1
|
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
|
2
|
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
|
3
|
Xu W, Qin X, Liu Y, Chen J, Wang Y. Advances in Enzyme-responsive Supramolecular In situ Self-assembled Peptide for Drug Delivery. Curr Drug Deliv 2025; 22:374-386. [PMID: 37496133 DOI: 10.2174/1567201820666230726151607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/17/2023] [Accepted: 06/21/2023] [Indexed: 07/28/2023]
Abstract
Because of low immunogenicity, ease of modification, and inherent biosafety, peptides have been well recognized as vehicles to deliver therapeutic agents to targeted regions with improved pharmacokinetic characteristics. Enzyme-responsive self-assembled peptides (ERSAPs) show superiority over their naive forms due to their enhanced targeting efficacy and long-retention property. In this review, we have summarized recent advances in the therapeutic application of ERSAPs, mainly focusing on their self-therapeutic properties and potential as vehicles to deliver different drugs.
Collapse
Affiliation(s)
- Wentao Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- Cancer Center, Department of Interventional Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiaowen Qin
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yang Liu
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jun Chen
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- Cancer Center, Department of Interventional Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuguang Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
4
|
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
|
5
|
Wang W, Sessler CD, Wang X, Liu J. In Situ Synthesis and Assembly of Functional Materials and Devices in Living Systems. Acc Chem Res 2024; 57:2013-2026. [PMID: 39007720 DOI: 10.1021/acs.accounts.4c00049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Integrating functional materials and devices with living systems enables novel methods for recording, manipulating, or augmenting organisms not accessible by traditional chemical, optical, or genetic approaches. (The term "device" refers to the fundamental components of complex electronic systems, such as transistors, capacitors, conductors, and electrodes.) Typically, these advanced materials and devices are synthesized, either through chemical or physical reactions, outside the biological systems (ex situ) before they are integrated. This is due in part to the more limited repertoire of biocompatible chemical transformations available for assembling functional materials in vivo. Given that most of the assembled bulk materials are impermeable to cell membranes and cannot go through the blood-brain barrier (BBB), the external synthesis poses challenges when trying to interface these materials and devices with cells precisely and in a timely manner and at the micro- and nanoscale─a crucial requirement for modulating cellular functions. In contrast to presynthesis in a separate location, in situ assembly, wherein small molecules or building blocks are directly assembled into functional materials within a biological system at the desired site of action, has offered a potential solution for spatiotemporal and genetic control of material synthesis and assembly. In this Account, we highlight recent advances in spatially and temporally targeted functional material synthesis and assembly in living cells, tissues and animals and provide perspective on how they may enable novel probing, modulation, or augmentation of fundamental biology. We discuss several strategies, starting from the traditional nontargeted methods to targeted assembly of functional materials and devices based on the endogenous markers of the biological system. We then focus on genetically targeted assembly of functional materials, which employs enzymatic catalysis centers expressed in living systems to assemble functional materials in specific molecular-defined cell types. We introduce the recent efforts of our group to modulate membrane capacitance and neuron excitability using in situ synthesized electrically functional polymers in a genetically targetable manner. These advances demonstrate the promise of in situ synthesis and assembly of functional materials and devices, including the optogenetic polymerization developed by our lab, to interface with cells in a cellular- or subcellular-specific manner by incorporating genetic and/or optical control over material assembly. Finally, we discuss remaining challenges, areas for improvement, potential applications to other biological systems, and novel methods for the in situ synthesis of functional materials that could be elevated by incorporating genetic or material design strategies. As researchers expand the toolkit of biocompatible in situ functional material synthetic techniques, we anticipate that these advancements could potentially offer valuable tools for exploring biological systems and developing therapeutic solutions.
Collapse
Affiliation(s)
- Wenbo Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, United States
| | - Chanan D Sessler
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Xiao Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, United States
| |
Collapse
|
6
|
Tan W, Zhang Q, Lee M, Lau W, Xu B. Enzymatic control of intermolecular interactions for generating synthetic nanoarchitectures in cellular environment. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2373045. [PMID: 39011064 PMCID: PMC11249168 DOI: 10.1080/14686996.2024.2373045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/10/2024] [Accepted: 06/23/2024] [Indexed: 07/17/2024]
Abstract
Nanoarchitectonics, as a technology to arrange nano-sized structural units such as molecules in a desired configuration, requires nano-organization, which usually relies on intermolecular interactions. This review briefly introduces the development of using enzymatic reactions to control intermolecular interactions for generating artificial nanoarchitectures in a cellular environment. We begin the discussion with the early examples and uniqueness of enzymatically controlled self-assembly. Then, we describe examples of generating intracellular nanostructures and their relevant applications. Subsequently, we discuss cases of forming nanostructures on the cell surface via enzymatic reactions. Following that, we highlight the use of enzymatic reactions for creating intercellular nanostructures. Finally, we provide a summary and outlook on the promises and future direction of this strategy. Our aim is to give an updated introduction to the use of enzymatic reaction in regulating intermolecular interactions, a phenomenon ubiquitous in biology but relatively less explored by chemists and materials scientists. Our goal is to stimulate new developments in this simple and versatile approach for addressing societal needs.
Collapse
Affiliation(s)
- Weiyi Tan
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Qiuxin Zhang
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Mikki Lee
- Department of Chemistry, Brandeis University, Waltham, MA, USA
- Department of Pharmacy and Pharmaceutical Sciences, National University ofSingapore, Singapore
| | - William Lau
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Shen S, Shao Y, Li C. Different types of cell death and their shift in shaping disease. Cell Death Discov 2023; 9:284. [PMID: 37542066 PMCID: PMC10403589 DOI: 10.1038/s41420-023-01581-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/16/2023] [Accepted: 07/26/2023] [Indexed: 08/06/2023] Open
Abstract
Cell death is the irreversible stop of life. It is also the basic physiological process of all organisms which involved in the embryonic development, organ maintenance and autoimmunity of the body. In recent years, we have gained more comprehension of the mechanism in cell death and have basically clarified the different types of "programmed cell death", such as apoptosis, necroptosis, autophagy, and pyroptosis, and identified some key genes in these processes. However, in these previous studies, the conversion between different cell death modes and their application in diseases are rarely explored. To sum up, although many valued discoveries have been discovered in the field of cell death in recent years, there are still many unknown problems to be solved in this field. Facts have proved that cell death is a very complex game, and a series of core players have the ability to destroy the delicate balance of the cell environment, from survival to death, from anti-inflammatory to pro-inflammatory. With the thorough research of the complex regulatory mechanism of cell death, there will certainly be exciting new research in this field in the next few years. The sake of this paper is to emphasize the complex mechanism of overturning the balance between different cell fates and provide relevant theoretical basis for the connection between cell death transformation and disease treatment in the future.
