1
|
Lin S, Li T, Zhang B, Wang P. Taurine rescues intervertebral disc degeneration by activating mitophagy through the PINK1/Parkin pathway. Biochem Biophys Res Commun 2024; 739:150587. [PMID: 39182353 DOI: 10.1016/j.bbrc.2024.150587] [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: 07/26/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Intervertebral disc degeneration (IDD) is a common cause of low back pain and disability. Recent studies have highlighted the critical role of mitochondrial dysfunction in the progression of IDD. In this study, we investigated the therapeutic potential of taurine in delaying IDD through the activation of mitophagy via the PINK1/Parkin pathway. Our in vitro and in vivo experiments demonstrate that taurine treatment significantly enhances mitophagy, reduces oxidative stress, delays cell senescence, and promotes the removal of damaged mitochondria in nucleus pulposus cells (NPC). Additionally, taurine-mediated activation of the PINK1/Parkin pathway leads to improved mitochondrial homeostasis and slows the progression of disc degeneration. These findings provide new insights into the protective effects of taurine and highlight its potential as a therapeutic agent for IDD.
Collapse
Affiliation(s)
- Shengyuan Lin
- Department of Orthopaedics, Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, 410199, China.
| | - Tao Li
- Department of Orthopaedics, Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, 410199, China
| | - Bin Zhang
- Department of Orthopaedics, Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, 410199, China
| | - Peng Wang
- Department of Orthopaedics, Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, 410199, China
| |
Collapse
|
2
|
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
|
3
|
Cai X, Xu W, Ren C, Zhang L, Zhang C, Liu J, Yang C. Recent progress in quantitative analysis of self-assembled peptides. EXPLORATION (BEIJING, CHINA) 2024; 4:20230064. [PMID: 39175887 PMCID: PMC11335468 DOI: 10.1002/exp.20230064] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/05/2023] [Indexed: 08/24/2024]
Abstract
Self-assembled peptides have been among the important biomaterials due to its excellent biocompatibility and diverse functions. Over the past decades, substantial progress and breakthroughs have been made in designing self-assembled peptides with multifaceted biomedical applications. The techniques for quantitative analysis, including imaging-based quantitative techniques, chromatographic technique and computational approach (molecular dynamics simulation), are becoming powerful tools for exploring the structure, properties, biomedical applications, and even supramolecular assembly processes of self-assembled peptides. However, a comprehensive review concerning these quantitative techniques remains scarce. In this review, recent progress in techniques for quantitative investigation of biostability, cellular uptake, biodistribution, self-assembly behaviors of self-assembled peptide etc., are summarized. Specific applications and roles of these techniques are highlighted in detail. Finally, challenges and outlook in this field are concluded. It is believed that this review will provide technical guidance for researchers in the field of peptide-based materials and pharmaceuticals, and facilitate related research for newcomers in this field.
Collapse
Affiliation(s)
- Xiaoyao Cai
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Wei Xu
- Department of PathologyCharacteristic Medical Center of Chinese People's Armed Police ForcesTianjinP. R. China
| | - Chunhua Ren
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Liping Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Congrou Zhang
- Metabolomics and Analytics Center, Leiden Academic Centre of Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Cuihong Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| |
Collapse
|
4
|
Yang Z, Wu Y, Liu H, He L, Deng X. AMYGNN: A Graph Convolutional Neural Network-Based Approach for Predicting Amyloid Formation from Polypeptides. J Chem Inf Model 2024; 64:1751-1762. [PMID: 38408296 DOI: 10.1021/acs.jcim.3c02035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
There has been an increasing interest in the use of amyloids for constructing various functional materials. The design of amyloid-associated functional materials requires the identification of the core peptide sequences as the fundamental building block. The existing computational methods are limited in terms of delineating polypeptides, the typical non-Euclidean structural data, and they fail to capture the dynamic interactions between amino acids due to ignoring the contextual information from surrounding amino acids. Here, we first propose the use of a state-of-the-art graph convolutional neural network for predicting the trends of amyloid formation from specific peptide sequences (AMYGNN) by abstracting each polypeptide as a graph, in which the constituting amino acids are viewed as nodes and edges characterizing the connections between pairs of amino acids are established when they meet a given distance threshold (Cα-Cα ≤ 5 Å). Our model achieves high performance with accuracy (0.9208), G-mean (0.9203), MCC (0.8417), and F1 (0.9235) in determining the characteristic peptide sequences to form amyloid. 32 of 534 crucial amino acid properties that greatly contribute to the formation of amyloids are ascertained, and the β-folding-like graph structure of a polypeptide is believed to be essential for the formation of amyloid. Our model enables the mapping of polypeptides with underlying interactions between amino acids and provides a quick and precise predictive framework for directing the construction of amyloid-associated functional materials.
Collapse
Affiliation(s)
- Zuojun Yang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Yuhan Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Hao Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Li He
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Xiaoyuan Deng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
5
|
He H, Yin J, Li M, Dessai CVP, Yi M, Teng X, Zhang M, Li Y, Du Z, Xu B, Cheng JX. Mapping enzyme activity in living systems by real-time mid-infrared photothermal imaging of nitrile chameleons. Nat Methods 2024; 21:342-352. [PMID: 38191931 PMCID: PMC11165695 DOI: 10.1038/s41592-023-02137-x] [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: 02/15/2023] [Accepted: 11/17/2023] [Indexed: 01/10/2024]
Abstract
Simultaneous spatial mapping of the activity of multiple enzymes in a living system can elucidate their functions in health and disease. However, methods based on monitoring fluorescent substrates are limited. Here, we report the development of nitrile (C≡N)-tagged enzyme activity reporters, named nitrile chameleons, for the peak shift between substrate and product. To image these reporters in real time, we developed a laser-scanning mid-infrared photothermal imaging system capable of imaging the enzymatic substrates and products at a resolution of 300 nm. We show that when combined, these tools can map the activity distribution of different enzymes and measure their relative catalytic efficiency in living systems such as cancer cells, Caenorhabditis elegans, and brain tissues, and can be used to directly visualize caspase-phosphatase interactions during apoptosis. Our method is generally applicable to a broad category of enzymes and will enable new analyses of enzymes in their native context.
Collapse
Affiliation(s)
- Hongjian He
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | - Jiaze Yin
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | - Mingsheng Li
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| | - Chinmayee Vallabh Prabhu Dessai
- Photonics Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Meihui Yi
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Xinyan Teng
- Photonics Center, Boston University, Boston, MA, USA
- Department of Chemistry, Boston University, Boston, MA, USA
| | - Meng Zhang
- Photonics Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Yueming Li
- Photonics Center, Boston University, Boston, MA, USA
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Zhiyi Du
- Photonics Center, Boston University, Boston, MA, USA
- Department of Chemistry, Boston University, Boston, MA, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Ji-Xin Cheng
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, USA.
- Photonics Center, Boston University, Boston, MA, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Department of Chemistry, Boston University, Boston, MA, USA.
| |
Collapse
|
6
|
Pilz M, Cavelius P, Qoura F, Awad D, Brück T. Lipopeptides development in cosmetics and pharmaceutical applications: A comprehensive review. Biotechnol Adv 2023; 67:108210. [PMID: 37460047 DOI: 10.1016/j.biotechadv.2023.108210] [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: 01/26/2023] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/25/2023]
Abstract
Lipopeptides are surface active, natural products of bacteria, fungi and green-blue algae origin, having diverse structures and functionalities. In analogy, a number of chemical synthesis techniques generated new designer lipopeptides with desirable features and functions. Lipopetides are self-assembly guided, supramolecular compounds which have the capacity of high-density presentation of the functional epitopes at the surface of the nanostructures. This feature contributes to their successful application in several industry sectors, including food, feed, personal care, and pharmaceutics. In this comprehensive review, the novel class of ribosomally synthesized lipopeptides is introduced alongside the more commonly occuring non-ribosomal lipopeptides. We highlight key representatives of the most researched as well as recently described lipopeptide families, with emphasis on structural features, self-assembly and associated functions. The common biological, chemical and hybrid production routes of lipopeptides, including prominent analogues and derivatives are also discussed. Furthermore, genetic engineering strategies aimed at increasing lipopeptide yields, diversity and biological activity are summarized and exemplified. With respect to application, this work mainly details the potential of lipopeptides in personal care and cosmetics industry as cleansing agents, moisturizer, anti-aging/anti-wrinkling, skin whitening and preservative agents as well as the pharmaceutical industry as anitimicrobial agents, vaccines, immunotherapy, and cancer drugs. Given that this review addresses human applications, we conclude on the topic of safety of lipopeptide formulations and their sustainable production.
