1
|
Lin M, Liu D, Gong Y, Shu L, Wang H, Zhang G, Li J, Gao Z, Sun J, Chen X. Bioactive Assembly Cofactor-Assisted Ursolic Acid Helix for Enhanced Anticancer Efficacy via In Situ Virus-like Transition. J Am Chem Soc 2025; 147:17010-17021. [PMID: 40354555 DOI: 10.1021/jacs.5c01214] [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: 05/14/2025]
Abstract
Natural bioactive pentacyclic triterpenoids, such as ursolic acid (UA), hold significant potential as anticancer agents. However, their clinical application is limited by their poor solubility and bioavailability. Herein, we developed a novel polypeptoid assembly cofactor-assisted nanoplatform designed to enhance UA's therapeutic efficacy through in situ self-assembly within the tumor microenvironment (TME). Bioactive polypeptoid polyelectrolytes, inspired by natural molecular chaperones, were utilized as assembly cofactors to guide UA's co-assembly into stimuli-responsive nanostructures. These polypeptoids provide precise control over the assembly process, improving stability and enabling reversible, pH-responsive transformations. Acid-responsive groups and the target molecule lactobionic acid further promote the specificity and efficacy of UA delivery. Under neutral conditions, the assemblies retain a helical fibrous structure, while in the acidic TME, they transform into virus-like clusters composed of assembly subunits, facilitating deeper tumor penetration. Once internalized, these nanoparticles escape into the cytoplasm and accumulate around the mitochondria, where the oxidation of thioether bonds triggers the release of UA and polypeptoids, causing mitochondrial damage and apoptosis. Some nanoparticles reassemble into fibrous structures intracellularly, extending their retention in tumor cells and potentially leading to mitochondria damage. Notably, the nanoplatform demonstrates excellent synergistic effects, achieving significantly higher therapeutic efficiency compared with individual components, including UA and polypeptoids. In vivo studies further confirmed the effectiveness, demonstrating significant tumor growth suppression and reduced metastasis. By integrating the therapeutic UA with bioactive polypeptoids under precise control, this synergistic platform represents a highly efficient and targeted approach to cancer therapy, offering a promising new opportunity for natural compounds for advanced nanomedicine.
Collapse
Affiliation(s)
- Min Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Dandan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Yiyu Gong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Lilei Shu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Helin Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Guojing Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Jiayi Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Zixin Gao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Jing Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Xuesi Chen
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022 Changchun, China
| |
Collapse
|
2
|
Wang A, Liu W, Jin X, Wu H, Zhang D, Han X, Liu Y, Li Z, Ding M, Li J, Tan H. Dynamics and Machine Learning Reveal the Link between Tripeptide Sequences and Evaporation-Driven Material Properties. NANO LETTERS 2025; 25:7560-7567. [PMID: 40289375 DOI: 10.1021/acs.nanolett.5c01415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Previous research showed that a peptide composed of three tyrosines (YYY) can turn into organic glass and cause strong adhesion between substrates via evaporation. However, the mechanisms of these processes remain unclear, and the exploration of applications of other peptide sequences is necessary. In this study, an optimized evaporation method was employed in molecular dynamics. It was found that YYY evaporation products possess abundant internal hydrogen bonds, which may facilitate the amorphous glass state formation. Moderate hydrophilicity of a peptide enhances molecular mobility and self-healing ability, while excessive hydrophilicity causes a water plasticizing effect. Stronger hydrophilicity also brings a larger curvature of evaporation products on polydimethylsiloxane (PDMS) substrate. A machine learning model was developed to predict the evaporation contact angle of peptide evaporation products and agrees well with the experiment. This research aims to improve understanding of peptide structure-function relationships and aid in designing custom organic optical devices based on peptide sequences.
Collapse
Affiliation(s)
- Ao Wang
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Med-X center for materials, Sichuan University, Chengdu 610065, China
| | - Wenkai Liu
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Med-X center for materials, Sichuan University, Chengdu 610065, China
| | - Xiaohan Jin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hecheng Wu
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Med-X center for materials, Sichuan University, Chengdu 610065, China
| | - Dongfei Zhang
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Med-X center for materials, Sichuan University, Chengdu 610065, China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yang Liu
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Med-X center for materials, Sichuan University, Chengdu 610065, China
| | - Zhen Li
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Med-X center for materials, Sichuan University, Chengdu 610065, China
| | - Mingming Ding
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Med-X center for materials, Sichuan University, Chengdu 610065, China
| | - Jiehua Li
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Med-X center for materials, Sichuan University, Chengdu 610065, China
| | - Hong Tan
- College of Polymer Science and Engineering, National Key Laboratory of Advanced Polymer Materials, Med-X center for materials, Sichuan University, Chengdu 610065, China
| |
Collapse
|
3
|
Yan JJ, Wang YY, Shi ZY, Ding YY, Wen HQ, Wu MP, Sun SC, Cai YF, Zhang Y. SIRT5 modulates mitochondria function via mitophagy and antioxidant mechanisms to facilitate oocyte maturation in mice. Int J Biol Macromol 2025; 306:141488. [PMID: 40015402 DOI: 10.1016/j.ijbiomac.2025.141488] [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: 01/03/2025] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Mitochondrial homeostasis, closely associated with mitophagy and antioxidant mechanisms, is essential for proper meiotic spindle assembly and chromosome segregation during oocyte maturation. SIRT5, known to modulate mitochondrial function under various conditions, has been shown to impact oocyte quality when inhibited, however, the precise mechanisms linking SIRT5 to mitochondrial homeostasis during meiotic progression remain unclear. In this study, we demonstrate that SIRT5 localizes predominantly at the periphery of the meiotic spindle and is enriched on chromosomes during oocyte maturation. Inhibition of SIRT5 led to significant meiotic defects, including disrupted spindle organization and chromosome misalignment. These defects were associated with increased histone acetylation, which impaired kinetochore-microtubule attachments. Moreover, SIRT5 inhibition resulted in mitochondrial dysfunction, subsequently elevating ROS levels and triggering oxidative stress, which further exacerbated meiotic abnormalities. Mechanistically, SIRT5 inhibition disrupted the balance of Parkin-dependent mitophagy by inducing ULK phosphorylation. Additionally, it activated the PI3K/Akt signaling pathway, which increased NADPH consumption and reduced GSH levels. Collectively, these findings reveal that SIRT5 plays dual roles in maintaining mitochondrial homeostasis during oocyte maturation: (1) by regulating Parkin-dependent mitophagy to prevent excessive mitochondrial clearance, and (2) by preserving the NADPH/GSH antioxidant system to ensure redox balance. These insights provide potential targets for improving oocyte quality and addressing mitochondrial dysfunction-related reproductive disorders in females.
Collapse
Affiliation(s)
- Jing-Jing Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yan-Yu Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhi-Yu Shi
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuan-Yuan Ding
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Hao-Quan Wen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Meng-Ping Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ya-Fei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
4
|
Zhang Y, Qian C, Chu C, Yang XZ, Wu Y, Cai L, Yao S, He W, Guo Z, Chen Y. Self-Assembly of Short Peptides Activates Specific ER-Phagy and Induces Pyroptosis for Enhanced Tumor Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202422874. [PMID: 40069115 DOI: 10.1002/anie.202422874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
Developing specific endoplasmic reticulum-autophagy (ER-phagy) inducers is highly desirable for discovering new ER-phagy receptors and elucidating the detailed ER-phagy mechanism and potential cancer immunotherapy. However, most of the current ER-phagy-inducing methods cause nonselective autophagy of other organelles. In this work, we report the design and synthesis of simple and stable short peptides (D-FFxFFs) that could specifically trigger ER-phagy, which further induces pyroptosis and activates the immune response against tumor cells. D-FFxFFs locate preferentially in ER and readily self-assemble to form nanosized misfolded protein mimics, which lead to distinct upregulation of dedicated ER-phagy receptors with no obvious autophagy of other organelles. Significant unfolded protein response (UPR) is activated via IRE1-JNK and PERK-ATF4 pathways. Interestingly, the persistent ER-phagy triggers ER Ca2+ release and a surge in mitochondrial Ca2+ levels, resulting in GSDMD-mediated pyroptosis other than apoptosis. The ER-phagy induces pyroptosis and activates a distinct antitumor immune response without evolving the acquired drug resistance. This work not only provides a powerful tool for investigating the mechanism and function of ER-phagy but also offers an appealing strategy for anticancer immunotherapy.
Collapse
Affiliation(s)
- Yunhua Zhang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Chengyuan Qian
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Chengyan Chu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Xiu-Zhi Yang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Yanping Wu
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Linxiang Cai
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Shankun Yao
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| | - Weijiang He
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
- Nanchuang (Jiangsu) Institute of Chemistry and Health, Nanjing, 210000, P.R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
- Nanchuang (Jiangsu) Institute of Chemistry and Health, Nanjing, 210000, P.R. China
| | - Yuncong Chen
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
- Nanchuang (Jiangsu) Institute of Chemistry and Health, Nanjing, 210000, P.R. China
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, Jiangsu, 210008, P.R. China
| |
Collapse
|
5
|
Wang L, Chen Z, Dai Z, Li M, Chen B, Wang B. Membrane-intercalating conjugated oligoelectrolytes for lipid membrane imaging. Biomater Sci 2025; 13:1923-1938. [PMID: 40084474 DOI: 10.1039/d5bm00028a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Membrane-intercalating conjugated oligoelectrolytes (MICOEs), a class of phospholipid bilayer mimics, demonstrate an exceptional ability to spontaneously integrate into biological membranes through a combination of electrostatic and hydrophobic interactions. This unique property makes them promising candidates for membrane imaging applications. Over the past decade, MICOEs have been successfully applied to imaging and tracking a wide range of biological membranes, including microbial membranes, mammalian plasma membranes, intracellular membranes, extracellular vesicles, and artificial liposomes. Recent advancements have shed light on the imaging mechanisms of MICOEs and highlighted their potential as fluorescent probes, with a focus on structural optimization to enhance their performance. Building on these developments, this review will explore the intercalation mechanisms of MICOEs, analyze the structure-activity relationships governing their molecular design and imaging capabilities, and discuss the future challenges and emerging opportunities for their application as advanced membrane dyes.
Collapse
Affiliation(s)
- Lingna Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Zehua Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Zhaohui Dai
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Meng Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Bo Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Bing Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| |
Collapse
|
6
|
Zhang Z, Liu X, Tian F, Wang Y, Hu B, Yu Z. Aspartate Isomerization Regulates in Situ Assembly of Peptides into Supramolecular Probes for Detection of Protein Carboxyl Methyltransferase in Bladder Cancer. NANO LETTERS 2025; 25:3923-3930. [PMID: 40016095 DOI: 10.1021/acs.nanolett.4c06452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Protein carboxyl methyltransferase (PCMT) restores aspartate isomers in proteins and plays a critical role in cancer prognosis. However, in vivo detection of PCMT remains challenging. Here, we report the aspartate isomerization-regulated in situ assembly of peptides into supramolecular probes within living cells for PCMT detection in bladder cancer. The peptide consists of alternating hydrophobic and hydrophilic residues and contains an isoAsp residue as a kinked site to prevent the facial amphiphilicity of the peptide. Exposure to PCMT converts isoAsp to Asp within the peptide, thereby promoting its assembly into nanofibers. Incorporation of 7-nitro-2,1,3-benzoxadiazole (NBD) into the nanofibers enables PCMT detection based on hydrophobicity-dependent fluorescence of NBD units. Both cellular and animal studies confirm the capability of supramolecular probes for efficient detection of PCMT. Our finding demonstrates an efficient strategy for regulating peptide assembly in living systems and thus provides a new tool for creation of biomedical agents in the future.
