds in inhibiting cell viability, a comprehensive investigation was performed using three distinct cell lines: MDA-MB-231 (human triple-negative breast cancer), MDA-MB-468 (human triple-negative breast cancer) and HEK 293 (human embryonic kidney). Remarkably, the results of our study revealed a substantial cytotoxic impact of these peptide gelators on the MDA-MB-231 and MDA-MB-468 cell lines in comparison to the HEK 293 cells. Caspase 3/7 activity is the possible mechanistic path to determine the apoptotic rates of the cell lines. This finding emphasizes the promising potential of these peptide-based gelators in targeting and suppressing the growth of human triple negative breast cancer cells, while showing non-cytotoxicity towards non-cancerous HEK 293 cells. In a nutshell, these peptide-based materials are coming to light as next generation biomaterials.
Collapse
Affiliation(s)
- Tanushree Mondal
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Akash Chatterjee
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Biswanath Hansda
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Biplab Mondal
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Prosenjit Sen
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| | - Arindam Banerjee
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata-700032, India.
| |
Collapse
|
44
|
Sletten ET, Fittolani G, Hribernik N, Dal Colle MCS, Seeberger PH, Delbianco M. Phosphates as Assisting Groups in Glycan Synthesis. ACS CENTRAL SCIENCE 2024; 10:138-142. [PMID: 38292611 PMCID: PMC10823511 DOI: 10.1021/acscentsci.3c00896] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 02/01/2024]
Abstract
In nature, phosphates are added to and cleaved from molecules to direct biological pathways. The concept was adapted to overcome limitations in the chemical synthesis of complex oligosaccharides. Phosphates were chemically placed on synthetic glycans to ensure site-specific enzymatic elongation by sialylation. In addition, the deliberate placement of phosphates helped to solubilize and isolate aggregating glycans. Upon traceless removal of the phosphates by enzymatic treatment with alkaline phosphatase, the native glycan structure was revealed, and the assembly of glycan nanostructures was triggered.
Collapse
Affiliation(s)
- Eric T. Sletten
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Giulio Fittolani
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Nives Hribernik
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Marlene C. S. Dal Colle
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
- Department
of Chemistry and Biochemistry, Freie Universität
Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Peter H. Seeberger
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
- Department
of Chemistry and Biochemistry, Freie Universität
Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Martina Delbianco
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
| |
Collapse
|
45
|
Meng F, Zhai X, Ma J, Li A, Wang X, Bai J. Enzyme-Induced Shape-Shifting Peptide Nanocarrier Coloaded with Paclitaxel and Dipyridamole Inhibits Platelet Function and Tumor Metastasis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:166-177. [PMID: 38143309 DOI: 10.1021/acsami.3c13855] [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: 12/26/2023]
Abstract
Tumor-associated platelets can bind to tumor cells and protect circulating tumor cells from NK-mediated immune surveillance. Tumor-associated platelets secrete cytokines to induce the epithelial-mesenchymal transition (EMT) in tumor cells, which promotes tumor metastasis. Combining chemotherapeutic agents with antiplatelet drugs can reduce the occurrence of metastasis, but the systemic application of chemotherapeutic agents and antiplatelet drugs is prone to causing serious side effects. Therefore, delivering drugs to the tumor microthrombus site for long-lasting inhibition is a problem that needs to be addressed. Here, we show that small molecule peptide nanoparticles containing the Cys-Arg-Glu-Lys-Ala (CREKA) peptide can deliver the platelet inhibitor dipyridamole (DIP) and the chemotherapeutic drug paclitaxel (PTX) to tumor tissues, thereby inhibiting tumor-associated platelet function while killing tumor cells. The drug-loaded nanoparticles PD/Pep1 inhibited platelet-tumor cell interactions, were effectively taken up by tumor cells, and underwent morphological transformation induced by alkaline phosphatase (ALP) to prolong the retention time of the drugs. After intravenous injection, PD/Pep1 can target tumors and inhibit tumor metastasis. Thus, this small molecule peptide nanoformulation provides a simple strategy for efficient drug delivery and shows promise as a novel cancer therapy platform.