Collapse
Affiliation(s)
- Melania Pilz
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany
| | - Philipp Cavelius
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany
| | - Farah Qoura
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany
| | - Dania Awad
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany.
| | - Thomas Brück
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany.
| |
Collapse
|
7
|
Santulli G, Kansakar U, Varzideh F, Mone P, Jankauskas SS, Lombardi A. Functional Role of Taurine in Aging and Cardiovascular Health: An Updated Overview. Nutrients 2023; 15:4236. [PMID: 37836520 PMCID: PMC10574552 DOI: 10.3390/nu15194236] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Taurine, a naturally occurring sulfur-containing amino acid, has attracted significant attention in recent years due to its potential health benefits. Found in various foods and often used in energy drinks and supplements, taurine has been studied extensively to understand its impact on human physiology. Determining its exact functional roles represents a complex and multifaceted topic. We provide an overview of the scientific literature and present an analysis of the effects of taurine on various aspects of human health, focusing on aging and cardiovascular pathophysiology, but also including athletic performance, metabolic regulation, and neurological function. Additionally, our report summarizes the current recommendations for taurine intake and addresses potential safety concerns. Evidence from both human and animal studies indicates that taurine may have beneficial cardiovascular effects, including blood pressure regulation, improved cardiac fitness, and enhanced vascular health. Its mechanisms of action and antioxidant properties make it also an intriguing candidate for potential anti-aging strategies.
Collapse
Affiliation(s)
- Gaetano Santulli
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Urna Kansakar
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| | - Fahimeh Varzideh
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Pasquale Mone
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; (F.V.); (P.M.)
| | - Stanislovas S. Jankauskas
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| | - Angela Lombardi
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (S.S.J.); (A.L.)
| |
Collapse
|
8
|
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: 8] [Impact Index Per Article: 4.0] [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
|
9
|
Bigo Simon A, Fores JR, Criado-Gonzalez M, Blandin L, Runser JY, Senger B, Fleith G, Schmutz M, Schurhammer R, Chaumont A, Schaaf P, Combet J, Jierry L. Mechanistic Insights into Hyaluronic Acid Induced Peptide Nanofiber Organization in Supramolecular Hydrogels. Biomacromolecules 2023; 24:3794-3805. [PMID: 37535455 DOI: 10.1021/acs.biomac.3c00445] [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: 08/05/2023]
Abstract
Composite hydrogels composed of low-molecular-weight peptide self-assemblies and polysaccharides are gaining great interest as new types of biomaterials. Interactions between polysaccharides and peptide self-assemblies are well reported, but a molecular picture of their impact on the resulting material is still missing. Using the phosphorylated tripeptide precursor Fmoc-FFpY (Fmoc, fluorenylmethyloxycarbonyl; F, phenylalanine; Y, tyrosine; p, phosphate group), we investigated how hyaluronic acid (HA) influences the enzyme-assisted self-assembly of Fmoc-FFY generated in situ in the presence of alkaline phosphatase (AP). In the absence of HA, Fmoc-FFY peptides are known to self-assemble in nanometer thick and micrometer long fibers. The presence of HA leads to the spontaneous formation of bundles of several micrometers thickness. Using fluorescence recovery after photobleaching (FRAP), we find that in the bundles both (i) HA colocalizes with the peptide self-assemblies and (ii) its presence in the bundles is highly dynamic. The attractive interaction between negatively charged peptide fibers and negatively charged HA chains is explained through molecular dynamic simulations that show the existence of hydrogen bonds. Whereas the Fmoc-FFY peptide self-assembly itself is not affected by the presence of HA, this polysaccharide organizes the peptide nanofibers in a nematic phase visible by small-angle X-ray scattering (SAXS). The mean distance d between the nanofibers decreases by increasing the HA concentration c, but remains always larger than the diameter of the peptide nanofibers, indicating that they do not interact directly with each other. At a high enough HA concentration, the nematic organization transforms into an ordered 2D hexagonal columnar phase with a nanofiber distance d of 117 Å. Depletion interaction generated by the polysaccharides can explain the experimental power law variation d ∼ c - 1 / 4 and is responsible for the bundle formation and organization. Such behavior is thus suggested for the first time on nano-objects using polymers partially adsorbing on self-assembled peptide nanofibers.
Collapse
Affiliation(s)
- Alexis Bigo Simon
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
- Université de Strasbourg, Faculté de Chimie, UMR7140, 1 rue Blaise Pascal, 67008 Strasbourg Cedex, France
| | - Jennifer Rodon Fores
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
- Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000 Strasbourg, France
| | - Miryam Criado-Gonzalez
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
- Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000 Strasbourg, France
| | - Lucille Blandin
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
| | - Jean-Yves Runser
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
- Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000 Strasbourg, France
| | - Bernard Senger
- Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000 Strasbourg, France
| | - Guillaume Fleith
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
| | - Marc Schmutz
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
| | - Rachel Schurhammer
- Université de Strasbourg, Faculté de Chimie, UMR7140, 1 rue Blaise Pascal, 67008 Strasbourg Cedex, France
| | - Alain Chaumont
- Université de Strasbourg, Faculté de Chimie, UMR7140, 1 rue Blaise Pascal, 67008 Strasbourg Cedex, France
| | - Pierre Schaaf
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
- Institut National de la Santé et de la Recherche Médicale, INSERM Unité 1121, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000 Strasbourg, France
- Université de Strasbourg, 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
| | - Jérôme Combet
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
| | - Loïc Jierry
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034 Strasbourg Cedex 2, BP 84047, France
| |
Collapse
|
10
|
Marciano Y, Nayeem N, Dave D, Ulijn RV, Contel M. N-Acetylation of Biodegradable Supramolecular Peptide Nanofilaments Selectively Enhances Their Proteolytic Stability for Targeted Delivery of Gold-Based Anticancer Agents. ACS Biomater Sci Eng 2023; 9:3379-3389. [PMID: 37192486 PMCID: PMC10699682 DOI: 10.1021/acsbiomaterials.3c00312] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Peptide materials are promising for various biomedical applications; however, a significant concern is their lack of stability and rapid degradation in vivo due to non-specific proteolysis. For materials specifically designed to respond to disease-specific proteases, it would be desirable to retain high susceptibility to target proteases while minimizing the impact of non-specific proteolysis. We describe N-terminal acetylation as a simple synthetic modification of amphiphilic self-assembling peptides that contain an MMP-9-cleavable segment and form soluble, nanoscale filaments. We found that the N-terminus capping of these peptides did not significantly impact their self-assembly behavior, critical aggregation concentration, or ability to encapsulate hydrophobic payloads. By contrast, their proteolytic stability in human plasma (especially for anionic peptide sequences) was considerably increased while susceptibility to hydrolysis by MMP-9 was retained when compared to non-acetylated peptides, especially during the first 12 h. We note, however, that due to the longer time scale required for in vitro studies (72 h), non-specific proteolysis of both anionic acetylated peptides leads to similar activity in vitro despite differing MMP-9 kinetics during the early stages. Overall, the enhanced stability against non-specific proteases, combined with the ability of these nanofilaments to enhance the effectiveness of gold-based drugs toward cancerous cells compared to healthy cells, brings these acetylated peptide filaments a step closer toward clinical translation.
