1
|
Li S, Liu H, Fang Y, Li Y, Zhou L, Chen D, Liang J, Wang H. Programming two-component peptide self-assembly by tuning the hydrophobic linker. Faraday Discuss 2025. [PMID: 40366063 DOI: 10.1039/d4fd00209a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Molecular self-assembly enables the formation of intricate networks through non-covalent interactions, serving as a key strategy for constructing structures ranging from molecules to macroscopic forms. While zero-dimensional and one-dimensional nanostructures have been widely achieved, two-dimensional nanostrip structures present unique advantages in biomedical and other applications due to their high surface area and potential for functionalization. However, their efficient design and precise regulation remain challenging. This study systematically explores how different hydrophobic amino acid linkers impact the microscopic morphology in two-component co-assembly systems with strong electrostatic interactions. The introduction of the AA linker resulted in distinctive 2D nanostrips, which stacked to form bilayer sheets, whereas VV, LL, and NleNle linkers formed one-dimensional fibers. In contrast, GG and PP linkers did not produce stable aggregates. Our findings highlight the role of intermolecular interactions in the development of 2D assemblies, providing new insights into the design and application of 2D materials.
Collapse
Affiliation(s)
- Sangshuang Li
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Huayang Liu
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Yu Fang
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Yaoting Li
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Laicheng Zhou
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Dinghao Chen
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Juan Liang
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| | - Huaimin Wang
- Department of Chemistry, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, 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
|
Sharma D, Dhiman I, Das S, Das DK, Pramanik DD, Dash SK, Pramanik A. Recent Advances in Therapeutic Peptides: Innovations and Applications in Treating Infections and Diseases. ACS OMEGA 2025; 10:17087-17107. [PMID: 40352490 PMCID: PMC12059905 DOI: 10.1021/acsomega.5c02077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025]
Abstract
Peptides have become a powerful frontier in modern medicine, offering a promising therapeutic solution for various diseases and advancing rapidly in pharmaceutical development. These small amino acid chains, with their innovative design, have attracted significant attention due to their versatility and high receptor specificity, which minimizes off-target effects, along with enhanced therapeutic efficacy, biodegradability, low toxicity, and minimal immunogenicity. They are being explored for use in several clinical domains, like metabolic diseases, immunomodulation, and cancer. Furthermore, antimicrobial peptides (AMPs) have grown to be a promising strategy to combat the worldwide challenge of antibiotic resistance, demonstrating promising results against multidrug-resistant organisms. Both natural and engineered peptides have been discovered and investigated, whereas numerous others are progressing toward clinical trials in a number of therapeutic domains. Recent improvements with surface modification, such as peptide engineering, peptide cyclization, PEGylation, and the utilization of synthetic amino acids to enhance their pharmacokinetic profiles and overcome the inherent disadvantages of these peptides have made it possible for the area to continue to advance. Moreover, their therapeutic potential has been further enhanced by innovative delivery methods, such as self-assembling peptides, nanocarriers, and alternate routes of administration. This Review critically states the potential of peptides as versatile therapeutics along with their modifications and advancements to drive the significant progress to treat infections and chronic diseases, along with their potential benefits and challenges.
Collapse
Affiliation(s)
- Deepshikha Sharma
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
| | - Isha Dhiman
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
| | - Swarnali Das
- Department
of Physiology, University of Gour Banga, Malda, West Bengal 732103, India
| | - Deepak Kumar Das
- Department
of Chemistry and Nanoscience, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Devlina Das Pramanik
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
| | - Sandeep Kumar Dash
- Department
of Physiology, University of Gour Banga, Malda, West Bengal 732103, India
| | - Arindam Pramanik
- Amity
Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201301, India
- School
of Medicine, University of Leeds, Leeds LS97TF, United Kingdom
| |
Collapse
|
4
|
Wang H, Zheng YT, Zhang J, Gao Y, Chen J, Cai P, Wang J, van Esch JH, Guo X, Li H, Wang Y. Synthesis of Abiotic Supramolecular Polymers Inside Living Cells via Organocatalysis-Mediated Self-Assembly. Angew Chem Int Ed Engl 2025; 64:e202500998. [PMID: 40059797 DOI: 10.1002/anie.202500998] [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: 01/13/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/18/2025]
Abstract
Cells execute mesmerizing functions using supramolecular polymers (SPs) formed through the self-assembly of biological precursors. Integration of the vast array of synthetic SPs with living cells would offer a powerful way to remold cellular functions and bridge the gap between synthetic materials and the biological realm, yet remains a challenge because of the lack of robust abiotic SP systems that can be triggered to self-assemble inside cells. Here, we report how fully abiotic SPs can be synthesized inside living cells via an organocatalysis-responsive self-assembly strategy, and how the in situ-generated SPs are capable of interfering and can interfere with cellular functions. The incorporation of a nucleophilic organocatalyst (CAT) into living cells accelerates the intracellular conversion of hydrazide (H) and aldehyde-derived precursors (A) to hydrazone-based monomers (HA3) that locally self-assemble into SPs. Interestingly, the in situ-generated SPs possess ignorable effects on cell viability and proliferation but remarkably hinder cell migration. Furthermore, the presence of SPs is found to retard intracellular diffusion and alter the organization of the actin cytoskeleton, both of which are suggested to be responsible for the hindered cellular migration. In considering the vastly wide range of synthetic SPs, tremendous non-natural cellular functionalities can be obtained by in situ-synthesizing SPs.
Collapse
Affiliation(s)
- Hucheng Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Ya-Ting Zheng
- School of Systems Science and Institute of Nonequilibrium Systems, Beijing Normal University, Beijing, 100875, P.R. China
| | - Jiahao Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Yuliang Gao
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Jingjing Chen
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Peiwen Cai
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Junyou Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Jan H van Esch
- Department of Chemical Engineering, Delft University of Technology, Delft, 2629 HZ, The Netherlands
| | - Xuhong Guo
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Hui Li
- School of Systems Science and Institute of Nonequilibrium Systems, Beijing Normal University, Beijing, 100875, P.R. China
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing, 100875, P.R. China
| | - Yiming Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, P.R. China
- Shanghai Key Laboratory for Intelligent Sensing and Detection Technology, East China University of Science and Technology, Shanghai, 200237, P.R. China
| |
Collapse
|
5
|
Qiao Y, Zia A, Shy A, Wu G, Chu M, Liu Z, Wang F, Xu B. Intrinsically Disordered Peptide Nanofibers from a Structured Motif Within Proteins. Angew Chem Int Ed Engl 2025:e202425456. [PMID: 40294067 DOI: 10.1002/anie.202425456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/12/2025] [Accepted: 04/28/2025] [Indexed: 04/30/2025]
Abstract
Intrinsically disordered regions (IDRs) are ubiquitous in proteins, orchestrating complex cellular signaling through higher-order protein assemblies. However, the properties and functions of intrinsically disordered peptide (IDP) assemblies are largely underexplored. This work unveiled a facile strategy for engineering IDP assemblies. We demonstrate that conjugating a structured motif derived from a protein's phosphorylation site to a self-assembling tripeptide unexpectedly yields self-assembled nanofibers with intrinsic disorder. Specifically, by using a glycine linker to attach a pentapeptide derived from a phosphorylation site within a random coil region of SRC kinase to the C-terminus of a widely used self-assembling enabler, we generated a phosphorylated octapeptide. The octapeptide exhibits cell compatibility and forms a hydrogel upon dephosphorylation of the phosphooctapeptide. Cryo-electron microscopy (cryo-EM) structural analysis of the nanofibers reveals that the peptides adopt two types of helical arrangements but exhibit intrinsic disorder at the periphery of the nanofibers. The hydrogels exhibit decreased protein adsorption with increasing peptide concentration. This study represents the first instance of a structured random coil within a protein transitioning into an intrinsically disordered state within self-assembled peptide nanofibers, expanding the pool of peptide sequences for IDPs and providing valuable insights for the engineering of peptide nanofibers with intrinsic disorder for the development of cell-compatible biomaterials.
Collapse
Affiliation(s)
- Yuchen Qiao
- Department of Chemistry, Brandeis University, 415 South St., Waltham, Massachusetts, 02454, USA
| | - Ayisha Zia
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, 35233, USA
| | - Adrianna Shy
- Department of Chemistry, Brandeis University, 415 South St., Waltham, Massachusetts, 02454, USA
| | - Grace Wu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, Massachusetts, 02454, USA
| | - Matthew Chu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, Massachusetts, 02454, USA
| | - Zhiyu Liu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, Massachusetts, 02454, USA
| | - Fengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, 35233, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, Massachusetts, 02454, USA
| |
Collapse
|
6
|
Shimizu N, Kanemitsu S, Umemura R, Yashiro T, Kawabata R, Nishimura K, Kawasaki S, Morita K, Aoi T, Maruyama T. Mechanistic Insights into the Apoptosis of Cancer Cells Induced by a Kinase-Responsive Peptide Amphiphile. Chemistry 2025; 31:e202403658. [PMID: 39876747 DOI: 10.1002/chem.202403658] [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: 10/02/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 01/30/2025]
Abstract
Organelle targeting is a useful approach in drug development for cancer therapy. Peptide amphiphiles are good candidates for targeting specific organelles because they can be engineered into a wide range of molecular structures, enabling customization for specific functional needs. We have developed a peptide amphiphile, C16-(EY)3, that can respond to tyrosine kinase activity and undergo phosphorylation inside cancer cells. C16-(EY)3 selectively induced apoptosis in cancer cells that overexpressed tyrosine kinase. The self-assembly of peptide amphiphiles on the endoplasmic reticulum (ER) membrane reduced the ER membrane fluidity and triggered ER stress. The mechanism of the cancer cell death induced by C16-(EY)3 was shown to involve phosphorylation by tyrosine kinase, ER stress induction, and the subsequent activation of caspase-4, -12, and -9, which ultimately triggered apoptosis through the activation of caspase-3 and -7. In vivo studies further validated the antitumor efficacy of C16-(EY)3, as transcutaneous administration of the peptide amphiphile inhibited tumor growth in mice. This study elucidated the mechanism of apoptosis induced by the peptide amphiphile, indicating the potential of peptide amphiphiles as organelle-targeting cancer therapeutics and providing a novel strategy for the development of selective and potent anticancer drugs.