Collapse
Affiliation(s)
- Zeyu Zhang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xin Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Feng Tian
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yushi Wang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Binbin Hu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zhilin Yu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
7
|
Sun M, He L, Chen R, Lv M, Chen ZS, Fan Z, Zhou Y, Qin J, Du J. Rational design of peptides to overcome drug resistance by metabolic regulation. Drug Resist Updat 2025; 79:101208. [PMID: 39914188 DOI: 10.1016/j.drup.2025.101208] [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: 10/13/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/24/2025]
Abstract
Chemotherapy is widely used clinically, however, its efficacy is often compromised by the development of drug resistance, which arises from prolonged administration of drugs or other stimuli. One of the driven causes of drug resistance in tumors or bacterial infections is metabolic reprogramming, which alters mitochondrial metabolism, disrupts metabolic pathways and causes ion imbalance. Bioactive peptide materials, due to their biocompatibility, diverse bioactivities, customizable sequences, and ease of modification, have shown promise in overcoming drug resistance. This review provides an in-depth analysis of metabolic reprogramming and associated microenvironmental changes that contribute to drug resistance in common tumors and bacterial infections, suggesting potential therapeutic targets. Additionally, we explore peptide-based materials for regulating metabolism and their potential synergic effect with other therapies, highlighting the mechanisms by which these peptides reverse drug resistance. Finally, we discuss future perspectives and the clinical challenges in peptide-based treatments, aiming to offer insights for overcoming drug-resistant diseases.
Collapse
Affiliation(s)
- Min Sun
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Le He
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ran Chen
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Mingchen Lv
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Zhen Fan
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yuxiao Zhou
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
| | - Jinlong Qin
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China.
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China; Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China.
| |
Collapse
|
8
|
Fiorentino F, Fabbrizi E, Mai A, Rotili D. Activation and inhibition of sirtuins: From bench to bedside. Med Res Rev 2025; 45:484-560. [PMID: 39215785 PMCID: PMC11796339 DOI: 10.1002/med.22076] [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: 05/25/2022] [Revised: 07/27/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024]
Abstract
The sirtuin family comprises seven NAD+-dependent enzymes which catalyze protein lysine deacylation and mono ADP-ribosylation. Sirtuins act as central regulators of genomic stability and gene expression and control key processes, including energetic metabolism, cell cycle, differentiation, apoptosis, and aging. As a result, all sirtuins play critical roles in cellular homeostasis and organism wellness, and their dysregulation has been linked to metabolic, cardiovascular, and neurological diseases. Furthermore, sirtuins have shown dichotomous roles in cancer, acting as context-dependent tumor suppressors or promoters. Given their central role in different cellular processes, sirtuins have attracted increasing research interest aimed at developing both activators and inhibitors. Indeed, sirtuin modulation may have therapeutic effects in many age-related diseases, including diabetes, cardiovascular and neurodegenerative disorders, and cancer. Moreover, isoform selective modulators may increase our knowledge of sirtuin biology and aid to develop better therapies. Through this review, we provide critical insights into sirtuin pharmacology and illustrate their enzymatic activities and biological functions. Furthermore, we outline the most relevant sirtuin modulators in terms of their modes of action, structure-activity relationships, pharmacological effects, and clinical applications.
Collapse
Affiliation(s)
- Francesco Fiorentino
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
| | - Emanuele Fabbrizi
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
| | - Antonello Mai
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
- Pasteur Institute, Cenci‐Bolognetti FoundationSapienza University of RomeRomeItaly
| | - Dante Rotili
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
| |
Collapse
|
9
|
Zhang N, Jiang N, Chen Q. Key Regulators of Parasite Biology Viewed Through a Post-Translational Modification Repertoire. Proteomics 2024:e202400120. [PMID: 39690890 DOI: 10.1002/pmic.202400120] [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: 07/29/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024]
Abstract
Parasites are the leading causes of morbidity and mortality in both humans and animals, imposing substantial socioeconomic burdens worldwide. Controlling parasitic diseases has become one of the key issues in achieving "One Health". Most parasites have sophisticated life cycles exhibiting progressive developmental stages, morphologies, and host-switching, which are controlled by various regulatory machineries including protein post-translational modifications (PTMs). PTMs have emerged as a key mechanism by which parasites modulate their virulence, developmental transitions, and environmental adaptations. PTMs are enzyme-mediated additions or removals of chemical groups that dynamically regulate the stability and functions of proteins and confer novel properties, playing vital roles in a variety of biological processes and cellular functions. In this review, we circumscribe how parasites utilize various PTMs to regulate their intricate lives, with a focus on the biological role of PTMs in parasite biology and pathogenesis.
Collapse
Affiliation(s)
- Naiwen Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| |
Collapse
|
10
|
Santillán F, Charron CL, Galarreta BC, Luyt LG. Tailored peptide nanomaterials for receptor targeted prostate cancer imaging. NANOSCALE 2024; 16:22001-22010. [PMID: 39514187 DOI: 10.1039/d4nr03273j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
We report the development of a peptide-based optical nanoprobe specifically tailored for prostate cancer imaging. The imaging probe is comprised of cyclic peptide nanotubes, formed via the aqueous co-assembly of four cyclic D,L-alternating octapeptides. The inherent properties of these cyclic building blocks have been carefully selected to enhance their efficacy in imaging applications, through the addition of a cancer targeting peptide and a fluorescent dye. Comprehensive characterization using scanning electron microscopy (FESEM) and low-voltage transmission electron microscopy (LV-TEM) confirms the formation of nanotubes through co-assembly of the cyclic peptides. The resulting nanotubes show an average diameter of 28 nm. Circular dichroism (CD) spectroscopy validates the formation of stable beta-sheet hydrogen bonding structures at both 20 and 37 °C, ensuring their suitability for biomedical applications. Evaluation of PSMA-binding specificity of the resulting peptide nanotubes is assessed using confocal fluorescence microscopy demonstrating receptor-mediated uptake in prostate cancer cells. We anticipate this strategy will provide the basis for the utilization of co-assembled systems for advancing molecular imaging techniques in prostate cancer and other cancers.
Collapse
Affiliation(s)
- Fátima Santillán
- Department of Chemistry, University of Western Ontario, 1151 Richmond St, London, Ontario N6A 5B7, Canada
| | - Carlie L Charron
- Department of Chemistry, Dalhousie University, 6274 Coburg Road, Halifax, Nova Scotia B3H 4R2, Canada
| | - Betty C Galarreta
- Departamento Académico de Ciencias - Sección Química, Pontificia Universidad Católica del Perú, Av. Universitaria 1801, San Miguel, Lima 15088, Perú
| | - Leonard G Luyt
- Departments of Chemistry and Departments of Medical Imaging and Oncology, University of Western Ontario, University of Western Ontario, Centre, 1151 Richmond St, London, ON, N6A 3K7, Canada
- Verspeeten Family Cancer Centre, London Health Sciences, 800 Commissioners Rd. E., London, ON, N6A 5W9, Canada.
| |
Collapse
|
11
|
Mo C, Zhang W, Zhu K, Du Y, Huang W, Wu Y, Song J. Advances in Injectable Hydrogels Based on Diverse Gelation Methods for Biomedical Imaging. SMALL METHODS 2024; 8:e2400076. [PMID: 38470225 DOI: 10.1002/smtd.202400076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/01/2024] [Indexed: 03/13/2024]
Abstract
The injectable hydrogels can deliver the loads directly to the predetermined sites and form reservoirs to increase the enrichment and retention of the loads in the target areas. The preparation and injection of injectable hydrogels involve the sol-gel transformation of hydrogels, which is affected by factors such as temperature, ions, enzymes, light, mechanics (self-healing property), and pH. However, tracing the injection, degradation, and drug release from hydrogels based on different ways of gelation is a major concern. To solve this problem, contrast agents are introduced into injectable hydrogels, enabling the hydrogels to be imaged under techniques such as fluorescence imaging, photoacoustic imaging, magnetic resonance imaging, and radionuclide imaging. This review details methods for causing the gelation of imageable hydrogels; discusses the application of injectable hydrogels containing contrast agents in various imaging techniques, and finally explores the potential and challenges of imageable hydrogels based on different modes of gelation.
Collapse
Affiliation(s)
- Chunxiang Mo
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 10010, China
| | - Weiyao Zhang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 10010, China
| | - Kang Zhu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 10010, China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100190, China
| | - Wei Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Ying Wu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 10010, China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 10010, China
| |
Collapse
|
12
|
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
|
13
|
Guo Y, Li P, Guo X, Yao C, Yang D. Synthetic Nanoassemblies for Regulating Organelles: From Molecular Design to Precision Therapeutics. ACS NANO 2024; 18:30224-30246. [PMID: 39441007 DOI: 10.1021/acsnano.4c10194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Each organelle referring to a complex multiorder architecture executes respective biological processes via its distinct spatial organization and internal microenvironment. As the assembly of biomolecules is the structural basis of living cells, creating synthetic nanoassemblies with specific physicochemical and morphological properties in living cells to interfere or couple with the natural organelle architectures has attracted great attention in precision therapeutics of cancers. In this review, we give an overview of the latest advances in the synthetic nanoassemblies for precise organelle regulation, including the formation mechanisms, triggering strategies, and biomedical applications in precision therapeutics. We summarize the emerging material systems, including polymers, peptides, and deoxyribonucleic acids (DNAs), and their respective intermolecular interactions for intercellular synthetic nanoassemblies, and highlight their design principles in constructing precursors that assemble into synthetic nanoassemblies targeting specific organelles in the complex cellular environment. We further showcase the developed intracellular synthetic nanoassemblies targeting specific organelles including mitochondria, the endoplasmic reticulum, lysosome, Golgi apparatus, and nucleus and describe their underlying mechanisms for organelle regulation and precision therapeutics for cancer. Last, the essential challenges in this field and prospects for future precision therapeutics of synthetic nanoassemblies are discussed. This review should facilitate the rational design of organelle-targeting synthetic nanoassemblies and the comprehensive recognition of organelles by materials and contribute to the deep understanding and application of the synthetic nanoassemblies for precision therapeutics.
Collapse
Affiliation(s)
- Yanfei Guo
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, P.R. China
| | - Peiran Li
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Xiaocui Guo
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Chi Yao
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Dayong Yang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, P.R. China
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| |
Collapse
|
14
|
Gan S, Yang L, Heng Y, Chen Q, Wang D, Zhang J, Wei W, Liu Z, Njoku DI, Chen JL, Hu Y, Sun H. Enzyme-Directed and Organelle-Specific Sphere-to-Fiber Nanotransformation Enhances Photodynamic Therapy in Cancer Cells. SMALL METHODS 2024; 8:e2301551. [PMID: 38369941 PMCID: PMC11579569 DOI: 10.1002/smtd.202301551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/27/2024] [Indexed: 02/20/2024]
Abstract
Employing responsive nanoplatforms as carriers for photosensitizers represents an effective strategy to overcome the challenges associated with photodynamic therapy (PDT), including poor solubility, low bioavailability, and high systemic toxicity. Drawing inspiration from the morphology transitions in biological systems, a general approach to enhance PDT that utilizes enzyme-responsive nanoplatforms is developed. The transformation of phosphopeptide/photosensitizer co-assembled nanoparticles is first demonstrated into nanofibers when exposed to cytoplasmic enzyme alkaline phosphatase. This transition is primarily driven by alkaline phosphatase-induced changes of the nanoparticles in the hydrophilic and hydrophobic balance, and intermolecular electrostatic interactions within the nanoparticles. The resulting nanofibers exhibit improved ability of generating reactive oxygen species (ROS), intracellular accumulation, and retention in cancer cells. Furthermore, the enzyme-responsive nanoplatform is expanded to selectively target mitochondria by mitochondria-specific enzyme sirtuin 5 (SIRT5). Under the catalysis of SIRT5, the succinylated peptide/photosensitizer co-assembled nanoparticles can be transformed into nanofibers specifically within the mitochondria. The resulting nanofibers exhibit excellent capability of modulating mitochondrial activity, enhanced ROS formation, and significant anticancer efficacy via PDT. Consequently, the enzyme-instructed in situ fibrillar transformation of peptide/photosensitizers co-assembled nanoparticles provides an efficient pathway to address the challenges associated with photosensitizers. It is envisaged that this approach will further expand the toolbox for enzyme-responsive biomaterials for cancer therapy.