Collapse
Affiliation(s)
- Yaron Marciano
- Department of Chemistry, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Advanced Science Research Center at The Graduate Center of the City University of New York, 85 Saint Nicholas Terrace, New York, NY 10031, USA
- PhD Program in Chemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Nazia Nayeem
- Department of Chemistry, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- PhD Program Biology, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Dhwanit Dave
- Advanced Science Research Center at The Graduate Center of the City University of New York, 85 Saint Nicholas Terrace, New York, NY 10031, USA
- PhD Program in Chemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, New York, NY 10065, USA
| | - Rein V. Ulijn
- Advanced Science Research Center at The Graduate Center of the City University of New York, 85 Saint Nicholas Terrace, New York, NY 10031, USA
- PhD Program in Chemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, New York, NY 10065, USA
| | - Maria Contel
- Department of Chemistry, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- PhD Program in Chemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- PhD Program Biology, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| |
Collapse
|
11
|
Yang S, Wang M, Wang T, Sun M, Huang H, Shi X, Duan S, Wu Y, Zhu J, Liu F. Self-assembled short peptides: Recent advances and strategies for potential pharmaceutical applications. Mater Today Bio 2023; 20:100644. [PMID: 37214549 PMCID: PMC10199221 DOI: 10.1016/j.mtbio.2023.100644] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/10/2023] [Accepted: 04/23/2023] [Indexed: 05/24/2023] Open
Abstract
Self-assembled short peptides have intrigued scientists due to the convenience of synthesis, good biocompatibility, low toxicity, inherent biodegradability and fast response to change in the physiological environment. Therefore, it is necessary to present a comprehensive summary of the recent advances in the last decade regarding the construction, route of administration and application of self-assembled short peptides based on the knowledge on their unique and specific ability of self-assembly. Herein, we firstly explored the molecular mechanisms of self-assembly of short peptides, such as non-modified amino acids, as well as Fmoc-modified, N-functionalized, and C-functionalized peptides. Next, cell penetration, fusion, and peptide targeting in peptide-based drug delivery were characterized. Then, the common administration routes and the potential pharmaceutical applications (drug delivery, antibacterial activity, stabilizers, imaging agents, and applications in bioengineering) of peptide drugs were respectively summarized. Last but not least, some general conclusions and future perspectives in the relevant fields were briefly listed. Although with certain challenges, great opportunities are offered by self-assembled short peptides to the fascinating area of drug development.
Collapse
Affiliation(s)
- Shihua Yang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
- Department of Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110102, China
| | - Mingge Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Tianye Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
- Department of Anus and Intestine Surgery, The First Hospital of Dalian Medical University, Dalian, 116000, China
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hanwei Huang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
- Department of Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110102, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Shijie Duan
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
- Department of Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110102, China
| | - Ying Wu
- Department of Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110102, China
| | - Jiaming Zhu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110001, China
- Department of Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110102, China
| |
Collapse
|
12
|
Harrison K, Mackay AS, Kambanis L, Maxwell JWC, Payne RJ. Synthesis and applications of mirror-image proteins. Nat Rev Chem 2023; 7:383-404. [PMID: 37173596 DOI: 10.1038/s41570-023-00493-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 05/15/2023]
Abstract
The homochirality of biomolecules in nature, such as DNA, RNA, peptides and proteins, has played a critical role in establishing and sustaining life on Earth. This chiral bias has also given synthetic chemists the opportunity to generate molecules with inverted chirality, unlocking valuable new properties and applications. Advances in the field of chemical protein synthesis have underpinned the generation of numerous 'mirror-image' proteins (those comprised entirely of D-amino acids instead of canonical L-amino acids), which cannot be accessed using recombinant expression technologies. This Review seeks to highlight recent work on synthetic mirror-image proteins, with a focus on modern synthetic strategies that have been leveraged to access these complex biomolecules as well as their applications in protein crystallography, drug discovery and the creation of mirror-image life.
Collapse
Affiliation(s)
- Katriona Harrison
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Angus S Mackay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Lucas Kambanis
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Joshua W C Maxwell
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia.
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
13
|
Hamley IW. Self-Assembly, Bioactivity, and Nanomaterials Applications of Peptide Conjugates with Bulky Aromatic Terminal Groups. ACS APPLIED BIO MATERIALS 2023; 6:384-409. [PMID: 36735801 PMCID: PMC9945136 DOI: 10.1021/acsabm.2c01041] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The self-assembly and structural and functional properties of peptide conjugates containing bulky terminal aromatic substituents are reviewed with a particular focus on bioactivity. Terminal moieties include Fmoc [fluorenylmethyloxycarbonyl], naphthalene, pyrene, naproxen, diimides of naphthalene or pyrene, and others. These provide a driving force for self-assembly due to π-stacking and hydrophobic interactions, in addition to the hydrogen bonding, electrostatic, and other forces between short peptides. The balance of these interactions leads to a propensity to self-assembly, even for conjugates to single amino acids. The hybrid molecules often form hydrogels built from a network of β-sheet fibrils. The properties of these as biomaterials to support cell culture, or in the development of molecules that can assemble in cells (in response to cellular enzymes, or otherwise) with a range of fascinating bioactivities such as anticancer or antimicrobial activity, are highlighted. In addition, applications of hydrogels as slow-release drug delivery systems and in catalysis and other applications are discussed. The aromatic nature of the substituents also provides a diversity of interesting optoelectronic properties that have been demonstrated in the literature, and an overview of this is also provided. Also discussed are coassembly and enzyme-instructed self-assembly which enable precise tuning and (stimulus-responsive) functionalization of peptide nanostructures.
Collapse
|
14
|
Tan W, Zhang Q, Hong P, Xu B. A Self-Assembling Probe for Imaging the States of Golgi Apparatus in Live Single Cells. Bioconjug Chem 2022; 33:1983-1988. [PMID: 35312281 PMCID: PMC9489815 DOI: 10.1021/acs.bioconjchem.2c00084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Despite the enormous progress in genomics and proteomics, it is still challenging to assess the states of organelles in living cells with high spatiotemporal resolution. Based on our recent finding of enzyme-instructed self-assembly of a thiophosphopeptide that targets the Golgi Apparatus (GA) instantly, we use the thiophosphopeptide, which is enzymatically responsive and redox active, as an integrative probe for revealing the state of the GA of live cells at the single cell level. By imaging the probe in the GA of live cells over time, our results show that the accumulation of the probe at the GA depends on cell types. By comparison to a conventional Golgi probe, this self-assembling probe accumulates at the GA much faster and are sensitive to the expression of alkaline phosphatases. In addition, subtle changes of the fluorophore results in slightly different GA responses. This work illustrates a novel class of active molecular probes that combine enzyme-instructed self-assembly and redox reaction for high-resolution imaging of the states of subcellular organelles over a large area and extended times.
Collapse
Affiliation(s)
- Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA
| | - Qiuxin Zhang
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA
| | - Pengyu Hong
- Department of Computer Science, Brandeis University, 415 South St., Waltham, MA 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA
| |
Collapse
|
15
|
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
|
16
|
Thermoresponsive Polymer Assemblies: From Molecular Design to Theranostics Application. Prog Polym Sci 2022. [DOI: 10.1016/j.progpolymsci.2022.101578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Yuan Y, Bulte JWM. Enzyme-mediated intratumoral self-assembly of nanotheranostics for enhanced imaging and tumor therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1786. [PMID: 35229485 PMCID: PMC9437863 DOI: 10.1002/wnan.1786] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/26/2021] [Accepted: 02/07/2022] [Indexed: 05/09/2023]
Abstract
Enzyme-mediated intratumoral self-assembled (EMISA) nanotheranostics represent a new class of smart agents for combined imaging and therapy of cancer. Cancer cells overexpress various enzymes that are essential for high metabolism, fast proliferation, and tissue invasion and metastasis. By conjugating small molecules that contain an enzyme-specific cleavage site to appropriate chemical linkers, it is possible to induce self-assembly of nanostructures in tumor cells having the target enzyme. This approach of injecting small theranostic molecules that eventually become larger nanotheranostics in situ avoids some of the major limitations that are encountered when injecting larger, pre-assembled nanotheranostics. The advantage of EMISA nanotheranostics include the avoidance of nonspecific uptake and rapid clearance by phagocytic cells, increased cellular accumulation, reduced drug efflux and prolonged cellular exposure time, all of which lead to an amplified imaging signal and therapeutic efficacy. We review here the different approaches that can be used for preparing EMISA-based organic, inorganic, or organic/inorganic hybrid nanotheranostics based on noncovalent interactions and/or covalent bonding. Imaging examples are shown for fluorescence imaging, nuclear imaging, photoacoustic imaging, Raman imaging, computed tomography imaging, bioluminescent imaging, and magnetic resonance imaging. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Biology-Inspired Nanomaterials > Peptide-Based Structures.