Collapse
Affiliation(s)
- Sikou Shen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Yina Shao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
10
|
Liang X, Zhang Y, Zhou J, Bu Z, Liu J, Zhang K. Tumor microenvironment-triggered intratumoral in situ construction of theranostic supramolecular self-assembly. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
|
11
|
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
|
12
|
Kim BJ. Enzyme-Instructed Self-Assembly of Peptides: From Concept to Representative Applications. Chem Asian J 2022; 17:e202200094. [PMID: 35213091 DOI: 10.1002/asia.202200094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/23/2022] [Indexed: 11/11/2022]
Abstract
Enzyme-instructed self-assembly, integrating enzymatic reaction and molecular self-assembly, has drawn noticeable attention over the last decade with the intension of being used in valuable applications. Recent advances in the field allow it possible to spatiotemporally control peptide self-assembly in cellular milieu, broadening the potential applications of peptide assemblies to cancer therapy and subcellular delivery. In this minireview, the concept of enzyme-instructed self-assembly of peptide, containing enzymatic trigger and spatiotemporal control, is described. Representative applications in cells are also discussed, followed by outlook on the field of enzyme-instructed self-assembly.
Collapse
Affiliation(s)
- Beom Jin Kim
- University of Ulsan, Chemistry, 12, Techno Industrial Complex-ro, 55 beon-gil, 4776, Ulsan, KOREA, REPUBLIC OF
| |
Collapse
|
13
|
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
|
14
|
Yi M, Guo J, He H, Tan W, Harmon N, Ghebreyessus K, Xu B. Phosphobisaromatic motifs enable rapid enzymatic self-assembly and hydrogelation of short peptides. SOFT MATTER 2021; 17:8590-8594. [PMID: 34545895 PMCID: PMC8600407 DOI: 10.1039/d1sm01221e] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Enzyme-instructed self-assembly (EISA) and hydrogelation is a versatile approach for generating soft materials. Most of the substrates for alkaline phosphatase catalysed EISA utilize phosphotyrosine (pTyr) as the enzymatic trigger for EISA and hydrogelation. Here we show the first example of phosphonaphthyl (pNP) and phosphobiphenyl (pBP) motifs acting as faster enzymatic triggers than phosphotyrosine for EISA and hydrogelation. This work illustrates novel enzyme triggers for rapid enzymatic self-assembly and hydrogelation.
Collapse
Affiliation(s)
- Meihui Yi
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA.
| | - Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA.
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA.
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA.
| | - Nya Harmon
- Department of Chemistry and Biochemistry, Hampton University, Hampton, VA, 23668, USA
| | - Kesete Ghebreyessus
- Department of Chemistry and Biochemistry, Hampton University, Hampton, VA, 23668, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA.
| |
Collapse
|
15
|
Liu N, Zhu L, Li Z, Liu W, Sun M, Zhou Z. In situ self-assembled peptide nanofibers for cancer theranostics. Biomater Sci 2021; 9:5427-5436. [PMID: 34319316 DOI: 10.1039/d1bm00782c] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Self-assembled nanofibers hold tremendous promise for cancer theranostics owing to their in situ assembly, spatiotemporal responsiveness, and diverse bioactivity. Herein, this review summarizes the recent advances of self-assembled peptide nanofibers and their applications in biological systems, focusing on the dynamic process of capturing cancer cells from the outside-in. (1) In situ self-assembly in response to pathological or physiological changes. (2) Diverse functions at different locations of tumors, such as forming thrombus in tumor vasculature, constructing a barrier on the cancer cell membrane, and disrupting the cancer organelles. Of note, with the assembly/aggregation induced residence (AIR) effect, the nanofibers could form a drug depot in situ for sustained release of chemotherapeutic drugs to increase their local concentration and prolong the residence time. Finally, perspectives toward future directions and challenges are presented to further understand and expand this exciting field.
Collapse
Affiliation(s)
- Ning Liu
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Lianghan Zhu
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Zhaoting Li
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Wenlong Liu
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Minjie Sun
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Zhanwei Zhou
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, P. R. China.
| |
Collapse
|
16
|
Zheng D, Liu J, Xie L, Wang Y, Ding Y, Peng R, Cui M, Wang L, Zhang Y, Zhang C, Yang Z. Enzyme-instructed and mitochondria-targeting peptide self-assembly to efficiently induce immunogenic cell death. Acta Pharm Sin B 2021; 12:2740-2750. [PMID: 35755291 PMCID: PMC9214332 DOI: 10.1016/j.apsb.2021.07.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/02/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022] Open
Abstract
Immunogenic cell death (ICD) plays a major role in cancer immunotherapy by stimulating specific T cell responses and restoring the antitumor immune system. However, effective type II ICD inducers without biotoxicity are still very limited. Herein, a tentative drug- or photosensitizer-free strategy was developed by employing enzymatic self-assembly of the peptide F-pY-T to induce mitochondrial oxidative stress in cancer cells. Upon dephosphorylation catalyzed by alkaline phosphatase overexpressed on cancer cells, the peptide F-pY-T self-assembled to form nanoparticles, which were subsequently internalized. These affected the morphology of mitochondria and induced serious reactive oxygen species production, causing the ICD characterized by the release of danger-associated molecular patterns (DAMPs). DAMPs enhanced specific immune responses by promoting the maturation of DCs and the intratumoral infiltration of tumor-specific T cells to eradicate tumor cells. The dramatic immunotherapeutic capacity could be enhanced further by combination therapy of F-pY-T and anti-PD-L1 agents without visible biotoxicity in the main organs. Thus, our results revealed an alternative strategy to induce efficient ICD by physically promoting mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Debin Zheng
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Jingfei Liu
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Limin Xie
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Yuhan Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Yinghao Ding
- College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Rong Peng
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Min Cui
- Department of Human Anatomy, Nanjing Medical University, Nanjing 211166, China
| | - Ling Wang
- College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Yongjie Zhang
- Department of Human Anatomy, Nanjing Medical University, Nanjing 211166, China
- Corresponding authors. Tel./fax: +86 25 86869485 (Yongjie Zhang); +86 22 23502875 (Chunqiu Zhang and Zhimou Yang).
| | - Chunqiu Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin 300071, China
- Corresponding authors. Tel./fax: +86 25 86869485 (Yongjie Zhang); +86 22 23502875 (Chunqiu Zhang and Zhimou Yang).