Collapse
Affiliation(s)
- Natsumi Shimizu
- Department of Chemical Science and Engineering, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| | - Sayuki Kanemitsu
- Department of Chemical Science and Engineering, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| | - Riku Umemura
- Department of Chemical Science and Engineering, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| | - Tomoko Yashiro
- Department of Chemical Science and Engineering, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| | - Ryoko Kawabata
- Department of Chemical Science and Engineering, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| | - Kanon Nishimura
- Department of Chemical Science and Engineering, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| | - Shinya Kawasaki
- Department of Chemical Science and Engineering, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| | - Kenta Morita
- Department of Chemical Science and Engineering, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
- Research Center for Membrane and Film Technology, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| | - Takashi Aoi
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, 7-5-2 Kusunokicho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tatsuo Maruyama
- Department of Chemical Science and Engineering, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
- Research Center for Membrane and Film Technology, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| |
Collapse
|
7
|
Wang Z, Liang H, Liu A, Li X, Guan L, Li L, He L, Whittaker AK, Yang B, Lin Q. Strength through unity: Alkaline phosphatase-responsive AIEgen nanoprobe for aggregation-enhanced multi-mode imaging and photothermal therapy of metastatic prostate cancer. CHINESE CHEM LETT 2025; 36:109765. [DOI: 10.1016/j.cclet.2024.109765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
8
|
Bharathidasan D, Maity C. Organelle-Specific Smart Supramolecular Materials for Bioimaging and Theranostics Application. Top Curr Chem (Cham) 2024; 383:1. [PMID: 39607460 DOI: 10.1007/s41061-024-00483-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
In cellular environments, certain synthetic molecules can form nanostructures via self-assembly, impacting molecular imaging, and biomedical applications. Control over the formation of these self-assembled nanostructures in subcellular organelle is challenging. By the action of stimuli, either present in the cellular environment or applied externally, in situ generation of molecular precursors can lead to accumulation and supramolecular nanostructure formation, resulting in efficient bioimaging. Here, we summarize smart fluorophore-based ordered nanostructure preparation at specific organelles for efficient bioimaging and therapeutic application towards cancer theranostics. We also present challenges and an outlook regarding intercellular self-assembly for theranostics application. Altogether, smart nanostructured materials with fluorescence read-outs at specific subcellular compartments would be beneficial in synthetic biology and precision therapeutics.
Collapse
Affiliation(s)
- Dineshkumar Bharathidasan
- (Organic)Material Science and Engineering Laboratory, Centre for Nanobiotechnology (CNBT), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamilnadu, 632014, India
| | - Chandan Maity
- (Organic)Material Science and Engineering Laboratory, Centre for Nanobiotechnology (CNBT), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamilnadu, 632014, India.
| |
Collapse
|
9
|
Guo J, Tan W, Xu B. Enzymatic self-assembly of short peptides for cell spheroid formation. J Mater Chem B 2024; 12:11210-11217. [PMID: 39370899 PMCID: PMC11540748 DOI: 10.1039/d4tb01154f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Cell spheroids, including organoids, serve as a valuable link between in vitro systems and in vivo animal models, offering powerful tools for studying cell biology in a three-dimensional environment. However, existing methods for generating cell spheroids are time consuming or difficult to scale up for large-scale production. Our recent study has revealed that transcytotic peptide assemblies, which transform from nanoparticles to nanofibers by enzymatic reactions, can create an intercellular fibril/gel, accelerating cell spheroid formation from a 2D cell culture or a cell suspension. While this finding presents an alternative approach for generating cell spheroids, the specific structural features required for efficient cell spheroid formation remain unclear. Based on our observation that a phosphotetrapeptide with a biphenyl cap at its N-terminus enables cell spheroid formation, we produced 10 variants of the original peptide. The variants explored modifications to the peptide backbone, length, electronic properties of the biphenyl capping group, and the type of phosphorylated amino acid residue. We then evaluated their ability for inducing cell spheroid formation. Our analysis revealed that, among the tested molecules, peptides with C-terminal phosphotyrosine, low critical micelle concentration, and dephosphorylation-guided nanoparticle to nanofiber morphological transition were the most effective in inducing the formation of cell spheroids. This work represents the first example to correlate the thermodynamic properties (e.g., self-assembling ability) and kinetic behavior (e.g., enzymatic dephosphorylation) of peptides with the efficacy of controlling intercellular interaction, thus offering valuable insights into using enzymatic self-assembly to generate peptide assemblies for biological applications.
Collapse
Affiliation(s)
- Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA.
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA.
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA.
| |
Collapse
|
10
|
Guo J, Zia A, Qiu Q, Norton M, Qiu K, Usuba J, Liu Z, Yi M, Rich-New ST, Hagan M, Fraden S, Han GD, Diao J, Wang F, Xu B. Cell-Free Nonequilibrium Assembly for Hierarchical Protein/Peptide Nanopillars. J Am Chem Soc 2024; 146:26102-26112. [PMID: 39255453 PMCID: PMC11669155 DOI: 10.1021/jacs.4c06775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Cells contain intricate protein nanostructures, but replicating them outside of cells presents challenges. One such example is the vertical fibronectin pillars observed in embryos. Here, we demonstrate the creation of cell-free vertical fibronectin pillar mimics using nonequilibrium self-assembly. Our approach utilizes enzyme-responsive phosphopeptides that assemble into nanotubes. Enzyme action triggers shape changes in peptide assemblies, driving the vertical growth of protein nanopillars into bundles. These bundles, with peptide nanotubes serving as a template to remodel fibronectin, can then recruit collagen, which forms aggregates or bundles depending on their types. Nanopillar formation relies on enzyme-catalyzed nonequilibrium self-assembly and is governed by the concentrations of enzyme, protein, peptide, the structure of the peptide, and peptide assembly morphologies. Cryo-EM reveals unexpected nanotube thinning and packing after dephosphorylation, indicating a complex sculpting process during assembly. Our study demonstrates a cell-free method for constructing intricate, multiprotein nanostructures with directionality and composition.
Collapse
Affiliation(s)
- Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Ayisha Zia
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Qianfeng Qiu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Michael Norton
- Department of Physics, Brandeis University, Waltham, MA, 02453, USA
| | - Kangqiang Qiu
- Department of Cancer Biology, Center for Chemical Imaging in Biomedicine, Advanced Cell Analysis Service Center, University of Cincinnati College of Medicine, Cincinnati OH, 45267, USA
| | - Junichi Usuba
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Zhiyu Liu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Meihui Yi
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Shane T. Rich-New
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Michael Hagan
- Department of Physics, Brandeis University, Waltham, MA, 02453, USA
| | - Seth Fraden
- Department of Physics, Brandeis University, Waltham, MA, 02453, USA
| | - Grace D. Han
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| | - Jiajie Diao
- Department of Cancer Biology, Center for Chemical Imaging in Biomedicine, Advanced Cell Analysis Service Center, University of Cincinnati College of Medicine, Cincinnati OH, 45267, USA
| | - Fengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- O’Neal Comprehensive Cancer Center University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA, 02453, USA
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
Coulter S, Pentlavalli S, An Y, Vora LK, Cross ER, Moore JV, Sun H, Schweins R, McCarthy HO, Laverty G. In Situ Forming, Enzyme-Responsive Peptoid-Peptide Hydrogels: An Advanced Long-Acting Injectable Drug Delivery System. J Am Chem Soc 2024; 146:21401-21416. [PMID: 38922296 PMCID: PMC11311241 DOI: 10.1021/jacs.4c03751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Long-acting drug delivery systems are promising platforms to improve patient adherence to medication by delivering drugs over sustained periods and removing the need for patients to comply with oral regimens. This research paper provides a proof-of-concept for the development of a new optimized in situ forming injectable depot based on a tetrabenzylamine-tetraglycine-d-lysine-O-phospho-d-tyrosine peptoid-D-peptide formulation ((NPhe)4GGGGk(AZT)y(p)-OH). The chemical versatility of the peptoid-peptide motif allows low-molecular-weight drugs to be precisely and covalently conjugated. After subcutaneous injection, a hydrogel depot forms from the solubilized peptoid-peptide-drug formulation in response to phosphatase enzymes present within the skin space. This system is able to deliver clinically relevant concentrations of a model drug, the antiretroviral zidovudine (AZT), for 35 days in Sprague-Dawley rats. Oscillatory rheology demonstrated that hydrogel formation began within ∼30 s, an important characteristic of in situ systems for reducing initial drug bursts. Gel formation continued for up to ∼90 min. Small-angle neutron scattering data reveal narrow-radius fibers (∼0.78-1.8 nm) that closely fit formation via a flexible cylinder elliptical model. The inclusion of non-native peptoid monomers and D-variant amino acids confers protease resistance, enabling enhanced biostability to be demonstrated in vitro. Drug release proceeds via hydrolysis of an ester linkage under physiological conditions, releasing the drug in an unmodified form and further reducing the initial drug burst. Subcutaneous administration of (NPhe)4GGGGk(AZT)y(p)-OH to Sprague-Dawley rats resulted in zidovudine blood plasma concentrations within the 90% maximal inhibitory concentration (IC90) range (30-130 ng mL-1) for 35 days.