Collapse
Affiliation(s)
- Yue Yuan
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui, China
| | - Jeff W. M. Bulte
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Localized Enzyme-Assisted Self-Assembly of low molecular weight hydrogelators. Mechanism, applications and perspectives. Adv Colloid Interface Sci 2022; 304:102660. [PMID: 35462266 DOI: 10.1016/j.cis.2022.102660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/28/2022] [Accepted: 03/31/2022] [Indexed: 01/31/2023]
Abstract
Nature uses systems of high complexity coordinated by the precise spatial and temporal control of associated processes, working from the molecular to the macroscopic scale. This living organization is mainly ensured by enzymatic actions. Herein, we review the concept of Localized Enzyme-Assisted Self-Assembly (LEASA). It is defined and presented as a straightforward and insightful strategy to achieve high levels of control in artificial systems. Indeed, the use of immobilized enzymes to drive self-assembly events leads not only to the local formation of supramolecular structures but also to tune their kinetics and their morphologies. The possibility to design tailored complex systems taking advantage of self-assembled networks through their inherent and emergent properties offers new perspectives for the design of novel, more adaptable materials. As a result, some applications have already been developed and are gathered in this review. Finally, challenges and perspectives of LEASA are introduced and discussed.
Collapse
|
19
|
Zhao S, Qiao X, Chen M, Li Y, Wang X, Xu Z, Wu Y, Luo X. d-Amino Acid-Based Antifouling Peptides for the Construction of Electrochemical Biosensors Capable of Assaying Proteins in Serum with Enhanced Stability. ACS Sens 2022; 7:1740-1746. [PMID: 35616064 DOI: 10.1021/acssensors.2c00518] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The susceptibility of peptides to proteolytic degradation in human serum significantly hindered the potential application of peptide-based antifouling biosensors for long-term assaying of clinical samples. Herein, a robust antifouling biosensor with enhanced stability was constructed based on peptides composed of d-amino acids (d-peptide) with prominent proteolytic resistance. The electrode was electropolymerized with poly(3,4-ehtylenedioxythiophene) and electrodeposited with Au nanoparticles (AuNPs), and the d-peptide was then immobilized onto the AuNPs, and a typical antibody specific for immunoglobulin M (IgM) was immobilized. Because of the effect of d-amino acids, the d-peptide-modified electrode surface showed prominent antifouling capability and high tolerance to enzymatic hydrolysis. Moreover, the d-peptide-modified electrode exhibited much stronger long-term stability, as well as antifouling ability in human serum than the electrode modified with normal peptides. The electrochemical biosensor exhibited a sensitive response to IgM linearly within the range of 100 pg mL-1 to 1.0 μg mL-1 and a very low detection limit down to 37 pg mL-1, and it was able to detect IgM in human serum with good accuracy. This work provided a new strategy to develop robust peptide-based biosensors to resist the proteolytic degradation for practical application in complex clinical samples.
Collapse
Affiliation(s)
- Shuju Zhao
- State Key Laboratory Base of Eco-chemical Engineering, Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science & Technology, Qingdao 266042, China
| | - Xiujuan Qiao
- State Key Laboratory Base of Eco-chemical Engineering, Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science & Technology, Qingdao 266042, China
| | - Min Chen
- State Key Laboratory Base of Eco-chemical Engineering, Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science & Technology, Qingdao 266042, China
| | - Yanxin Li
- State Key Laboratory Base of Eco-chemical Engineering, Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science & Technology, Qingdao 266042, China
| | - Xin Wang
- State Key Laboratory Base of Eco-chemical Engineering, Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science & Technology, Qingdao 266042, China
| | - Zhenying Xu
- State Key Laboratory Base of Eco-chemical Engineering, Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science & Technology, Qingdao 266042, China
| | - Yumin Wu
- State Key Laboratory Base of Eco-chemical Engineering, Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science & Technology, Qingdao 266042, China
| | - Xiliang Luo
- State Key Laboratory Base of Eco-chemical Engineering, Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science & Technology, Qingdao 266042, China
| |
Collapse
|
20
|
Abstract
Supramolecular assemblies are essential components of living organisms. Cellular scaffolds, such as the cytoskeleton or the cell membrane, are formed via secondary interactions between proteins or lipids and direct biological processes such as metabolism, proliferation and transport. Inspired by nature’s evolution of function through structure formation, a range of synthetic nanomaterials has been developed in the past decade, with the goal of creating non-natural supramolecular assemblies inside living mammalian cells. Given the intricacy of biological pathways and the compartmentalization of the cell, different strategies can be employed to control the assembly formation within the highly crowded, dynamic cellular environment. In this Review, we highlight emerging molecular design concepts aimed at creating precursors that respond to endogenous stimuli to build nanostructures within the cell. We describe the underlying reaction mechanisms that can provide spatial and temporal control over the subcellular formation of synthetic nanostructures. Showcasing recent advances in the development of bioresponsive nanomaterials for intracellular self-assembly, we also discuss their impact on cellular function and the challenges associated with establishing structure–bioactivity relationships, as well as their relevance for the discovery of novel drugs and imaging agents, to address the shortfall of current solutions to pressing health issues. ![]()
Creating artificial nanostructures inside living cells requires the careful design of molecules that can transform into active monomers within a complex cellular environment. This Review explores the recent development of bioresponsive precursors for the controlled formation of intracellular supramolecular assemblies.
Collapse
|
21
|
Zhang Q, Tan W, Xu B. Enzymatic Noncovalent Synthesis for Targeting Subcellular Organelles. Chempluschem 2022; 87:e202200060. [PMID: 35420712 PMCID: PMC9508291 DOI: 10.1002/cplu.202200060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/23/2022] [Indexed: 11/09/2022]
Abstract
Enzymatic noncovalent synthesis (ENS) exploits enzymatic reactions to produce spatially organized higher-order supramolecular assemblies that modulate cellular processes. While ENS is a general mechanism to create higher-order assemblies of proteins for diverse cellular functions, the exploration of ENS of other bioactive molecules, such as peptides or small organic molecules, is rather limited. Since ENS generates non-diffusive supramolecular assemblies locally, it provides a unique approach to targeting subcellular organelles. In this Review, we highlight the recent progress of the application of ENS of peptide assemblies for targeting subcellular organelles. After a brief introduction of the concept of ENS, we introduce the case of generating artificial filaments by ENS in cell cytosol, then discuss the use of ENS for targeting endoplasmic reticulum, mitochondria, Golgi apparatus, and lysosomes, and finally we describe the targeting of nucleus by ENS. We hope to illustrate the promise of ENS, as a localized molecular process in an open system, for understanding diseases, controlling cell behaviors, and developing new therapeutics.
Collapse
Affiliation(s)
- Qiuxin Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Weiyi Tan
- 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
|
22
|
Wang W, Wang S. Cell-based biocomposite engineering directed by polymers. LAB ON A CHIP 2022; 22:1042-1067. [PMID: 35244136 DOI: 10.1039/d2lc00067a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Biological cells such as bacterial, fungal, and mammalian cells always exploit sophisticated chemistries and exquisite micro- and nano-structures to execute life activities, providing numerous templates for engineering bioactive and biomorphic materials, devices, and systems. To transform biological cells into functional biocomposites, polymer-directed cell surface engineering and intracellular functionalization have been developed over the past two decades. Polymeric materials can be easily adopted by various cells through polymer grafting or in situ hydrogelation and can successfully bridge cells with other functional materials as interfacial layers, thus achieving the manufacture of advanced biocomposites through bioaugmentation of living cells and transformation of cells into templated materials. This review article summarizes the recent progress in the design and construction of cell-based biocomposites by polymer-directed strategies. Furthermore, the applications of cell-based biocomposites in broad fields such as cell research, biomedicine, and bioenergy are discussed. Last, we provide personal perspectives on challenges and future trends in this interdisciplinary area.