Collapse
Affiliation(s)
- Shenglong Gan
- Department of Chemistry and COSDAF (Centre of Super‐Diamond and Advanced Films) City University of Hong KongHong Kong999077China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057China
| | - Liu Yang
- College of Chemistry and Chemical EngineeringCentral South UniversityChangshaHunan410083China
| | - Yiyuan Heng
- Department of Chemistry and COSDAF (Centre of Super‐Diamond and Advanced Films) City University of Hong KongHong Kong999077China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057China
| | - Qingxin Chen
- Department of Chemistry and COSDAF (Centre of Super‐Diamond and Advanced Films) City University of Hong KongHong Kong999077China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057China
| | - Dongqing Wang
- Department of Chemistry and COSDAF (Centre of Super‐Diamond and Advanced Films) City University of Hong KongHong Kong999077China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057China
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical GeneticsDepartment of Laboratory MedicineSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610072China
| | - Jie Zhang
- Department of Chemistry and COSDAF (Centre of Super‐Diamond and Advanced Films) City University of Hong KongHong Kong999077China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057China
| | - Wenyu Wei
- Department of Chemistry and COSDAF (Centre of Super‐Diamond and Advanced Films) City University of Hong KongHong Kong999077China
| | - Zhiyang Liu
- Department of Chemistry and COSDAF (Centre of Super‐Diamond and Advanced Films) City University of Hong KongHong Kong999077China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057China
| | - Demian Ifeanyi Njoku
- Department of Applied ScienceHong Kong Metropolitan UniversityHo Man TinKowloonHong Kong999077China
| | - Jian Lin Chen
- Department of Applied ScienceHong Kong Metropolitan UniversityHo Man TinKowloonHong Kong999077China
| | - Yi Hu
- State Key Laboratory of ComplexSevereand Rare DiseasesBiomedical Engineering Facility of National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijing100730China
| | - Hongyan Sun
- Department of Chemistry and COSDAF (Centre of Super‐Diamond and Advanced Films) City University of Hong KongHong Kong999077China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057China
| |
Collapse
|
15
|
Wang H, Bai S, Gu G, Zhang C, Wang Y. Chemical Reaction Steers Spatiotemporal Self-Assembly of Supramolecular Hydrogels. Chempluschem 2024; 89:e202400396. [PMID: 38923325 DOI: 10.1002/cplu.202400396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
Supramolecular structures are widespread in living system, which are usually spatiotemporally regulated by sophisticated metabolic processes to enable vital biological functions. Inspired by living system, tremendous efforts have been made to realize spatiotemporal control over the self-assembly of supramolecular materials in synthetic scenario by coupling chemical reaction with molecular self-assembly process. In this review, we focused on the works related to supramolecular hydrogels that are regulated in space and time using chemical reaction. Firstly, we summarized how spatially controlled self-assembly of supramolecular hydrogels can be achieved via chemical reaction-instructed self-assembly, and the application of such a self-assembly methodology in biotherapy was discussed as well. Second, we reviewed dynamic supramolecular hydrogels dictated by chemical reaction networks that can evolve their structures and properties against time. Third, we discussed the recent progresses in the control of the self-assembly of supramolecular hydrogels in both space and time though a reaction-diffusion-coupled self-assembly approach. Finally, we provided a perspective on the further development of spatiotemporally controlled supramolecular hydrogels using chemical reaction in the future.
Collapse
Affiliation(s)
- Hucheng Wang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Shengyu Bai
- School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Guanyao Gu
- School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Chunyu Zhang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yiming Wang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Shanghai Key Laboratory for Intelligent Sensing and Detection Technology, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|
16
|
You J, Guo Y, Dong Z. Polypeptides-Based Nanocarriers in Tumor Therapy. Pharmaceutics 2024; 16:1192. [PMID: 39339228 PMCID: PMC11435007 DOI: 10.3390/pharmaceutics16091192] [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: 07/16/2024] [Revised: 09/07/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer remains a worldwide problem, and new treatment strategies are being actively developed. Peptides have the characteristics of good biocompatibility, strong targeting, functional diversity, modifiability, membrane permeable ability, and low immunogenicity, and they have been widely used to construct targeted drug delivery systems (DDSs). In addition, peptides, as endogenous substances, have a high affinity, which can not only regulate immune cells but also work synergistically with drugs to kill tumor cells, demonstrating significant potential for application. In this review, the latest progress of polypeptides-based nanocarriers in tumor therapy has been outlined, focusing on their applications in killing tumor cells and regulating immune cells. Additionally, peptides as carriers were found to primarily provide a transport function, which was also a subject of interest to us. At the end of the paper, the shortcomings in the construction of peptide nano-delivery system have been summarized, and possible solutions are proposed therein. The application of peptides provides a promising outlook for cancer treatment, and we hope this article can provide in-depth insights into possible future avenues of exploration.
Collapse
Affiliation(s)
- Juhua You
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yifei Guo
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Zhengqi Dong
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| |
Collapse
|
17
|
Song N, Tian F, Zou Y, Yu Z. Self-Assembly in Living Cells: Bottom-Up Syntheses in Natural Factory. ACS APPLIED MATERIALS & INTERFACES 2024; 16:45821-45829. [PMID: 39177358 DOI: 10.1021/acsami.4c10653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
In situ self-assembly in living systems is referred to as the processes that regulate assembly by stimuli-responsive reactions at target sites under physiological conditions. Due to the advantages of precisely forming well-defined nanostructures at pathological lesions, in situ-formed assemblies with tailored bioactivity are promising for the development of next-generation biomedical agents. In this Perspective, we summarize the progress of in situ self-assembly of peptides in living cells with an emphasis on the state-of-the-art strategies regulating assembly processes, establishing complexity within assembly systems, and exploiting their applications in biomedicines. We also provide our forward conceiving perspectives on the challenges in the development of in situ assembly in living cells to demonstrate its great potential in creating biomaterials for healthcare in the future.
Collapse
Affiliation(s)
- Na Song
- Shandong Provincial Engineering Research Center of Novel Pharmaceutical Excipients and Controlled Release Preparations, School of Pharmacy, Dezhou University, Dezhou 253000, China
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Feng Tian
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yixuan Zou
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zhilin Yu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, China
- Haihe Laboratory of Synthetic Biology, Tianjin 300308, China
| |
Collapse
|
18
|
Ok HW, Jin S, Park G, Jana B, Ryu JH. Folic Acid-Functionalized β-Cyclodextrin for Delivery of Organelle-Targeted Peptide Chemotherapeutics in Cancer. Mol Pharm 2024; 21:4498-4509. [PMID: 39069731 DOI: 10.1021/acs.molpharmaceut.4c00400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Recent emphasis on the design of drug delivery systems typically involves the effective transport of a pharmaceutical substance to the disease site with the desired therapeutic efficacy and minimal cytotoxicity. Organelle-targeted peptides have become an integral part of designing an important class of prodrug/prodrug assemblies for new supramolecular therapeutics owing to their favorable biocompatibility, synthetic ease, tunability of their aggregation behavior, and desired functionalization for site-specificity. However, it is still limited due to the low selectivity. We designed a folic acid-functionalized β-cyclodextrin (FA-CD) as a delivery platform for specific and selective delivery of organelle-targeted (such as microtubule, lysosome, and mitochondria) peptide chemotherapeutics to the folate receptor (FR) overexpressing cancer cell lines. Low toxicity was found for the FA-CD and organelle-targeted peptide inclusion complex in FR-negative normal cells, but superior inhibition of tumor growth with no in vivo toxicity was found for the inclusion complex in the xenograft tumor model.
Collapse
Affiliation(s)
- Hae Won Ok
- 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
| | - Gaeun Park
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Batakrishna Jana
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur 741246, India
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
19
|
Su H, Rong G, Li L, Cheng Y. Subcellular targeting strategies for protein and peptide delivery. Adv Drug Deliv Rev 2024; 212:115387. [PMID: 38964543 DOI: 10.1016/j.addr.2024.115387] [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: 04/19/2024] [Revised: 06/15/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Cytosolic delivery of proteins and peptides provides opportunities for effective disease treatment, as they can specifically modulate intracellular processes. However, most of protein-based therapeutics only have extracellular targets and are cell-membrane impermeable due to relatively large size and hydrophilicity. The use of organelle-targeting strategy offers great potential to overcome extracellular and cell membrane barriers, and enables localization of protein and peptide therapeutics in the organelles. Although progresses have been made in the recent years, organelle-targeted protein and peptide delivery is still challenging and under exploration. We reviewed recent advances in subcellular targeted delivery of proteins/peptides with a focus on targeting mechanisms and strategies, and highlight recent examples of active and passive organelle-specific protein and peptide delivery systems. This emerging platform could open a new avenue to develop more effective protein and peptide therapeutics.
Collapse
Affiliation(s)
- Hao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Guangyu Rong
- Department of Ophthalmology and Vision Science, Shanghai Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, 200030, China
| | - Longjie Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Yiyun Cheng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
20
|
Kim S, Lee Y, Seu MS, Sim Y, Ryu JH. Enzyme-instructed intramitochondrial polymerization for enhanced anticancer treatment without the development of drug-resistance. J Control Release 2024; 373:189-200. [PMID: 39002798 DOI: 10.1016/j.jconrel.2024.07.029] [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: 04/19/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Intracellular polymerization in living cells motivated chemists to generate polymeric structures with a multitude of possibilities to interact with biomacromolecules. However, out-of-control of the intracellular chemical reactions would be an obstacle restricting its application, providing the toxicity of non-targeted cells. Here, we reported intracellular thioesterase-mediated polymerization for selectively occurring polymerization using disulfide bonds in cancer cells. The acetylated monomers did not form disulfide bonds even under an oxidative environment, but they could polymerize into the polymeric structure after cleavage of acetyl groups only when encountered activity of thioesterase enzyme. Furthermore, acetylated monomers could be self-assembled with doxorubicin, providing doxorubicin loaded micelles for efficient intracellular delivery of drug and monomers. Since thioesterase enzymes were overexpressed in cancer cells specifically, the micelles were disrupted under activity of the enzyme and the polymerization could occur selectively in the cancer mitochondria. The resulting polymeric structures disrupted the mitochondrial membrane, thus activating the cellular death of cancer cells with high selectivity. This strategy selectively targets diverse cancer cells involving drug-resistant cells over normal cells. Moreover, the mitochondria targeting strategy overcomes the development of drug resistance even with repeated treatment. This approach provides a way for selective intracellular polymerization with desirable anticancer treatment.
