1
|
Cong Y, Li X, Hong H. Current strategies for senescence treatment: Focused on theranostic performance of nanomaterials. J Control Release 2025; 382:113710. [PMID: 40220869 DOI: 10.1016/j.jconrel.2025.113710] [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: 01/30/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/14/2025]
Abstract
Age-related diseases imposed heavy burdens to the healthcare systems globally, while cell senescence served as one fundamental molecular/cellular basis for these diseases. How to tackle the senescence-relevant problems is a hotspot for biomedical research. In this review article, the hallmarks and molecular pathways of cell senescence were firstly discussed, followed by the introduction of the current anti-senescence strategies, including senolytics and senomorphics. With suitable physical or chemical properties, multiple types of nanomaterials were used successfully in senescence therapeutics, as well as senescence detection. Based on the accumulating knowledges for senescence, the rules of how to use these nanoplatforms more efficiently against senescence were also summarized, including but not limited to surface modification, material-cargo interactions, factor responsiveness etc. The comparison of these "senescence-selective" nanoplatforms to other treatment options (prodrugs, ADCs, PROTACs, CART etc.) was also given. Learning from the past, nanotechnology can add more choice for treating age-related diseases, and provide more (diagnostic) information to further our understanding of senescence process.
Collapse
Affiliation(s)
- Yiyang Cong
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Medical School of Nanjing University, Nanjing 210093, China
| | - Xiaoyang Li
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Medical School of Nanjing University, Nanjing 210093, China
| | - Hao Hong
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Medical School of Nanjing University, Nanjing 210093, China.
| |
Collapse
|
2
|
Qi Y, Wang F, Liu J, Wang C, Liu Y. Enzyme-mediated hydrogelation for biomedical applications: A review. Int J Biol Macromol 2025; 311:143379. [PMID: 40258561 DOI: 10.1016/j.ijbiomac.2025.143379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/11/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025]
Abstract
Hydrogels possess significant potential for biomedical applications due to their flexibility and biocompatibility. However, current physical or chemical methods for their preparation often fail to balance biocompatibility and mechanical properties, limiting their application scope. Enzymatic preparation of hydrogels offers advantages including mild reaction conditions, absence of toxic substances, and superior biocompatibility. This review focuses on the enzymatic preparation systems of hydrogels and its application in the fast-growing biomedical field. Firstly, the mechanisms of enzyme-mediated hydrogel preparation can be categorized into four classes: enzyme cross-linking, enzyme polymerization, enzyme-mediated self-assembly of small molecular gelators, and enzyme-induced pH changes. Hydrogels prepared through the first two mechanisms retain the mechanical advantages of chemically cross-linked hydrogels while preserving the inherent biocompatibility. Additionally, hydrogels prepared via the latter two mechanisms exhibit rapid responses to external stimuli similar to physically crosslinked hydrogels while maintaining high biocompatibility. Furthermore, we discuss their application in biomedical scope and analyze the correlation between the mechanism of enzyme-mediated hydrogels and their respective application domains. Finally, the current challenges faced by enzymatically mediated hydrogelation are summarized; notably that enzymes incorporated and immobilized during hydrogel preparation remain active, resulting in catalytic activity exhibited by these enzymatically mediated hydrogels, which broadens their potential applications.
Collapse
Affiliation(s)
- Yue Qi
- Green Papermaking and Resource Recycling National Key Laboratory, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, PR China
| | - Fangfang Wang
- Green Papermaking and Resource Recycling National Key Laboratory, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, PR China.
| | - Junliang Liu
- Qilu Pharmaceutical Co., LTD., Jinan 250104, PR China
| | - Chunyang Wang
- Green Papermaking and Resource Recycling National Key Laboratory, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, PR China
| | - Yu Liu
- Green Papermaking and Resource Recycling National Key Laboratory, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, PR China.
| |
Collapse
|
3
|
Xu W, Qin X, Liu Y, Chen J, Wang Y. Advances in Enzyme-responsive Supramolecular In situ Self-assembled Peptide for Drug Delivery. Curr Drug Deliv 2025; 22:374-386. [PMID: 37496133 DOI: 10.2174/1567201820666230726151607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/17/2023] [Accepted: 06/21/2023] [Indexed: 07/28/2023]
Abstract
Because of low immunogenicity, ease of modification, and inherent biosafety, peptides have been well recognized as vehicles to deliver therapeutic agents to targeted regions with improved pharmacokinetic characteristics. Enzyme-responsive self-assembled peptides (ERSAPs) show superiority over their naive forms due to their enhanced targeting efficacy and long-retention property. In this review, we have summarized recent advances in the therapeutic application of ERSAPs, mainly focusing on their self-therapeutic properties and potential as vehicles to deliver different drugs.
Collapse
Affiliation(s)
- Wentao Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- Cancer Center, Department of Interventional Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiaowen Qin
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yang Liu
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jun Chen
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- Cancer Center, Department of Interventional Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuguang Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Jha SK, De Rubis G, Devkota SR, Zhang Y, Adhikari R, Jha LA, Bhattacharya K, Mehndiratta S, Gupta G, Singh SK, Panth N, Dua K, Hansbro PM, Paudel KR. Cellular senescence in lung cancer: Molecular mechanisms and therapeutic interventions. Ageing Res Rev 2024; 97:102315. [PMID: 38679394 DOI: 10.1016/j.arr.2024.102315] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/03/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Lung cancer stands as the primary contributor to cancer-related fatalities worldwide, affecting both genders. Two primary types exist where non-small cell lung cancer (NSCLC), accounts for 80-85% and SCLC accounts for 10-15% of cases. NSCLC subtypes include adenocarcinoma, squamous cell carcinoma, and large cell carcinoma. Smoking, second-hand smoke, radon gas, asbestos, and other pollutants, genetic predisposition, and COPD are lung cancer risk factors. On the other hand, stresses such as DNA damage, telomere shortening, and oncogene activation cause a prolonged cell cycle halt, known as senescence. Despite its initial role as a tumor-suppressing mechanism that slows cell growth, excessive or improper control of this process can cause age-related diseases, including cancer. Cellular senescence has two purposes in lung cancer. Researchers report that senescence slows tumor growth by constraining multiplication of impaired cells. However, senescent cells also demonstrate the pro-inflammatory senescence-associated secretory phenotype (SASP), which is widely reported to promote cancer. This review will look at the role of cellular senescence in lung cancer, describe its diagnostic markers, ask about current treatments to control it, look at case studies and clinical trials that show how senescence-targeting therapies can be used in lung cancer, and talk about problems currently being faced, and possible solutions for the same in the future.
Collapse
Affiliation(s)
- Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Shankar Raj Devkota
- Monash Biomedicine Discovery Institute, and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Yali Zhang
- School of Chemical Engineering, University of Adelaide, Adelaide 5005, Australia
| | - Radhika Adhikari
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Laxmi Akhileshwar Jha
- Naraina Vidya Peeth Group of Institutions, Faculty of Pharmacy, Dr. A. P. J. Abdul Kalam Technical University, Lucknow, Uttar Pradesh 0208020, India
| | - Kunal Bhattacharya
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam 781026, India; Royal School of Pharmacy, The Assam Royal Global University, Guwahati, Assam 781035, India
| | - Samir Mehndiratta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Nisha Panth
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia.
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia.
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia.
| |
Collapse
|
7
|
Yu K, Zhou P, Wang M, Zou P, Wang H, Liu Y, Xie M. β-Galactosidase-guided self-assembled 68Ga nanofibers probe for micro-PET tumor imaging. Bioorg Med Chem Lett 2024; 104:129727. [PMID: 38582132 DOI: 10.1016/j.bmcl.2024.129727] [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: 10/17/2023] [Revised: 03/12/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
β-galactosidase (β-gal) has high activity in various malignancies, which is suitable for targeted positron emission tomography (PET) imaging. Meanwhile, β-gal can successfully guide the formation of nanofibers, which enhances the intensity of imaging and extends the imaging time. Herein, we designed a β-galactosidase-guided self-assembled PET imaging probe [68Ga]Nap-NOTA-1Gal. We envisage that β-gal could recognize and cleave the target site, bringing about self-assembling to form nanofibers, thereby enhancing the PET imaging effect. The targeting specificity of [68Ga]Nap-NOTA-1Gal for detecting β-gal activity was examined using the control probe [68Ga]Nap-NOTA-1. Micro-PET imaging showed that tumor regions of [68Ga]Nap-NOTA-1Gal were visible after injection. And the tumor uptake of [68Ga]Nap-NOTA-1Gal was higher than [68Ga]Nap-NOTA-1 at all-time points. Our results demonstrated that the [68Ga]Nap-NOTA-1Gal can be used for the purpose of a new promising PET probe for helping diagnose cancer with high levels of β-gal activity.