Collapse
Affiliation(s)
- Wenshuo Wang
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
- Shandong Energy Institute, Qingdao, 266101, China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
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
|
24
|
Shen Y, Wang Y, Hamley IW, Qi W, Su R, He Z. Chiral self-assembly of peptides: Toward the design of supramolecular polymers with enhanced chemical and biological functions. Prog Polym Sci 2021. [DOI: 10.1016/j.progpolymsci.2021.101469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
25
|
O'Neill CL, Shrimali PC, Clapacs ZE, Files MA, Rudra JS. Peptide-based supramolecular vaccine systems. Acta Biomater 2021; 133:153-167. [PMID: 34010691 PMCID: PMC8497425 DOI: 10.1016/j.actbio.2021.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/01/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022]
Abstract
Currently approved replication-competent and inactivated vaccines are limited by excessive reactogenicity and poor safety profiles, while subunit vaccines are often insufficiently immunogenic without co-administering exogenous adjuvants. Self-assembling peptide-, peptidomimetic-, and protein-based biomaterials offer a means to overcome these challenges through their inherent modularity, multivalency, and biocompatibility. As these scaffolds are biologically derived and present antigenic arrays reminiscent of natural viruses, they are prone to immune recognition and are uniquely capable of functioning as self-adjuvanting vaccine delivery vehicles that improve humoral and cellular responses. Beyond this intrinsic immunological advantage, the wide range of available amino acids allows for facile de novo design or straightforward modifications to existing sequences. This has permitted the development of vaccines and immunotherapies tailored to specific disease models, as well as generalizable platforms that have been successfully applied to prevent or treat numerous infectious and non-infectious diseases. In this review, we briefly introduce the immune system, discuss the structural determinants of coiled coils, β-sheets, peptide amphiphiles, and protein subunit nanoparticles, and highlight the utility of these materials using notable examples of their innate and adaptive immunomodulatory capacity. STATEMENT OF SIGNIFICANCE: Subunit vaccines have recently gained considerable attention due to their favorable safety profiles relative to traditional whole-cell vaccines; however, their reduced efficacy requires co-administration of reactogenic adjuvants to boost immune responses. This has led to collaborative efforts between engineers and immunologists to develop nanomaterial-based vaccination platforms that can elicit protection without deleterious side effects. Self-assembling peptidic biomaterials are a particularly attractive approach to this problem, as their structure and function can be controlled through primary sequence design and their capacity for multivalent presentation of antigens grants them intrinsic self-adjuvanticity. This review introduces the various architectures adopted by self-assembling peptides and discusses their application as modulators of innate and adaptive immunity.
Collapse
Affiliation(s)
- Conor L O'Neill
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| | - Paresh C Shrimali
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| | - Zoe E Clapacs
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| | - Megan A Files
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United States.
| | - Jai S Rudra
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| |
Collapse
|
26
|
Jiang Q, Liu X, Liang G, Sun X. Self-assembly of peptide nanofibers for imaging applications. NANOSCALE 2021; 13:15142-15150. [PMID: 34494635 DOI: 10.1039/d1nr04992e] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Pathological stimuli-responsive self-assembly of peptide nanofibers enables selective accumulation of imaging agent cargos in the stimuli-rich regions of interest. It provides enhanced imaging signals, biocompatibility, and tumor/disease accessibility and retention, thereby promoting smart, precise, and sensitive tumor/disease imaging both in vitro and in vivo. Considering the remarkable significance and recent encouraging breakthroughs of self-assembled peptide nanofibers in tumor/disease diagnosis, this reivew is herein proposed. We emphasize the recent advances particularly in the past three years, and provide an outlook in this field.
Collapse
Affiliation(s)
- Qiaochu Jiang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China.
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China.
| | - Gaolin Liang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China.
| | - Xianbao Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China.
| |
Collapse
|
27
|
Yao Q, Gao S, Wu C, Lin T, Gao Y. Enzymatic non-covalent synthesis of supramolecular assemblies as a general platform for bioorthogonal prodrugs activation to combat drug resistance. Biomaterials 2021; 277:121119. [PMID: 34492583 DOI: 10.1016/j.biomaterials.2021.121119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/10/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022]
Abstract
Multi-drug resistance (MDR) is one of the leading causes of the anticancer failures. Besides the blockage of the MDR pathways, the development of more potent drugs is with urgent needs, but has been postponed mainly due to an imbalance between safety and efficacy. The recent development of the bioorthogonal prodrug activation strategy has shown immense potential to balance safety and efficacy, while recent studies only focused on few drug entities such as doxorubicin and monomethyl auristatin E, leaving the vast collection of toxins undetermined. Here we have enumerated typical molecular entities ranging from food and drug administration (FDA) approved drugs to a heated antibody drug conjugates (ADC) warhead and a trichothecene toxin to demonstrate that the bioorthogonal caging and specific activation could serve as a general design to increase the therapeutic index of bioactive molecules. These prodrugs can be efficiently activated on-demand by the bioorthogonal activators whose distribution was regulated by the cancer cell specific enzymatic non-covalent synthesis of supramolecular self-assemblies. The prodrug activation not only enhanced the synergistic therapeutic effect within a broad range of dose ratios but also allowed the convenient switching of drug identities to successfully combat MDR tumor in vivo. In general, this strategy might serve as a general platform, which can be readily applicable to enlarge the therapeutic window for various bioactive molecules. We envision that the spatiotemporal controlled bioorthogonal prodrug activation would facilitate the discovery of anticancer drugs.
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, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuo Gao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Chengling Wu
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Ting Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, 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, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
28
|
Yao Q, Wu C, Chen J, Zhao Y, Gao Y. Enzyme-instructed supramolecular assemblies promote intracellular boron accumulation for boron neutron capture therapy. NANOTECHNOLOGY 2021; 32:435602. [PMID: 34280913 DOI: 10.1088/1361-6528/ac15ca] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/19/2021] [Indexed: 06/13/2023]
Abstract
Selective accumulation of boron agents in cancer cells is of critical importance for BNCT. Here we involve enzyme-instructed supramolecular assembly (EISA) to facilitate the accumulation of a typical boron agent borylphenylalanine (BPA) in cancer cells. By covalently conjugating BPA to the phosphorylated assembly precursor, the boron-bearing precursors undergo phosphatase-catalyzed dephosphorylation to yield assembly molecules, which then self-assemble to form nanomaterials. Due to the up-regulated phosphatase activity of cancer cells, kinetic preference allows the EISA to accumulate boron in HeLa cells selectively. Interestingly, by attaching BPA on the backbone or side-chain of precursor, the boron-bearing isomers show different assembly propensity with time-dependent morphology change, which leads to the differentiated accumulation of boron inside cells. Overall, the optimized boron-bearing assembly precursor could significantly improve the boron accumulation compared with BPA in cancer cells. In this study, we have demonstrated a convenient method to introduce boron agents to cancer cells. We envision that the EISA-mediated accumulation of boron will be helpful in the design of boron agents to facilitate BNCT treatment.