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin 300071, China
- Corresponding authors. Tel./fax: +86 25 86869485 (Yongjie Zhang); +86 22 23502875 (Chunqiu Zhang and Zhimou Yang).
| |
Collapse
|
17
|
Yang D, Kim BJ, He H, Xu B. Enzymatically Forming Cell Compatible Supramolecular Assemblies of Tryptophan-Rich Short Peptides. Pept Sci (Hoboken) 2021; 113:e24173. [PMID: 35445163 PMCID: PMC9017786 DOI: 10.1002/pep2.24173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/04/2020] [Indexed: 10/27/2023]
Abstract
Here we report a new type of tryptophan-rich short peptides, which act as hydrogelators, form supramolecular assemblies via enzymatic dephosphorylation, and exhibit cell compatibility. The facile synthesis of the peptides starts with the production of phosphotyrosine, then uses solid phase peptide synthesis (SPPS) to build the phosphopeptides that contain multiple tryptophan residues. Besides exhibiting excellent solubility, these phosphopeptides, unlike the previously reported cytotoxic phenylalanine-rich phosphopeptides, are largely compatible toward mammalian cells. Our preliminary mechanistic study suggests that the tryptophan-rich peptides, instead of forming pericellular assemblies, largely accumulate in lysosomes. Such lysosomal localization may account for their cell compatibility. Moreover, these tryptophan-rich peptides are able to transiently reduce the cytotoxicity of phenylalanine-rich peptide assemblies. This rather unexpected result implies that tryptophan may act as a useful aromatic building block for developing cell compatible supramolecular assemblies for soft materials and find applications for protecting cells from cytotoxic peptide assemblies.
Collapse
Affiliation(s)
- Dongsik Yang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Beom Jin Kim
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| |
Collapse
|
18
|
Chen J, Zhao Y, Yao Q, Gao Y. Pathological environment directed in situ peptidic supramolecular assemblies for nanomedicines. Biomed Mater 2021; 16:022011. [PMID: 33630754 DOI: 10.1088/1748-605x/abc2e9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peptidic self-assembly provides a powerful method to build biomedical materials with integrated functions. In particular, pathological environment instructed peptidic supramolecular have gained great progress in treating various diseases. Typically, certain pathology related factors convert hydrophilic precursors to corresponding more hydrophobic motifs to assemble into supramolecular structures. Herein, we would like to review the recent progress of nanomedicines based on the development of instructed self-assembly against several specific disease models. Firstly we introduce the cancer instructed self-assembly. These assemblies have exhibited great inhibition efficacy, as well as enhanced imaging contrast, against cancer models both in vitro and in vivo. Then we discuss the infection instructed peptidic self-assembly. A number of different molecular designs have demonstrated the potential antibacterial application with satisfied efficiency for peptidic supramolecular assemblies. Further, we discuss the application of instructed peptidic self-assembly for other diseases including neurodegenerative disease and vaccine. The assemblies have succeeded in down-regulating abnormal Aβ aggregates and immunotherapy. In summary, the self-assembly precursors are typical two-component molecules with (1) a self-assembling motif and (2) a cleavable trigger responsive to the pathological environment. Upon cleavage, the self-assembly occurs selectively in pathological loci whose targeting capability is independent from active targeting. Bearing the novel targeting regime, we envision that the pathological conditions instructed peptidic self-assembly will lead a paradigm shift on biomedical materials.
Collapse
Affiliation(s)
- Jiali Chen
- CAS Center of Excellence for Nanoscience, Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yan Zhao
- CAS Center of Excellence for Nanoscience, Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Qingxin Yao
- CAS Center of Excellence for Nanoscience, Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yuan Gao
- CAS Center of Excellence for Nanoscience, Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
19
|
Kim BJ, Fang Y, He H, Xu B. Trypsin-Instructed Self-Assembly on Endoplasmic Reticulum for Selectively Inhibiting Cancer Cells: Dedicated to Professor George M. Whitesides on the occasion of his 80th birthday. Adv Healthc Mater 2021; 10:e2000416. [PMID: 32342647 PMCID: PMC7725443 DOI: 10.1002/adhm.202000416] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/07/2020] [Indexed: 12/15/2022]
Abstract
Selectively targeting the endoplasmic reticulum (ER) of cancer cells, though promising a new strategy for cancer therapy, remains underdeveloped. Enzyme-instructed self-assembly (EISA) is emerging as a promising approach for selectively targeting ER of cancer cells. This work reports an easily accessible branched peptide that consists of a D-tetrapeptide backbone and a branch with the sequence of KYDKKKKDG (K: lysine; Y: tyrosine; D: aspratic acid; G: glycine), being an EISA substrate of typsin-1 (PRSS1), selectively inhibits cancer cells. Depending on the type of cells, the level of PRSS1 expression dictates the cytotoxicity of the branched peptide. Moreover, immunostaining and fluorescent imaging reveal that PRSS1 overexpresses on the ER of a high-grade serous ovarian cancer cell line (OVSAHO). The overexpression of PRSS1 renders the branched peptide to exhibit high selectivity against OVSAHO by the in situ formation of the peptide assemblies on the ER of OVSAHO cells, which causes ER stress and eventual cell death. This work, illustrating trypsin-guided EISA for inhibiting cancer cells by enzymatic reaction on ER for the first time, offers a new way to target the subcellular organelles of cancer cells for potential cancer therapy.