Collapse
Affiliation(s)
- Sophie
M. Coulter
- Biofunctional
Nanomaterials Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, Co. Antrim BT9 7BL, N. Ireland
| | - Sreekanth Pentlavalli
- Biofunctional
Nanomaterials Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, Co. Antrim BT9 7BL, N. Ireland
| | - Yuming An
- Biofunctional
Nanomaterials Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, Co. Antrim BT9 7BL, N. Ireland
| | - Lalitkumar K. Vora
- Biofunctional
Nanomaterials Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, Co. Antrim BT9 7BL, N. Ireland
| | - Emily R. Cross
- Biofunctional
Nanomaterials Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, Co. Antrim BT9 7BL, N. Ireland
| | - Jessica V. Moore
- Biofunctional
Nanomaterials Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, Co. Antrim BT9 7BL, N. Ireland
| | - Han Sun
- Biofunctional
Nanomaterials Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, Co. Antrim BT9 7BL, N. Ireland
| | - Ralf Schweins
- Large
Scale Structures Group, Institut Laue −
Langevin, 71 Avenue des Martyrs, CS 20156, Grenoble
Cedex 9, 38042, France
| | - Helen O. McCarthy
- Biofunctional
Nanomaterials Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, Co. Antrim BT9 7BL, N. Ireland
| | - Garry Laverty
- Biofunctional
Nanomaterials Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, Co. Antrim BT9 7BL, N. Ireland
| |
Collapse
|
15
|
Carney E, Ghasem Zadeh Moslabeh F, Kang SY, Bunnell BA, Lee MY, Habibi N. Self-assembling peptides induced by eyes absent enzyme to boost the efficacy of doxorubicin therapy in drug-resistant breast cancer cells. Heliyon 2024; 10:e33629. [PMID: 39071664 PMCID: PMC11283099 DOI: 10.1016/j.heliyon.2024.e33629] [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: 01/29/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024] Open
Abstract
Enzyme-induced self-assembly (EISA) is a recently developed nanotechnology technique in which small molecules are induced by cellular enzymes self-assembling into nanostructures inside cancer cells. This technique can boost the efficacy of chemotherapy drugs by avoiding drug efflux, inhibiting the cells' DNA repair mechanisms, and targeting the mitochondria. In this work, we study the self-assembly of a short peptide and its fluorescence analogue induced by Eyes absent (EYA) tyrosine phosphatases to boost the efficacy of doxorubicin (DOX) therapy in drug-resistant types of breast cancer cells, MDA-MB-231 and MCF-7. The peptides Fmoc-FF-YP and NBD-FF-YP were synthesized with the solid-phase peptide synthesis (SPPS) method and analyzed with HPLC and MALDI-TOF. Dynamic light scattering was used to determine the size distribution of peptides exposed to the EYA enzyme in vitro. The presence of EYA enzymes in breast cancer cells was confirmed using the western blotting assay. The intracellular location of the peptide self-assembly was studied by imaging fluorescence NBD-tagged peptides. The efficacy of the peptide alone and with DOX was determined against MCF-7 and MDA-MB-231 using MTT and LIVE-DEAD assays. Nucleus and cytoplasm F-actin (Phalloidin) staining was used to determine cell morphology changes in response to the combination therapy of peptides/DOX. At an optimal concentration, the peptides are not toxic to the cells; however, they boost the efficacy of DOX against drug-resistant breast cancer cells. We used state-of-the-art computer-aided techniques to predict the molecular structure of peptides and their interactions with EYA. This study demonstrates an approach for incorporating non-cytotoxic components into DOX combination therapy, thereby avoiding increased systemic burden or adverse effects.
Collapse
Affiliation(s)
- Emily Carney
- Nanomedicine Lab, Department of Biomedical Engineering, University of North Texas, Texas, United States
| | | | - Soo-Yeon Kang
- Bioprinting Lab, Department of Biomedical Engineering, University of North Texas, Texas, United States
| | - Bruce A. Bunnell
- Department of Microbiology, Immunology & Genetics, University of North Texas Health Science Center, Fort Worth, United States
| | - Moo-Yeal Lee
- Bioprinting Lab, Department of Biomedical Engineering, University of North Texas, Texas, United States
| | - Neda Habibi
- Nanomedicine Lab, Department of Biomedical Engineering, University of North Texas, Texas, United States
| |
Collapse
|
16
|
Carney E, Habibi N. Enzyme-Instructed Self-Assembly of Peptides Induced by Tyrosine Phosphatase in Breast Cancer Cells. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039731 DOI: 10.1109/embc53108.2024.10782764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Enzyme induced self-assembly (EISA) leverages the expression of locally diseased enzymes within tumors to facilitate the self-assembly of small molecules into larger nanostructures capable of targeting cancer cells. Eye absent (EYA) is a tyrosine phosphatase enzyme found to be overexpressed in specific types of drug-resistant breast cancer cells. Utilizing this overexpression, we propose the synthesis of small peptides that can undergo self-assembly induced by the EYA tyrosine phosphatase. Our research demonstrates the presence of the EYA protein in Triple-Negative Breast Cancer cells by using the western blot. We then study the interaction between EYA's Tyr-phosphatase and the peptides P1and P2, and validate the successful intracellular self-assembly of fluorescence-tagged peptides in TNBC cells. These findings are reinforced by viability assays demonstrating the efficacy of the combination therapy of peptides and anticancer drugs for decreasing viability of TNBC cells. This paves the way for investigations into the discovery of new therapeutics for breast cancers.
Collapse
|
17
|
Sun J, Huang X, Shi R, Ji T, Ding J, Chen X. Enzyme-triggered orthotopic network formation from poly(amino acid) nanoparticle to suppress tumor growth and metastases. NANO TODAY 2024; 56:102222. [DOI: 10.1016/j.nantod.2024.102222] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
|
18
|
Adorinni S, Gentile S, Bellotto O, Kralj S, Parisi E, Cringoli MC, Deganutti C, Malloci G, Piccirilli F, Pengo P, Vaccari L, Geremia S, Vargiu AV, De Zorzi R, Marchesan S. Peptide Stereochemistry Effects from p Ka-Shift to Gold Nanoparticle Templating in a Supramolecular Hydrogel. ACS NANO 2024; 18:3011-3022. [PMID: 38235673 DOI: 10.1021/acsnano.3c08004] [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: 01/19/2024]
Abstract
The divergent supramolecular behavior of a series of tripeptide stereoisomers was elucidated through spectroscopic, microscopic, crystallographic, and computational techniques. Only two epimers were able to effectively self-organize into amphipathic structures, leading to supramolecular hydrogels or crystals, respectively. Despite the similarity between the two peptides' turn conformations, stereoconfiguration led to different abilities to engage in intramolecular hydrogen bonding. Self-assembly further shifted the pKa value of the C-terminal side chain. As a result, across the pH range 4-6, only one epimer predominated sufficiently as a zwitterion to reach the critical molar fraction, allowing gelation. By contrast, the differing pKa values and higher dipole moment of the other epimer favored crystallization. The four stereoisomers were further tested for gold nanoparticle (AuNP) formation, with the supramolecular hydrogel being the key to control and stabilize AuNPs, yielding a nanocomposite that catalyzed the photodegradation of a dye. Importantly, the AuNP formation occurred without the use of reductants other than the peptide, and the redox chemistry was investigated by LC-MS, NMR, and infrared scattering-type near field optical microscopy (IR s-SNOM). This study provides important insights for the rational design of simple peptides as minimalistic and green building blocks for functional nanocomposites.
Collapse
Affiliation(s)
- Simone Adorinni
- Chemical Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
| | - Serena Gentile
- Chemical Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
| | - Ottavia Bellotto
- Chemical Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
| | - Slavko Kralj
- Materials Synthesis Department, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Evelina Parisi
- Chemical Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
| | - Maria C Cringoli
- Chemical Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
| | - Caterina Deganutti
- Chemical Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
| | - Giuliano Malloci
- Physics Department, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Federica Piccirilli
- Elettra Sincrotrone Trieste, 34149 Basovizza, Italy
- Area Science Park, Padriciano 99, 34149 Trieste, Italy
| | - Paolo Pengo
- Chemical Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
| | - Lisa Vaccari
- Elettra Sincrotrone Trieste, 34149 Basovizza, Italy
| | - Silvano Geremia
- Chemical Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
| | - Attilio V Vargiu
- Physics Department, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Rita De Zorzi
- Chemical Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
| | - Silvia Marchesan
- Chemical Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy
- Unit of Trieste, INSTM, 34127 Trieste, Italy
| |
Collapse
|
19
|
Kumar Pradhan M, Suresh Puthenpurackal S, Srivastava A. Enzymatic Dimerization-Induced Self-Assembly of Alanine-Tyramine Conjugates into Versatile, Uniform, Enzyme-Loaded Organic Nanoparticles. Angew Chem Int Ed Engl 2024; 63:e202314960. [PMID: 37992201 DOI: 10.1002/anie.202314960] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
Herein, we report a novel enzymatic dimerization-induced self-assembly (e-DISA) procedure that converts alanine-tyramine conjugates into highly uniform enzyme-loaded nanoparticles (NPs) or nanocontainers by the action of horseradish peroxidase (HRP) in an aqueous medium under ambient conditions. The NP formation was possible with both enantiomers of alanine, and the average diameter could be varied from 150 nm to 250 nm (with a 5-12 % standard deviation of as-prepared samples) depending on the precursor concentration. About 60 % of the added HRP enzyme was entrapped within the NPs and was subsequently utilized for post-synthetic modification of the NPs with phenolic compounds such as tyramine or tannic acid. One-pot multi-enzyme entrapment of glucose oxidase (GOx) and peroxidase (HRP) within the NPs was also achieved. These GOx-HRP loaded NPs allowed multimodal detection of glucose, including that present in human saliva, with a limit of detection (LoD) of 740 nM through fluorimetry. The NPs exhibited good cytocompatibility and were stable to changes in pH (acidic to basic), temperature, ultrasonication, and even the presence of organic solvent (EtOH) to a certain extent, since they are stabilized by intermolecular hydrogen bonding, π-π, and CH-π interactions. The proposed e-DISA procedure can be widely expanded through the design of diverse enzyme-responsive precursors.