Collapse
Affiliation(s)
- 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
| | - Chengling Wu
- 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
| | - 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
| | - 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
| | - 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
|
29
|
Jiang C, Huang H, Kang X, Yang L, Xi Z, Sun H, Pluth MD, Yi L. NBD-based synthetic probes for sensing small molecules and proteins: design, sensing mechanisms and biological applications. Chem Soc Rev 2021; 50:7436-7495. [PMID: 34075930 PMCID: PMC8763210 DOI: 10.1039/d0cs01096k] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Compounds with a nitrobenzoxadiazole (NBD) skeleton exhibit prominent useful properties including environmental sensitivity, high reactivity toward amines and biothiols (including H2S) accompanied by distinct colorimetric and fluorescent changes, fluorescence-quenching ability, and small size, all of which facilitate biomolecular sensing and self-assembly. Amines are important biological nucleophiles, and the unique activity of NBD ethers with amines has allowed for site-specific protein labelling and for the detection of enzyme activities. Both H2S and biothiols are involved in a wide range of physiological processes in mammals, and misregulation of these small molecules is associated with numerous diseases including cancers. In this review, we focus on NBD-based synthetic probes as advanced chemical tools for biomolecular sensing. Specifically, we discuss the sensing mechanisms and selectivity of the probes, the design strategies for multi-reactable multi-quenching probes, and the associated biological applications of these important constructs. We also highlight self-assembled NBD-based probes and outline future directions for NBD-based chemosensors. We hope that this comprehensive review will facilitate the development of future probes for investigating and understanding different biological processes and aid the development of potential theranostic agents.
Collapse
Affiliation(s)
- Chenyang Jiang
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| | - Haojie Huang
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| | - Xueying Kang
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| | - Liu Yang
- Department of Chemistry and Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China.
| | - Zhen Xi
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, College of Chemistry, National Pesticide Engineering Research Center, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Hongyan Sun
- Department of Chemistry and Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China. and Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| | - Michael D Pluth
- Department of Chemistry and Biochemistry, Materials Science Institute, Knight Campus for Accelerating Scientific Impact, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA.
| | - Long Yi
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| |
Collapse
|
30
|
Tan W, Zhang Q, Wang J, Yi M, He H, Xu B. Enzymatic Assemblies of Thiophosphopeptides Instantly Target Golgi Apparatus and Selectively Kill Cancer Cells*. Angew Chem Int Ed Engl 2021; 60:12796-12801. [PMID: 33783926 PMCID: PMC8159897 DOI: 10.1002/anie.202102601] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/19/2021] [Indexed: 01/01/2023]
Abstract
Changing an oxygen atom of the phosphoester bond in phosphopeptides by a sulfur atom enables instantly targeting Golgi apparatus (GA) and selectively killing cancer cells by enzymatic self-assembly. Specifically, conjugating cysteamine S-phosphate to the C-terminal of a self-assembling peptide generates a thiophosphopeptide. Being a substrate of alkaline phosphatase (ALP), the thiophosphopeptide undergoes rapid ALP-catalyzed dephosphorylation to form a thiopeptide that self-assembles. The thiophosphopeptide enters cells via caveolin-mediated endocytosis and macropinocytosis and instantly accumulates in GA because of dephosphorylation and formation of disulfide bonds in Golgi by themselves and with Golgi proteins. Moreover, the thiophosphopeptide potently and selectively inhibits cancer cells (HeLa) with the IC50 (about 3 μM), which is an order of magnitude more potent than that of the parent phosphopeptide.
Collapse
Affiliation(s)
- Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Qiuxin Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Jiaqing Wang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Meihui Yi
- 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
|
31
|
Kubota R, Tanaka W, Hamachi I. Microscopic Imaging Techniques for Molecular Assemblies: Electron, Atomic Force, and Confocal Microscopies. Chem Rev 2021; 121:14281-14347. [DOI: 10.1021/acs.chemrev.0c01334] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ryou Kubota
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Wataru Tanaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- JST-ERATO, Hamachi Innovative Molecular Technology for Neuroscience, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8530, Japan
| |
Collapse
|
32
|
Tan W, Zhang Q, Wang J, Yi M, He H, Xu B. Enzymatic Assemblies of Thiophosphopeptides Instantly Target Golgi Apparatus and Selectively Kill Cancer Cells**. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102601] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Weiyi Tan
- Department of Chemistry Brandeis University 415 South Street Waltham MA 02453 USA
| | - Qiuxin Zhang
- Department of Chemistry Brandeis University 415 South Street Waltham MA 02453 USA
| | - Jiaqing Wang
- Department of Chemistry Brandeis University 415 South Street Waltham MA 02453 USA
| | - Meihui Yi
- 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
|
33
|
Zhou Y, Chen Y, Tan Y, Hu R, Niu M. An NRP1/MDM2-Targeted D-Peptide Supramolecular Nanomedicine for High-Efficacy and Low-Toxic Liver Cancer Therapy. Adv Healthc Mater 2021; 10:e2002197. [PMID: 33690977 DOI: 10.1002/adhm.202002197] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/31/2021] [Indexed: 01/15/2023]
Abstract
Supramolecular nanomedicines based on self-assembly of D-peptides have been of great interest as potential candidates for cancer therapy. Neuropilin-1 (NRP1) and mouse double minute 2 (MDM2) have been considered as the anticancer targets because of their overexpression in cancers. However, NRP1/MDM2-targeted D-peptide supramolecular nanomedicines remain unreported. Here, a potent anticancer D-peptide supramolecular nanomedicine targeting NRP1 and MDM2, termed as NMTP-5, is identified by using structure-based virtual screening techniques. NMTP-5 exhibits good biostability and strong cellular uptake performance. Moreover, NMTP-5 displays strong anticancer activity to SK-Hep-1 cells in vitro and in vivo, with no apparent host toxicity. This work demonstrates that NMTP-5 can be used as a potential chemotherapeutic agent for the treatment of liver cancer.
Collapse
Affiliation(s)
- Yunjiang Zhou
- Key Laboratory of Drug Quality Control and Pharmacovigilance Ministry of Education State Key Laboratory of Natural Medicines School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Yaxin Chen
- Key Laboratory of Drug Quality Control and Pharmacovigilance Ministry of Education State Key Laboratory of Natural Medicines School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Yingying Tan
- Key Laboratory of Drug Quality Control and Pharmacovigilance Ministry of Education State Key Laboratory of Natural Medicines School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Rong Hu
- Key Laboratory of Drug Quality Control and Pharmacovigilance Ministry of Education State Key Laboratory of Natural Medicines School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Miao‐Miao Niu
- Key Laboratory of Drug Quality Control and Pharmacovigilance Ministry of Education State Key Laboratory of Natural Medicines School of Basic Medicine and Clinical Pharmacy China Pharmaceutical University Nanjing 210009 China
| |
Collapse
|
34
|
Li G, Hu X, Wu X, Zhang Y. Microtubule-Targeted Self-Assembly Triggers Prometaphase-Metaphase Oscillations Suppressing Tumor Growth. NANO LETTERS 2021; 21:3052-3059. [PMID: 33756080 DOI: 10.1021/acs.nanolett.1c00233] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Microtubules are highly strategic targets of cancer therapies. Small molecule antimitotic agents are so far the best chemotherapeutic medication in cancer treatment. However, the high rate of neuropathy and drug resistance limit their clinical usage. Inspired by the multicomponent-targeting feature of molecular self-assembly (MSA) overcoming drug resistance, we synthesized peptide-based rotor molecules that self-assemble in response to the surrounding environment to target the microtubule array. The MSAs self-adjust morphologically in response to the pH change and viscosity variations during Golgi-endosome trafficking, escape trafficking cargos, and eventually bind to the microtubule array physically in a nonspecific manner. Such unrefined nano-bio interactions suppress regional tubulin polymerization triggering atypical prometaphase--metaphase oscillations to inhibit various cancer cells proliferating without inducing obvious neurotoxicity. The MSA also exerts potent antiproliferative effects in the subcutaneous cervix cancer xenograft tumor model equivalent to Cisplatin, better than the classic antimitotic drug Taxol.