Collapse
Affiliation(s)
- Beom Jin Kim
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Yu Fang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| |
Collapse
|
20
|
Regulation of tumor microenvironment for pancreatic cancer therapy. Biomaterials 2021; 270:120680. [PMID: 33588140 DOI: 10.1016/j.biomaterials.2021.120680] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/04/2021] [Accepted: 01/14/2021] [Indexed: 02/05/2023]
Abstract
Pancreatic cancer (PC) is one kind of the most lethal malignancies worldwide, owing to its insidious symptoms, early metastases, and negative responses to current therapies. With an increasing understanding of pathology, the tumor microenvironment (TME) plays a significant role in ineffective treatment and poor prognosis of PC. Thus, a growing number of studies have focused on whether components of the TME could be effective targets for PC therapy. Biomaterials have been widely applied in cancer therapy, and numerous organic or inorganic biomaterials for TME regulation have been developed to inhibit the growth and metastasis of PC, as well as reverse therapeutic resistance. In this review, we discuss various biomaterials utilized to treat PC based on different components of the TME, including, but not limited to, extracellular matrix (ECM), abnormal tumor vascularization, and tumor-associated immune cells, as well as other unconventional therapeutic strategies. Besides, the perspectives on the underlying future of theranostic nanomedicines for PC therapy are also presented.
Collapse
|
21
|
Ren H, Li R, Chen Z, Li L, Wang H. Modification Methods and Applications of Self-Assembly Peptides. CHINESE J ORG CHEM 2021. [DOI: 10.6023/cjoc202104020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
22
|
Shy AN, Wang H, Feng Z, Xu B. Heterotypic Supramolecular Hydrogels Formed by Noncovalent Interactions in Inflammasomes. Molecules 2020; 26:E77. [PMID: 33375296 PMCID: PMC7795891 DOI: 10.3390/molecules26010077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 01/04/2023] Open
Abstract
The advance of structural biology has revealed numerous noncovalent interactions between peptide sequences in protein structures, but such information is less explored for developing peptide materials. Here we report the formation of heterotypic peptide hydrogels by the two binding motifs revealed by the structures of an inflammasome. Specifically, conjugating a self-assembling motif to the positively or negatively charged peptide sequence from the ASCPYD filaments of inflammasome produces the solutions of the peptides. The addition of the peptides of the oppositely charged and complementary peptides to the corresponding peptide solution produces the heterotypic hydrogels. Rheology measurement shows that ratios of the complementary peptides affect the viscoelasticity of the resulted hydrogel. Circular dichroism indicates that the addition of the complementary peptides results in electrostatic interactions that modulate self-assembly. Transmission electron microscopy reveals that the ratio of the complementary peptides controls the morphology of the heterotypic peptide assemblies. This work illustrates a rational, biomimetic approach that uses the structural information from the protein data base (PDB) for developing heterotypic peptide materials via self-assembly.
Collapse
Affiliation(s)
| | | | | | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA; (A.N.S.); (H.W.); (Z.F.)
| |
Collapse
|
23
|
Abstract
Enzymatic reactions and noncovalent (i.e., supramolecular) interactions are two fundamental nongenetic attributes of life. Enzymatic noncovalent synthesis (ENS) refers to a process where enzymatic reactions control intermolecular noncovalent interactions for spatial organization of higher-order molecular assemblies that exhibit emergent properties and functions. Like enzymatic covalent synthesis (ECS), in which an enzyme catalyzes the formation of covalent bonds to generate individual molecules, ENS is a unifying theme for understanding the functions, morphologies, and locations of molecular ensembles in cellular environments. This review intends to provide a summary of the works of ENS within the past decade and emphasize ENS for functions. After comparing ECS and ENS, we describe a few representative examples where nature uses ENS, as a rule of life, to create the ensembles of biomacromolecules for emergent properties/functions in a myriad of cellular processes. Then, we focus on ENS of man-made (synthetic) molecules in cell-free conditions, classified by the types of enzymes. After that, we introduce the exploration of ENS of man-made molecules in the context of cells by discussing intercellular, peri/intracellular, and subcellular ENS for cell morphogenesis, molecular imaging, cancer therapy, and other applications. Finally, we provide a perspective on the promises of ENS for developing molecular assemblies/processes for functions. This review aims to be an updated introduction for researchers who are interested in exploring noncovalent synthesis for developing molecular science and technologies to address societal needs.
Collapse
Affiliation(s)
- Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Meihui Yi
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Adrianna N Shy
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| |
Collapse
|
24
|
Jayapaul J, Schröder L. Probing Reversible Guest Binding with Hyperpolarized 129Xe-NMR: Characteristics and Applications for Cucurbit[ n]urils. Molecules 2020; 25:E957. [PMID: 32093412 PMCID: PMC7070628 DOI: 10.3390/molecules25040957] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 01/01/2023] Open
Abstract
Cucurbit[n]urils (CB[n]s) are a family of macrocyclic host molecules that find various applications in drug delivery, molecular switching, and dye displacement assays. The CB[n]s with n = 5-7 have also been studied with 129Xe-NMR. They bind the noble gas with a large range of exchange rates. Starting with insights from conventional direct detection of bound Xe, this review summarizes recent achievements with chemical exchange saturation transfer (CEST) detection of efficiently exchanging Xe in various CB[n]-based supramolecular systems. Unprecedented sensitivity has been reached by combining the CEST method with hyperpolarized Xe, the production of which is also briefly described. Applications such as displacement assays for enzyme activity detection and rotaxanes as emerging types of Xe biosensors are likewise discussed in the context of biomedical applications and pinpoint future directions for translating this field to preclinical studies.
Collapse
Affiliation(s)
| | - Leif Schröder
- Molecular Imaging, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany;
| |
Collapse
|
25
|
Restu WK, Yamamoto S, Nishida Y, Ienaga H, Aoi T, Maruyama T. Hydrogel formation by short D-peptide for cell-culture scaffolds. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110746. [PMID: 32279773 DOI: 10.1016/j.msec.2020.110746] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/27/2020] [Accepted: 02/14/2020] [Indexed: 10/25/2022]
Abstract
The present study reports that a short oligopeptide D-P1, consisting of only five D-amino acids, self-assembled into entangled nanofibers to form a hydrogel that functioned as a scaffold for cell cultures. D-P1 (Ac-D-Phe-D-Phe-D-Phe-Gly-D-Lys) gelated aqueous buffer solution and water at a minimum gelation concentration of 0.5 wt%. The circular dichroism (CD) measurements demonstrated the formation of a β-sheet structure in the self-assembly of D-P1. We investigated the gelation properties and CD spectra of both the D- and L-forms of the oligopeptide, and found only a minimal difference between them. The D-P1 hydrogel was resistant to a protease, whereas the L-P1 hydrogel was rapidly degraded. Both oligopeptides exhibited nontoxic properties to human cancer cells and embryoid bodies (EBs) derived from human-induced pluripotent stem cells. Additionally, we succeeded in forming spheroids of HeLa cells on the D-P1 hydrogel, which indicates the potential of this hydrogel for 3-dimensional cell culture.