Collapse
Affiliation(s)
- Manas Kumar Pradhan
- Department of Chemistry, IISER Bhopal, Bhopal, 462066, Madhya Pradesh, India
| | | | - Aasheesh Srivastava
- Department of Chemistry, IISER Bhopal, Bhopal, 462066, Madhya Pradesh, India
| |
Collapse
|
20
|
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
|
21
|
Abstract
Higher-order or supramolecular protein assemblies, usually regulated by enzymatic reactions, are ubiquitous and essential for cellular functions. This evolutionary fact has provided a rigorous scientific foundation, as well as an inspiring blueprint, for exploring supramolecular assemblies of man-made molecules that are responsive to biological cues as a novel class of therapeutics for biomedicine. Among the emerging man-made supramolecular structures, peptide assemblies, formed by enzyme reactions or other stimuli, have received most of the research attention and advanced most rapidly.In this Account, we will review works that apply enzyme-instructed self-assembly (EISA) to generate intracellular peptide assemblies for developing a new kind of biomedicine, especially in the field of novel cancer nanomedicines and modulating cell morphogenesis. As a versatile and cell-compatible approach, EISA can generate nondiffusive peptide assemblies locally; thus, it provides a unique approach to target subcellular organelles with exceptional cell selectivity. We have arranged this Account in the following way: after introducing the concept, simplicity, and uniqueness of EISA, we discuss the EISA-formed intracellular peptide assemblies, including artificial filaments, in the cell cytosol. Then, we describe the representative examples targeting subcellular organelles, such as mitochondria, endoplasmic reticulum, Golgi apparatus, lysosomes, and the nucleus, by enzyme-instructed intracellular peptide assemblies for potential cancer therapeutics. After that, we highlight the recent exploration of the transcytosis of peptide assemblies for controlling cell morphogenesis. Finally, we provide a brief outlook of enzyme-instructed intracellular peptide assemblies. This Account aims to illustrate the promise of EISA-generated intracellular peptide assemblies in understanding diseases, controlling cell behaviors, and developing new therapeutics from a class of less explored molecular entities, which are substrates of enzymes and become building blocks of self-assembly after the enzymatic reactions.
Collapse
Affiliation(s)
- Zhiyu Liu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02453, United States
| | - Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02453, United States
| | - Yuchen Qiao
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02453, United States
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02453, United States
| |
Collapse
|
22
|
Wu C, Wang M, Sun J, Jia Y, Zhu X, Liu G, Zhu Y, Guan Y, Zhang Z, Pang X. Peptide-drug co-assembling: A potent armament against cancer. Theranostics 2023; 13:5322-5347. [PMID: 37908727 PMCID: PMC10614680 DOI: 10.7150/thno.87356] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/19/2023] [Indexed: 11/02/2023] Open
Abstract
Cancer is still one of the major problems threatening human health and the therapeutical efficacies of available treatment choices are often rather low. Due to their favorable biocompatibility, simplicity of modification, and improved therapeutic efficacy, peptide-based self-assembled delivery systems have undergone significant evolution. Physical encapsulation and covalent conjugation are two common approaches to load drugs for peptide assembly-based delivery, which are always associated with drug leaks in the blood circulation system or changed pharmacological activities, respectively. To overcome these difficulties, a more elegant peptide-based assembly strategy is desired. Notably, peptide-mediated co-assembly with drug molecules provides a new method for constructing nanomaterials with improved versatility and structural stability. The co-assembly strategy can be used to design various nanostructures for cancer therapy, such as nanotubes, nanofibrils, hydrogels, and nanovesicles. Recently, these co-assembled nanostructures have gained tremendous attention for their unique superiorities in tumor therapy. This article describes the classification of assembled peptides, driving forces for co-assembly, and specifically, the design methodologies for various drug molecules in co-assembly. It also highlights recent research on peptide-mediated co-assembled delivery systems for cancer therapy. Finally, it summarizes the pros and cons of co-assembly in cancer therapy and offers some suggestions for conquering the challenges in this field.
Collapse
Affiliation(s)
- Can Wu
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Manman Wang
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Jinpan Sun
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Yongyan Jia
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Xiali Zhu
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Gaizhi Liu
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Yanhui Zhu
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Yanbin Guan
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Zhenqiang Zhang
- Academy of Chinese Medicine Science, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Xin Pang
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| |
Collapse
|
23
|
Cavallaro PA, De Santo M, Belsito EL, Longobucco C, Curcio M, Morelli C, Pasqua L, Leggio A. Peptides Targeting HER2-Positive Breast Cancer Cells and Applications in Tumor Imaging and Delivery of Chemotherapeutics. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2476. [PMID: 37686984 PMCID: PMC10490457 DOI: 10.3390/nano13172476] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023]
Abstract
Breast cancer represents the most common cancer type and one of the major leading causes of death in the female worldwide population. Overexpression of HER2, a transmembrane glycoprotein related to the epidermal growth factor receptor, results in a biologically and clinically aggressive breast cancer subtype. It is also the primary driver for tumor detection and progression and, in addition to being an important prognostic factor in women diagnosed with breast cancer, HER2 is a widely known therapeutic target for drug development. The aim of this review is to provide an updated overview of the main approaches for the diagnosis and treatment of HER2-positive breast cancer proposed in the literature over the past decade. We focused on the different targeting strategies involving antibodies and peptides that have been explored with their relative outcomes and current limitations that need to be improved. The review also encompasses a discussion on targeted peptides acting as probes for molecular imaging. By using different types of HER2-targeting strategies, nanotechnology promises to overcome some of the current clinical challenges by developing novel HER2-guided nanosystems suitable as powerful tools in breast cancer imaging, targeting, and therapy.
Collapse
Affiliation(s)
- Palmira Alessia Cavallaro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Marzia De Santo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Emilia Lucia Belsito
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Camilla Longobucco
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Manuela Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Catia Morelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| | - Luigi Pasqua
- Department of Environmental Engineering, University of Calabria, Via P. Bucci, 87036 Rende, Italy
| | - Antonella Leggio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (P.A.C.); (M.D.S.); (E.L.B.); (C.L.); (M.C.); (C.M.)
| |
Collapse
|
24
|
Guo J, Wang F, Huang Y, He H, Tan W, Yi M, Egelman EH, Xu B. Cell spheroid creation by transcytotic intercellular gelation. NATURE NANOTECHNOLOGY 2023; 18:1094-1104. [PMID: 37217766 PMCID: PMC10525029 DOI: 10.1038/s41565-023-01401-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/14/2023] [Indexed: 05/24/2023]
Abstract
Cell spheroids bridge the discontinuity between in vitro systems and in vivo animal models. However, inducing cell spheroids by nanomaterials remains an inefficient and poorly understood process. Here we use cryogenic electron microscopy to determine the atomic structure of helical nanofibres self-assembled from enzyme-responsive D-peptides and fluorescent imaging to show that the transcytosis of D-peptides induces intercellular nanofibres/gels that potentially interact with fibronectin to enable cell spheroid formation. Specifically, D-phosphopeptides, being protease resistant, undergo endocytosis and endosomal dephosphorylation to generate helical nanofibres. On secretion to the cell surface, these nanofibres form intercellular gels that act as artificial matrices and facilitate the fibrillogenesis of fibronectins to induce cell spheroids. No spheroid formation occurs without endo- or exocytosis, phosphate triggers or shape switching of the peptide assemblies. This study-coupling transcytosis and morphological transformation of peptide assemblies-demonstrates a potential approach for regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Jiaqi Guo
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Fengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA.
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Yimeng Huang
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Meihui Yi
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA.
| | - Bing Xu
- Department of Chemistry, Brandeis University, Waltham, MA, USA.
| |
Collapse
|
25
|
Qiao Y, Xu B. Peptide Assemblies for Cancer Therapy. ChemMedChem 2023; 18:e202300258. [PMID: 37380607 PMCID: PMC10613339 DOI: 10.1002/cmdc.202300258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 06/30/2023]
Abstract
Supramolecular assemblies made by the self-assembly of peptides are finding an increasing number of applications in various fields. While the early exploration of peptide assemblies centered on tissue engineering or regenerative medicine, the recent development has shown that peptide assemblies can act as supramolecular medicine for cancer therapy. This review covers the progress of applying peptide assemblies for cancer therapy, with the emphasis on the works appeared over the last five years. We start with the introduction of a few seminal works on peptide assemblies, then discuss the combination of peptide assemblies with anticancer drugs. Next, we highlight the use of enzyme-controlled transformation or shapeshifting of peptide assemblies for inhibiting cancer cells and tumors. After that, we provide the outlook for this exciting field that promises new kind of therapeutics for cancer therapy.
Collapse
Affiliation(s)
- Yuchen Qiao
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| |
Collapse
|
26
|
Yan Z, Liu Y, Zhao L, Hu J, Du Y, Peng X, Liu Z. In situ stimulus-responsive self-assembled nanomaterials for drug delivery and disease treatment. MATERIALS HORIZONS 2023; 10:3197-3217. [PMID: 37376926 DOI: 10.1039/d3mh00592e] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
The individual motifs that respond to specific stimuli for the self-assembly of nanomaterials play important roles. In situ constructed nanomaterials are formed spontaneously without human intervention and have promising applications in bioscience. However, due to the complex physiological environment of the human body, designing stimulus-responsive self-assembled nanomaterials in vivo is a challenging problem for researchers. In this article, we discuss the self-assembly principles of various nanomaterials in response to the tissue microenvironment, cell membrane, and intracellular stimuli. We propose the applications and advantages of in situ self-assembly in drug delivery and disease diagnosis and treatment, with a focus on in situ self-assembly at the lesion site, especially in cancer. Additionally, we introduce the significance of introducing exogenous stimulation to construct self-assembly in vivo. Based on this foundation, we put forward the prospects and possible challenges in the field of in situ self-assembly. This review uncovers the relationship between the structure and properties of in situ self-assembled nanomaterials and provides new ideas for innovative drug molecular design and development to solve the problems in the targeted delivery and precision medicine.