Collapse
Affiliation(s)
- Fanhu Meng
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Xiaoqing Zhai
- School of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Jihong Ma
- School of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Aimei Li
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| | - Xizhen Wang
- Medical Imaging Center, Affiliated Hospital of Weifang Medical University, Weifang 261053, China
| | - Jingkun Bai
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
46
|
Wang Y, Xie L, Li X, Wang L, Yang Z. Chemo-immunotherapy by dual-enzyme responsive peptide self-assembling abolish melanoma. Bioact Mater 2024; 31:549-562. [PMID: 37746663 PMCID: PMC10511343 DOI: 10.1016/j.bioactmat.2023.09.006] [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: 06/25/2023] [Revised: 09/10/2023] [Accepted: 09/10/2023] [Indexed: 09/26/2023] Open
Abstract
Herein, we designed Comp. 1 to simultaneously respond to two enzymes: alkaline phosphatase and matrix metalloproteinase 2, which is commonly found in highly malignant cancer cell lines containing B16-F10 murine melanoma cells and CT26 murine colon carcinoma cells. We used the regional differences in the expression levels of dual-markers to accurately release immune molecule IND into tumor microenvironment for the activation of anti-tumor related immune effects, while in-situ self-assembly occurs. The dual-enzyme response process can further regulate the peptide precursors' self-assembly in the form of short rod-shaped nanofibers, enabling the delivery of the loaded chemotherapeutic drug HCPT into the cancer cells and further allowing the peptide assemblies to escape from lysosomes and return to cytoplasm in the form of tiny nanoparticles to induce apoptosis of cancer cells. This process does not occur in the single-positive breast cancer cell line MCF-7 or the normal hepatocytes cell line LO2, indicating the selectivity of the cancer cells exhibited using our strategy. In vivo studies revealed that Comp. 1 can effectively cooperate with chemotherapy to enhance the immunotherapy effect and induce immune responses associated with elevated pro-inflammatory cytokines in vivo to inhibit malignant tumors growth. Our dual-enzyme responsive chemo-immunotherapy strategy feasible in anti-tumor treatment, provides a new avenue for regulating peptide self-assembly to adapt to diverse tumor properties and may eventually be used for the development of novel multifunctional anti-tumor nanomedicines.
Collapse
Affiliation(s)
- Yuhan Wang
- Tianjin Key Laboratory of Inflammation Biology, Department of Pharmacology, School of Basic Medicine, Tianjin Medical University, Tianjin, 300070, PR China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| | - Limin Xie
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| | - Xinxin Li
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| | - Ling Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300071, PR China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| |
Collapse
|
47
|
Zhou J, Cai Y, Li T, Zhou H, Dong H, Wu X, Li Z, Wang W, Yuan D, Li Y, Shi J. Aflibercept Loaded Eye-Drop Hydrogel Mediated with Cell-Penetrating Peptide for Corneal Neovascularization Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302765. [PMID: 37679056 DOI: 10.1002/smll.202302765] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Corneal neovascularization (CoNV) is a major cause of visual impairment worldwide. Currently, available treatment options have limited efficacy and are associated with adverse effects due to biological barriers and clearance mechanisms. To address this challenge, a novel topical delivery system is developed-Gel 2_1&Eylea-an aflibercept-loaded eye-drop hydrogel mediated with cell-penetrating peptide 1. Gel 2_1&Eylea demonstrates superior membrane permeability, increased stability, and prolonged drug retention time on the ocular surface, and thus may improve drug efficacy. In a rabbit CoNV model, Gel 2_1&Eylea significantly reduces the density of neovascularization with no adverse effects on normal corneoscleral limbal vessels, demonstrating high efficacy and biocompatibility. This work identifies a promising treatment for CoNV which has the potential to benefit other ocular neovascular diseases.