Collapse
Affiliation(s)
- Witta Kartika Restu
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan; Research Center for Chemistry, Indonesian Institute of Sciences, Kawasan Puspiptek Serpong, Tangerang Selatan, Banten 15314, Indonesia
| | - Shota Yamamoto
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan
| | - Yuki Nishida
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodaicho, Nada-ku, Kobe 657-8501, Japan
| | - Hirotoshi Ienaga
- Department of iPS cell Applications, Graduate School of Medicine, Kobe University, 7-5-1 Kusunokicho, Chuo-ku, Kobe 650-0017, Japan
| | - Takashi Aoi
- Department of iPS cell Applications, Graduate School of Medicine, Kobe University, 7-5-1 Kusunokicho, Chuo-ku, Kobe 650-0017, 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
|
26
|
Abstract
Enzymatic reactions and self-assembly are two fundamental attributes of cells. It is not surprising that one can use enzyme-instructed self-assembly (EISA)-the integration of enzymatic transformation and molecular self-assembly-to modulate the emergent properties of supramolecular assemblies for controlling cell behaviors. The exploration of EISA for developing cancer therapy and imaging has made considerable progress over the last five years. In this Topical Review, we discuss these exciting results and the future promise of EISA. After describing several key studies to illustrate the progress of EISA in developing cancer therapy, we discuss the use of EISA for molecular imaging. Then, we give the outlook of EISA for developing supramolecular anticancer medicine that inhibits multiple hallmark capabilities of cancer.
Collapse
Affiliation(s)
- Beom Jin Kim
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| |
Collapse
|
27
|
|
28
|
Shy AN, Kim BJ, Xu B. Enzymatic Noncovalent Synthesis of Supramolecular Soft Matter for Biomedical Applications. MATTER 2019; 1:1127-1147. [PMID: 32104791 PMCID: PMC7043404 DOI: 10.1016/j.matt.2019.09.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Enzymatic noncovalent synthesis (ENS), a process that integrates enzymatic reactions and supramolecular (i.e., noncovalent) interactions for spatial organization of higher-order molecular assemblies, represents an emerging research area at the interface of physical and biological sciences. This review provides a few representative examples of ENS in the context of supramolecular soft matter. After a brief comparison of enzymatic covalent and noncovalent synthesis, we discuss ENS of man-made molecules for generating supramolecular nanostructures (e.g., supramolecular hydrogels) in cell-free conditions. Then, we introduce ENS in a cellular environment. To illustrate the unique merits for applications, we discuss intercellular, peri- or intracellular, and subcellular ENS for cell morphogenesis, molecular imaging, cancer therapy, and targeted delivery. Finally, we provide an outlook on the potential of ENS. We hope that this review offers a new perspective for scientists who develop supramolecular soft matter to address societal needs at various frontiers.
Collapse
Affiliation(s)
- Adrianna N. Shy
- Department of Chemistry, Brandeis University, Waltham, MA 02453, USA
| | - Beom Jin Kim
- Department of Chemistry, Brandeis University, Waltham, MA 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, Waltham, MA 02453, USA
| |
Collapse
|
29
|
Yao Q, Huang Z, Liu D, Chen J, Gao Y. Enzyme-Instructed Supramolecular Self-Assembly with Anticancer Activity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1804814. [PMID: 30444545 DOI: 10.1002/adma.201804814] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/18/2018] [Indexed: 06/09/2023]
Abstract
Cancer remains one of the leading causes of death, which has continuously stimulated the development of numerous functional biomaterials with anticancer activities. Herein is reviewed one recent trend of biomaterials focusing on the advances in enzyme-instructed supramolecular self-assembly (EISA) with anticancer activity. EISA relies on enzymatic transformations to convert designed small-molecular precursors into corresponding amphiphilic residues that can form assemblies in living systems. EISA has shown some advantages in controlling cell fate from three aspects. 1) Based on the abnormal activity of specific enzymes, EISA can differentiate cancer cells from normal cells. In contrast to the classical ligand-receptor recognition, the targeting capability of EISA relies on dynamic control of the self-assembly process. 2) The interactions between EISA and cellular components directly disrupt cellular processes or pathways, resulting in cell death phenotypes. 3) EISA spatiotemporally controls the distribution of therapeutic agents, which boosts drug delivery efficiency. Therefore, with regard to the development of EISA, the aim is to provide a perspective on the future directions of research into EISA as anticancer theranostics.
Collapse
Affiliation(s)
- Qingxin Yao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Zhentao Huang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Dongdong Liu
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Jiali Chen
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Yuan Gao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
30
|
Jeena MT, Jeong K, Go EM, Cho Y, Lee S, Jin S, Hwang SW, Jang JH, Kang CS, Bang WY, Lee E, Kwak SK, Kim S, Ryu JH. Heterochiral Assembly of Amphiphilic Peptides Inside the Mitochondria for Supramolecular Cancer Therapeutics. ACS NANO 2019; 13:11022-11033. [PMID: 31508938 DOI: 10.1021/acsnano.9b02522] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Self-assembly of peptides containing both l- and d-isomers often results in nanostructures with enhanced properties compared to their enantiomeric analogues, such as faster kinetics of formation, higher mechanical strength, and enzymatic stability. However, occurrence and consequences of the heterochiral assembly in the cellular microenvironment are unknown. In this study, we monitored heterochiral assembly of amphiphilic peptides inside the cell, specifically mitochondria of cancer cells, resulting in nanostructures with refined morphological and biological properties owing to the superior interaction between the backbones of opposite chirality. We have designed a mitochondria penetrating tripeptide containing a diphenyl alanine building unit, named as Mito-FF due to their mitochondria targeting ability. The short peptide amphiphile, Mito-FF co-assembled with its mirror pair, Mito-ff, induced superfibrils of around 100 nm in diameter and 0.5-1 μm in length, while enantiomers formed only narrow fibers of 10 nm in diameter. The co-administration of Mito-FF and Mito-ff in the cell induced drastic mitochondrial disruption both in vitro and in vivo. The experimental and theoretical analyses revealed that pyrene capping played a major role in inducing superfibril morphology upon the co-assembly of racemic peptides. This work shows the impact of chirality control over the peptide self-assembly inside the biological system, thus showing a potent strategy for fabricating promising peptide biomaterials by considering chirality as a design modality.