Collapse
Affiliation(s)
- Ziling Yan
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, P. R. China
| | - Licheng Zhao
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, P. R. China
| | - Jiaxin Hu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
| | - Yimin Du
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
| | - Xingxing Peng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
- Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan Province, P. R. China
| |
Collapse
|
27
|
Jana B, Jin S, Go EM, Cho Y, Kim D, Kim S, Kwak SK, Ryu JH. Intra-Lysosomal Peptide Assembly for the High Selectivity Index against Cancer. J Am Chem Soc 2023; 145:18414-18431. [PMID: 37525328 DOI: 10.1021/jacs.3c04467] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Lysosomes remain powerful organelles and important targets for cancer therapy because cancer cell proliferation is greatly dependent on effective lysosomal function. Recent studies have shown that lysosomal membrane permeabilization induces cell death and is an effective way to treat cancer by bypassing the classical caspase-dependent apoptotic pathway. However, most lysosome-targeted anticancer drugs have very low selectivity for cancer cells. Here, we show intra-lysosomal self-assembly of a peptide amphiphile as a powerful technique to overcome this problem. We designed a peptide amphiphile that localizes in the cancer lysosome and undergoes cathepsin B enzyme-instructed supramolecular assembly. This localized assembly induces lysosomal swelling, membrane permeabilization, and damage to the lysosome, which eventually causes caspase-independent apoptotic death of cancer cells without conventional chemotherapeutic drugs. It has specific anticancer effects and is effective against drug-resistant cancers. Moreover, this peptide amphiphile exhibits high tumor targeting when attached to a tumor-targeting ligand and causes significant inhibition of tumor growth both in cancer and drug-resistant cancer xenograft models.
Collapse
Affiliation(s)
- Batakrishna Jana
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seongeon Jin
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Eun Min Go
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Yumi Cho
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Dohyun Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sangpil Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sang Kyu Kwak
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
28
|
Fan P, Guan Y, Zhang X, Wang J, Xu Y, Song B, Zhang S, Wang H, Liu Y, Qiao ZY. Cell membrane-specific self-assembly of peptide nanomedicine induces tumor immunogenic death to enhance cancer therapy. NANOSCALE HORIZONS 2023; 8:1226-1234. [PMID: 37366007 DOI: 10.1039/d3nh00173c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Immunogenic cell death (ICD), as an unusual cell death pattern, mediates cancer cells to release a series of damage-associated molecular patterns (DAMPs), and is widely used in the field of cancer immunotherapy. Injuring the cell membrane can serve as a novel ICD initiation strategy. In this study, a peptide nanomedicine (PNpC) is designed using the fragment CM11 of cecropin, which is effective in disrupting cell membranes because of its α-helical structure. PNpC self-assembles in situ in the presence of high levels of alkaline phosphatase (ALP) on the tumor cell membrane, transforming from nanoparticles to nanofibers, which reduces the cellular internalization of the nanomedicine and increases the interaction between CM11 and tumor cell membranes. Both in vitro and in vivo results indicate that PNpC plays a significant role in killing tumor cells by triggering ICD. The ICD induced by the destruction of the cancer cell membrane is accompanied by the release of DAMPs, which promotes the maturation of DCs and facilitates the presentation of tumor-associated antigens (TAA), resulting in the infiltration of CD8+ T cells. We believe that PNpC can trigger ICD while killing cancer cells, providing a new reference for cancer immunotherapy.
Collapse
Affiliation(s)
- Pengsheng Fan
- College of Marine Life Science, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, China.
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Yinghua Guan
- College of Marine Life Science, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, China.
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Xiaoying Zhang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Jiaqi Wang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Yinsheng Xu
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Benli Song
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Suling Zhang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Hao Wang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Ya Liu
- College of Marine Life Science, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, China.
| | - Zeng-Ying Qiao
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| |
Collapse
|
29
|
Zhou Y, Li Q, Wu Y, Li X, Zhou Y, Wang Z, Liang H, Ding F, Hong S, Steinmetz NF, Cai H. Molecularly Stimuli-Responsive Self-Assembled Peptide Nanoparticles for Targeted Imaging and Therapy. ACS NANO 2023; 17:8004-8025. [PMID: 37079378 DOI: 10.1021/acsnano.3c01452] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Self-assembly has emerged as an extensively used method for constructing biomaterials with sizes ranging from nanometers to micrometers. Peptides have been extensively investigated for self-assembly. They are widely applied owing to their desirable biocompatibility, biodegradability, and tunable architecture. The development of peptide-based nanoparticles often requires complex synthetic processes involving chemical modification and supramolecular self-assembly. Stimuli-responsive peptide nanoparticles, also termed "smart" nanoparticles, capable of conformational and chemical changes in response to stimuli, have emerged as a class of promising materials. These smart nanoparticles find a diverse range of biomedical applications, including drug delivery, diagnostics, and biosensors. Stimuli-responsive systems include external stimuli (such as light, temperature, ultrasound, and magnetic fields) and internal stimuli (such as pH, redox environment, salt concentration, and biomarkers), facilitating the generation of a library of self-assembled biomaterials for biomedical imaging and therapy. Thus, in this review, we mainly focus on peptide-based nanoparticles built by self-assembly strategy and systematically discuss their mechanisms in response to various stimuli. Furthermore, we summarize the diverse range of biomedical applications of peptide-based nanomaterials, including diagnosis and therapy, to demonstrate their potential for medical translation.
Collapse
Affiliation(s)
- Yang Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Qianqian Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Ye Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Xinyu Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Ya Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Zhu Wang
- Department of Urology, Affiliated People's Hospital of Longhua Shenzhen, Southern Medical University, 38 Jinglong Jianshe Road, Shenzhen, Guangdong 518109, PR China
| | - Hui Liang
- Department of Urology, Affiliated People's Hospital of Longhua Shenzhen, Southern Medical University, 38 Jinglong Jianshe Road, Shenzhen, Guangdong 518109, PR China
| | - Feiqing Ding
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Sheng Hong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Nicole F Steinmetz
- Department of NanoEngineering, Department of Biongineering, Department of Radiology, Moores Cancer Center, Center for Nano-ImmunoEngineering, Center for Engineering in Cancer, Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, California 92093, United States
| | - Hui Cai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| |
Collapse
|
30
|
Kumar V, Ozguney B, Vlachou A, Chen Y, Gazit E, Tamamis P. Peptide Self-Assembled Nanocarriers for Cancer Drug Delivery. J Phys Chem B 2023; 127:1857-1871. [PMID: 36812392 PMCID: PMC10848270 DOI: 10.1021/acs.jpcb.2c06751] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/24/2022] [Indexed: 02/24/2023]
Abstract
The design of novel cancer drug nanocarriers is critical in the framework of cancer therapeutics. Nanomaterials are gaining increased interest as cancer drug delivery systems. Self-assembling peptides constitute an emerging novel class of highly attractive nanomaterials with highly promising applications in drug delivery, as they can be used to facilitate drug release and/or stability while reducing side effects. Here, we provide a perspective on peptide self-assembled nanocarriers for cancer drug delivery and highlight the aspects of metal coordination, structure stabilization, and cyclization, as well as minimalism. We review particular challenges in nanomedicine design criteria and, finally, provide future perspectives on addressing a portion of the challenges via self-assembling peptide systems. We consider that the intrinsic advantages of such systems, along with the increasing progress in computational and experimental approaches for their study and design, could possibly lead to novel classes of single or multicomponent systems incorporating such materials for cancer drug delivery.
Collapse
Affiliation(s)
- Vijay
Bhooshan Kumar
- The
Shmunis School of Biomedicine and Cancer Research, George S. Wise
Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Busra Ozguney
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
| | - Anastasia Vlachou
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
| | - Yu Chen
- The
Shmunis School of Biomedicine and Cancer Research, George S. Wise
Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ehud Gazit
- The
Shmunis School of Biomedicine and Cancer Research, George S. Wise
Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Department
of Materials Science and Engineering, Iby and Aladar Fleischman Faculty
of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol
School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Phanourios Tamamis
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
- Department
of Materials Science and Engineering, Texas
A&M University, College
Station, Texas 77843-3003, United States
| |
Collapse
|
31
|
Hamley IW. Self-Assembly, Bioactivity, and Nanomaterials Applications of Peptide Conjugates with Bulky Aromatic Terminal Groups. ACS APPLIED BIO MATERIALS 2023; 6:384-409. [PMID: 36735801 PMCID: PMC9945136 DOI: 10.1021/acsabm.2c01041] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The self-assembly and structural and functional properties of peptide conjugates containing bulky terminal aromatic substituents are reviewed with a particular focus on bioactivity. Terminal moieties include Fmoc [fluorenylmethyloxycarbonyl], naphthalene, pyrene, naproxen, diimides of naphthalene or pyrene, and others. These provide a driving force for self-assembly due to π-stacking and hydrophobic interactions, in addition to the hydrogen bonding, electrostatic, and other forces between short peptides. The balance of these interactions leads to a propensity to self-assembly, even for conjugates to single amino acids. The hybrid molecules often form hydrogels built from a network of β-sheet fibrils. The properties of these as biomaterials to support cell culture, or in the development of molecules that can assemble in cells (in response to cellular enzymes, or otherwise) with a range of fascinating bioactivities such as anticancer or antimicrobial activity, are highlighted. In addition, applications of hydrogels as slow-release drug delivery systems and in catalysis and other applications are discussed. The aromatic nature of the substituents also provides a diversity of interesting optoelectronic properties that have been demonstrated in the literature, and an overview of this is also provided. Also discussed are coassembly and enzyme-instructed self-assembly which enable precise tuning and (stimulus-responsive) functionalization of peptide nanostructures.