Collapse
Affiliation(s)
- Kangxia Yu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Peng Zhou
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Meimei Wang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Pei Zou
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Hongyong Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yaling Liu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China.
| | - Minhao Xie
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China.
| |
Collapse
|
8
|
Jin P, Duan X, Li L, Zhou P, Zou C, Xie K. Cellular senescence in cancer: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e542. [PMID: 38660685 PMCID: PMC11042538 DOI: 10.1002/mco2.542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 02/28/2024] [Accepted: 03/07/2024] [Indexed: 04/26/2024] Open
Abstract
Aging exhibits several hallmarks in common with cancer, such as cellular senescence, dysbiosis, inflammation, genomic instability, and epigenetic changes. In recent decades, research into the role of cellular senescence on tumor progression has received widespread attention. While how senescence limits the course of cancer is well established, senescence has also been found to promote certain malignant phenotypes. The tumor-promoting effect of senescence is mainly elicited by a senescence-associated secretory phenotype, which facilitates the interaction of senescent tumor cells with their surroundings. Targeting senescent cells therefore offers a promising technique for cancer therapy. Drugs that pharmacologically restore the normal function of senescent cells or eliminate them would assist in reestablishing homeostasis of cell signaling. Here, we describe cell senescence, its occurrence, phenotype, and impact on tumor biology. A "one-two-punch" therapeutic strategy in which cancer cell senescence is first induced, followed by the use of senotherapeutics for eliminating the senescent cells is introduced. The advances in the application of senotherapeutics for targeting senescent cells to assist cancer treatment are outlined, with an emphasis on drug categories, and the strategies for their screening, design, and efficient targeting. This work will foster a thorough comprehension and encourage additional research within this field.
Collapse
Affiliation(s)
- Ping Jin
- State Key Laboratory for Conservation and Utilization of Bio‐Resources in Yunnan, School of Life SciencesYunnan UniversityKunmingYunnanChina
| | - Xirui Duan
- Department of OncologySchool of MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Lei Li
- Department of Anorectal SurgeryHospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ping Zhou
- Department of OncologySchool of MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| | - Cheng‐Gang Zou
- State Key Laboratory for Conservation and Utilization of Bio‐Resources in Yunnan, School of Life SciencesYunnan UniversityKunmingYunnanChina
| | - Ke Xie
- Department of OncologySchool of MedicineSichuan Academy of Medical Sciences and Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuanChina
| |
Collapse
|
9
|
Jha LA, Kumar B, Jha SK, Paudel KR. Futuristic senolytic drug incorporated nanomedicine therapy to treat osteoarthritis. Nanomedicine (Lond) 2024; 19:837-840. [PMID: 38426446 DOI: 10.2217/nnm-2023-0348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Affiliation(s)
- Laxmi Akhileshwar Jha
- Naraina Vidya Peeth Group of Institutions, Faculty of Pharmacy, Dr. A. P. J. Abdul Kalam Technical University, Kanpur, 0208020, Uttar Pradesh, India
| | - Bhupendra Kumar
- Department of Biological Sciences & Bioengineering (BSBE), Indian Institute of Technology, Kanpur, 208016, Uttar Pradesh, India
| | - Saurav Kumar Jha
- Department of Biological Sciences & Bioengineering (BSBE), Indian Institute of Technology, Kanpur, 208016, Uttar Pradesh, India
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute & University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, 2007, Australia
| |
Collapse
|
10
|
Zhang H, Xu T, Liang J, Wu B, Yang X, Wang J, Zhou Z, Wang H. Controlling Supramolecular Fiber Formation of Nucleopeptide by Guanosine Triphosphate. Biomacromolecules 2023; 24:5678-5686. [PMID: 37934694 DOI: 10.1021/acs.biomac.3c00674] [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: 11/09/2023]
Abstract
Cells use dynamic self-assembly to construct functional structures for maintaining cellular homeostasis. However, using a natural biological small molecule to mimic this phenomenon remains challenging. This work reports the dynamic microfiber formation of nucleopeptide driven by guanosine triphosphate, the small molecule that controls microtubule polymerization in living cells. Deactivation of GTP by enzyme dissociates the fibers, which could be reactivated by adding GTP. Molecular dynamic simulation unveils the mystery of microfiber formation of GBM-1 and GTP. Moreover, the microfiber formation can also be controlled by diffusion-driven GTP gradients across a semipermeable membrane in bulk conditions and the microfluidic method in the defined droplets. This study provides a new platform to construct dynamic self-assembly materials of molecular building blocks driven by GTP.
Collapse
Affiliation(s)
- Hongyue Zhang
- Department of Chemistry, Zhejiang University, Hangzhou 310027, P. R. China
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| | - Tengyan Xu
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| | - Juan Liang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| | - Bihan Wu
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| | - Xuejiao Yang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| | - Jing Wang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| | - Ziao Zhou
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| | - Huaimin Wang
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Institute of Natural Sciences, Westlake Institute for Advanced Study, Westlake University, No. 600 Dunyu Road, Hangzhou 310024, Zhejiang Province, China
| |
Collapse
|
11
|
Li Q, Wang Z, Shi N, Qi Y, Yao W, Yu J, Lu Y. Application and prospect of the therapeutic strategy of inhibiting cellular senescence combined with pro-regenerative biomaterials in regenerative medicine. SMART MEDICINE 2023; 2:e20230030. [PMID: 39188301 PMCID: PMC11235619 DOI: 10.1002/smmd.20230030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/20/2023] [Indexed: 08/28/2024]
Abstract
Complete regeneration of damaged tissues/organs has always been the ultimate challenge in regenerative medicine. Aging has long been considered the basis of age-related diseases, as senescent cells gradually accumulate in tissues with increasing age, tissues exhibit aging and normal physiological functions are inhibited. In recent years, in damaged tissues, scholars have found that the number of cells with features of cellular senescence continues to increase over time. The accumulation of senescent cells severely hinders the healing of damaged tissues. Furthermore, by clearing senescent cells or inhibiting the aging microenvironment, damaged tissues regained their original regenerative and repair capabilities. On the other hand, various biomaterials have been proved to have good biocompatibility and can effectively support cell regeneration after injury. Combining the two solutions, inhibiting the cellular senescence in damaged tissues and establishing a pro-regenerative environment through biomaterial technology gradually reveals a new, unexpected treatment strategy applied to the field of regenerative medicine. In this review, we first elucidate the main characteristics of senescent cells from morphological, functional and molecular levels, and discuss in detail the process of accumulation of senescent cells in tissues. Then, we will explore in depth how the accumulation of senescent cells after damage affects tissue repair and regeneration at different stages. Finally, we will turn to how to promote tissue regeneration and repair in the field of regenerative medicine by inhibiting cellular senescence combined with biomaterial technology. Our goal is to understand the relationship between cellular senescence and tissue regeneration through this new perspective, and provide valuable references for the development of new therapeutic strategies in the future.
Collapse
Affiliation(s)
- Qianyi Li
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Pôle Sino‐Français de Recherches en Sciences du Vivant et G´enomiqueShanghaiChina
- International Laboratory in Cancer, Aging and HematologyShanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur UniversityShanghaiChina
| | | | | | - Yang Qi
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenfei Yao
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jie Yu
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yiming Lu
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Pôle Sino‐Français de Recherches en Sciences du Vivant et G´enomiqueShanghaiChina
- International Laboratory in Cancer, Aging and HematologyShanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur UniversityShanghaiChina
- Division of Critical CareNanxiang Hospital of Jiading DistrictShanghaiChina
| |
Collapse
|
12
|
Wang F, Xia W, Zhang M, Wu R, Song X, Hao Y, Feng Y, Zhang L, Li D, Kang W, Liu C, Liu L. Engineering of antimicrobial peptide fibrils with feedback degradation of bacterial-secreted enzymes. Chem Sci 2023; 14:10914-10924. [PMID: 37829030 PMCID: PMC10566480 DOI: 10.1039/d3sc01089a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 09/06/2023] [Indexed: 10/14/2023] Open
Abstract
Proteins and peptides can assemble into functional amyloid fibrils with distinct architectures. These amyloid fibrils can fulfil various biological functions in living organisms, and then be degraded. By incorporating an amyloidogenic segment and enzyme-cleavage segment together, we designed a peptide (enzyme-cleavage amyloid peptides (EAP))-based functional fibril which could be degraded specifically by gelatinase. To gain molecular insights into the assembly and degradation of EAP fibrils, we determined the atomic structure of the EAP fibril using cryo-electron microscopy. The amyloidogenic segment of EAP adopted a β-strand conformation and mediated EAP-fibril formation mainly via steric zipper-like interactions. The enzyme-cleavage segment was partially involved in self-assembly, but also exhibited high flexibility in the fibril structure, with accessibility to gelatinase binding and degradation. Moreover, we applied the EAP fibril as a tunable scaffold for developing degradable self-assembled antimicrobial fibrils (SANs) by integrating melittin and EAP together. SANs exhibited superior activity for killing bacteria, and significantly improved the stability and biocompatibility of melittin. SANs were eliminated automatically by the gelatinase secreted from targeted bacteria. Our work provides a new strategy for rational design of functional fibrils with a feedback regulatory loop for optimizing the biocompatibility and biosafety of designed fibrils. Our work may aid further developments of "smart" peptide-based biomaterials for biomedical applications.