Collapse
Affiliation(s)
- Jianan Zhou
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Yuting Cai
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Tingting Li
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Haixiang Zhou
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Huilei Dong
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Xia Wu
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
- Shenzhen International Institute for Biomedical Research, Longhua District, Shenzhen, Guangdong, 518116, China
| | - Zenghui Li
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Wenjie Wang
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Dan Yuan
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Yun Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Junfeng Shi
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| |
Collapse
|
48
|
Wang H, Mills J, Sun B, Cui H. Therapeutic Supramolecular Polymers: Designs and Applications. Prog Polym Sci 2024; 148:101769. [PMID: 38188703 PMCID: PMC10769153 DOI: 10.1016/j.progpolymsci.2023.101769] [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] [Indexed: 01/09/2024]
Abstract
The self-assembly of low-molecular-weight building motifs into supramolecular polymers has unlocked a new realm of materials with distinct properties and tremendous potential for advancing medical practices. Leveraging the reversible and dynamic nature of non-covalent interactions, these supramolecular polymers exhibit inherent responsiveness to their microenvironment, physiological cues, and biomolecular signals, making them uniquely suited for diverse biomedical applications. In this review, we intend to explore the principles of design, synthesis methodologies, and strategic developments that underlie the creation of supramolecular polymers as carriers for therapeutics, contributing to the treatment and prevention of a spectrum of human diseases. We delve into the principles underlying monomer design, emphasizing the pivotal role of non-covalent interactions, directionality, and reversibility. Moreover, we explore the intricate balance between thermodynamics and kinetics in supramolecular polymerization, illuminating strategies for achieving controlled sizes and distributions. Categorically, we examine their exciting biomedical applications: individual polymers as discrete carriers for therapeutics, delving into their interactions with cells, and in vivo dynamics; and supramolecular polymeric hydrogels as injectable depots, with a focus on their roles in cancer immunotherapy, sustained drug release, and regenerative medicine. As the field continues to burgeon, harnessing the unique attributes of therapeutic supramolecular polymers holds the promise of transformative impacts across the biomedical landscape.
Collapse
Affiliation(s)
- Han Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jason Mills
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Boran Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Nanomedicine, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
49
|
Zhang X, Wang J, Zhang Y, Yang Z, Gao J, Gu Z. Synthesizing biomaterials in living organisms. Chem Soc Rev 2023; 52:8126-8164. [PMID: 37921625 DOI: 10.1039/d2cs00999d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Living organisms fabricate biomacromolecules such as DNA, RNA, and proteins by the self-assembly process. The research on the mechanism of biomacromolecule formation also inspires the exploration of in vivo synthesized biomaterials. By elaborate design, artificial building blocks or precursors can self-assemble or polymerize into functional biomaterials within living organisms. In recent decades, these so-called in vivo synthesized biomaterials have achieved extensive applications in cell-fate manipulation, disease theranostics, bioanalysis, cellular surface engineering, and tissue regeneration. In this review, we classify strategies for in vivo synthesis into non-covalent, covalent, and genetic types. The development of these approaches is based on the chemical principles of supramolecular chemistry and synthetic chemistry, biological cues such as enzymes and microenvironments, and the means of synthetic biology. By summarizing the design principles in detail, some insights into the challenges and opportunities in this field are provided to enlighten further research.
Collapse
Affiliation(s)
- Xiangyang Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Junxia Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
50
|
Guo L, Yang J, Wang H, Yi Y. Multistage Self-Assembled Nanomaterials for Cancer Immunotherapy. Molecules 2023; 28:7750. [PMID: 38067480 PMCID: PMC10707962 DOI: 10.3390/molecules28237750] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Advances in nanotechnology have brought innovations to cancer therapy. Nanoparticle-based anticancer drugs have achieved great success from bench to bedside. However, insufficient therapy efficacy due to various physiological barriers in the body remains a key challenge. To overcome these biological barriers and improve the therapeutic efficacy of cancers, multistage self-assembled nanomaterials with advantages of stimuli-responsiveness, programmable delivery, and immune modulations provide great opportunities. In this review, we describe the typical biological barriers for nanomedicines, discuss the recent achievements of multistage self-assembled nanomaterials for stimuli-responsive drug delivery, highlighting the programmable delivery nanomaterials, in situ transformable self-assembled nanomaterials, and immune-reprogramming nanomaterials. Ultimately, we perspective the future opportunities and challenges of multistage self-assembled nanomaterials for cancer immunotherapy.
Collapse
Affiliation(s)
- Lamei Guo
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Jinjun Yang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Yu Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| |
Collapse
|