Collapse
|
32
|
Wu J, Liu Y, Cao M, Zheng N, Ma H, Ye X, Yang N, Liu Z, Liao W, Sun L. Cancer-Responsive Multifunctional Nanoplatform Based on Peptide Self-Assembly for Highly Efficient Combined Cancer Therapy by Alleviating Hypoxia and Improving the Immunosuppressive Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:5667-5678. [PMID: 36651290 DOI: 10.1021/acsami.2c20388] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Hypoxia, as a main feature of the tumor microenvironment, has greatly limited the efficacy of photodynamic therapy (PDT), as well as its clinical application. Here, a multifunctional composite nanoplatform, the peptide/Ce6/MnO2 nanocomposite (RKCM), has been constructed to alleviate tumor hypoxia and increase the efficacy of PDT using rationally designed peptide fibrils to encapsulate chlorin e6 (Ce6) inside and to mineralize MnO2 nanoparticles on the surface. As a result, RKCM significantly improved the PDT efficacy by increasing reactive oxygen species (ROS) generation, decreasing tumor cell viability, and inhibiting tumor growth and metastasis. Besides, decreased HIF-1α expression and increased immune-activated cell infiltration were also observed in RKCM/laser treatment xenograft. Mechanically, (1) Ce6 can induce singlet oxygen (1O2) generation under laser irradiation to give photodynamic therapy (PDT); (2) MnO2 can react with H2O2 in situ to supply additional O2 to alleviate tumor hypoxia; and (3) the released Mn2+ ions can induce a Fenton-like reaction to generate •OH for chemical dynamic therapy (CDT). Moreover, RKCM/laser treatment also presented with an abscopal effect to block the occurrence of lung metastasis by remolding the pre-metastasis immune microenvironment. With these several aspects working together, the peptide/Ce6/MnO2 nanoplatform can achieve highly efficient tumor therapy. Such a strategy based on peptide self-assembly provides a promising way to rationally design a cancer-responsive multifunctional nanoplatform for highly efficient combined cancer therapy by alleviating hypoxia and improving the immune microenvironment.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Thoracic Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| | - Yang Liu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Meiwen Cao
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Nannan Zheng
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Hongchao Ma
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Xiandong Ye
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Nanyan Yang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhihong Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Oncology, Air Force Medical Center of PLA, Air Force Medical University, Beijing 100089, China
| |
Collapse
|
33
|
Lin F, Jia C, Wu FG. Intracellular Enzyme-Instructed Self-Assembly of Peptides (IEISAP) for Biomedical Applications. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196557. [PMID: 36235094 PMCID: PMC9571778 DOI: 10.3390/molecules27196557] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/13/2022]
Abstract
Despite the remarkable significance and encouraging breakthroughs of intracellular enzyme-instructed self-assembly of peptides (IEISAP) in disease diagnosis and treatment, a comprehensive review that focuses on this topic is still desirable. In this article, we carefully review the advances in the applications of IEISAP, including the development of various bioimaging techniques, such as fluorescence imaging, photoacoustic imaging, magnetic resonance imaging, positron-emission tomography imaging, radiation imaging, and multimodal imaging, which are successfully leveraged in visualizing cancer tissues and cells, bacteria, and enzyme activity. We also summarize the utilization of IEISAP in disease treatments, including anticancer, antibacterial, and antiinflammation applications, among others. We present the design, action modes, structures, properties, functions, and performance of IEISAP materials, such as nanofibers, nanoparticles, nanoaggregates, and hydrogels. Finally, we conclude with an outlook towards future developments of IEISAP materials for biomedical applications. It is believed that this review may foster the future development of IEISAP with better performance in the biomedical field.
Collapse
|
34
|
Morita K, Nishimura K, Yamamoto S, Shimizu N, Yashiro T, Kawabata R, Aoi T, Tamura A, Maruyama T. In Situ Synthesis of an Anticancer Peptide Amphiphile Using Tyrosine Kinase Overexpressed in Cancer Cells. JACS AU 2022; 2:2023-2028. [PMID: 36186562 PMCID: PMC9516706 DOI: 10.1021/jacsau.2c00301] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 06/16/2023]
Abstract
Cell-selective killing using molecular self-assemblies is an emerging concept for cancer therapy. Reported molecular self-assemblies are triggered by hydrolysis of well-designed molecules inside or outside cancer cells. This hydrolysis can occur in cancer and normal cells because of the abundance of water in living systems. Here, we report the in situ synthesis of a self-assembling molecule using a tyrosine kinase overexpressed in cancer cells. We designed a tyrosine-containing peptide amphiphile (C16-E4Y) that is transformed into a phosphorylated peptide amphiphile (C16-E4pY) by the overexpressed tyrosine kinase. Phosphorylation of C16-E4Y promoted self-assembly to form nanofibers in cancer cells. C16-E4Y exhibited selective cytotoxicity toward cancer cells overexpressing the tyrosine kinase. Self-assembled C16-E4pY induced endoplasmic reticulum stress that caused apoptotic cell death. Animal experiments revealed that C16-E4Y has antitumor activity. These results show that an enzyme overexpressed in cancer cells is available for intracellular synthesis of an antitumor self-assembling drug that is cell-selective.
Collapse
Affiliation(s)
- Kenta Morita
- Department
of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan
| | - Kanon Nishimura
- Department
of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan
| | - Shota Yamamoto
- Department
of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan
| | - Natsumi Shimizu
- Department
of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan
| | - Tomoko Yashiro
- Department
of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan
| | - Ryoko Kawabata
- Department
of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan
| | - Takashi Aoi
- Graduate
School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuou-ku, Kobe 650-0017, Japan
| | - Atsuo Tamura
- Department
of Chemistry, Graduate School of Science, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan
| | - Tatsuo Maruyama
- Department
of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan
| |
Collapse
|
35
|
Zheng J, Song X, Yang Z, Yin C, Luo W, Yin C, Ni Y, Wang Y, Zhang Y. Self-assembly hydrogels of therapeutic agents for local drug delivery. J Control Release 2022; 350:898-921. [PMID: 36089171 DOI: 10.1016/j.jconrel.2022.09.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 10/14/2022]
Abstract
Advanced drug delivery systems are of vital importance to enhance therapeutic efficacy. Among various recently developed formulations, self-assembling hydrogels composed of therapeutic agents have shown promising potential for local drug delivery owing to their excellent biocompatibility, high drug-loading efficiency, low systemic toxicity, and sustained drug release behavior. In particular, therapeutic agents self-assembling hydrogels with well-defined nanostructures are beneficial for direct delivery to the target site via injection, not only improving drug availability, but also extending their retention time and promoting cellular uptake. In brief, the self-assembly approach offers better opportunities to improve the precision of pharmaceutical treatment and achieve superior treatment efficacies. In this review, we intend to cover the recent developments in therapeutic agent self-assembling hydrogels. First, the molecular structures, self-assembly mechanisms, and application of self-assembling hydrogels are systematically outlined. Then, we summarize the various self-assembly strategies, including the single therapeutic agent, metal-coordination, enzyme-instruction, and co-assembly of multiple therapeutic agents. Finally, the potential challenges and future perspectives are discussed. We hope that this review will provide useful insights into the design and preparation of therapeutic agent self-assembling hydrogels.
Collapse
Affiliation(s)
- Jun Zheng
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Xianwen Song
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Zhaoyu Yang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chao Yin
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Weikang Luo
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chunyang Yin
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yaqiong Ni
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yang Wang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Yi Zhang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China.
| |
Collapse
|
36
|
Dynamic assembly and biocatalysis-selected gelation endow self-compartmentalized multienzyme superactivity. Sci China Chem 2022. [DOI: 10.1007/s11426-022-1330-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
37
|
Cheng Y, Clark AE, Zhou J, He T, Li Y, Borum RM, Creyer MN, Xu M, Jin Z, Zhou J, Yim W, Wu Z, Fajtová P, O’Donoghue AJ, Carlin AF, Jokerst JV. Protease-Responsive Peptide-Conjugated Mitochondrial-Targeting AIEgens for Selective Imaging and Inhibition of SARS-CoV-2-Infected Cells. ACS NANO 2022; 16:12305-12317. [PMID: 35878004 PMCID: PMC9344892 DOI: 10.1021/acsnano.2c03219] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/11/2022] [Indexed: 05/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a serious threat to human health and lacks an effective treatment. There is an urgent need for both real-time tracking and precise treatment of the SARS-CoV-2-infected cells to mitigate and ultimately prevent viral transmission. However, selective triggering and tracking of the therapeutic process in the infected cells remains challenging. Here, we report a main protease (Mpro)-responsive, mitochondrial-targeting, and modular-peptide-conjugated probe (PSGMR) for selective imaging and inhibition of SARS-CoV-2-infected cells via enzyme-instructed self-assembly and aggregation-induced emission (AIE) effect. The amphiphilic PSGMR was constructed with tunable structure and responsive efficiency and validated with recombinant proteins, cells transfected with Mpro plasmid or infected by SARS-CoV-2, and a Mpro inhibitor. By rational construction of AIE luminogen (AIEgen) with modular peptides and Mpro, we verified that the cleavage of PSGMR yielded gradual aggregation with bright fluorescence and enhanced cytotoxicity to induce mitochondrial interference of the infected cells. This strategy may have value for selective detection and treatment of SARS-CoV-2-infected cells.