Collapse
Affiliation(s)
- Fenghua Wang
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University Zhenjiang Jiangsu 212013 China
- College of Aeronautical Engineering, Jiangsu Aviation Vocational and Technical College Zhenjiang Jiangsu 212134 China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences Shanghai 201210 China
| | - Mingming Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences Shanghai 201210 China
| | - Rongrong Wu
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University Zhenjiang Jiangsu 212013 China
| | - Xiaolu Song
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University Zhenjiang Jiangsu 212013 China
| | - Yun Hao
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University Zhenjiang Jiangsu 212013 China
| | - Yonghai Feng
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University Zhenjiang Jiangsu 212013 China
| | - Liwei Zhang
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University Zhenjiang Jiangsu 212013 China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University Shanghai 200030 China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University Shanghai 200240 China
| | - Wenyan Kang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai 200025 China
- Department of Neurology, Ruijin Hainan Hospital, Shanghai Jiao Tong University School of Medicine (Boao Research Hospital) Hainan 571434 China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences Shanghai 201210 China
- Department of Neurology, Ruijin Hainan Hospital, Shanghai Jiao Tong University School of Medicine (Boao Research Hospital) Hainan 571434 China
- State Key Laboratory of Bio-Organic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences Shanghai 200032 China
| | - Lei Liu
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University Zhenjiang Jiangsu 212013 China
| |
Collapse
|
13
|
Qiao Y, Xu B. Peptide Assemblies for Cancer Therapy. ChemMedChem 2023; 18:e202300258. [PMID: 37380607 PMCID: PMC10613339 DOI: 10.1002/cmdc.202300258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 06/30/2023]
Abstract
Supramolecular assemblies made by the self-assembly of peptides are finding an increasing number of applications in various fields. While the early exploration of peptide assemblies centered on tissue engineering or regenerative medicine, the recent development has shown that peptide assemblies can act as supramolecular medicine for cancer therapy. This review covers the progress of applying peptide assemblies for cancer therapy, with the emphasis on the works appeared over the last five years. We start with the introduction of a few seminal works on peptide assemblies, then discuss the combination of peptide assemblies with anticancer drugs. Next, we highlight the use of enzyme-controlled transformation or shapeshifting of peptide assemblies for inhibiting cancer cells and tumors. After that, we provide the outlook for this exciting field that promises new kind of therapeutics for cancer therapy.
Collapse
Affiliation(s)
- Yuchen Qiao
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| |
Collapse
|
14
|
Advances in Self-Assembled Peptides as Drug Carriers. Pharmaceutics 2023; 15:pharmaceutics15020482. [PMID: 36839803 PMCID: PMC9964150 DOI: 10.3390/pharmaceutics15020482] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/19/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
In recent years, self-assembled peptide nanotechnology has attracted a great deal of attention for its ability to form various regular and ordered structures with diverse and practical functions. Self-assembled peptides can exist in different environments and are a kind of medical bio-regenerative material with unique structures. These materials have good biocompatibility and controllability and can form nanoparticles, nanofibers and hydrogels to perform specific morphological functions, which are widely used in biomedical and material science fields. In this paper, the properties of self-assembled peptides, their influencing factors and the nanostructures that they form are reviewed, and the applications of self-assembled peptides as drug carriers are highlighted. Finally, the prospects and challenges for developing self-assembled peptide nanomaterials are briefly discussed.
Collapse
|
15
|
Lozano-Torres B, García-Fernández A, Domínguez M, Sancenón F, Blandez JF, Martínez-Máñez R. β-Galactosidase-Activatable Nile Blue-Based NIR Senoprobe for the Real-Time Detection of Cellular Senescence. Anal Chem 2022; 95:1643-1651. [PMID: 36580602 PMCID: PMC9850349 DOI: 10.1021/acs.analchem.2c04766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cellular senescence is a stable cell cycle arrest in response to stress or other damage stimuli to maintain tissue homeostasis. However, the accumulation of senescent cells can lead to the progression of various senescence-related disorders. In this paper, we describe the development of a β-galactosidase-activatable near-infrared (NIR) senoprobe, NBGal, for the detection of senescent cells based on the use of the FDA-approved Nile blue (NB) fluorophore. NBGal was validated in chemotherapeutic-induced senescence cancer models in vitro using SK-Mel 103 and 4T1 cell lines. In vivo monitoring of cellular senescence was evaluated in orthotopic triple-negative breast cancer-bearing mice treated with palbociclib to induce senescence. In all cases, NBGal exhibited a selective tracking of senescent cells mainly ascribed to the overexpressed β-galactosidase enzyme responsible for hydrolyzing the NBGal probe generating the highly emissive NB fluorophore. In this way, NBGal has proven to be a qualitative, rapid, and minimally invasive probe that allows the direct detection of senescent cells in vivo.
Collapse
Affiliation(s)
- Beatriz Lozano-Torres
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica
de València, Centro de Investigación Príncipe
Felipe, C/ Eduardo Primo
Yúfera 3, Valencia 46012, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain
| | - Alba García-Fernández
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica
de València, Centro de Investigación Príncipe
Felipe, C/ Eduardo Primo
Yúfera 3, Valencia 46012, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain,Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe, Av. Fernando Abril Martorell,
10, Torre A 7a̲ planta, Valencia 46026, Spain
| | - Marcia Domínguez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain
| | - Félix Sancenón
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica
de València, Centro de Investigación Príncipe
Felipe, C/ Eduardo Primo
Yúfera 3, Valencia 46012, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain,Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe, Av. Fernando Abril Martorell,
10, Torre A 7a̲ planta, Valencia 46026, Spain,
| | - Juan F. Blandez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain,Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe, Av. Fernando Abril Martorell,
10, Torre A 7a̲ planta, Valencia 46026, Spain,
| | - Ramón Martínez-Máñez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica
de València, Centro de Investigación Príncipe
Felipe, C/ Eduardo Primo
Yúfera 3, Valencia 46012, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain,Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe, Av. Fernando Abril Martorell,
10, Torre A 7a̲ planta, Valencia 46026, Spain,
| |
Collapse
|
16
|
Choi JY, Yee SF, Tchangalova T, Yang G, Fisher JP. Recent Advances in Senotherapeutics Delivery. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:1223-1234. [PMID: 35451328 PMCID: PMC9805860 DOI: 10.1089/ten.teb.2021.0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/12/2022] [Indexed: 01/13/2023]
Abstract
Accumulation of senescent cells (SnCs) in various tissue types has been connected to an occurrence of different age-related diseases that are indicated by its own tissue-specific hallmarks. Discovery of novel senolytic compounds that target major cellular mechanisms to inhibit the level of SnCs within the specific tissues or organs has been an emerging field in the age-related disease research. Although the positive effect of senolytics in global suppression of SnCs has been well studied in the past, effective tissue-specific delivery strategy of senotherapeutics before clinical application needs to be further investigated. In this review, we discuss the latest biological insights to currently available senotherapeutic options and explore the impactful in vitro tissue-engineered models possibly as a testbed for replicable testing of tissue-specific potency of senolytics. Impact statement Senotherapy, the inhibition of accumulated senescent cells, is recognized as a significantly impactful way to treat various human diseases. However, there is limited comprehensive reviews on this topic. This review provides in-depth discussion on diverse delivery strategies of senolytic agents and latest updates on a novel senotherapeutic research.