Collapse
Affiliation(s)
- Guanying Li
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xunwu Hu
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Xia Wu
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Ye Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| |
Collapse
|
35
|
Kwek G, Do TC, Lu X, Lin J, Xing B. Scratching the Surface of Unventured Possibilities with In Situ Self-Assembly: Protease-Activated Developments for Imaging and Therapy. ACS APPLIED BIO MATERIALS 2021; 4:2192-2216. [PMID: 35014345 DOI: 10.1021/acsabm.0c01340] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In situ self-assembly has attracted increasing research interest for applications in imaging and therapy in recent years. Particularly for protease-activated developments, inspiration is drawn from the innate specificity of their catalytic activities, rapid discovery of the various roles they play in the proliferation of certain diseases, and inherent susceptibility of small molecule peptide conjugates to proteolytic digestion in vivo. The overexpression of a disease-related protease of interest can be exploited as an endogenous stimulus for site-specific self-assembly to largely amplify a molecular event happening at the cellular level. This holds great potential for applications in early stage disease detection, long-term disease monitoring, and sustained therapeutic effects. This review summarizes the recent developments in protease-activated self-assemblies for imaging and therapeutic applications toward the manifestation of tumors, bacterial infections, neurodegenerative disorders, and wound recovery.
Collapse
Affiliation(s)
- Germain Kwek
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, 637371 Singapore
| | - Thang Cong Do
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, 637371 Singapore
| | - Xiaoling Lu
- International Nanobody Research Centre of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Bengang Xing
- Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, 637371 Singapore.,School of Chemical & Biomedical Engineering, Nanyang Technological University, 637549 Singapore
| |
Collapse
|
36
|
Liu J, Wu C, Dai G, Feng F, Chi Y, Xu K, Zhong W. Molecular self-assembly of a tyroservatide-derived octapeptide and hydroxycamptothecin for enhanced therapeutic efficacy. NANOSCALE 2021; 13:5094-5102. [PMID: 33650607 DOI: 10.1039/d0nr08741f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Tyroservatide (YSV) belongs to a class of bioactive peptides that have drawn considerable attention in the field of drug discovery, yet it displays limited potency and often requires a millimolar concentration to execute its cellular functions. To enhance the potency of the drug through a self-assembling strategy, we designed and synthesized a series of octapeptides through conjugation of YSV with a pentapeptide sequence bearing alternating hydrophobic and hydrophilic amino acids to promote their self-assembling capabilities. Initial screening for hydrogelation gave a novel octapeptide (denoted as 1-YSV hereafter) that was capable of self-assembling under physiological conditions to afford supramolecular nanofibers with enhanced anti-cancer efficacy compared to YSV itself. Interestingly, 1-YSV formed a robust co-assembly with the anticancer drug hydroxycamptothecin (HCPT) to afford 1-YSV/HCPT hydrogel, which not only greatly improved the viscoelastic properties of hydrogels, but also stabilized HCPT in the hydrogel matrix and avoided the agglomeration of drug molecules. Compared to HCPT solution, the hydrogel formulation of 1-YSV/HCPT demonstrated better efficacy against the proliferation of non-small cell lung cancer A549 cells both in vitro and in vivo. Finally, thanks to the pure amino acid-based composition, the 1-YSV/HCPT formulation exhibited excellent biocompatibility, giving a low hemolytic rate to red blood cells, with mild local tissue reactions and negligible systematic toxicities in mice.
Collapse
Affiliation(s)
- Jing Liu
- Department of Chemistry, China Pharmaceutical University, Nanjing, P. R. China
| | - Can Wu
- Department of Chemistry, China Pharmaceutical University, Nanjing, P. R. China
| | - Guoru Dai
- Department of Chemistry, China Pharmaceutical University, Nanjing, P. R. China
| | - Feng Feng
- Department of Chemistry, China Pharmaceutical University, Nanjing, P. R. China
| | - Yuquan Chi
- Department of Chemistry, China Pharmaceutical University, Nanjing, P. R. China
| | - Keming Xu
- Department of Chemistry, China Pharmaceutical University, Nanjing, P. R. China and Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing, P. R. China.
| | - Wenying Zhong
- Department of Chemistry, China Pharmaceutical University, Nanjing, P. R. China and Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing, P. R. China. and Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
Qin M, Zhang Y, Xing C, Yang L, Zhao C, Dou X, Feng C. Effect of Stereochemistry on Chirality and Gelation Properties of Supramolecular Self-Assemblies. Chemistry 2021; 27:3119-3129. [PMID: 33225542 DOI: 10.1002/chem.202004533] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Indexed: 01/01/2023]
Abstract
Although chiral nanostructures have been fabricated at various structural levels, the transfer and amplification of chirality from molecules to supramolecular self-assemblies are still puzzling, especially for heterochiral molecules. Herein, four series of C2 -symmetrical dipeptide-based derivatives bearing various amino acid sequences and different chiralities are designed and synthesized. The transcription and amplification of molecular chirality to supramolecular assemblies are achieved. The results show that supramolecular chirality is only determined by the amino acid adjacent to the benzene core, irrespective of the absolute configuration of the C-terminal amino acid. In addition, molecular chirality also has a significant influence on the gelation behavior. For the diphenylalanine-based gelators, the homochiral gelators can be gelled through a conventional heating-cooling process, whereas heterochiral gelators form translucent stable gels under sonication. The racemic gels possess higher mechanical properties than those of the pure enantiomers. All of these results contribute to an increasing knowledge over control of the generation of specific chiral supramolecular structures and the development of new optimized strategies to achieve functional supramolecular organogels through heterochiral and racemic systems.
Collapse
Affiliation(s)
- Minggao Qin
- State Key Lab of Metal Matrix Composites, School of, Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Yaqian Zhang
- State Key Lab of Metal Matrix Composites, School of, Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Chao Xing
- State Key Lab of Metal Matrix Composites, School of, Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Li Yang
- State Key Lab of Metal Matrix Composites, School of, Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Changli Zhao
- State Key Lab of Metal Matrix Composites, School of, Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Xiaoqiu Dou
- State Key Lab of Metal Matrix Composites, School of, Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| | - Chuanliang Feng
- State Key Lab of Metal Matrix Composites, School of, Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P.R. China
| |
Collapse
|
40
|
Zhang W, Zhai Z, Li S, Lin X, Bai W, Ding N, Zhang Y, Tong J, Sun J, Gao C. In situ formation of tetraphenylethylene nano-structures on microgels inside living cells via reduction-responsive self-assembly. NANOSCALE 2021; 13:138-149. [PMID: 33350429 DOI: 10.1039/d0nr06661c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Controlling the assembly of synthetic molecules in living systems is of significance for their adaptive applications. However, it is difficult to achieve, especially for composite self-assemblies, due to the complexity and dynamic change of the intracellular environment, and there exist technical difficulties for the direct visualization of organic and polymer self-assemblies. Herein, we demonstrate a novel strategy for the in situ formation of self-assembled micro-nano composite structures in a cell milieu using reduction-responsive microgels (MGs) as a platform. The MGs were prepared by a templating and crosslinking method using a synthetic amphiphlic polymer as the basic material and porous CaCO3 microparticles as the template. The aggregation-induced emission (AIE) tetraphenylethylene moieties and reduction-labile disulfide bonds in the MGs were employed as the self-assembly building blocks and triggering sites for the intracellular self-assembly, respectively. In the presence of reductive agents such as glutathione, nano-spikes were gradually formed on the MGs. After the MGs were internalized by cells, the in situ formation of microgel/nano-spike composite structures was evidenced by the enhanced fluorescence intensity and was further confirmed by direct transmission electron microscopy observation. This work provides an effective strategy to cope with the challenging task of achieving and probing controlled self-assembly in a cell milieu, leading to new insights into investigating biological self-assembly and promoting the development of micro-/nanomaterials by learning from nature.
Collapse
Affiliation(s)
- Wenbo Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Verma N, Thapa K, Dua K. Material and strategies used in oncology drug delivery. ADVANCED DRUG DELIVERY SYSTEMS IN THE MANAGEMENT OF CANCER 2021:47-62. [DOI: 10.1016/b978-0-323-85503-7.00015-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
42
|
Deng Y, Zhan W, Liang G. Intracellular Self-Assembly of Peptide Conjugates for Tumor Imaging and Therapy. Adv Healthc Mater 2021; 10:e2001211. [PMID: 32902191 DOI: 10.1002/adhm.202001211] [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: 07/11/2020] [Revised: 08/19/2020] [Indexed: 12/20/2022]
Abstract
Intracellular self-assembly (ISA) is a versatile and powerful strategy for in situ constructing sophisticated and functional supramolecular nanostructures, which has been widely applied in biomedicine and biomedical engineering. Among the common building blocks for ISA, peptides have attracted increasingly attention due to their intrinsic bioactivity, biocompatibility, and biodegradability. Particularly, by conjugating functional motifs (e.g., probes or drugs) to peptides to yield the peptide conjugates, the latter show enhanced stability and efficiency, and probably new functions. In recent years, employing ISA of peptide conjugates for tumor imaging and treatment has achieved great progresses. Therefore, the recent progress of ISA of peptide conjugates is summarized in this progress report. Moreover, several examples of ISA of peptide conjugates for other important imaging or therapeutic applications are also introduced. Finally, a brief perspective on remaining challenges and potential directions for future research in this area is presented.