Collapse
Affiliation(s)
- Yong Cheng
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alex E. Clark
- Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Jiajing Zhou
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Tengyu He
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yi Li
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Raina M. Borum
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Matthew N. Creyer
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ming Xu
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhicheng Jin
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jingcheng Zhou
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wonjun Yim
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhuohong Wu
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Pavla Fajtová
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anthony J. O’Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Aaron F. Carlin
- Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Jesse V. Jokerst
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
38
|
Asokan-Sheeja H, Yang S, A Adones A, Chen W, B Fulton B, K Chintapula U, T Nguyen K, J Lovely C, A Brautigam C, Nam K, Dong H. Self‐assembling Peptides with Internal Ionizable Unnatural Amino Acids: A New and General Approach to pH‐responsive Peptide Materials. Chem Asian J 2022; 17:e202200724. [DOI: 10.1002/asia.202200724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/11/2022] [Indexed: 11/06/2022]
Affiliation(s)
| | - Su Yang
- The University of Texas at Arlington Chemistry UNITED STATES
| | - Ashley A Adones
- The University of Texas at Arlington Chemistry UNITED STATES
| | - Weike Chen
- The University of Texas at Arlington Chemistry UNITED STATES
| | | | | | - Kytai T Nguyen
- The University of Texas at Arlington Bioengineering UNITED STATES
| | - Carl J Lovely
- The University of Texas at Arlington Chemistry UNITED STATES
| | - Chad A Brautigam
- UT Southwestern: The University of Texas Southwestern Medical Center Biophysics UNITED STATES
| | - Kwangho Nam
- The University of Texas at Arlington Chemistry UNITED STATES
| | - He Dong
- University of Texas at Arlington Chemistry 700 Planetarium Place 76019 Arlington UNITED STATES
| |
Collapse
|
39
|
Macdougall LJ, Hoffman TE, Kirkpatrick BE, Fairbanks BD, Bowman CN, Spencer SL, Anseth KS. Intracellular Crowding by Bio-Orthogonal Hydrogel Formation Induces Reversible Molecular Stasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202882. [PMID: 35671709 PMCID: PMC9377388 DOI: 10.1002/adma.202202882] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Indexed: 05/22/2023]
Abstract
To survive extreme conditions, certain animals enter a reversible protective stasis through vitrification of the cytosol by polymeric molecules such as proteins and polysaccharides. In this work, synthetic gelation of the cytosol in living cells is used to induce reversible molecular stasis. Through the sequential lipofectamine-mediated transfection of complementary poly(ethylene glycol) macromers into mammalian cells, intracellular crosslinking occurs through bio-orthogonal strain-promoted azide-alkyne cycloaddition click reactions. This achieves efficient polymer uptake with minimal cell death (99% viable). Intracellular crosslinking decreases DNA replication and protein synthesis, and increases the quiescent population by 2.5-fold. Real-time tracking of single cells containing intracellular crosslinked polymers identifies increases in intermitotic time (15 h vs 19 h) and decreases in motility (30 µm h-1 vs 15 µm h-1 ). The cytosol viscosity increases threefold after intracellular crosslinking and results in disordered cytoskeletal structure in addition to the disruption of cellular coordination in a scratch assay. By incorporating photodegradable nitrobenzyl moieties into the polymer backbone, the effects of intracellular crosslinking are reversed upon exposure to light, thereby restoring proliferation (80% phospho-Rb+ cells), protein translation, and migration. Reversible intracellular crosslinking provides a novel method for dynamic manipulation of intracellular mechanics, altering essential processes that determine cellular function.
Collapse
Affiliation(s)
- Laura J Macdougall
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Timothy E Hoffman
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Bruce E Kirkpatrick
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Medical Scientist Training Program, School of Medicine, University of Colorado, Aurora, CO, 80045, USA
| | - Benjamin D Fairbanks
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- Material Science and Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Christopher N Bowman
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Material Science and Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Sabrina L Spencer
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Material Science and Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
40
|
Song J, Zhang Q, Li G, Zhang Y. Constructing ECM-like Structure on the Plasma Membrane via Peptide Assembly to Regulate the Cellular Response. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:8733-8747. [PMID: 35839338 DOI: 10.1021/acs.langmuir.2c00711] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
This feature article introduces the design of self-assembling peptides that serve as the basic building blocks for the construction of extracellular matrix (ECM)-like structure in the vicinity of the plasma membrane. By covalently conjugating a bioactive motif, such as membrane protein binding ligand or enzymatic responsive building block, with a self-assembling motif, especially the aromatic peptide, a self-assembling peptide that retains bioactivity is obtained. Instructed by the target membrane protein or enzyme, the bioactive peptides self-assemble into ECM-like structure exerting various stimuli to regulate the cellular response via intracellular signaling, especially mechanotransduction. By briefly summarizing the properties and applications (e.g., wound healing, controlling cell motility and cell fate) of these peptides, we intend to illustrate the basic requirements and promises of the peptide assembly as a true bottom-up approach in the construction of artificial ECM.
Collapse
Affiliation(s)
- Jiaqi Song
- Department of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi'an Jiaotong University, Shaanxi 710061, P. R. China
| | - Qizheng Zhang
- Active Soft Matter Group, CAS Songshan Lake Materials Laboratory, Dongguan 523808, China
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Guanying Li
- Department of Biophysics, School of Basic Medical Sciences, Health Science Centre, Xi'an Jiaotong University, Shaanxi 710061, P. R. China
| | - Ye Zhang
- Active Soft Matter Group, CAS Songshan Lake Materials Laboratory, Dongguan 523808, China
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| |
Collapse
|
41
|
Wang H, Monroe M, Leslie F, Flexner C, Cui H. Supramolecular nanomedicines through rational design of self-assembling prodrugs. Trends Pharmacol Sci 2022; 43:510-521. [PMID: 35459589 DOI: 10.1016/j.tips.2022.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 01/23/2023]
Abstract
Advancements in the development of nanomaterials have led to the creation of a plethora of functional constructs as drug delivery vehicles to address many dire medical needs. The emerging prodrug strategy provides an alternative solution to create nanomedicines of extreme simplicity by directly using the therapeutic agents as molecular building blocks. This Review outlines different prodrug-based drug delivery systems, highlights the advantages of the prodrug strategy for therapeutic delivery, and demonstrates how combinations of different functionalities - such as stimuli responsiveness, targeting propensity, and multidrug conjugation - can be incorporated into designed prodrug delivery systems. Furthermore, we discuss the opportunities and challenges facing this rapidly growing field.
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
| | - Maya Monroe
- 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
| | - Faith Leslie
- 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
| | - Charles Flexner
- Divisions of Clinical Pharmacology and Infectious Diseases, Johns Hopkins University School of Medicine and Bloomberg School of Public Health, Baltimore, MD 21287, 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; Center of Nanomedicine, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
42
|
Qian X, Xu X, Wu Y, Wang J, Li J, Chen S, Wen J, Li Y, Zhang Z. Strategies of engineering nanomedicines for tumor retention. J Control Release 2022; 346:193-211. [PMID: 35447297 DOI: 10.1016/j.jconrel.2022.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 01/29/2023]
Abstract
The retention of therapeutic agents in solid tumors at sufficient concentration and duration is crucial for their antitumor effects. Given the important contribution of nanomedicines to oncology, we herein summarized two major strategies of nanomedicines for tumor retention, such as transformation- and interactions-mediated strategies. The transformation-mediated retention strategy was achieved by enlarging particle size of nanomedicines or modulating the morphology into fibrous structures, while the interactions-mediated retention strategy was accomplished by modulating nanomedicines to promote their interactions with versatile cells or components in tumors. Moreover, we provide some considerations and perspectives of tumor-retaining nanomedicines for effective cancer therapy.
Collapse
Affiliation(s)
- Xindi Qian
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoxuan Xu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Wu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiaoying Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shuo Chen
- School of Pharmacy, the University of Auckland, Auckland 1142, New Zealand
| | - Jingyuan Wen
- School of Pharmacy, the University of Auckland, Auckland 1142, New Zealand
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China.; University of Chinese Academy of Sciences, Beijing 100049, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China.
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China.; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
43
|
Wang XJ, Cheng J, Zhang LY, Zhang JG. Self-assembling peptides-based nano-cargos for targeted chemotherapy and immunotherapy of tumors: recent developments, challenges, and future perspectives. Drug Deliv 2022; 29:1184-1200. [PMID: 35403517 PMCID: PMC9004497 DOI: 10.1080/10717544.2022.2058647] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Xue-Jun Wang
- Department of General Surgery, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, China
| | - Jian Cheng
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital of Hangzhou Medical College), Hangzhou, China
| | - Le-Yi Zhang
- Department of General Surgery, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, China
| | - Jun-Gang Zhang
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital of Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
44
|
Wu B, Zhao S, Yang X, Zhou L, Ma Y, Zhang H, Li W, Wang H. Biomimetic Heterodimerization of Tetrapeptides to Generate Liquid Crystalline Hydrogel in A Two-Component System. ACS NANO 2022; 16:4126-4138. [PMID: 35230089 DOI: 10.1021/acsnano.1c09860] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Anisotropic structures made by hierarchical self-assembly and crystallization play an essential role in the living system. However, the spontaneous formation of liquid crystalline hydrogel of low molecular weight organic molecules with controlled properties remains challenging. This work describes a rational design of tetrapeptide without N-terminal modification and chemical conjugation that utilizes intermolecular interactions to drive the formation of nanofiber bundles in a two-component system, which could not be accessed by a single component. The diameter of nanofibers can be simply controlled by varying the enantiomer of electrostatic pairs. Mutation of lysine (K) to arginine (R) results in an over 30-fold increase of mechanical property. Mechanistic studies using different techniques unravel the mechanism of self-assembly and formation of anisotropic liquid crystalline domains. All-atom molecular dynamics simulations reveal that the mixture of heterochiral peptides self-assembles into a nanofiber with a larger width compared to the homochiral assemblies due to the different stacking pattern and intermolecular interactions. The intermolecular interactions show an obvious increase by substituting the K with R, facilitating a more stable assembly and further altering the assembly mechanics and bulk material properties. Moreover, we also demonstrated that the hydrogel properties can be easily controlled by incorporating a light-responsive group. This work provides a method to generate the liquid crystalline hydrogel from isotropic monomers.