Collapse
Affiliation(s)
- Ji Young Choi
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, Maryland, USA
- NIBIB/NIH Center of Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - Samantha F. Yee
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, Maryland, USA
| | - Tzvetelina Tchangalova
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, Maryland, USA
| | - Guang Yang
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, Maryland, USA
| | - John P. Fisher
- Tissue Engineering and Biomaterials Laboratory, Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, Maryland, USA
- NIBIB/NIH Center of Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
17
|
Cheng C, Sun Q, Wang X, He B, Jiang T. Enzyme-manipulated hydrogelation of small molecules for biomedical applications. Acta Biomater 2022; 151:88-105. [PMID: 35970483 DOI: 10.1016/j.actbio.2022.08.016] [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: 05/17/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022]
Abstract
Enzyme-manipulated hydrogelation based on self-assembly of small molecules is an attractive methodology for development of functional biomaterials. Upon the catalysis of enzymes, small-molecular precursors are converted into assemblable building blocks, which arrange into high-ordered nanofibers via non-covalent interactions at the molecular level, and further trap water to form hydrogels at the macroscopic level. Such approach has numerous advantages of region- and enantioselectivity, and mild reaction conditions for encapsulation of biomedications or cells that are fragile to environmental change. In addition to the common applications as drug reservoirs or cell scaffolds, the utilization of endogenous enzymes as stimuli to initiate self-assembly in the living cells and tissue is considered as an intelligent spatiotemporally controllable hydrogelation strategy for biomedical applications. The enzyme-instructed in situ self-assembly and hydrogelation can modulate the cell behavior, and even present therapeutic bioactivities, which provides a new perspective in the field of disease treatment. In this review, we categorize distinct enzymatic stimuli and elaborate substrate design, catalytic characteristics, and mechanisms of self-assembly and hydrogelation. The biomedical applications in drug delivery, tissue engineering, bioimaging, and in situ gelation-produced bioactivity are outlined. Advantages and limitations regarding the state-of-the-art enzyme-driven hydrogelation technologies and future perspectives are also discussed. STATEMENT OF SIGNIFICANCE: Hydrogel is a semi-solid soft material containing a large amount of water. Due to the features of adjustable flexibility, extremely porous architecture, and the high similarity of structure to natural extracellular matrices, the hydrogel has broad application prospects in biomedicine. In recent 20 years, enzyme-manipulated hydrogelation based on self-assembly of small molecules has developed rapidly as an attractive methodology for the construction of functional biomaterials. Upon the catalysis of enzymes, small-molecular precursors are converted into assemblable building blocks, which arrange into high-ordered nanofibers via non-covalent interactions at the molecular level, and further trap water to form hydrogels at the macroscopic level. This review summarized the characteristics of enzymatic hydrogel, as well as the traditional application and emerging prospect of enzyme-instructed self-assembly and hydrogelation.
Collapse
Affiliation(s)
- Cheng Cheng
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Qingyun Sun
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Xiuping Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Bingfang He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China.
| | - Tianyue Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
18
|
Macdougall LJ, Hoffman TE, Kirkpatrick BE, Fairbanks BD, Bowman CN, Spencer SL, Anseth KS. Intracellular Crowding by Bio-Orthogonal Hydrogel Formation Induces Reversible Molecular Stasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202882. [PMID: 35671709 PMCID: PMC9377388 DOI: 10.1002/adma.202202882] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Indexed: 05/22/2023]
Abstract
To survive extreme conditions, certain animals enter a reversible protective stasis through vitrification of the cytosol by polymeric molecules such as proteins and polysaccharides. In this work, synthetic gelation of the cytosol in living cells is used to induce reversible molecular stasis. Through the sequential lipofectamine-mediated transfection of complementary poly(ethylene glycol) macromers into mammalian cells, intracellular crosslinking occurs through bio-orthogonal strain-promoted azide-alkyne cycloaddition click reactions. This achieves efficient polymer uptake with minimal cell death (99% viable). Intracellular crosslinking decreases DNA replication and protein synthesis, and increases the quiescent population by 2.5-fold. Real-time tracking of single cells containing intracellular crosslinked polymers identifies increases in intermitotic time (15 h vs 19 h) and decreases in motility (30 µm h-1 vs 15 µm h-1 ). The cytosol viscosity increases threefold after intracellular crosslinking and results in disordered cytoskeletal structure in addition to the disruption of cellular coordination in a scratch assay. By incorporating photodegradable nitrobenzyl moieties into the polymer backbone, the effects of intracellular crosslinking are reversed upon exposure to light, thereby restoring proliferation (80% phospho-Rb+ cells), protein translation, and migration. Reversible intracellular crosslinking provides a novel method for dynamic manipulation of intracellular mechanics, altering essential processes that determine cellular function.
Collapse
Affiliation(s)
- Laura J Macdougall
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Timothy E Hoffman
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Bruce E Kirkpatrick
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Medical Scientist Training Program, School of Medicine, University of Colorado, Aurora, CO, 80045, USA
| | - Benjamin D Fairbanks
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- Material Science and Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Christopher N Bowman
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Material Science and Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Sabrina L Spencer
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303, USA
- Material Science and Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
19
|
Zhang L, Pitcher LE, Yousefzadeh MJ, Niedernhofer LJ, Robbins PD, Zhu Y. Cellular senescence: a key therapeutic target in aging and diseases. J Clin Invest 2022; 132:e158450. [PMID: 35912854 PMCID: PMC9337830 DOI: 10.1172/jci158450] [Citation(s) in RCA: 284] [Impact Index Per Article: 94.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cellular senescence is a hallmark of aging defined by stable exit from the cell cycle in response to cellular damage and stress. Senescent cells (SnCs) can develop a characteristic pathogenic senescence-associated secretory phenotype (SASP) that drives secondary senescence and disrupts tissue homeostasis, resulting in loss of tissue repair and regeneration. The use of transgenic mouse models in which SnCs can be genetically ablated has established a key role for SnCs in driving aging and age-related disease. Importantly, senotherapeutics have been developed to pharmacologically eliminate SnCs, termed senolytics, or suppress the SASP and other markers of senescence, termed senomorphics. Based on extensive preclinical studies as well as small clinical trials demonstrating the benefits of senotherapeutics, multiple clinical trials are under way. This Review discusses the role of SnCs in aging and age-related diseases, strategies to target SnCs, approaches to discover and develop senotherapeutics, and preclinical and clinical advances of senolytics.
Collapse
Affiliation(s)
- Lei Zhang
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Louise E. Pitcher
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Matthew J. Yousefzadeh
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, and
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
20
|
Su L, Hendrikse SIS, Meijer EW. Supramolecular glycopolymers: How carbohydrates matter in structure, dynamics, and function. Curr Opin Chem Biol 2022; 69:102171. [PMID: 35749930 DOI: 10.1016/j.cbpa.2022.102171] [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: 04/08/2022] [Revised: 05/14/2022] [Accepted: 05/18/2022] [Indexed: 11/18/2022]
Abstract
Supramolecular glycopolymers exhibiting inherent dynamicity, tunability, and adaptivity allow us to arrive at a deeper understanding of multivalent carbohydrate-carbohydrate interactions and carbohydrate-protein interactions, both being essential to key biological events. The impacts of the carbohydrate segments in these supramolecular glycopolymers towards their structure, dynamics, and function as biomaterials are addressed in this minireview. Bottlenecks and challenges are discussed, and we speculate about possible future directions.
Collapse
Affiliation(s)
- Lu Su
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands; Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Simone I S Hendrikse
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands; Department of Chemical Engineering, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - E W Meijer
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands; School of Chemistry and UNSW RNA Institute, The University of New South Wales Sydney, NSW 2052, Australia.
| |
Collapse
|
21
|
Dergham M, Lin S, Geng J. Supramolecular Self-Assembly in Living Cells. Angew Chem Int Ed Engl 2022; 61:e202114267. [PMID: 35037350 DOI: 10.1002/anie.202114267] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Indexed: 12/17/2022]
Abstract
Supramolecular interactions rely on non-covalent forces, such as hydrophobic effects, hydrogen-bonding, and electrostatic interactions, which govern many intracellular biological pathways. In cellulo supramolecular self-assembly is mainly based on host-guest interactions, changes in pH, enzymes, and polymerization-induced self-assembly to accurately induce various unnatural reactions without disturbing natural biological processes. This process can produce synthetic biocompatible macromolecules to control cell properties and regulate biological functions, such as cell proliferation and differentiation. This Minireview focuses on the latest reports in the field of in cellulo supramolecular self-assembly and anticipates future advances regarding its activation in response to internal and external stimuli, such as pH changes, reactive oxygen species, and enzymes, as well as external light illumination.
Collapse
Affiliation(s)
- Mohamed Dergham
- Centre for Polymers in Medicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Nanshan, 518055, China.,University of Chinese Academy of Science, Beijing, 100049, China
| | - Shanmeng Lin
- Centre for Polymers in Medicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Nanshan, 518055, China
| | - Jin Geng
- Centre for Polymers in Medicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Nanshan, 518055, China
| |
Collapse
|
22
|
Abstract
Supramolecular assemblies are essential components of living organisms. Cellular scaffolds, such as the cytoskeleton or the cell membrane, are formed via secondary interactions between proteins or lipids and direct biological processes such as metabolism, proliferation and transport. Inspired by nature’s evolution of function through structure formation, a range of synthetic nanomaterials has been developed in the past decade, with the goal of creating non-natural supramolecular assemblies inside living mammalian cells. Given the intricacy of biological pathways and the compartmentalization of the cell, different strategies can be employed to control the assembly formation within the highly crowded, dynamic cellular environment. In this Review, we highlight emerging molecular design concepts aimed at creating precursors that respond to endogenous stimuli to build nanostructures within the cell. We describe the underlying reaction mechanisms that can provide spatial and temporal control over the subcellular formation of synthetic nanostructures. Showcasing recent advances in the development of bioresponsive nanomaterials for intracellular self-assembly, we also discuss their impact on cellular function and the challenges associated with establishing structure–bioactivity relationships, as well as their relevance for the discovery of novel drugs and imaging agents, to address the shortfall of current solutions to pressing health issues. ![]()
Creating artificial nanostructures inside living cells requires the careful design of molecules that can transform into active monomers within a complex cellular environment. This Review explores the recent development of bioresponsive precursors for the controlled formation of intracellular supramolecular assemblies.