Collapse
Affiliation(s)
- Sangpil Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Yeji Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Min-Seok Seu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Youjung Sim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| |
Collapse
|
21
|
Olov N, Nour S, Harris AR, Li D, Cook M, Williams RJ, Cheeseman S, Nisbet DR. Using Nanoscale Passports To Understand and Unlock Ion Channels as Gatekeepers of the Cell. ACS NANO 2024; 18:22709-22733. [PMID: 39136685 DOI: 10.1021/acsnano.4c05654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Natural ion channels are proteins embedded in the cell membrane that control many aspects of cell and human physiology by acting as gatekeepers, regulating the flow of ions in and out of cells. Advances in nanotechnology have influenced the methods for studying ion channels in vitro, as well as ways to unlock the delivery of therapeutics by modulating them in vivo. This review provides an overview of nanotechnology-enabled approaches for ion channel research with a focus on the synthesis and applications of synthetic ion channels. Further, the uses of nanotechnology for therapeutic applications are critically analyzed. Finally, we provide an outlook on the opportunities and challenges at the intersection of nanotechnology and ion channels. This work highlights the key role of nanoscale interactions in the operation and modulation of ion channels, which may prompt insights into nanotechnology-enabled mechanisms to study and exploit these systems in the near future.
Collapse
Affiliation(s)
- Nafiseh Olov
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| | - Shirin Nour
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Polymer Science Group, Department of Chemical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Alexander R Harris
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| | - Dan Li
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Mark Cook
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Department of Medicine, St Vincent's Hospital, Melbourne, Fitzroy, VIC 3065, Australia
| | - Richard J Williams
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Centre for Sustainable Bioproducts, Deakin University, Waurn Ponds, VIC 3217, Australia
- IMPACT, School of Medicine, Deakin University, Waurn Ponds, VIC 3217, Australia
| | - Samuel Cheeseman
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| | - David R Nisbet
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| |
Collapse
|
22
|
Tian F, Guo RC, Wu C, Liu X, Zhang Z, Wang Y, Wang H, Li G, Yu Z. Assembly of Glycopeptides in Living Cells Resembling Viral Infection for Cargo Delivery. Angew Chem Int Ed Engl 2024; 63:e202404703. [PMID: 38655625 DOI: 10.1002/anie.202404703] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
Self-assembly in living cells represents one versatile strategy for drug delivery; however, it suffers from the limited precision and efficiency. Inspired by viral traits, we here report a cascade targeting-hydrolysis-transformation (THT) assembly of glycosylated peptides in living cells holistically resembling viral infection for efficient cargo delivery and combined tumor therapy. We design a glycosylated peptide via incorporating a β-galactose-serine residue into bola-amphiphilic sequences. Co-assembling of the glycosylated peptide with two counterparts containing irinotecan (IRI) or ligand TSFAEYWNLLSP (PMI) results in formation of the glycosylated co-assemblies SgVEIP, which target cancer cells via β-galactose-galectin-1 association and undergo galactosidase-induced morphological transformation. While GSH-reduction causes release of IRI from the co-assemblies, the PMI moieties release p53 and facilitate cell death via binding with protein MDM2. Cellular experiments show membrane targeting, endo-/lysosome-mediated internalization and in situ formation of nanofibers in cytoplasm by SgVEIP. This cascade THT process enables efficient delivery of IRI and PMI into cancer cells secreting Gal-1 and overexpressing β-galactosidase. In vivo studies illustrate enhanced tumor accumulation and retention of the glycosylated co-assemblies, thereby suppressing tumor growth. Our findings demonstrate an in situ assembly strategy mimicking viral infection, thus providing a new route for drug delivery and cancer therapy in the future.
Collapse
Affiliation(s)
- Feng Tian
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Ruo-Chen Guo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Chunxia Wu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Xin Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Zeyu Zhang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Yamei Wang
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Science and Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Hao Wang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Gongyu Li
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Science and Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Zhilin Yu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
- Haihe Laboratory of Synthetic Biology, 21 West 15th Avenue, Tianjin, 300308, China
| |
Collapse
|
23
|
Tan W, Zhang Q, Lee M, Lau W, Xu B. Enzymatic control of intermolecular interactions for generating synthetic nanoarchitectures in cellular environment. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2373045. [PMID: 39011064 PMCID: PMC11249168 DOI: 10.1080/14686996.2024.2373045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/10/2024] [Accepted: 06/23/2024] [Indexed: 07/17/2024]
Abstract
Nanoarchitectonics, as a technology to arrange nano-sized structural units such as molecules in a desired configuration, requires nano-organization, which usually relies on intermolecular interactions. This review briefly introduces the development of using enzymatic reactions to control intermolecular interactions for generating artificial nanoarchitectures in a cellular environment. We begin the discussion with the early examples and uniqueness of enzymatically controlled self-assembly. Then, we describe examples of generating intracellular nanostructures and their relevant applications. Subsequently, we discuss cases of forming nanostructures on the cell surface via enzymatic reactions. Following that, we highlight the use of enzymatic reactions for creating intercellular nanostructures. Finally, we provide a summary and outlook on the promises and future direction of this strategy. Our aim is to give an updated introduction to the use of enzymatic reaction in regulating intermolecular interactions, a phenomenon ubiquitous in biology but relatively less explored by chemists and materials scientists. Our goal is to stimulate new developments in this simple and versatile approach for addressing societal needs.
Collapse
Affiliation(s)
- Weiyi Tan
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Qiuxin Zhang
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Mikki Lee
- Department of Chemistry, Brandeis University, Waltham, MA, USA
- Department of Pharmacy and Pharmaceutical Sciences, National University ofSingapore, Singapore
| | - William Lau
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, Waltham, MA, USA
| |
Collapse
|
24
|
Singh IR, Aggarwal N, Srivastava S, Panda JJ, Mishra J. Small Peptide-Based Nanodelivery Systems for Cancer Therapy and Diagnosis. J Pharmacol Exp Ther 2024; 390:30-44. [PMID: 37977815 DOI: 10.1124/jpet.123.001845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023] Open
Abstract
Developing nano-biomaterials with tunable topology, size, and surface characteristics has shown tremendously favorable benefits in various biologic and clinical applications. Among various nano-biomaterials, peptide-based drug delivery systems offer multiple merits over other synthetic systems due to their enhanced bio- and cytocompatibility and desirable biochemical and biophysical properties. Currently, around 100 peptide-based drugs are clinically available for numerous therapeutic purposes. In conjugation with chemotherapeutic moieties, peptides demonstrate a remarkable ability to reduce nonspecific drug effects by improving drug targetability at cancer sites. This review encompasses a wide-ranging role played by different peptide-based nanostructures in cancer theranostics. Section 1 introduces the rising concern about cancer as a disease and further describes peptide-based nanomaterials as biomedical agents to tackle the ailment. The subsequent section explores the mechanistic pathways behind the self-assembly of peptides to form hierarchically distinct assemblies. The crux of our review lies in an exhaustive exploration of the applications of various types of peptide-based nanostructures in cancer therapy and diagnosis. SIGNIFICANCE STATEMENT: Peptide-based drug delivery systems possess superior biocompatibility, biochemical, and biophysical properties compared to other synthetic alternatives. The development of these nano-biomaterials with customizable topology, size, and surface characteristics have shown promising outcomes in biomedical contexts. Peptides in conjunction with chemotherapeutic agents exhibit the ability to enhance drug targetability at cancer sites, reducing nonspecific drug effects. This comprehensive review emphasizes the pivotal role of diverse peptide-based nanostructures as cancer theranostics, elucidating their potential in revolutionizing cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Imocha Rajkumar Singh
- Chemical Biology Unit, Institute of Nano Science and Technology, Mohali, India (I.R.S., N.A., S.S., J.J.P.) and School of Biosciences, RIMT University, Mandi Gobindgarh, India (J.M.)
| | - Nidhi Aggarwal
- Chemical Biology Unit, Institute of Nano Science and Technology, Mohali, India (I.R.S., N.A., S.S., J.J.P.) and School of Biosciences, RIMT University, Mandi Gobindgarh, India (J.M.)
| | - Swapnil Srivastava
- Chemical Biology Unit, Institute of Nano Science and Technology, Mohali, India (I.R.S., N.A., S.S., J.J.P.) and School of Biosciences, RIMT University, Mandi Gobindgarh, India (J.M.)
| | - Jiban Jyoti Panda
- Chemical Biology Unit, Institute of Nano Science and Technology, Mohali, India (I.R.S., N.A., S.S., J.J.P.) and School of Biosciences, RIMT University, Mandi Gobindgarh, India (J.M.)
| | - Jibanananda Mishra
- Chemical Biology Unit, Institute of Nano Science and Technology, Mohali, India (I.R.S., N.A., S.S., J.J.P.) and School of Biosciences, RIMT University, Mandi Gobindgarh, India (J.M.)
| |
Collapse
|
25
|
Liu H, Wang H. From cells to subcellular organelles: Next-generation cancer therapy based on peptide self-assembly. Adv Drug Deliv Rev 2024; 209:115327. [PMID: 38703895 DOI: 10.1016/j.addr.2024.115327] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 05/06/2024]
Abstract
Due to the editability, functionality, and excellent biocompatibility of peptides, in situ self-assembly of peptides in cells is a powerful strategy for biomedical applications. Subcellular organelle targeting of peptides assemblies enables more precise drug delivery, enhances selectivity to disease cells, and mitigates drug resistance, providing an effective strategy for disease diagnosis and therapy. This reviewer first introduces the triggering conditions, morphological changes, and intracellular locations of self-assembling peptides. Then, the functions of peptide assemblies are summarized, followed by a comprehensive understanding of the interactions between peptide assemblies and subcellular organelles. Finally, we provide a brief outlook and the remaining challenges in this field.
Collapse
Affiliation(s)
- Huayang Liu
- Department of Chemistry, School of Science, Westlake University, No. 600 Dunyu Road, Sandun Town, Hangzhou 310024, Zhejiang Province, China; Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Huaimin Wang
- Department of Chemistry, School of Science, Westlake University, No. 600 Dunyu Road, Sandun Town, Hangzhou 310024, Zhejiang Province, China; Institute of Natural Sciences, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China.
| |
Collapse
|
26
|
Zhang Q, Tan W, Liu Z, Zhang Y, Wei WS, Fraden S, Xu B. Unnatural Peptide Assemblies Rapidly Deplete Cholesterol and Potently Inhibit Cancer Cells. J Am Chem Soc 2024; 146:12901-12906. [PMID: 38701349 PMCID: PMC11223060 DOI: 10.1021/jacs.4c03101] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Cholesterol-rich membranes play a pivotal role in cancer initiation and progression, necessitating innovative approaches to target these membranes for cancer inhibition. Here we report the first case of unnatural peptide (1) assemblies capable of depleting cholesterol and inhibiting cancer cells. Peptide 1 self-assembles into micelles and is rapidly taken up by cancer cells, especially when combined with an acute cholesterol-depleting agent (MβCD). Click chemistry has confirmed that 1 depletes cell membrane cholesterol. It localizes in membrane-rich organelles, including the endoplasmic reticulum, Golgi apparatus, and lysosomes. Furthermore, 1 potently inhibits malignant cancer cells, working synergistically with cholesterol-lowering agents. Control experiments have confirmed that C-terminal capping and unnatural amino acid residues (i.e., BiP) are essential for both cholesterol depletion and potent cancer cell inhibition. This work highlights unnatural peptide assemblies as a promising platform for targeting the cell membrane in controlling cell fates.