Collapse
Affiliation(s)
| | - Keunsoo Jeong
- Center for Theragnosis , Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea
| | | | - Yuri Cho
- Center for Theragnosis , Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology , Korea University , Seoul 02841 , Republic of Korea
| | - Seokyung Lee
- Center for Theragnosis , Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea
| | | | - Suk-Won Hwang
- KU-KIST Graduate School of Converging Science and Technology , Korea University , Seoul 02841 , Republic of Korea
| | - Joo Hee Jang
- Division of Applied RI , Korea Institute of Radiological and Medical Sciences , Seoul 01812 , Republic of Korea
| | - Chi Soo Kang
- Division of Applied RI , Korea Institute of Radiological and Medical Sciences , Seoul 01812 , Republic of Korea
| | - Woo-Young Bang
- School of Materials Science and Engineering , Gwangju Institute of Science and Technology , Gwangju 61005 , Republic of Korea
| | - Eunji Lee
- School of Materials Science and Engineering , Gwangju Institute of Science and Technology , Gwangju 61005 , Republic of Korea
| | | | - Sehoon Kim
- Center for Theragnosis , Korea Institute of Science and Technology , Seoul 02792 , Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology , Korea University , Seoul 02841 , Republic of Korea
| | | |
Collapse
|
31
|
Feng Z, Han X, Wang H, Tang T, Xu B. Enzyme-Instructed Peptide Assemblies Selectively Inhibit Bone Tumors. Chem 2019; 5:2442-2449. [PMID: 31552305 PMCID: PMC6758912 DOI: 10.1016/j.chempr.2019.06.020] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alkaline phosphatases (ALP) contribute to immunosuppression in solid tumors, but they, unfortunately, are "undruggable". Here we report enzyme-instructed assembly of peptides for selectively inhibiting the tumors that overexpress ALP. We developed a precursor with two parts; an amphiphilic, self-assembling peptides joined to a hydrophilic block (i.e., tyrosine phosphate). ALP, overexpressed on and in osteosarcoma cancer cells (e.g., Saos-2), cleaves the phosphates from the tyrosine residue of the precursor and triggers the self-assembly of the resulting peptides. Being selectively formed on and inside the cancer cells, the peptide assemblies induce the cancer cell death and efficiently inhibit the tumor growth in an orthotopic osteosarcoma mice model without harming normal organs. Accordingly, the peptide assemblies significantly improve the survival ratio of metastatic tumor bearing mice. Without relying on inhibiting ALP, this approach integrates enzyme reaction and molecular self-assembly for generating peptide fibrils as potential anticancer therapeutics.
Collapse
Affiliation(s)
- Zhaoqianqi Feng
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
- These authors contributed equally to this work
| | - Xiuguo Han
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- These authors contributed equally to this work
| | - Huaimin Wang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
- Lead Contact
| |
Collapse
|
32
|
Wang H, Feng Z, Xu B. Assemblies of Peptides in a Complex Environment and their Applications. Angew Chem Int Ed Engl 2019; 58:10423-10432. [PMID: 30903643 PMCID: PMC6656613 DOI: 10.1002/anie.201814552] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Indexed: 01/28/2023]
Abstract
Using peptide assemblies with emergent properties to achieve elaborate functions has attracted increasing attention in recent years. Besides tailoring the self-assembly of peptides in vitro, peptide research is advancing into a new and exciting frontier: the rational design of peptide assemblies (or their derivatives) for biological functions in a complex environment. This Minireview highlights recent developments in peptide assemblies and their applications in biological systems. After introducing the unique merits of peptide assemblies, we discuss the recent progress in designing peptides (or peptide derivatives) for self-assembly with conformational control. Then, we describe biological functions of peptide assemblies, with an emphasis on approach-instructed assembly for spatiotemporal control of peptide assemblies, in the cellular context. Finally, we discuss the future promises and challenges of this exciting area of chemistry.
Collapse
Affiliation(s)
- Huaimin Wang
- Department of chemistry, Brandeis University, 415 South St, Waltham, MA 02454, USA
| | - Zhaoqianqi Feng
- 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
|
33
|
Huang Z, Yao Q, Wei S, Chen J, Gao Y. Enzyme-Instructed Self-assembly in Biological Milieu for Theranostics Purpose. Curr Med Chem 2019; 26:1351-1365. [DOI: 10.2174/0929867324666170921104010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 06/19/2017] [Accepted: 06/19/2017] [Indexed: 12/12/2022]
Abstract
Precision medicine is in an urgent need for public healthcare. Among the past
several decades, the flourishing development in nanotechnology significantly advances
the realization of precision nanomedicine. Comparing to well-documented nanoparticlebased
strategy, in this review, we focus on the strategy using enzyme instructed selfassembly
(EISA) in biological milieu for theranostics purpose. In principle, the design of
small molecules for EISA requires two aspects: (1) the substrate of enzyme of interest;
and (2) self-assembly potency after enzymatic conversion. This strategy has shown its irreplaceable
advantages in nanomedicne, specifically for cancer treatments and Vaccine
Adjuvants. Interestingly, all the reported examples rely on only one kind of enzymehydrolase.
Therefore, we envision that the application of EISA strategy just begins and
will lead to a new paradigm in nanomedicine.