Collapse
|
23
|
Yang L, Liu G, Chen Q, Wan Y, Liu Z, Zhang J, Huang C, Xu Z, Li S, Lee CS, Zhang L, Sun H. An Activatable NIR Probe for the Detection and Elimination of Senescent Cells. Anal Chem 2022; 94:5425-5431. [PMID: 35319866 DOI: 10.1021/acs.analchem.2c00239] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cellular senescence is involved in diverse physiological processes. Accumulation of senescent cells can lead to numerous age-related diseases. Therefore, it is of great significance to develop chemical tools to effectively detect and eliminate senescent cells. Till date, a dual functional probe that could detect and eliminate senescent cells has yet been accomplished. Herein, a β-gal-activated probe, MB-βgal, based on the methylene blue (MB) fluorophore, was designed to detect and eliminate senescent cells. In the absence of β-gal, the probe showed no fluorescence and its 1O2 production efficiency was suppressed simultaneously. On the other hand, MB-βgal could be specifically activated by the high level of β-gal in senescent cells, thus, releasing free MB with near-infrared (NIR) fluorescence and high 1O2 production efficiency under light irradiation. MB-βgal demonstrated a fast response, high sensitivity, and high selectivity in detecting β-gal in an aqueous solution and was further applied to visualization and ablation of senescent cells. As a proof of concept, the dual functions of MB-βgal were successfully demonstrated in senescent HeLa cells and mouse embryonic fibroblast cells.
Collapse
Affiliation(s)
- Liu Yang
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, People's Republic of China.,Department of Chemistry and COSDAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, People's Republic of China
| | - Guopan Liu
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, People's Republic of China.,Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China
| | - Qingxin Chen
- Department of Chemistry and COSDAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, People's Republic of China
| | - Yingpeng Wan
- Department of Chemistry and COSDAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China
| | - Zhiyang Liu
- Department of Chemistry and COSDAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, People's Republic of China
| | - Jie Zhang
- Department of Chemistry and COSDAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, People's Republic of China
| | - Chen Huang
- Department of Chemistry and COSDAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, People's Republic of China
| | - Zhiqiang Xu
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, People's Republic of China
| | - Shengliang Li
- Department of Chemistry and COSDAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China.,College of Pharmaceutical Sciences, Soochow University Suzhou, 215123, People's Republic of China
| | - Chun-Sing Lee
- Department of Chemistry and COSDAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China
| | - Liang Zhang
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, People's Republic of China.,Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China
| | - Hongyan Sun
- Department of Chemistry and COSDAF (Centre of Super-Diamond and Advanced Films), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong 999077, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, People's Republic of China
| |
Collapse
|
24
|
Morsli S, Doherty GJ, Muñoz-Espín D. Activatable senoprobes and senolytics: Novel strategies to detect and target senescent cells. Mech Ageing Dev 2022; 202:111618. [PMID: 34990647 DOI: 10.1016/j.mad.2021.111618] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 01/10/2023]
Abstract
Pharmacologically active compounds that manipulate cellular senescence (senotherapies) have recently shown great promise in multiple pre-clinical disease models, and some of them are now being tested in clinical trials. Despite promising proof-of-principle evidence, there are known on- and off-target toxicities associated with these compounds, and therefore more refined and novel strategies to improve their efficacy and specificity for senescent cells are being developed. Preferential release of drugs and macromolecular formulations within senescent cells has been predominantly achieved by exploiting one of the most widely used biomarkers of senescence, the increase in lysosomal senescence-associated β-galactosidase (SA-β-gal) activity, a common feature of most reported senescent cell types. Galacto-conjugation is a versatile therapeutic and detection strategy to facilitate preferential targeting of senescent cells by using a variety of existing formulations, including modular systems, nanocarriers, activatable prodrugs, probes, and small molecules. We discuss the benefits and drawbacks of these specific senescence targeting tools and how the strategy of galacto-conjugation might be utilised to design more specific and sophisticated next-generation senotherapeutics, as well as theranostic agents. Finally, we discuss some innovative strategies and possible future directions for the field.
Collapse
Affiliation(s)
- Samir Morsli
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK
| | - Gary J Doherty
- Department of Oncology, Box 193, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK.
| | - Daniel Muñoz-Espín
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK.
| |
Collapse
|
25
|
Kim BJ. Enzyme-Instructed Self-Assembly of Peptides: From Concept to Representative Applications. Chem Asian J 2022; 17:e202200094. [PMID: 35213091 DOI: 10.1002/asia.202200094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/23/2022] [Indexed: 11/11/2022]
Abstract
Enzyme-instructed self-assembly, integrating enzymatic reaction and molecular self-assembly, has drawn noticeable attention over the last decade with the intension of being used in valuable applications. Recent advances in the field allow it possible to spatiotemporally control peptide self-assembly in cellular milieu, broadening the potential applications of peptide assemblies to cancer therapy and subcellular delivery. In this minireview, the concept of enzyme-instructed self-assembly of peptide, containing enzymatic trigger and spatiotemporal control, is described. Representative applications in cells are also discussed, followed by outlook on the field of enzyme-instructed self-assembly.
Collapse
Affiliation(s)
- Beom Jin Kim
- University of Ulsan, Chemistry, 12, Techno Industrial Complex-ro, 55 beon-gil, 4776, Ulsan, KOREA, REPUBLIC OF
| |
Collapse
|
26
|
Wei X, Li M, Zheng Z, Ma J, Gao Y, Chen L, Peng Y, Yu S, Yang L. Senescence in chronic wounds and potential targeted therapies. BURNS & TRAUMA 2022; 10:tkab045. [PMID: 35187179 PMCID: PMC8853744 DOI: 10.1093/burnst/tkab045] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/15/2021] [Accepted: 11/19/2021] [Indexed: 01/10/2023]
Abstract
Chronic wounds (e.g. diabetic wounds, pressure wounds, vascular ulcers, etc.) do not usually heal in a timely and orderly manner but rather last for years and may lead to irreversible adverse events, resulting in a substantial financial burden for patients and society. Recently, a large amount of evidence has proven that cellular senescence has a crucial influence on chronic nonhealing wounds. As a defensive mechanism, cell senescence is a manner of cell-cycle arrest with increased secretory phenotype to resist death, preventing cells from stress-induced damage in cancer and noncancer diseases. A growing amount of research has advanced the perception of cell senescence in various chronic wounds and focuses on pathological and physiological processes and therapies targeting senescent cells. However, previous reviews have failed to sum up novel understandings of senescence in chronic wounds and emerging strategies targeting senescence. Herein, we discuss the characteristics and mechanisms of cellular senescence and the link between senescence and chronic wounds as well as some novel antisenescence strategies targeting other diseases that may be applied for chronic wounds.
Collapse
Affiliation(s)
- Xuerong Wei
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Minxiong Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, 200011, Shanghai, China
| | - Zijun Zheng
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Jun Ma
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Yujie Peng
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Shengxiang Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangzhou, 510515, China
| |
Collapse
|
27
|
Dergham M, Lin S, Geng J. Supramolecular Self‐assembly in Living Cells. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202114267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Mohamed Dergham
- Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Biomedicine and Biotechnology CHINA
| | - Shanmeng Lin
- Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Biomedicine and Biotechnology CHINA
| | - Jin Geng
- Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Institute of Biomedicine and Biotechnology Xuyuan Road 518055 Shenzhen CHINA
| |
Collapse
|
28
|
Chen G, Zhang J, Sheng M, Zhang S, Wu Q, Liu L, Yu B, Kou J. Serum of limb remote ischemic postconditioning inhibits fMLP-triggered activation and reactive oxygen species releasing of rat neutrophils. Redox Rep 2021; 26:176-183. [PMID: 34663202 PMCID: PMC8530488 DOI: 10.1080/13510002.2021.1982515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Objectives The study explores the protective role of the peripheral serum of limb remote ischemic postconditioning (LRIP) in reducing the reactive oxygen species (ROS) levels and neutrophil activation, which are responsible for the deleterious reperfusion injury. Methods LRIP was induced in Sprague–Dawley rats by three cycles of 5 min occlusion /5 min reperfusion on the left hind limb. The blood samples were collected before LRIP or 0 and 1 h after LRIP (named SerumSham, SerumLRIP0, SerumLRIP1, respectively). The effects of LRIP serum on ROS level and neutrophils activation were determined. The expression of MyD88-TRAF6-MAPKs and PI3K/AKT pathways in neutrophils were examined. Results When compared with SerumSham, SerumLRIP0 and SerumLRIP1 significantly reduced the ROS released from neutrophils activated by fMLP. Meanwhile, the mRNA expression levels of NADPH oxidase subunit p22phox and multiple ROS-producing related key proteins, such as NADPH oxidase subunit p47phox ser 304, ser 345. MyD88, p-ERK, p-JNK and p-P38 expression of neutrophils were downregulated by SerumLRIP0 and SerumLRIP1. SerumLRIP1 also downregulated p47phox mRNA expression and tumor necrosis factor receptor-associated factor 6 (TRAF6) protein expression. Conclusion LRIP serum protects against ROS level and neutrophils activation involving the MyD88-TRAF6-MAPKs. This finding provides new insight into the understanding of LRIP mechanisms.