Collapse
Affiliation(s)
- Bihan Wu
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province China
| | - Shuang Zhao
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Institute of Advanced Technology, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province China
| | - Xuejiao Yang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province China
| | - Laicheng Zhou
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province China
| | - Yang Ma
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province China
| | - Hongyue Zhang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province China
| | - Wenbin Li
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Institute of Advanced Technology, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province China
| | - Huaimin Wang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province China
| |
Collapse
|
45
|
Le‐Vinh B, Akkuş‐Dağdeviren ZB, Le NN, Nazir I, Bernkop‐Schnürch A. Alkaline Phosphatase: A Reliable Endogenous Partner for Drug Delivery and Diagnostics. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100219] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Bao Le‐Vinh
- Department of Pharmaceutical Technology Institute of Pharmacy University of Innsbruck Innrain 80/82 Innsbruck 6020 Austria
- Department of Industrial Pharmacy Faculty of Pharmacy University of Medicine and Pharmacy at Ho Chi Minh City Ho Chi Minh City 700000 Viet Nam
| | - Zeynep Burcu Akkuş‐Dağdeviren
- Department of Pharmaceutical Technology Institute of Pharmacy University of Innsbruck Innrain 80/82 Innsbruck 6020 Austria
| | - Nguyet‐Minh Nguyen Le
- Department of Pharmaceutical Technology Institute of Pharmacy University of Innsbruck Innrain 80/82 Innsbruck 6020 Austria
- Department of Industrial Pharmacy Faculty of Pharmacy University of Medicine and Pharmacy at Ho Chi Minh City Ho Chi Minh City 700000 Viet Nam
| | - Imran Nazir
- Department of Pharmacy COMSATS University Islamabad Abbottabad Campus Abbottabad 22060 Pakistan
| | - Andreas Bernkop‐Schnürch
- Department of Pharmaceutical Technology Institute of Pharmacy University of Innsbruck Innrain 80/82 Innsbruck 6020 Austria
| |
Collapse
|
46
|
Yi M, Tan W, Guo J, Xu B. Enzymatic noncovalent synthesis of peptide assemblies generates multimolecular crowding in cells for biomedical applications. Chem Commun (Camb) 2021; 57:12870-12879. [PMID: 34817487 PMCID: PMC8711086 DOI: 10.1039/d1cc05565h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Enzymatic noncovalent synthesis enables the spatiotemporal control of multimolecular crowding in cells, thus offering a unique opportunity for modulating cellular functions. This article introduces some representative enzymes and molecular building blocks for generating peptide assemblies as multimolecular crowding in cells, highlights the relevant biomedical applications, such as anticancer therapy, molecular imaging, trafficking proteins, genetic engineering, artificial intracellular filaments, cell morphogenesis, and antibacterial, and briefly discusses the promises of ENS as a multistep molecular process in biology and medicine.
Collapse
Affiliation(s)
- Meihui Yi
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA.
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA.
| | - Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA.
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA.
| |
Collapse
|
47
|
Tong KC, Wan PK, Lok CN, Che CM. Dynamic supramolecular self-assembly of platinum(ii) complexes perturbs an autophagy-lysosomal system and triggers cancer cell death. Chem Sci 2021; 12:15229-15238. [PMID: 34976343 PMCID: PMC8635173 DOI: 10.1039/d1sc02841c] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/17/2021] [Indexed: 12/27/2022] Open
Abstract
Self-assembly of platinum(ii) complexes to form supramolecular structures/nanostructures due to intermolecular ligand π-π stacking and metal-ligand dispersive interactions is widely used to develop functional molecular materials, but the application of such non-covalent molecular interactions has scarcely been explored in medical science. Herein is described the unprecedented biological properties of platinum(ii) complexes relevant to induction of cancer cell death via manifesting such intermolecular interactions. With conjugation of a glucose moiety to the planar platinum(ii) terpyridyl scaffold, the water-soluble complex [Pt(tpy)(C[triple bond, length as m-dash]CArOGlu)](CF3SO3) (1a, tpy = 2,2':6',2''-terpyridine, Glu = glucose) is able to self-assemble into about 100 nm nanoparticles in physiological medium, be taken up by lung cancer cells via energy-dependent endocytosis, and eventually transform into other superstructures distributed in endosomal/lysosomal and mitochondrial compartments apparently following cleavage of the glycosidic linkage. Accompanying the formation of platinum-containing superstructures are increased autophagic vacuole formation, lysosomal membrane permeabilization, and mitochondrial membrane depolarization, as well as anti-tumor activity of 1a in a mouse xenograft model. These findings highlight the dynamic, multi-stage extracellular and intracellular supramolecular self-assembly of planar platinum(ii) complexes driven by modular intermolecular interactions with potential anti-cancer application.
Collapse
Affiliation(s)
- Ka-Chung Tong
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China .,Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong China
| | - Pui-Ki Wan
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China .,Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong China
| | - Chun-Nam Lok
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China .,Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong China
| | - Chi-Ming Che
- State Key Laboratory of Synthetic Chemistry, Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China .,Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong China
| |
Collapse
|
48
|
Wang J, Liu J, Yang Z. Recent advances in peptide-based nanomaterials for targeting hypoxia. NANOSCALE ADVANCES 2021; 3:6027-6039. [PMID: 36133944 PMCID: PMC9418673 DOI: 10.1039/d1na00637a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/01/2021] [Indexed: 06/16/2023]
Abstract
Hypoxia is a prominent feature of many severe diseases such as malignant tumors, ischemic strokes, and rheumatoid arthritis. The lack of oxygen has a paramount impact on angiogenesis, invasion, metastasis, and chemotherapy resistance. The potential of hypoxia as a therapeutic target has been increasingly recognized over the last decade. In order to treat these disease states, peptides have been extensively investigated due to their advantages in safety, target specificity, and tumor penetrability. Peptides can overcome difficulties such as low drug/energy delivery efficiency, hypoxia-induced drug resistance, and tumor nonspecificity. There are three main strategies for targeting hypoxia through peptide-based nanomaterials: (i) using peptide ligands to target cellular environments unique to hypoxic conditions, such as cell surface receptors that are upregulated in cells under hypoxic conditions, (ii) utilizing peptide linkers sensitive to the hypoxic microenvironment that can be cleaved to release therapeutic or diagnostic payloads, and (iii) a combination of the above where targeting peptides will localize the system to a hypoxic environment for it to be selectively cleaved to release its payload, forming a dual-targeting system. This review focuses on recent developments in the design and construction of novel peptide-based hypoxia-targeting nanomaterials, followed by their mechanisms and potential applications in diagnosis and treatment of hypoxic diseases. In addition, we address challenges and prospects of how peptide-based hypoxia-targeting nanomaterials can achieve a wider range of clinical applications.
Collapse
Affiliation(s)
- Jun Wang
- School of Pharmacy, Jining Medical University Rizhao 276800 China
| | - Jing Liu
- School of Pharmacy, Jining Medical University Rizhao 276800 China
| | - Zhongxing Yang
- School of Pharmacy, Jining Medical University Rizhao 276800 China
| |
Collapse
|
49
|
Yang X, Lu H, Tao Y, Zhou L, Wang H. Spatiotemporal Control over Chemical Assembly in Living Cells by Integration of Acid-Catalyzed Hydrolysis and Enzymatic Reactions. Angew Chem Int Ed Engl 2021; 60:23797-23804. [PMID: 34473893 DOI: 10.1002/anie.202109729] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Indexed: 02/04/2023]
Abstract
Spatiotemporal control of chemical assembly in living cells remains challenging. We have now developed an efficient and general platform to precisely control the formation of assemblies in living cells. We introduced an O-[bis(dimethylamino)phosphono]tyrosine protection strategy in the self-assembly motif as the Trojan horse, whereby the programmed precursors resist hydrolysis by phosphatases on and inside cells because the unmasking of the enzymatic cleavage site occurs selectively in the acidic environment of lysosomes. After demonstrating the multistage self-assembly processes in vitro by liquid chromatography/mass spectrometry (LC-MS), cryogenic electron microscopy (Cryo-EM), and circular dichroism (CD), we investigated the formation of site-specific self-assembly in living cells using confocal laser scanning microscopy (CLSM), LC-MS, and biological electron microscopy (Bio-EM). Controlling chemical assembly in living systems spatiotemporally may have applications in supramolecular chemistry, materials science, synthetic biology, and chemical biology.
Collapse
Affiliation(s)
- Xuejiao Yang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou, 310024, Zhejiang Province, China
| | - Honglei Lu
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou, 310024, Zhejiang Province, China
| | - Yinghua Tao
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou, 310024, Zhejiang Province, China
| | - Laicheng Zhou
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou, 310024, Zhejiang Province, China
| | - Huaimin Wang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou, 310024, Zhejiang Province, China.,Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| |
Collapse
|
50
|
Yang X, Lu H, Tao Y, Zhou L, Wang H. Spatiotemporal Control over Chemical Assembly in Living Cells by Integration of Acid‐Catalyzed Hydrolysis and Enzymatic Reactions. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202109729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Xuejiao Yang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province School of Science Westlake University Institute of Natural Sciences Westlake Institute for Advanced Study 18 Shilongshan Road Hangzhou 310024 Zhejiang Province China
| | - Honglei Lu
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province School of Science Westlake University Institute of Natural Sciences Westlake Institute for Advanced Study 18 Shilongshan Road Hangzhou 310024 Zhejiang Province China
| | - Yinghua Tao
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province School of Science Westlake University Institute of Natural Sciences Westlake Institute for Advanced Study 18 Shilongshan Road Hangzhou 310024 Zhejiang Province China
| | - Laicheng Zhou
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province School of Science Westlake University Institute of Natural Sciences Westlake Institute for Advanced Study 18 Shilongshan Road Hangzhou 310024 Zhejiang Province China
| | - Huaimin Wang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province School of Science Westlake University Institute of Natural Sciences Westlake Institute for Advanced Study 18 Shilongshan Road Hangzhou 310024 Zhejiang Province China
- Westlake Laboratory of Life Sciences and Biomedicine School of Life Sciences Westlake University Hangzhou Zhejiang China
| |
Collapse
|