Collapse
Affiliation(s)
- Zhentao Huang
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing 100190, China
| | - Qingxin Yao
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing 100190, China
| | - Simin Wei
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing 100190, China
| | - Jiali Chen
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing 100190, China
| | - Yuan Gao
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, No. 11 Zhongguancun Beiyitiao, Beijing 100190, China
| |
Collapse
|
34
|
Wang H, Feng Z, Xu B. Assemblies of Peptides in a Complex Environment and their Applications. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201814552] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Huaimin Wang
- Department of Chemistry Brandeis University 415 South St Waltham MA 02454 USA
| | - Zhaoqianqi Feng
- 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
|
35
|
Feng Z, Wang H, Xu B. Instructed Assembly of Peptides for Intracellular Enzyme Sequestration. J Am Chem Soc 2018; 140:16433-16437. [PMID: 30452246 DOI: 10.1021/jacs.8b10542] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Liquid-like droplets of biomacromolecules are emerging as a fundamental mechanism of cellular signaling, but designing synthetic mimics to form such membraneless organelles remains unexplored. Here we report the use of supramolecular assemblies of small peptides, as a mimic of biomacromolecular condensates, for intracellular sequestration of enzymes on endoplasmic reticulum (ER). Specifically, integrating a short peptide with naproxen (a nonsteroidal anti-inflammatory drug (NSAID) and a ligand of cyclooxygenase-2 (COX-2)) generates an enzymatic substrate that acts as a precursor for instructed assembly. Slowly dephosphorylating the precursors by phosphatases forms the corresponding hydrogelators in a cellular environment, which results in the supramolecular assemblies on ER. Consisting of the precursor and the hydrogelator molecules, the assemblies enable the sequestration of COX-2 and protein-tyrosine phosphatase 1B (PTP1B) on ER. Further structure-activity investigation reveals that the colocalization of COX-2 and PTP1B relies on the NSAID motif, the phosphotyrosine, and the enzymatic dephosphorylation of the precursor. This work, for the first time, illustrates the use of supramolecular processes for associating enzymes in cells and may provide insights for understanding intracellular liquid condensates and a new strategy for modulating protein-protein interactions.
Collapse
Affiliation(s)
- Zhaoqianqi Feng
- Department of Chemistry , Brandeis University , 415 South Street , Waltham , Massachusetts 02454 , United States
| | - Huaimin Wang
- Department of Chemistry , Brandeis University , 415 South Street , Waltham , Massachusetts 02454 , United States
| | - Bing Xu
- Department of Chemistry , Brandeis University , 415 South Street , Waltham , Massachusetts 02454 , United States
| |
Collapse
|
36
|
Advances in transformable drug delivery systems. Biomaterials 2018; 178:546-558. [DOI: 10.1016/j.biomaterials.2018.03.056] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/21/2018] [Accepted: 03/31/2018] [Indexed: 12/14/2022]
|
37
|
Cong Y, Qiao ZY, Wang H. Molecular Self-Assembly Constructed in Physiological Conditions for Cancer Diagnosis and Therapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yong Cong
- CAS Center for Excellence in Nanoscience; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; No. 11 Beiyitiao, Zhongguancun Beijing 100190 China
| | - Zeng-Ying Qiao
- CAS Center for Excellence in Nanoscience; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; No. 11 Beiyitiao, Zhongguancun Beijing 100190 China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology; No. 11 Beiyitiao, Zhongguancun Beijing 100190 China
| |
Collapse
|
38
|
Fan Q, Ji Y, Wang J, Wu L, Li W, Chen R, Chen Z. Self-assembly behaviours of peptide-drug conjugates: influence of multiple factors on aggregate morphology and potential self-assembly mechanism. ROYAL SOCIETY OPEN SCIENCE 2018; 5:172040. [PMID: 29765659 PMCID: PMC5936924 DOI: 10.1098/rsos.172040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/13/2018] [Indexed: 05/30/2023]
Abstract
Peptide-drug conjugates (PDCs) as self-assembly prodrugs have the unique and specific features to build one-component nanomedicines. Supramolecular structure based on PDCs could form various morphologies ranging from nanotube, nanofibre, nanobelt to hydrogel. However, the assembly process of PDCs is too complex to predict or control. Herein, we investigated the effects of extrinsic factors on assembly morphology and the possible formation of nanostructures based on PDCs. To this end, we designed a PDC consisting of hydrophobic drug (S)-ketoprofen (Ket) and valine-glutamic acid dimeric repeats peptide (L-VEVE) to study their assembly behaviour. Our results showed that the critical assembly concentration of Ket-L-VEVE was 0.32 mM in water to form various nanostructures which experienced from micelle, nanorod, nanofibre to nanoribbon. The morphology was influenced by multiple factors including molecular design, assembly time, pH and hydrogen bond inhibitor. On the basis of experimental results, we speculated the possible assembly mechanism of Ket-L-VEVE. The π-π stacking interaction between Ket molecules could serve as an anchor, and hydrogen bonded-induced β-sheets and hydrophilic/hydrophobic balance between L-VEVE peptide play structure-directing role in forming filament-like or nanoribbon morphology. This work provides a new sight to rationally design and precisely control the nanostructure of PDCs based on aromatic fragment.
Collapse
Affiliation(s)
| | | | | | | | | | - Rui Chen
- Authors for correspondence: Rui Chen e-mail:
| | - Zhipeng Chen
- Authors for correspondence: Zhipeng Chen e-mail:
| |
Collapse
|
39
|
Du W, Hu X, Wei W, Liang G. Intracellular Peptide Self-Assembly: A Biomimetic Approach for in Situ Nanodrug Preparation. Bioconjug Chem 2018; 29:826-837. [PMID: 29316785 DOI: 10.1021/acs.bioconjchem.7b00798] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Most nanodrugs are preprepared by encapsulating or loading the drugs with nanocarriers (e.g., dendrimers, liposomes, micelles, and polymeric nanoparticles). However, besides the low bioavailability and fast excretion of the nanodrugs in vivo, nanocarriers often exhibit in vitro and in vivo cytotoxicity, oxidative stress, and inflammation. Self-assembly is a ubiquitous process in biology where it plays important roles and underlies the formation of a wide variety of complex biological structures. Inspired by some cellular nanostructures (e.g., actin filaments, microtubules, vesicles, and micelles) in biological systems which are formed via molecular self-assembly, in recent decades, scientists have utilized self-assembly of oligomeric peptide under specific physiological or pathological environments to in situ construct nanodrugs for lesion-targeted therapies. On one hand, peptide-based nanodrugs always have some excellent intrinsic chemical (specificity, intrinsic bioactivity, biodegradability) and physical (small size, conformation) properties. On the other hand, stimuli-regulated intracellular self-assembly of nanodrugs is quite an efficient way to accumulate the drugs in lesion location and can realize an in situ slow release of the drugs. In this review article, we provided an overview on recent design principles for intracellular peptide self-assembly and illustrate how these principles have been applied for the in situ preparation of nanodrugs at the lesion location. In the last part, we list some challenges underlying this strategy and their possible solutions. Moreover, we envision the future possible theranostic applications of this strategy.