Collapse
Affiliation(s)
- Gangling Chen
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China.,State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jiangwei Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China.,State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Mingyue Sheng
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China.,State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Sanli Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China.,State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Qi Wu
- State Key Laboratory of Natural Medicines, Research Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Lei Liu
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Boyang Yu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China.,State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Junping Kou
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China.,State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
29
|
Jiang C, Huang H, Kang X, Yang L, Xi Z, Sun H, Pluth MD, Yi L. NBD-based synthetic probes for sensing small molecules and proteins: design, sensing mechanisms and biological applications. Chem Soc Rev 2021; 50:7436-7495. [PMID: 34075930 PMCID: PMC8763210 DOI: 10.1039/d0cs01096k] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Compounds with a nitrobenzoxadiazole (NBD) skeleton exhibit prominent useful properties including environmental sensitivity, high reactivity toward amines and biothiols (including H2S) accompanied by distinct colorimetric and fluorescent changes, fluorescence-quenching ability, and small size, all of which facilitate biomolecular sensing and self-assembly. Amines are important biological nucleophiles, and the unique activity of NBD ethers with amines has allowed for site-specific protein labelling and for the detection of enzyme activities. Both H2S and biothiols are involved in a wide range of physiological processes in mammals, and misregulation of these small molecules is associated with numerous diseases including cancers. In this review, we focus on NBD-based synthetic probes as advanced chemical tools for biomolecular sensing. Specifically, we discuss the sensing mechanisms and selectivity of the probes, the design strategies for multi-reactable multi-quenching probes, and the associated biological applications of these important constructs. We also highlight self-assembled NBD-based probes and outline future directions for NBD-based chemosensors. We hope that this comprehensive review will facilitate the development of future probes for investigating and understanding different biological processes and aid the development of potential theranostic agents.
Collapse
Affiliation(s)
- Chenyang Jiang
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| | - Haojie Huang
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| | - Xueying Kang
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| | - Liu Yang
- Department of Chemistry and Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China.
| | - Zhen Xi
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, College of Chemistry, National Pesticide Engineering Research Center, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Hongyan Sun
- Department of Chemistry and Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China. and Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| | - Michael D Pluth
- Department of Chemistry and Biochemistry, Materials Science Institute, Knight Campus for Accelerating Scientific Impact, Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA.
| | - Long Yi
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Lab of Bioprocess, Beijing University of Chemical Technology (BUCT), Beijing 100029, China.
| |
Collapse
|
30
|
Zhu L, Shi Y, Xiong Y, Ba L, Li Q, Qiu M, Zou Z, Peng G. Emerging self-assembling peptide nanomaterial for anti-cancer therapy. J Biomater Appl 2021; 36:882-901. [PMID: 34180306 DOI: 10.1177/08853282211027882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recently it is mainly focused on anti-tumor comprehensive treatments like finding target tumor cells or activating immune cells to inhibit tumor recurrence and metastasis. At present, chemotherapy and molecular-targeted drugs can inhibit tumor cell growth to a certain extent. However, multi-drug resistance and immune escape often make it difficult for new drugs to achieve expected effects. Peptide hydrogel nanoparticles is a new type of biological material with functional peptide chains as the core and self-assembling peptide (SAP) as the framework. It has a variety of significant biological functions, including effective local inflammation suppression and non-drug-resistant cell killing. Besides, it can induce immune activation more persistently in an adjuvant independent manner when compared with simple peptides. Thus, SAP nanomaterial has great potential in regulating cell physiological functions, drug delivery and sensitization, vaccine design and immunotherapy. Not only that, it is also a potential way to focus on some specific proteins and cells through peptides, which has already been examined in previous research. A full understanding of the function and application of SAP nanoparticles can provide a simple and practical strategy for the development of anti-tumor drugs and vaccine design, which contributes to the historical transition of peptide nanohydrogels from bench to bedside and brings as much survival benefits as possible to cancer patients.
Collapse
Affiliation(s)
- Lisheng Zhu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangyang Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Xiong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Ba
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuting Li
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengjun Qiu
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenwei Zou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Peng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
|
32
|
Wang J, Li H, Xu B. Biological functions of supramolecular assemblies of small molecules in cellular environment. RSC Chem Biol 2021; 2:289-305. [PMID: 34423303 PMCID: PMC8341129 DOI: 10.1039/d0cb00219d] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/05/2021] [Indexed: 12/29/2022] Open
Abstract
Like biomacromolecules, certain small molecules (e.g., aggregators) are able to self-assemble in aqueous phase to form nanoscale aggregates. Though it is well-established that the aggregates may interact with enzymes in vitro, the study of the biological activities of the assemblies of small molecules in cellular environment is only at its beginning. This review summarizes the recent progresses in exploring the biological functions of supramolecular assemblies of small molecules (SASMs). We first discuss the use of SASMs to inhibit pathogenic cells, such as cancer cells and bacteria. The use of SASMs to target different parts of cancer cells, such as pericellular space, cytosol, and subcellular organelles, and to combine with other bioactive entities (e.g., proteins and clinically used drugs), is particularly promising for addressing the challenge of acquired multidrug resistance in cancer therapy. Then, we describe the use of SASMs to sustain physiological functions of normal cells, that is, promoting cells proliferation and differentiation for tissue regeneration. After that, we show the use of SASMs as a basic tool to research cell behaviors, for instance, identifying the specific cells, improving enzyme probes, revealing membrane dynamics, enhancing molecular imaging, and mimicking context-dependent signaling. Finally, we give the outlook of the research of SASMs. We expect that this review, by highlighting the biological functions of SASMs, provides a starting point to explore the chemical biology of SASMs.
Collapse
Affiliation(s)
- Jingyu Wang
- School of Biomedical Engineering and Technology, Tianjin Medical UniversityTianjin 300070P. R. China
| | - Hui Li
- School of Biomedical Engineering and Technology, Tianjin Medical UniversityTianjin 300070P. R. China
| | - Bing Xu
- Department of Chemistry, Brandeis UniversityWalthamMassachusetts 02454USA
| |
Collapse
|
33
|
Morris J, Bietsch J, Bashaw K, Wang G. Recently Developed Carbohydrate Based Gelators and Their Applications. Gels 2021; 7:24. [PMID: 33652820 PMCID: PMC8006029 DOI: 10.3390/gels7010024] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/13/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Carbohydrate based low molecular weight gelators have been an intense subject of study over the past decade. The self-assembling systems built from natural products have high significance as biocompatible materials and renewable resources. The versatile structures available from naturally existing monosaccharides have enriched the molecular libraries that can be used for the construction of gelators. The bottom-up strategy in designing low molecular weight gelators (LMWGs) for a variety of applications has been adopted by many researchers. Rational design, along with some serendipitous discoveries, has resulted in multiple classes of molecular gelators. This review covers the literature from 2017-2020 on monosaccharide based gelators, including common hexoses, pentoses, along with some disaccharides and their derivatives. The structure-based design and structure to gelation property relationships are reviewed first, followed by stimuli-responsive gelators. The last section focuses on the applications of the sugar based gelators, including their utilization in environmental remediation, ion sensing, catalysis, drug delivery and 3D-printing. We will also review the available LMWGs and their structure correlations to the desired properties for different applications. This review aims at elucidating the design principles and structural features that are pertinent to various applications and hope to provide certain guidelines for researchers that are working at the interface of chemistry, biochemistry, and materials science.