Collapse
Affiliation(s)
- Qiuxin Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Zhiyu Liu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Yichi Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Wei-Shao Wei
- Martin A. Fisher School of Physics, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Seth Fraden
- Martin A. Fisher School of Physics, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| |
Collapse
|
27
|
Hu JJ, Lin N, Zhang Y, Xia F, Lou X. Nanofibers in Organelles: From Structure Design to Biomedical Applications. Angew Chem Int Ed Engl 2024; 63:e202313139. [PMID: 37889872 DOI: 10.1002/anie.202313139] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 10/29/2023]
Abstract
Nanofibers are one of the most important morphologies of molecular self-assemblies, the formation of which relies on the diverse intermolecular interactions of fibrous-forming units. In the past decade, rapid advances have been made in the biomedical application of nanofibers, such as bioimaging and tumor treatment. An important topic to be focused on is not only the nanofiber formation mechanism but also where it forms, because different destinations could have different influences on cells and its formation could be triggered by unique stimuli in organelles. It is therefore necessary and timely to summarize the nanofibers assembled in organelles. This minireview discusses the formation mechanism, triggering strategies, and biomedical applications of nanofibers, which may facilitate the rational design of nanofibers, improve our understanding of the relationship between nanofiber properties and organelle characteristics, allow a comprehensive recognition of organelles affected by materials, and enhance the therapeutic efficiency of nanofibers.
Collapse
Affiliation(s)
- Jing-Jing Hu
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Niya Lin
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Yunfan Zhang
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Fan Xia
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Xiaoding Lou
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| |
Collapse
|
28
|
Li F, Tang R, Kang Y, Cui X, Wang Y, Yang X. Fluorescent composite based on peptide nanotubes activating coumarin 6 for sensitive detection of new coccine in food samples. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 305:123492. [PMID: 37844452 DOI: 10.1016/j.saa.2023.123492] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/18/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
New coccine (NC), as a kind of common colorant, has been frequently used in our daily life. Herein, the fluorescent composite (PNTs@C6) prepared by the hydrophobic non-covalent interaction between peptide nanotubes and coumarin 6 (C6) was designed for the determination of NC. Due to the activation of C6 by peptide nanotubes, the composite exhibits strong green fluorescence emission, which can be selectively quenched by NC through the inner filter effect. Therefore, a new fluorescent method based on the PNTs@C6 composite for NC detection was constructed. Under optimal conditions, the fluorescence quenching of the sensor exhibits a good linear relationship with the concentration of NC in the range of 0.01-10 μM and the limit of detection is 3.6 nM. Furthermore, the strategy shows simplicity, rapid response and high selectivity and has been successfully applied to the detection of NC in food samples.
Collapse
Affiliation(s)
- Fang Li
- College of Chemistry and Chemical Engineering, Chemical Synthesis and Pollution Control Key Laboratory of Sichuan Province, China West Normal University, Nanchong 637000, China
| | - Rong Tang
- College of Chemistry and Chemical Engineering, Chemical Synthesis and Pollution Control Key Laboratory of Sichuan Province, China West Normal University, Nanchong 637000, China
| | - Yujie Kang
- College of Chemistry and Chemical Engineering, Chemical Synthesis and Pollution Control Key Laboratory of Sichuan Province, China West Normal University, Nanchong 637000, China
| | - Xiaoyan Cui
- Nanchong Food and Drug Inspection Institute, Nanchong 637000, China
| | - Ya Wang
- College of Chemistry and Chemical Engineering, Chemical Synthesis and Pollution Control Key Laboratory of Sichuan Province, China West Normal University, Nanchong 637000, China.
| | - Xiupei Yang
- College of Chemistry and Chemical Engineering, Chemical Synthesis and Pollution Control Key Laboratory of Sichuan Province, China West Normal University, Nanchong 637000, China.
| |
Collapse
|
29
|
Wang Y, Xie L, Li X, Wang L, Yang Z. Chemo-immunotherapy by dual-enzyme responsive peptide self-assembling abolish melanoma. Bioact Mater 2024; 31:549-562. [PMID: 37746663 PMCID: PMC10511343 DOI: 10.1016/j.bioactmat.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/10/2023] [Accepted: 09/10/2023] [Indexed: 09/26/2023] Open
Abstract
Herein, we designed Comp. 1 to simultaneously respond to two enzymes: alkaline phosphatase and matrix metalloproteinase 2, which is commonly found in highly malignant cancer cell lines containing B16-F10 murine melanoma cells and CT26 murine colon carcinoma cells. We used the regional differences in the expression levels of dual-markers to accurately release immune molecule IND into tumor microenvironment for the activation of anti-tumor related immune effects, while in-situ self-assembly occurs. The dual-enzyme response process can further regulate the peptide precursors' self-assembly in the form of short rod-shaped nanofibers, enabling the delivery of the loaded chemotherapeutic drug HCPT into the cancer cells and further allowing the peptide assemblies to escape from lysosomes and return to cytoplasm in the form of tiny nanoparticles to induce apoptosis of cancer cells. This process does not occur in the single-positive breast cancer cell line MCF-7 or the normal hepatocytes cell line LO2, indicating the selectivity of the cancer cells exhibited using our strategy. In vivo studies revealed that Comp. 1 can effectively cooperate with chemotherapy to enhance the immunotherapy effect and induce immune responses associated with elevated pro-inflammatory cytokines in vivo to inhibit malignant tumors growth. Our dual-enzyme responsive chemo-immunotherapy strategy feasible in anti-tumor treatment, provides a new avenue for regulating peptide self-assembly to adapt to diverse tumor properties and may eventually be used for the development of novel multifunctional anti-tumor nanomedicines.
Collapse
Affiliation(s)
- Yuhan Wang
- Tianjin Key Laboratory of Inflammation Biology, Department of Pharmacology, School of Basic Medicine, Tianjin Medical University, Tianjin, 300070, PR China
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| | - Limin Xie
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| | - Xinxin Li
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| | - Ling Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300071, PR China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, National Institute of Functional Materials, Nankai University, Tianjin, 300071, PR China
| |
Collapse
|
30
|
Zhang Q, Yu S, Shang J, He S, Liu X, Wang F. Spatiotemporally Programmed Disassembly of Multifunctional Integrated DNAzyme Nanoplatfrom for Amplified Intracellular MicroRNA Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305672. [PMID: 37670211 DOI: 10.1002/smll.202305672] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/12/2023] [Indexed: 09/07/2023]
Abstract
The sensing performance of DNAzymes in live cells is tremendously hampered by the inefficient and inhomogeneous delivery of DNAzyme probes and their incontrollable off-site activation, originating from their susceptibility to nuclease digestion. This requires the development of a more compact and robust DNAzyme-delivering system with site-specific DNAzyme activation property. Herein, a highly compact and robust Zn@DDz nanoplatform is constructed by integrating the unimolecular microRNA-responsive DNA-cleaving DNAzyme (DDz) probe with the requisite DNAzyme Zn2+ -ion cofactors, and the amplified intracellular imaging of microRNA via the spatiotemporally programmed disassembly of Zn@DDz nanoparticles is achieved. The multifunctional Zn@DDz nanoplatform is simply composed of a structurally blocked self-hydrolysis DDz probe and the inorganic Zn2+ -ion bridge, with high loading capacity, and can effectively deliver the initially catalytic inert DDz probe and Zn2+ into living cells with enhanced stabilities. Upon their entry into the acidic microenvironment of living cells, the self-sufficient Zn@DDz nanoparticle is disassembled to release DDz probe and simultaneously supply Zn2+ -ion cofactors. Then, endogenous microRNA-21 catalyzes the reconfiguration and activation of DDz for generating the amplified readout signal with multiply guaranteed imaging performance. Thus, this work paves an effective way for promoting DNAzyme-based biosensing systems in living cells, and shows great promise in clinical diagnosis.
Collapse
Affiliation(s)
- Qingqing Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, P. R. China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
- Research Institute of Shenzhen, Wuhan University, Shenzhen, 518057, P. R. China
| | - Shanshan Yu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, P. R. China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
- Research Institute of Shenzhen, Wuhan University, Shenzhen, 518057, P. R. China
| | - Jinhua Shang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, P. R. China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
- Research Institute of Shenzhen, Wuhan University, Shenzhen, 518057, P. R. China
| | - Shizhen He
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, P. R. China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
- Research Institute of Shenzhen, Wuhan University, Shenzhen, 518057, P. R. China
| | - Xiaoqing Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, P. R. China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
- Research Institute of Shenzhen, Wuhan University, Shenzhen, 518057, P. R. China
| | - Fuan Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430072, P. R. China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, P. R. China
- Research Institute of Shenzhen, Wuhan University, Shenzhen, 518057, P. R. China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, 430072, China
| |
Collapse
|
31
|
Li M, Yu H, Li Y, Li X, Huang S, Liu X, Weng G, Xu L, Hou T, Guo DS, Wang Y. Rational design of supramolecular self-assembly sensor for living cell imaging of HDAC1 and its application in high-throughput screening. Biosens Bioelectron 2023; 242:115716. [PMID: 37820557 DOI: 10.1016/j.bios.2023.115716] [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: 07/23/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
Supramolecular chemistry offers new insights in bioimaging, but specific tracking of enzyme in living cells via supramolecular host-guest reporter pair remains challenging, largely due to the interference caused by the complex cellular environment on the binding between analytes and hosts. Here, by exploiting the principle of supramolecular tandem assay (STA) and the classic host-guest reporter pair (p-sulfonatocalix[4]arene (SC4A) and lucigenin (LCG)) and rationally designing artificial peptide library to screen sequence with high affinity of the target enzyme, we developed a "turn-on" fluorescent sensing system for intracellular imaging of histone deacetylase 1 (HDAC1), which is a potential therapeutic target for various diseases, including cancer, neurological, and cardiovascular diseases. Based on computational simulations and experimental validations, we verified that the deacetylated peptide by HDAC1 competed LCG, freeing it from the SC4A causing fluorescence increase. Enzyme kinetics experiments were further conducted to prove that this assay could detect HDAC1 specifically with high sensitivity (the LOD value is 0.015 μg/mL, ten times lower than the published method). This system was further applied for high-throughput screening of HDAC1 inhibitors over a natural compound library containing 147 compounds, resulting in the identification of a novel HDAC1 down-regulator (Ginsenoside RK3). Our results demonstrated the sensitivity and robustness of the assay system towards HDAC1. It should serve as a valuable tool for biochemical studies and drug screening.