Collapse
Affiliation(s)
- Wei Du
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry , University of Science and Technology of China , 96 Jinzhai Road , Hefei , Anhui 230026 , China
| | - Xiaomu Hu
- Department of Medicinal Chemistry, School of Pharmacy , The Fourth Military Medical University , Changle West Road 169 , Xi'an , Shanxi 710032 , China
| | - Weichen Wei
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry , University of Science and Technology of China , 96 Jinzhai Road , Hefei , Anhui 230026 , China
| | - Gaolin Liang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry , University of Science and Technology of China , 96 Jinzhai Road , Hefei , Anhui 230026 , China
| |
Collapse
|
40
|
Huang Z, Gao Y. Enzyme-Instructed Self-assembly of Small Peptides In Vivo for Biomedical Application. IN VIVO SELF-ASSEMBLY NANOTECHNOLOGY FOR BIOMEDICAL APPLICATIONS 2018. [DOI: 10.1007/978-981-10-6913-0_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
41
|
Abstract
In this review, supramolecular catalysis refers to the integration of the catalytic process with molecular self-assembly driven by noncovalent interactions, and dynamic assemblies are the assemblies that form and dissipate reversibly. Cells extensively employ supramolecular catalysis and dynamic assemblies for controlling their complex functions. The dynamic generation of supramolecular assemblies of small molecules has made considerable progress in the last decade, though the disassembly processes remain underexplored. Here, we discuss the regulation of dynamic assemblies via self-assembly and disassembly processes for therapeutics and diagnostics. We first briefly introduce the self-assembly and disassembly processes in the context of cells, which provide the rationale for designing approaches to control the assemblies. Then, we describe recent advances in designing and regulating the self-assembly and disassembly of small molecules, especially for molecular imaging and anticancer therapeutics. Finally, we provide a perspective on future directions of the research on supramolecular catalysis and dynamic assemblies for medicine.
Collapse
Affiliation(s)
- Zhaoqianqi Feng
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | | | | | | |
Collapse
|
42
|
Feng Z, Wang H, Chen X, Xu B. Self-Assembling Ability Determines the Activity of Enzyme-Instructed Self-Assembly for Inhibiting Cancer Cells. J Am Chem Soc 2017; 139:15377-15384. [PMID: 28990765 PMCID: PMC5669277 DOI: 10.1021/jacs.7b07147] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
![]()
Enzyme-instructed
self-assembly (EISA) represents a dynamic continuum
of supramolecular nanostructures that selectively inhibits cancer
cells via simultaneously targeting multiple hallmark capabilities
of cancer, but how to design the small molecules for EISA from the
vast molecular space remains an unanswered question. Here we show
that the self-assembling ability of small molecules controls the anticancer
activity of EISA. Examining the EISA precursor analogues consisting
of an N-capped d-tetrapeptide, a phosphotyrosine residue,
and a diester or a diamide group, we find that, regardless of the
stereochemistry and the regiochemistry of their tetrapeptidic backbones,
the anticancer activities of these precursors largely match their
self-assembling abilities. Additional mechanistic studies confirm
that the assemblies of the small peptide derivatives result in cell
death, accompanying significant rearrangement of cytoskeletal proteins
and plasma membranes. These results imply that the diester or diamide
derivatives of the d-tetrapeptides self-assemble pericellularly,
as well as intracellularly, to result in cell death. As the first
case to correlate thermodynamic properties (e.g., self-assembling
ability) of small molecules with the efficacy of a molecule process
against cancer cells, this work provides an important insight for
developing a molecular dynamic continuum for potential cancer therapy,
as well as understanding the cytotoxicity of pathogenic assemblies.
Collapse
Affiliation(s)
- Zhaoqianqi Feng
- Department of Chemistry, Brandeis University , 415 South Street, Waltham, Massachusetts 02453, United States
| | - Huaimin Wang
- Department of Chemistry, Brandeis University , 415 South Street, Waltham, Massachusetts 02453, United States
| | - Xiaoyi Chen
- 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
|
43
|
Zhou J, Li J, Du X, Xu B. Supramolecular biofunctional materials. Biomaterials 2017; 129:1-27. [PMID: 28319779 PMCID: PMC5470592 DOI: 10.1016/j.biomaterials.2017.03.014] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/09/2017] [Accepted: 03/10/2017] [Indexed: 12/27/2022]
Abstract
This review discusses supramolecular biofunctional materials, a novel class of biomaterials formed by small molecules that are held together via noncovalent interactions. The complexity of biology and relevant biomedical problems not only inspire, but also demand effective molecular design for functional materials. Supramolecular biofunctional materials offer (almost) unlimited possibilities and opportunities to address challenging biomedical problems. Rational molecular design of supramolecular biofunctional materials exploit powerful and versatile noncovalent interactions, which offer many advantages, such as responsiveness, reversibility, tunability, biomimicry, modularity, predictability, and, most importantly, adaptiveness. In this review, besides elaborating on the merits of supramolecular biofunctional materials (mainly in the form of hydrogels and/or nanoscale assemblies) resulting from noncovalent interactions, we also discuss the advantages of small peptides as a prevalent molecular platform to generate a wide range of supramolecular biofunctional materials for the applications in drug delivery, tissue engineering, immunology, cancer therapy, fluorescent imaging, and stem cell regulation. This review aims to provide a brief synopsis of recent achievements at the intersection of supramolecular chemistry and biomedical science in hope of contributing to the multidisciplinary research on supramolecular biofunctional materials for a wide range of applications. We envision that supramolecular biofunctional materials will contribute to the development of new therapies that will ultimately lead to a paradigm shift for developing next generation biomaterials for medicine.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Jie Li
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Xuewen Du
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA.
| |
Collapse
|