Collapse
Affiliation(s)
| | | | | | - Guijun Wang
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA 23529, USA; (J.M.); (J.B.); (K.B.)
| |
Collapse
|
34
|
Wen Z, Zhan J, Li H, Xu G, Ma S, Zhang J, Li Z, Ou C, Yang Z, Cai Y, Chen M. Dual-ligand supramolecular nanofibers inspired by the renin-angiotensin system for the targeting and synergistic therapy of myocardial infarction. Theranostics 2021; 11:3725-3741. [PMID: 33664858 PMCID: PMC7914367 DOI: 10.7150/thno.53644] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Rationale: The compensatory activation of the renin-angiotensin system (RAS) after myocardial infarction (MI) plays a crucial role in the pathogenesis of heart failure. Most existing studies on this subject focus on mono- or dual-therapy of blocking the RAS, which exhibit limited efficacy and often causes serious adverse reactions. Few studies have been conducted on targeted therapy based on the activated RAS post-MI. Thus, the development of multiple-functional nanomedicine with concurrent targeting ability and synergistic therapeutic effect against RAS may show great promise in improving cardiac function post-MI. Methods: We utilized a cooperative self-assembly strategy constructing supramolecular nanofibers— telmisartan-doped co-assembly nanofibers (TDCNfs) to counter-regulate RAS through targeted delivery and combined therapy. TDCNfs were prepared through serial steps of solvent exchange, heating incubation, gelation, centrifugation, and lyophilization, in which the telmisartan was doped in the self-assembly process of Ang1-7 to obtain the co-assembly nanofibers wherein they act as both therapeutic agents and target-guide agents. Results: TDCNfs exhibited the desired binding affinity to the two different receptors, AT1R and MasR. Through the dual ligand-receptor interactions to mediate the coincident downstream pathways, TDCNfs not only displayed favorably targeted properties to hypoxic cardiomyocytes, but also exerted synergistic therapeutic effects in apoptosis reduction, inflammatory response alleviation, and fibrosis inhibition in vitro and in vivo, significantly protecting cardiac function and mitigating post-MI adverse outcomes. Conclusion: A dual-ligand nanoplatform was successfully developed to achieve targeted and synergistic therapy against cardiac deterioration post-MI. We envision that the integration of multiple therapeutic agents through supramolecular self-assembly would offer new insight for the systematic and targeted treatment of cardiovascular diseases.
Collapse
|
35
|
Ren H, Li R, Chen Z, Li L, Wang H. Modification Methods and Applications of Self-Assembly Peptides. CHINESE J ORG CHEM 2021. [DOI: 10.6023/cjoc202104020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
36
|
Adamczyk-Grochala J, Lewinska A. Nano-Based Theranostic Tools for the Detection and Elimination of Senescent Cells. Cells 2020; 9:E2659. [PMID: 33322013 PMCID: PMC7764355 DOI: 10.3390/cells9122659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
The progressive accumulation of apoptosis-resistant and secretory active senescent cells (SCs) in animal and human aged tissues may limit lifespan and healthspan and lead to age-related diseases such as cancer, neurodegenerative disorders, and metabolic syndrome. Thus, SCs are suggested targets in anti-aging therapy. In the last two decades, a number of nanomaterials have gained much attention as innovative tools in theranostic applications due to their unique properties improving target visualization, drug and gene delivery, controlled drug release, effective diagnosis, and successful therapy. Although the healthcare industry has focused on a plethora of applications of nanomaterials, it remains elusive how nanomaterials may modulate cellular senescence, a hallmark of aging. In this review paper, we consider novel nanotechnology-based strategies for healthspan promotion and the prevention of age-related dysfunctions that are based on the delivery of therapeutic compounds capable to preferentially killing SCs (nano-senolytics) and/or modulating a proinflammatory secretome (nano-senomorphics/nano-senostatics). Recent examples of SC-targeted nanomaterials and the mechanisms underlying different aspects of the nanomaterial-mediated senolysis are presented and discussed.
Collapse
Affiliation(s)
- Jagoda Adamczyk-Grochala
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Anna Lewinska
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| |
Collapse
|
37
|
Guo J, Tian C, Xu B. Biomaterials based on noncovalent interactions of small molecules. EXCLI JOURNAL 2020; 19:1124-1140. [PMID: 33088250 PMCID: PMC7573174 DOI: 10.17179/excli2020-2656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 07/27/2020] [Indexed: 11/10/2022]
Abstract
Unlike conventional materials that covalent bonds connecting atoms as the major force to hold the materials together, supramolecular biomaterials rely on noncovalent intermolecular interactions to assemble. The reversibility and biocompatibility of supramolecular biomaterials render them with diverse range of functions and lead to rapid development in the past two decades. This review focuses on the noncovalent and enzymatic control of supramolecular biomaterials, with the introduction to various triggering mechanism to initiate self-assembly. Representative applications of supramolecular biomaterials are highlighted in four categories: tissue engineering, cancer therapy, drug delivery, and molecular imaging. By introducing various applications, we intend to show enzymatic control and noncovalent interactions as a powerful tool for achieving spatiotemporal control of biomaterials both invitro and in vivo for biomedicine.
Collapse
Affiliation(s)
- Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA
| | - Changhao Tian
- Department of Physics, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, China
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02453, USA
| |
Collapse
|
38
|
Kaur H, Sharma P, Patel N, Pal VK, Roy S. Accessing Highly Tunable Nanostructured Hydrogels in a Short Ionic Complementary Peptide Sequence via pH Trigger. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:12107-12120. [PMID: 32988205 DOI: 10.1021/acs.langmuir.0c01472] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Creating diverse nanostructures from a single gelator through modulating the self-assembly pathway has been gaining much attention in recent years. To this direction, we are exploring the effect of modulation of pH as a potential self-assembly pathway in governing the physicochemical properties of the final gel phase material. In this context, we used a classical nongelator with the ionic complementary sequence FEFK, which was rationally conjugated to an aromatic group naphthoxyacetic acid (Nap) at the N-terminal end to tune its gelation behavior. Interestingly, the presence of oppositely charged amino acids in the peptide amphiphile resulted in pH-responsive behavior, leading to the formation of hydrogels over a wide pH range (2.0-12.0); however, their structures differ significantly at the nanoscale. Thus, by simply manipulating the overall charge over the exposed surface of the peptide amphiphiles as a function of pH, we were able to access diverse self-assembled nanostructures within a single gelator domain. The charged state of the gelator at the extreme pH (2.0, 12.0) led to a thinner fiber formation, in contrast to the thicker fibers observed near the physiological pH owing to charge neutralization, thus promoting the lateral association. Such variation in molecular packing was found to be further reflected in the variable mechanical strengths of the peptide hydrogels obtained at different pH values. Moreover, the gelation of the peptide at physiological pH offers an additional advantage to explore this hydrogel as a cell culture scaffold. We anticipate that our study on controlling the self-assembly pathway of the ionic complementary peptide amphiphile can be an elegant approach to access diverse self-assembled materials, which can expand the zone of its applicability as a stimuli-responsive biomaterial.
Collapse
Affiliation(s)
- Harsimran Kaur
- Habitat Centre, Institute of Nano Science and Technology, Sector 64, Phase 10, Mohali, Punjab 160062, India
| | - Pooja Sharma
- Habitat Centre, Institute of Nano Science and Technology, Sector 64, Phase 10, Mohali, Punjab 160062, India
| | - Nidhi Patel
- Habitat Centre, Institute of Nano Science and Technology, Sector 64, Phase 10, Mohali, Punjab 160062, India
| | - Vijay Kumar Pal
- Habitat Centre, Institute of Nano Science and Technology, Sector 64, Phase 10, Mohali, Punjab 160062, India
| | - Sangita Roy
- Habitat Centre, Institute of Nano Science and Technology, Sector 64, Phase 10, Mohali, Punjab 160062, India
| |
Collapse
|
39
|
Abstract
Enzymatic reactions and noncovalent (i.e., supramolecular) interactions are two fundamental nongenetic attributes of life. Enzymatic noncovalent synthesis (ENS) refers to a process where enzymatic reactions control intermolecular noncovalent interactions for spatial organization of higher-order molecular assemblies that exhibit emergent properties and functions. Like enzymatic covalent synthesis (ECS), in which an enzyme catalyzes the formation of covalent bonds to generate individual molecules, ENS is a unifying theme for understanding the functions, morphologies, and locations of molecular ensembles in cellular environments. This review intends to provide a summary of the works of ENS within the past decade and emphasize ENS for functions. After comparing ECS and ENS, we describe a few representative examples where nature uses ENS, as a rule of life, to create the ensembles of biomacromolecules for emergent properties/functions in a myriad of cellular processes. Then, we focus on ENS of man-made (synthetic) molecules in cell-free conditions, classified by the types of enzymes. After that, we introduce the exploration of ENS of man-made molecules in the context of cells by discussing intercellular, peri/intracellular, and subcellular ENS for cell morphogenesis, molecular imaging, cancer therapy, and other applications. Finally, we provide a perspective on the promises of ENS for developing molecular assemblies/processes for functions. This review aims to be an updated introduction for researchers who are interested in exploring noncovalent synthesis for developing molecular science and technologies to address societal needs.