Collapse
Affiliation(s)
- Min Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huijuan Yu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Nankai University, Tianjin 300071, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yiran Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xin Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shiqing Huang
- Fluorescence Research Group, Singapore University of Technology and Design, 8 Somapah Road 487372, Singapore
| | - Xiaogang Liu
- Fluorescence Research Group, Singapore University of Technology and Design, 8 Somapah Road 487372, Singapore
| | - Gaoqi Weng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Nankai University, Tianjin 300071, China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Future Health Laboratory Innovation Center of Yangtze River Delta Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
32
|
Guo RC, Wang N, Wang W, Zhang Z, Luo W, Wang Y, Du H, Xu Y, Li G, Yu Z. Artificial Peptide-Protein Necrosomes Promote Cell Death. Angew Chem Int Ed Engl 2023; 62:e202314578. [PMID: 37870078 DOI: 10.1002/anie.202314578] [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: 09/28/2023] [Revised: 10/12/2023] [Accepted: 10/23/2023] [Indexed: 10/24/2023]
Abstract
The presence of disordered region or large interacting surface within proteins significantly challenges the development of targeted drugs, commonly known as the "undruggable" issue. Here, we report a heterogeneous peptide-protein assembling strategy to selectively phosphorylate proteins, thereby activating the necroptotic signaling pathway and promoting cell necroptosis. Inspired by the structures of natural necrosomes formed by receptor interacting protein kinases (RIPK) 1 and 3, the kinase-biomimetic peptides are rationally designed by incorporating natural or D -amino acids, or connecting D -amino acids in a retro-inverso (DRI) manner, leading to one RIPK3-biomimetic peptide PR3 and three RIPK1-biomimetic peptides. Individual peptides undergo self-assembly into nanofibrils, whereas mixing RIPK1-biomimetic peptides with PR3 accelerates and enhances assembly of PR3. In particular, RIPK1-biomimetic peptide DRI-PR1 exhibits reliable binding affinity with protein RIPK3, resulting in specific cytotoxicity to colon cancer cells that overexpress RIPK3. Mechanistic studies reveal the increased phosphorylation of RIPK3 induced by RIPK1-biomimetic peptides, elucidating the activation of the necroptotic signaling pathway responsible for cell death without an obvious increase in secretion of inflammatory cytokines. Our findings highlight the potential of peptide-protein hybrid aggregation as a promising approach to address the "undruggable" issue and provide alternative strategies for overcoming cancer resistance in the future.
Collapse
Affiliation(s)
- Ruo-Chen Guo
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Ning Wang
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Science and Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Weishu Wang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zeyu Zhang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Wendi Luo
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yushi Wang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Haiqin Du
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Yifei Xu
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Gongyu Li
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Science and Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhilin Yu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
33
|
Zhang X, Wang J, Zhang Y, Yang Z, Gao J, Gu Z. Synthesizing biomaterials in living organisms. Chem Soc Rev 2023; 52:8126-8164. [PMID: 37921625 DOI: 10.1039/d2cs00999d] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Living organisms fabricate biomacromolecules such as DNA, RNA, and proteins by the self-assembly process. The research on the mechanism of biomacromolecule formation also inspires the exploration of in vivo synthesized biomaterials. By elaborate design, artificial building blocks or precursors can self-assemble or polymerize into functional biomaterials within living organisms. In recent decades, these so-called in vivo synthesized biomaterials have achieved extensive applications in cell-fate manipulation, disease theranostics, bioanalysis, cellular surface engineering, and tissue regeneration. In this review, we classify strategies for in vivo synthesis into non-covalent, covalent, and genetic types. The development of these approaches is based on the chemical principles of supramolecular chemistry and synthetic chemistry, biological cues such as enzymes and microenvironments, and the means of synthetic biology. By summarizing the design principles in detail, some insights into the challenges and opportunities in this field are provided to enlighten further research.
Collapse
Affiliation(s)
- Xiangyang Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Junxia Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
34
|
Kim S, Chae JB, Kim D, Park CW, Sim Y, Lee H, Park G, Lee J, Hong S, Jana B, Kim C, Chung H, Ryu JH. Supramolecular Senolytics via Intracellular Oligomerization of Peptides in Response to Elevated Reactive Oxygen Species Levels in Aging Cells. J Am Chem Soc 2023; 145:21991-22008. [PMID: 37664981 DOI: 10.1021/jacs.3c06898] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Senolytics, which eliminate senescent cells from tissues, represent an emerging therapeutic strategy for various age-related diseases. Most senolytics target antiapoptotic proteins, which are overexpressed in senescent cells, limiting specificity and inducing severe side effects. To overcome these limitations, we constructed self-assembling senolytics targeting senescent cells with an intracellular oligomerization system. Intracellular aryl-dithiol-containing peptide oligomerization occurred only inside the mitochondria of senescent cells due to selective localization of the peptides by RGD-mediated cellular uptake into integrin αvβ3-overexpressed senescent cells and elevated levels of reactive oxygen species, which can be used as a chemical fuel for disulfide formation. This oligomerization results in an artificial protein-like nanoassembly with a stable α-helix secondary structure, which can disrupt the mitochondrial membrane via multivalent interactions because the mitochondrial membrane of senescent cells has weaker integrity than that of normal cells. These three specificities (integrin αvβ3, high ROS, and weak mitochondrial membrane integrity) of senescent cells work in combination; therefore, this intramitochondrial oligomerization system can selectively induce apoptosis of senescent cells without side effects on normal cells. Significant reductions in key senescence markers and amelioration of retinal degeneration were observed after elimination of the senescent retinal pigment epithelium by this peptide senolytic in an age-related macular degeneration mouse model and in aged mice, and this effect was accompanied by improved visual function. This system provides a strategy for the treatment of age-related diseases using supramolecular senolytics.
Collapse
Affiliation(s)
- Sangpil Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jae-Byoung Chae
- Department of Ophthalmology, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Dohyun Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Chul-Woo Park
- Department of Ophthalmology, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Youjung Sim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hyungwoo Lee
- Department of Ophthalmology, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Gaeun Park
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jaeeun Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seongho Hong
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Batakrishna Jana
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Chaekyu Kim
- Fusion Biotechnology, Ulsan 44919, Republic of Korea
| | - Hyewon Chung
- Department of Ophthalmology, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
35
|
Pacheco-Liñán P, Alonso-Moreno C, Ocaña A, Ripoll C, García-Gil E, Garzón-Ruíz A, Herrera-Ochoa D, Blas-Gómez S, Cohen B, Bravo I. Formation of Highly Emissive Anthracene Excimers for Aggregation-Induced Emission/Self-Assembly Directed (Bio)imaging. ACS APPLIED MATERIALS & INTERFACES 2023; 15:44786-44795. [PMID: 37699547 PMCID: PMC11165449 DOI: 10.1021/acsami.3c10823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023]
Abstract
AIEgens have emerged as a promising alternative to molecular rotors in bioimaging applications. However, transferring the concept of aggregation-induced emission (AIE) from solution to living systems remains a challenge. Given the highly heterogeneous nature and the compartmentalization of the cell, different approaches are needed to control the self-assembly within the crowded intricate cellular environment. Herein, we report for the first time the self-assembly mechanism of an anthracene-guanidine derivative (AG) forming the rare and highly emissive T-shaped dimer in breast cancer cell lines as a proof of concept. This process is highly sensitive to the local environment in terms of polarity, viscosity, and/or water quantity that should enable the use of the AG as a fluorescence lifetime imaging biosensor for intracellular imaging of cellular structures and the monitoring of intracellular state parameters. Different populations of the monomer and T-shaped and π-π dimers were observed in the cell membrane, cytoplasm, and nucleoplasm, related to the local viscosity and presence of water. The T-shaped dimer is formed preferentially in the nucleus because of the higher density and viscosity compared to the cytoplasm. The present results should serve as a precursor for the development of new design strategies for molecular systems for a wide range of applications such as cell viscosity, density, or temperature sensing and imaging.
Collapse
Affiliation(s)
- Pedro
J. Pacheco-Liñán
- Unidad
nanoDrug. Facultad de Farmacia de Albacete, Universidad de Castilla-La
Mancha, 02008 Albacete, Spain
| | - Carlos Alonso-Moreno
- Unidad
nanoDrug. Facultad de Farmacia de Albacete, Universidad de Castilla-La
Mancha, 02008 Albacete, Spain
- Centro
Regional de Investigaciones Biomédicas (CRIB), 02008 Albacete, Spain
- Centro
de Innovación en Química Avanzada (ORFEO-CINQA), Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Alberto Ocaña
- Experimental
Therapeutics Unit, Hospital clínico
San Carlos, IdISSC and CIBERONC, 28040 Madrid, Spain
- Unidad
de Investigación del Complejo Hospitalario Universitario de
Albacete. Oncología Traslacional, 02008 Albacete, Spain
| | - Consuelo Ripoll
- Unidad
nanoDrug. Facultad de Farmacia de Albacete, Universidad de Castilla-La
Mancha, 02008 Albacete, Spain
| | - Elena García-Gil
- Unidad
de Investigación del Complejo Hospitalario Universitario de
Albacete. Oncología Traslacional, 02008 Albacete, Spain
| | - Andrés Garzón-Ruíz
- Unidad
nanoDrug. Facultad de Farmacia de Albacete, Universidad de Castilla-La
Mancha, 02008 Albacete, Spain
| | - Diego Herrera-Ochoa
- Unidad
nanoDrug. Facultad de Farmacia de Albacete, Universidad de Castilla-La
Mancha, 02008 Albacete, Spain
| | - Sofía Blas-Gómez
- Unidad
nanoDrug. Facultad de Farmacia de Albacete, Universidad de Castilla-La
Mancha, 02008 Albacete, Spain
| | - Boiko Cohen
- Departamento
de Química Física, Facultad de Ciencias Ambientales
y Bioquímica, and Instituto de Nanociencia, Nanotecnología
y Materiales Moleculares (INAMOL), Universidad
de Castilla-La Mancha, Avenida Carlos III, S/N, 45071 Toledo, Spain
| | - Iván Bravo
- Unidad
nanoDrug. Facultad de Farmacia de Albacete, Universidad de Castilla-La
Mancha, 02008 Albacete, Spain
- Centro
Regional de Investigaciones Biomédicas (CRIB), 02008 Albacete, Spain
| |
Collapse
|
36
|
Jana B, Jin S, Go EM, Cho Y, Kim D, Kim S, Kwak SK, Ryu JH. Intra-Lysosomal Peptide Assembly for the High Selectivity Index against Cancer. J Am Chem Soc 2023; 145:18414-18431. [PMID: 37525328 DOI: 10.1021/jacs.3c04467] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Lysosomes remain powerful organelles and important targets for cancer therapy because cancer cell proliferation is greatly dependent on effective lysosomal function. Recent studies have shown that lysosomal membrane permeabilization induces cell death and is an effective way to treat cancer by bypassing the classical caspase-dependent apoptotic pathway. However, most lysosome-targeted anticancer drugs have very low selectivity for cancer cells. Here, we show intra-lysosomal self-assembly of a peptide amphiphile as a powerful technique to overcome this problem. We designed a peptide amphiphile that localizes in the cancer lysosome and undergoes cathepsin B enzyme-instructed supramolecular assembly. This localized assembly induces lysosomal swelling, membrane permeabilization, and damage to the lysosome, which eventually causes caspase-independent apoptotic death of cancer cells without conventional chemotherapeutic drugs. It has specific anticancer effects and is effective against drug-resistant cancers. Moreover, this peptide amphiphile exhibits high tumor targeting when attached to a tumor-targeting ligand and causes significant inhibition of tumor growth both in cancer and drug-resistant cancer xenograft models.