Collapse
Affiliation(s)
- Yu Deng
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University 2 Sipailou Road Nanjing Jiangsu 210096 China
| | - Wenjun Zhan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University 2 Sipailou Road Nanjing Jiangsu 210096 China
| | - Gaolin Liang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering Southeast University 2 Sipailou Road Nanjing Jiangsu 210096 China
| |
Collapse
|
43
|
Wang A, Mao Q, Zhao M, Ye S, Fang J, Cui C, Zhao Y, Zhang Y, Zhang Y, Zhou F, Shi H. pH/Reduction Dual Stimuli-Triggered Self-Assembly of NIR Theranostic Probes for Enhanced Dual-Modal Imaging and Photothermal Therapy of Tumors. Anal Chem 2020; 92:16113-16121. [PMID: 33275417 DOI: 10.1021/acs.analchem.0c03800] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Tumor microenvironment plays a pivotal role in the growth and metastasis of tumors, and has become a promising target for precise diagnosis and treatment of tumors. Herein, a novel smart NIR theranostic probe Cy-1 that can simultaneously respond to low intracellular pH and reductive glutathione (GSH) is reported. This probe has demonstrated to be able to intermolecularly undergo a biologically compatible CBT-Cys condensation reaction to selectively form large nanoaggregates in the tumor microenvironment resulting in its enhanced accumulation and retention in the tumor, which as a consequence significantly improves the sensitivity of NIR/photoacoustic imaging and photothermal therapeutic efficacy of tumors in living mice. We thus believe that this dual stimuli-mediated self-assembly strategy may offer a promising and universal platform for cancer theranostics.
Collapse
Affiliation(s)
- Anna Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Qiulian Mao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Meng Zhao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Shuyue Ye
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Jing Fang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Chaoxiang Cui
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Yan Zhao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Yuqi Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Yu Zhang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, P. R. China
| | - Feng Zhou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou 215006, P. R. China
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
44
|
Kubota R, Torigoe S, Liu S, Hamachi I. In Situ Real-time Confocal Imaging of a Self-assembling Peptide-grafted Polymer Showing pH-responsive Hydrogelation. CHEM LETT 2020. [DOI: 10.1246/cl.200513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Ryou Kubota
- Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Shogo Torigoe
- Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Shuang Liu
- Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Itaru Hamachi
- Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- JST-ERATO, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8530, Japan
| |
Collapse
|
45
|
Guo J, Tian C, Xu B. Biomaterials based on noncovalent interactions of small molecules. EXCLI JOURNAL 2020; 19:1124-1140. [PMID: 33088250 PMCID: PMC7573174 DOI: 10.17179/excli2020-2656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 07/27/2020] [Indexed: 11/10/2022]
Abstract
Unlike conventional materials that covalent bonds connecting atoms as the major force to hold the materials together, supramolecular biomaterials rely on noncovalent intermolecular interactions to assemble. The reversibility and biocompatibility of supramolecular biomaterials render them with diverse range of functions and lead to rapid development in the past two decades. This review focuses on the noncovalent and enzymatic control of supramolecular biomaterials, with the introduction to various triggering mechanism to initiate self-assembly. Representative applications of supramolecular biomaterials are highlighted in four categories: tissue engineering, cancer therapy, drug delivery, and molecular imaging. By introducing various applications, we intend to show enzymatic control and noncovalent interactions as a powerful tool for achieving spatiotemporal control of biomaterials both invitro and in vivo for biomedicine.
Collapse
Affiliation(s)
- Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA
| | - Changhao Tian
- Department of Physics, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, China
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA
| |
Collapse
|
46
|
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
|
47
|
Zhang S, Zhang Y. Promoting Dual-Targeting Anticancer Effect by Regulating the Dynamic Intracellular Self-Assembly. ACS APPLIED MATERIALS & INTERFACES 2020; 12:41105-41112. [PMID: 32819089 DOI: 10.1021/acsami.0c12271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Despite the promise of nanomedicine in the fight against complex diseases, the enthusiasm for its pharmaceutical development is backed by the elevated costs associated with the R&D process. Therefore, as a compromise solution, nanotechnology was mainly applied as a drug delivery system to improve bioavailability and controllability of pharmaceutical drugs. Attempting to break the restrictions without elevating potential costs, we multiply the functions of excipients in the nanodelivery system by endowing subcellular-targeting ability. To prove the concept, fluorescent endoplasmic reticulum-targeted short peptides were covalently connected to chemotherapy medication chlorambucil achieving enhanced drug-loading efficiency. Via visualized intracellular dynamic enzyme-catalyzed hydrolysis, the ER-targeting excipient and nucleus-targeting chlorambucil are released simultaneously, achieving a synergistic anticancer effect and elucidating the influence of intracellular self-assembly transition on enzymatic reactions.
Collapse
Affiliation(s)
- Shijin Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna Son, Okinawa 904-0495, Japan
| | - Ye Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna Son, Okinawa 904-0495, Japan
| |
Collapse
|
48
|
He H, Liu S, Wu D, Xu B. Enzymatically Formed Peptide Assemblies Sequestrate Proteins and Relocate Inhibitors to Selectively Kill Cancer Cells. Angew Chem Int Ed Engl 2020; 59:16445-16450. [PMID: 32521103 PMCID: PMC7844580 DOI: 10.1002/anie.202006290] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Indexed: 12/24/2022]
Abstract
Herein, we show that an enzymatic reaction can generate peptide assemblies that sequestrate proteins to selectively kill cancer cells. A phosphopeptide bearing the antagonistic motif (AVPI) to the inhibitors of apoptotic proteins (IAPs) enters cancer cells and normal cells by caveolin-dependent endocytosis and macropinocytosis, respectively. The AVPI-bearing peptide assemblies sequestrates IAPs and releases bortezomib (BTZ), a proteasome inhibitor, in the cytosol of cancer cells, but rescues the normal cells (namely, HS-5 cells) by trafficking the BTZ into lysosomes. Alkaline phosphatase (ALP) acts as a context-dependent signal for trafficking the peptide/BTZ assemblies and selectively induces the death of the cancer cells. The assemblies of AVPI exhibit enhanced proteolytic resistance. This work, which utilizes the difference in endocytic uptake of enzymatically formed peptide assemblies to selectively kill cancer cells, promises a new way to develop selective cancer therapeutics.
Collapse
Affiliation(s)
- Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Shuang Liu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Difei Wu
- 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
|
49
|
Li Y, Cao X, Tian C, Zheng JS. Chemical protein synthesis-assisted high-throughput screening strategies for d-peptides in drug discovery. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2020.04.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
50
|
Baillet J, Gaubert A, Verget J, Latxague L, Barthélémy P. β-Galactosidase instructed self-assembly of supramolecular bolaamphiphiles hydrogelators. SOFT MATTER 2020; 16:7648-7651. [PMID: 32657300 DOI: 10.1039/d0sm01055c] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
β-Galactosidase instructed supramolecular assemblies of Low Molecular Weight Gelators (LMWGs) derived from glyconucleo-bolaamphiphiles have been designed. These precursors, comprising galactose sensitive units at both polar heads, showed the formation of hydrogels upon the action of β-galactosidase.
Collapse
Affiliation(s)
- Julie Baillet
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Alexandra Gaubert
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Julien Verget
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Laurent Latxague
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Philippe Barthélémy
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| |
Collapse
|