Collapse
Affiliation(s)
- Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Meihui Yi
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Adrianna N Shy
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, Massachusetts 02454, United States
| |
Collapse
|
40
|
Baillet J, Gaubert A, Verget J, Latxague L, Barthélémy P. β-Galactosidase instructed self-assembly of supramolecular bolaamphiphiles hydrogelators. SOFT MATTER 2020; 16:7648-7651. [PMID: 32657300 DOI: 10.1039/d0sm01055c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
β-Galactosidase instructed supramolecular assemblies of Low Molecular Weight Gelators (LMWGs) derived from glyconucleo-bolaamphiphiles have been designed. These precursors, comprising galactose sensitive units at both polar heads, showed the formation of hydrogels upon the action of β-galactosidase.
Collapse
Affiliation(s)
- Julie Baillet
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Alexandra Gaubert
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Julien Verget
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Laurent Latxague
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Philippe Barthélémy
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| |
Collapse
|
41
|
Baillet J, Gaubert A, Bassani DM, Verget J, Latxague L, Barthélémy P. Supramolecular gels derived from nucleoside based bolaamphiphiles as a light-sensitive soft material. Chem Commun (Camb) 2020; 56:3397-3400. [PMID: 32091070 DOI: 10.1039/d0cc00336k] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Light-sensitive Low Molecular Weight Gelators (LMWGs) derived from glyconucleoside bolaamphiphiles containing a stilbene unit displayed gelation abilities in hydroalcoholic mixtures. These materials showed a gel-sol transition under UV irradiation thanks to E-Z isomerization of stilbene and could find potential applications as drug delivery systems.
Collapse
Affiliation(s)
- Julie Baillet
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Alexandra Gaubert
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Dario M Bassani
- University of Bordeaux, CNRS, Bordeaux INP, ISM, UMR 5255, F-33400 Talence, France
| | - Julien Verget
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Laurent Latxague
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| | - Philippe Barthélémy
- University of Bordeaux, INSERM U1212, UMR CNRS 5320, F-33076 Bordeaux, France.
| |
Collapse
|
42
|
Pal VK, Jain R, Roy S. Tuning the Supramolecular Structure and Function of Collagen Mimetic Ionic Complementary Peptides via Electrostatic Interactions. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:1003-1013. [PMID: 31865708 DOI: 10.1021/acs.langmuir.9b02941] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Collagen, the most abundant component of natural ECM, has attracted interest of scientific communities to replicate its multihierarchical self-assembling structure. Recent developments in collagen mimetic peptides were inclined toward the production of self-assembling short peptides capable of mimicking complex higher order structures with tunable mechanical properties. Here, we report for the first time, the crucial molecular design of oppositely charged collagen mimetic shortest bioactive pentapeptide sequences, as a minimalistic building block for development of next-generation biomaterials. Our rational design involves synthesis of two pentapeptides, where the fundamental molecular motif of collagen, that is, Gly-X-Y has been mutated at the central position with positively charged, lysine, and negatively charged, aspartate, residues. Depending on their overall surface charge, these peptides showed high propensity to form self-supporting hydrogel either at acidic or basic pH, which limits their biomedical applications. Interestingly, simple mixing of the two peptides was found to induce the coassembly of these designed peptides, which drives the formation of self-supporting hydrogel at physiological pH and thus enhanced the potential of exploring these peptides for biomedical purposes. This coassembly of ionic peptides was accompanied by the enhancement in the mechanical stiffness of the gels and reduction in overall zeta potential of the combined hydrogel, which provides the evidence for additional electrostatic interactions. Furthermore, the thixotropic nature of these gels offers an additional advantage of exploration of designer biomaterials as injectable gels. The nanofibers of coassembled hydrogel were found to be highly biocompatible to the fibroblast cells compared to the individual peptides, which was evident from their cytotoxicity studies. We anticipate that our rational design of ECM protein mimics in the form of short bioactive peptides will contribute significantly to the development of novel biomaterials and play a crucial role in the field of tissue engineering and regenerative medicines.
Collapse
Affiliation(s)
- Vijay Kumar Pal
- Institute of Nano Science and Technology , Habitat Centre, Sector 64, Phase 10 , Mohali , Punjab 160062 , India
| | - Rashmi Jain
- Institute of Nano Science and Technology , Habitat Centre, Sector 64, Phase 10 , Mohali , Punjab 160062 , India
| | - Sangita Roy
- Institute of Nano Science and Technology , Habitat Centre, Sector 64, Phase 10 , Mohali , Punjab 160062 , India
| |
Collapse
|
43
|
Zou P, Chen WT, Sun T, Gao Y, Li LL, Wang H. Recent advances: peptides and self-assembled peptide-nanosystems for antimicrobial therapy and diagnosis. Biomater Sci 2020; 8:4975-4996. [DOI: 10.1039/d0bm00789g] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Bacterial infections, especially the refractory treatment of drug-resistant bacteria, are one of the greatest threats to human health. Self-assembling peptide-based strategies can specifically detect the bacteria at the site of infection in the body and kill it.
Collapse
Affiliation(s)
- Pengfei Zou
- CAS Center for Excellence in Nanoscience
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
- National Center for Nanoscience and Technology (NCNST)
- Center of Materials Science and Optoelectronics Engineering
- University of Chinese Academy of Sciences
| | - Wen-Ting Chen
- Department of Chemistry and the Department of Physics and Astronomy
- University of Waterloo
- Waterloo
- Canada
| | - Tongyi Sun
- School of Life Science and Technology
- Shandong Key Laboratory of Proteins and Peptides Pharmaceutical Engineering
- Shandong Universities Key Laboratory of Biopharmaceuticals
- Weifang Medical University
- Weifang
| | - Yuanyuan Gao
- School of Pharmacy
- Weifang Medical University
- Weifang
- China
| | - Li-Li Li
- CAS Center for Excellence in Nanoscience
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
- National Center for Nanoscience and Technology (NCNST)
- Center of Materials Science and Optoelectronics Engineering
- University of Chinese Academy of Sciences
| | - Hao Wang
- CAS Center for Excellence in Nanoscience
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
- National Center for Nanoscience and Technology (NCNST)
- Center of Materials Science and Optoelectronics Engineering
- University of Chinese Academy of Sciences
| |
Collapse
|
44
|
Zheng D, Chen Y, Ai S, Zhang R, Gao Z, Liang C, Cao L, Chen Y, Hong Z, Shi Y, Wang L, Li X, Yang Z. Tandem Molecular Self-Assembly Selectively Inhibits Lung Cancer Cells by Inducing Endoplasmic Reticulum Stress. RESEARCH 2019; 2019:4803624. [PMID: 31912037 PMCID: PMC6944487 DOI: 10.34133/2019/4803624] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/13/2019] [Indexed: 12/31/2022]
Abstract
The selective formation of nanomaterials in cancer cells and tumors holds great promise for cancer diagnostics and therapy. Until now, most strategies rely on a single trigger to control the formation of nanomaterials in situ. The combination of two or more triggers may provide for more sophisticated means of manipulation. In this study, we rationally designed a molecule (Comp. 1) capable of responding to two enzymes, alkaline phosphatase (ALP), and reductase. Since the A549 lung cancer cell line showed elevated levels of extracellular ALP and intracellular reductase, we demonstrated that Comp. 1 responded in a stepwise fashion to those two enzymes and displayed a tandem molecular self-assembly behavior. The selective formation of nanofibers in the mitochondria of the lung cancer cells led to the disruption of the mitochondrial membrane, resulting in an increased level of reactive oxygen species (ROS) and the release of cytochrome C (Cyt C). ROS can react with proteins, resulting in endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). This severe ER stress led to disruption of the ER, formation of vacuoles, and ultimately, apoptosis of the A549 cells. Therefore, Comp. 1 could selectively inhibit lung cancer cells in vitro and A549 xenograft tumors in vivo. Our study provides a novel strategy for the selective formation of nanomaterials in lung cancer cells, which is powerful and promising for the diagnosis and treatment of lung cancer.
Collapse
Affiliation(s)
- Debin Zheng
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Yumiao Chen
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Sifan Ai
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Renshu Zhang
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, and Wenzhou Institute of Biomaterials and Engineering, CNITECH, CAS, Wenzhou 325035, China
| | - Zhengfeng Gao
- College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Chunhui Liang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Li Cao
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Yaoxia Chen
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Zhangyong Hong
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Yang Shi
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Ling Wang
- College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Xingyi Li
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, and Wenzhou Institute of Biomaterials and Engineering, CNITECH, CAS, Wenzhou 325035, China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|