Collapse
Affiliation(s)
- Batakrishna Jana
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seongeon Jin
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Eun Min Go
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Yumi Cho
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Dohyun Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sangpil Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sang Kyu Kwak
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
37
|
Zhan W, Gao G, Liu Z, Liu X, Xu L, Wang M, Xu HD, Tang R, Cao J, Sun X, Liang G. Enzymatic Self-Assembly of Adamantane-Peptide Conjugate for Combating Staphylococcus aureus Infection. Adv Healthc Mater 2023; 12:e2203283. [PMID: 36880480 DOI: 10.1002/adhm.202203283] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/28/2023] [Indexed: 03/08/2023]
Abstract
Staphylococcus aureus (S. aureus) remains a leading cause of bacterial infections. However, eradication of S. aureus infections with common antibiotics is increasingly difficult due to outbreaks of drug resistance. Therefore, new antibiotic classes and antibacterial strategies are urgently in demand. Herein, it is shown that an adamantane-peptide conjugate, upon dephosphorylation by alkaline phosphatase (ALP) constitutively expressed on S. aureus, generates fibrous assemblies in situ to combat S. aureus infection. By attaching adamantane to a phosphorylated tetrapeptide Nap-Phe-Phe-Lys-Tyr(H2 PO3 )-OH, the rationally designed adamantane-peptide conjugate Nap-Phe-Phe-Lys(Ada)-Tyr(H2 PO3 )-OH (Nap-FYp-Ada) is obtained. Upon bacterial ALP activation, Nap-FYp-Ada is dephosphorylated and self-assembles into nanofibers on the surface of S. aureus. As revealed by cell assays, the assemblies of adamantane-peptide conjugates interact with cell lipid membrane and thereby disrupt membrane integrity to kill S. aureus. Animal experiments further demonstrate the excellent potential of Nap-FYp-Ada in the treatment of S. aureus infection in vivo. This work provides an alternative approach to design antimicrobial agents.
Collapse
Affiliation(s)
- Wenjun Zhan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Ge Gao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Zhiyu Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Lingling Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Manli Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Hai-Dong Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Runqun Tang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Jingyuan Cao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Xianbao Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| | - Gaolin Liang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing, 210096, P. R. China
| |
Collapse
|
38
|
Wang Y, Wang P, Li C. Fluorescence microscopic platforms imaging mitochondrial abnormalities in neurodegenerative diseases. Adv Drug Deliv Rev 2023; 197:114841. [PMID: 37088402 DOI: 10.1016/j.addr.2023.114841] [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: 01/07/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 04/25/2023]
Abstract
Neurodegenerative diseases (NDs) are progressive disorders that cause the degeneration of neurons. Mitochondrial dysfunction is a common symptom in NDs and plays a crucial role in neuronal loss. Mitochondrial abnormalities can be observed in the early stages of NDs and evolve throughout disease progression. Visualizing mitochondrial abnormalities can help understand ND progression and develop new therapeutic strategies. Fluorescence microscopy is a powerful tool for dynamically imaging mitochondria due to its high sensitivity and spatiotemporal resolution. This review discusses the relationship between mitochondrial dysfunction and ND progression, potential biomarkers for imaging dysfunctional mitochondria, advances in fluorescence microscopy for detecting organelles, the performance of fluorescence probes in visualizing ND-associated mitochondria, and the challenges and opportunities for developing new generations of fluorescence imaging platforms for monitoring mitochondria in NDs.
Collapse
Affiliation(s)
- Yicheng Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy; Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pengwei Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy; Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy; Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University Shanghai 201203, China.
| |
Collapse
|
39
|
Zhou Y, Li Q, Wu Y, Li X, Zhou Y, Wang Z, Liang H, Ding F, Hong S, Steinmetz NF, Cai H. Molecularly Stimuli-Responsive Self-Assembled Peptide Nanoparticles for Targeted Imaging and Therapy. ACS NANO 2023; 17:8004-8025. [PMID: 37079378 DOI: 10.1021/acsnano.3c01452] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Self-assembly has emerged as an extensively used method for constructing biomaterials with sizes ranging from nanometers to micrometers. Peptides have been extensively investigated for self-assembly. They are widely applied owing to their desirable biocompatibility, biodegradability, and tunable architecture. The development of peptide-based nanoparticles often requires complex synthetic processes involving chemical modification and supramolecular self-assembly. Stimuli-responsive peptide nanoparticles, also termed "smart" nanoparticles, capable of conformational and chemical changes in response to stimuli, have emerged as a class of promising materials. These smart nanoparticles find a diverse range of biomedical applications, including drug delivery, diagnostics, and biosensors. Stimuli-responsive systems include external stimuli (such as light, temperature, ultrasound, and magnetic fields) and internal stimuli (such as pH, redox environment, salt concentration, and biomarkers), facilitating the generation of a library of self-assembled biomaterials for biomedical imaging and therapy. Thus, in this review, we mainly focus on peptide-based nanoparticles built by self-assembly strategy and systematically discuss their mechanisms in response to various stimuli. Furthermore, we summarize the diverse range of biomedical applications of peptide-based nanomaterials, including diagnosis and therapy, to demonstrate their potential for medical translation.
Collapse
Affiliation(s)
- Yang Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Qianqian Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Ye Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Xinyu Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Ya Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Zhu Wang
- Department of Urology, Affiliated People's Hospital of Longhua Shenzhen, Southern Medical University, 38 Jinglong Jianshe Road, Shenzhen, Guangdong 518109, PR China
| | - Hui Liang
- Department of Urology, Affiliated People's Hospital of Longhua Shenzhen, Southern Medical University, 38 Jinglong Jianshe Road, Shenzhen, Guangdong 518109, PR China
| | - Feiqing Ding
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Sheng Hong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| | - Nicole F Steinmetz
- Department of NanoEngineering, Department of Biongineering, Department of Radiology, Moores Cancer Center, Center for Nano-ImmunoEngineering, Center for Engineering in Cancer, Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, California 92093, United States
| | - Hui Cai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchang Road, Guangming District, Shenzhen 518107, China
| |
Collapse
|
40
|
Sun Y, Lyu B, Yang C, He B, Zhang H, Wang X, Zhang Q, Dai W. An enzyme-responsive and transformable PD-L1 blocking peptide-photosensitizer conjugate enables efficient photothermal immunotherapy for breast cancer. Bioact Mater 2023; 22:47-59. [PMID: 36203955 PMCID: PMC9519467 DOI: 10.1016/j.bioactmat.2022.08.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/02/2022] [Accepted: 08/15/2022] [Indexed: 12/07/2022] Open
Abstract
Mild photothermal therapy combined with immune checkpoint blockade has received increasing attention for the treatment of advanced or metastatic cancers due to its good therapeutic efficacy. However, it remains a challenge to facilely integrate the two therapies and make it potential for clinical translation. This work designed a peptide-photosensitizer conjugate (PPC), which consisted of a PD-L1 antagonist peptide (CVRARTR), an MMP-2 specific cleavable sequence, a self-assembling motif, and the photosensitizer Purpurin 18. The single-component PPC can self-assemble into nanospheres which is suitable for intravenous injection. The PPC nanosphere is cleaved by MMP-2 when it accumulates in tumor sites, thereby initiating the cancer-specific release of the antagonist peptide. Simultaneously, the nanospheres gradually transform into co-assembled nanofibers, which promotes the retention of the remaining parts within the tumor. In vivo studies demonstrated that PPC nanospheres under laser irradiation promote the infiltration of cytotoxic T lymphocytes and maturation of DCs, which sensitize 4T1 tumor cells to immune checkpoint blockade therapy. Therefore, PPC nanospheres inhibit tumor growth efficiently both in situ and distally and blocked the formation of lung metastases. The present study provides a simple and efficient integrated strategy for breast cancer photoimmunotherapy. A peptide-photosensitizer conjugate (PPC) with self-assembled ability. Self-assembled PPC realized enzyme-responsive PD-L1 blocking peptide release. Shape transformation from nanospheres to co-assembled nanofibers. Efficient integrated strategy for breast cancer photoimmunotherapy.
Collapse
Affiliation(s)
- Yanan Sun
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang, 050017, China
| | - Bochen Lyu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Chang Yang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Corresponding author.
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Corresponding author.
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Corresponding author.
| |
Collapse
|
41
|
Liu Y, Wu Y, Luo Z, Li M. Designing supramolecular self-assembly nanomaterials as stimuli-responsive drug delivery platforms for cancer therapy. iScience 2023; 26:106279. [PMID: 36936787 PMCID: PMC10014307 DOI: 10.1016/j.isci.2023.106279] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Stimuli-responsive nanomaterials have attracted substantial interest in cancer therapy, as they hold promise to deliver anticancer agents to tumor sites in a precise and on-demand manner. Interestingly, supramolecular chemistry is a burgeoning discipline that entails the reversible bonding between components at the molecular and nanoscale levels, and the recent advances in this area offer the possibility to design nanotherapeutics with improved controllability and functionality for cancer therapy. Herein, we provide a comprehensive summary of typical non-covalent interaction modes, which primarily include hydrophobic interaction, hydrogel bonding, host-guest interaction, π-π stacking, and electrostatic interaction. Special emphasis is placed on the implications of these interaction modes to design novel stimuli-responsive drug delivery principles and concepts, aiming to enhance the spatial, temporal, and dosage precision of drug delivery to cancer cells. Finally, future perspectives are discussed to highlight current challenges and future opportunities in self-assembly-based stimuli-responsive drug delivery nanotechnologies for cancer therapy.
Collapse
Affiliation(s)
- Yingqi Liu
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yunyun Wu
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing 400042, China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
42
|
Fabbrizi E, Fiorentino F, Carafa V, Altucci L, Mai A, Rotili D. Emerging Roles of SIRT5 in Metabolism, Cancer, and SARS-CoV-2 Infection. Cells 2023; 12:cells12060852. [PMID: 36980194 PMCID: PMC10047932 DOI: 10.3390/cells12060852] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Sirtuin 5 (SIRT5) is a predominantly mitochondrial enzyme catalyzing the removal of glutaryl, succinyl, malonyl, and acetyl groups from lysine residues through a NAD+-dependent deacylase mechanism. SIRT5 is an important regulator of cellular homeostasis and modulates the activity of proteins involved in different metabolic pathways such as glycolysis, tricarboxylic acid (TCA) cycle, fatty acid oxidation, electron transport chain, generation of ketone bodies, nitrogenous waste management, and reactive oxygen species (ROS) detoxification. SIRT5 controls a wide range of aspects of myocardial energy metabolism and plays critical roles in heart physiology and stress responses. Moreover, SIRT5 has a protective function in the context of neurodegenerative diseases, while it acts as a context-dependent tumor promoter or suppressor. In addition, current research has demonstrated that SIRT5 is implicated in the SARS-CoV-2 infection, although opposing conclusions have been drawn in different studies. Here, we review the current knowledge on SIRT5 molecular actions under both healthy and diseased settings, as well as its functional effects on metabolic targets. Finally, we revise the potential of SIRT5 as a therapeutic target and provide an overview of the currently reported SIRT5 modulators, which include both activators and inhibitors.
Collapse
Affiliation(s)
- Emanuele Fabbrizi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesco Fiorentino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Vincenzo Carafa
- Department of Precision Medicine, Università degli Studi della Campania “L. Vanvitelli”, 80138 Naples, Italy
- BIOGEM, 83031 Ariano Irpino, Italy
| | - Lucia Altucci
- Department of Precision Medicine, Università degli Studi della Campania “L. Vanvitelli”, 80138 Naples, Italy
- BIOGEM, 83031 Ariano Irpino, Italy
- IEOS—Istituto per l’Endocrinologia e Oncologia Sperimentale, CNR, 80131 Naples, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
- Pasteur Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (A.M.); (D.R.); Tel.: +39-0649913392 (A.M.); +39-0649913237 (D.R.); Fax: +39-0649693268 (A.M.)
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (A.M.); (D.R.); Tel.: +39-0649913392 (A.M.); +39-0649913237 (D.R.); Fax: +39-0649693268 (A.M.)
| |
Collapse
|
43
|
Song Y, Zhang Z, Cao Y, Yu Z. Stimulus-Responsive Amino Acids Behind In Situ Assembled Bioactive Peptide Materials. Chembiochem 2023; 24:e202200497. [PMID: 36278304 DOI: 10.1002/cbic.202200497] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/18/2022] [Indexed: 02/04/2023]
Abstract
In situ self-assembly of peptides into well-defined nanostructures represents one of versatile strategies for creation of bioactive materials within living cells with great potential in disease diagnosis and treatment. The intimate relationship between amino acid sequences and the assembling propensity of peptides has been thoroughly elucidated over the past few decades. This has inspired development of various controllable self-assembling peptide systems based on stimuli-responsive naturally occurring or non-canonical amino acids, including redox-, pH-, photo-, enzyme-responsive amino acids. This review attempts to summarize the recent progress achieved in manipulating in situ self-assembly of peptides by controllable reactions occurring to amino acids. We will highlight the systems containing non-canonical amino acids developed in our laboratory during the past few years, primarily including acid/enzyme-responsive 4-aminoproline, redox-responsive (seleno)methionine, and enzyme-responsive 2-nitroimidazolyl alanine. Utilization of the stimuli-responsive assembling systems in creation of bioactive materials will be specifically introduced to emphasize their advantages for addressing the concerns lying in disease theranostics. Eventually, we will provide the perspectives for the further development of stimulus-responsive amino acids and thereby demonstrating their great potential in development of next-generation biomaterials.
Collapse
Affiliation(s)
- Yanqiu Song
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, P. R. China
| | - Zeyu Zhang
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, P. R. China
| | - Yawei Cao
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, P. R. China
| | - Zhilin Yu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, P. R. China.,Haihe Laboratory of Synthetic Biology, 21 West 15th Avenue, Tianjin, 300308, P. R. China
| |
Collapse
|
44
|
Wei W, Zhang J, Xu Z, Liu Z, Huang C, Cheng K, Meng L, Matsuda Y, Hao Q, Zhang H, Sun H. Universal Strategy to Develop Fluorogenic Probes for Lysine Deacylase/Demethylase Activity and Application in Discriminating Demethylation States. ACS Sens 2023; 8:28-39. [PMID: 36602906 DOI: 10.1021/acssensors.2c01345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Dynamically controlling the post-translational modification of the ε-amino groups of lysine residues is critical for regulating many cellular events. Increasing studies have revealed that many important diseases, including cancer and neurological disorders, are associated with the malfunction of lysine deacylases and demethylases. Developing fluorescent probes that are capable of detecting lysine deacylase and demethylase activity is highly useful for interrogating their roles in epigenetic regulation and diseases. Due to the distinct substrate recognition of these epigenetic eraser enzymes, designing a universal strategy for detecting their activity poses substantial difficulty. Moreover, designing activity-based probes for differentiating their demethylation states is even more challenging and still remains largely unexplored. Herein, we report a universal strategy to construct probes that can detect the enzymatic activity of epigenetic "erasers" through NBD-based long-distance intramolecular reactions. The probes can be easily prepared by installing the O-NBD group at the C-terminal residue of specific peptide substrates by click chemistry. Based on this strategy, detecting the activity of lysine deacetylase, desuccinylase, or demethylase with superior sensitivity and selectivity has been successfully achieved through single-step probe development. Furthermore, the demethylase probe based on this strategy is capable of distinguishing different demethylation states by both absorption and fluorescence lifetime readout. We envision that these newly developed probes will provide powerful tools to facilitate drug discovery in epigenetics in the future.
Collapse
Affiliation(s)
- Wenyu Wei
- Department of Chemistry and COSADAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen518057, China
| | - Jie Zhang
- Department of Chemistry and COSADAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen518057, China
| | - Zhiqiang Xu
- Department of Chemistry and COSADAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen518057, China
| | - Zhiyang Liu
- Department of Chemistry and COSADAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen518057, China
| | - Chen Huang
- Department of Chemistry and COSADAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen518057, China
| | - Ke Cheng
- Department of Chemistry and COSADAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen518057, China
| | - Lingkuan Meng
- Department of Chemistry and COSADAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen518057, China
| | - Yudai Matsuda
- Department of Chemistry and COSADAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong999077, China
| | - Quan Hao
- Department of Physiology, University of Hong Kong, Pok Fu Lam, Hong Kong999077, China
| | - Huatang Zhang
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, Guangdong510006, China
| | - Hongyan Sun
- Department of Chemistry and COSADAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen518057, China
| |
Collapse
|
45
|
Yang L, Chen Q, Wang Z, Zhang H, Sun H. Small-molecule fluorescent probes for plasma membrane staining: Design, mechanisms and biological applications. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Jiang X, Yang C, Qiu J, Ma D, Xu C, Hu S, Han W, Yuan B, Lu Y. Nanomolar LL-37 induces permeability of a biomimetic mitochondrial membrane. NANOSCALE 2022; 14:17654-17660. [PMID: 36413063 DOI: 10.1039/d2nr05409d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
LL-37, the only human host cathelicidin peptide, is proposed to be able to induce host cell apoptosis through mitochondrial membrane permeabilization (MMP). Detailed pathways of the LL-37-triggered MMP are however still disputed. It is generally believed that cationic peptides permeate a membrane mostly in conditions of micromolar peptide concentrations and negatively charged membranes, which are not usually satisfied in the mitochondrial circumstance. Herein, using a variety of single-molecule techniques, we show that nanomolar LL-37 specifically induces permeability of a phosphoethanolamine (PE)-rich biomimetic mitochondrial membrane in a protein-independent manner. The insertion dynamics of single LL-37 molecules exhibit different metastable states in bilayers composed of different lipids. Moreover, the PE lipids significantly facilitate adsorption and accumulation of LL-37 on the PE-rich bilayer, and produce deeper insertion of peptide oligomers, especially tetramers, into the bilayer. This work offers an alternative pathway of the LL-37-triggered MMP and apoptosis.
Collapse
Affiliation(s)
- Xin Jiang
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China.
| | - Chenguang Yang
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Qiu
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China.
| | - Dongfei Ma
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China.
| | - Cheng Xu
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China.
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, Jiangsu 215006, China
| | - Shuxin Hu
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weijing Han
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China.
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China.
| | - Ying Lu
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
47
|
Kim D, Kim S, Park G, Choi H, Ryu JH. Spatiotemporal Self-Assembly of Peptide Amphiphiles by Carbonic Anhydrase IX-Targeting Induces Cancer-Lysosomal Membrane Disruption. JACS AU 2022; 2:2539-2547. [PMID: 36465549 PMCID: PMC9709935 DOI: 10.1021/jacsau.2c00422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 06/17/2023]
Abstract
To achieve spatiotemporal control, an enzyme-instructed self-assembly system is widely used, but this approach typically has a small effect on cellular fate. In this study, we show that the intralysosomal assembly by a carbonic anhydrase IX (CAIX)-targeting peptide amphiphile (Pep-AT) can control cellular fate with a low therapeutic dose by tuning the surface charge based on pH change. Pep-AT self-assembles into a fibrous aggregate with a negative surface charge in an extracellular environment near CAIX. During endocytosis, it changes into a nanofiber with a positive surface charge at the lysosome. Then, it can disrupt the lysosomal membrane and induce cellular apoptosis. This study demonstrates that a spatiotemporal assembly induced by a cancer enzyme and specific organelle can control the cellular fate of cancer.
Collapse
|
48
|
Bolding JE, Martín‐Gago P, Rajabi N, Gamon LF, Hansen TN, Bartling CRO, Strømgaard K, Davies MJ, Olsen CA. Aryl Fluorosulfate Based Inhibitors That Covalently Target the SIRT5 Lysine Deacylase. Angew Chem Int Ed Engl 2022; 61:e202204565. [PMID: 36130196 PMCID: PMC9828517 DOI: 10.1002/anie.202204565] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Indexed: 01/12/2023]
Abstract
The sirtuin enzymes are a family of lysine deacylases that regulate gene transcription and metabolism. Sirtuin 5 (SIRT5) hydrolyzes malonyl, succinyl, and glutaryl ϵ-N-carboxyacyllysine posttranslational modifications and has recently emerged as a vulnerability in certain cancers. However, chemical probes to illuminate its potential as a pharmacological target have been lacking. Here we report the harnessing of aryl fluorosulfate-based electrophiles as an avenue to furnish covalent inhibitors that target SIRT5. Alkyne-tagged affinity-labeling agents recognize and capture overexpressed SIRT5 in cultured HEK293T cells and can label SIRT5 in the hearts of mice upon intravenous injection of the compound. This work demonstrates the utility of aryl fluorosulfate electrophiles for targeting of SIRT5 and suggests this as a means for the development of potential covalent drug candidates. It is our hope that these results will serve as inspiration for future studies investigating SIRT5 and general sirtuin biology in the mitochondria.
Collapse
Affiliation(s)
- Julie E. Bolding
- Center for Biopharmaceuticals & Department of Drug Design and PharmacologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2DK-2100CopenhagenDenmark
| | - Pablo Martín‐Gago
- Center for Biopharmaceuticals & Department of Drug Design and PharmacologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2DK-2100CopenhagenDenmark
| | - Nima Rajabi
- Center for Biopharmaceuticals & Department of Drug Design and PharmacologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2DK-2100CopenhagenDenmark
| | - Luke F. Gamon
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenBlegdamsvej 3DK-2200CopenhagenDenmark
| | - Tobias N. Hansen
- Center for Biopharmaceuticals & Department of Drug Design and PharmacologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2DK-2100CopenhagenDenmark
| | - Christian R. O. Bartling
- Center for Biopharmaceuticals & Department of Drug Design and PharmacologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2DK-2100CopenhagenDenmark
| | - Kristian Strømgaard
- Center for Biopharmaceuticals & Department of Drug Design and PharmacologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2DK-2100CopenhagenDenmark
| | - Michael J. Davies
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenBlegdamsvej 3DK-2200CopenhagenDenmark
| | - Christian A. Olsen
- Center for Biopharmaceuticals & Department of Drug Design and PharmacologyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2DK-2100CopenhagenDenmark
| |
Collapse
|
49
|
Liu S, Zhang Q, He H, Yi M, Tan W, Guo J, Xu B. Intranuclear Nanoribbons for Selective Killing of Osteosarcoma Cells. Angew Chem Int Ed Engl 2022; 61:e202210568. [PMID: 36102872 PMCID: PMC9869109 DOI: 10.1002/anie.202210568] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Indexed: 01/26/2023]
Abstract
Herein, we show intranuclear nanoribbons formed upon dephosphorylation of leucine-rich L- or D-phosphopeptide catalyzed by alkaline phosphatase (ALP) to selectively kill osteosarcoma cells. Being dephosphorylated by ALP, the peptides are first transformed into micelles and then converted into nanoribbons. The peptides/assemblies first aggregate on cell membranes, then enter cells via endocytosis, and finally accumulate in nuclei (mainly in nucleoli). Proteomics analysis suggests that the assemblies interact with histone proteins. The peptides kill osteosarcoma cells rapidly and are nontoxic to normal cells. Moreover, the repeated stimulation of the osteosarcoma cells by the peptides sensitizes the cancer cells rather than inducing resistance. This work not only illustrates a novel mechanism for nucleus targeting, but may also pave a new way for selectively killing osteosarcoma cells and minimizing drug resistance.
Collapse
Affiliation(s)
- Shuang Liu
- School of Materials Science and Engineering, Wuhan University of Technology, 122 Luoshi Road, Wuhan, Hubei, 430070, China
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| | - Qiuxin Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02454, USA
| | - Meihui Yi
- 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
| | - Jiaqi Guo
- 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
|
50
|
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
|