1
|
Dai X, Mei K, Liu J, Sun B, Qiu N. Redox-responsive ferulic acid-biotin conjugate: Design, synthesis, and enhanced anticancer efficacy. Bioorg Med Chem Lett 2025; 122:130209. [PMID: 40147804 DOI: 10.1016/j.bmcl.2025.130209] [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: 12/19/2024] [Revised: 02/18/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
In this study, ferulic acid (FA) was conjugated with biotin via a disulfide bond to improve its anticancer activity. The resulting conjugate (FA-SS-Bio) was characterized by proton nuclear magnetic resonance (1H NMR) and exhibited an amorphous structure, in contrast to the crystalline nature of FA. FA-SS-Bio demonstrated accelerated drug release under reductive and oxidative conditions. Biotinylation significantly increased cell uptake of the drug in biotin receptor (BR)-positive HeLa and MCF-7 cells, as confirmed by cellular uptake studies and molecular docking, which revealed strong biotin-BR interactions. Additionally, the cytotoxicity of FA-SS-Bio was significantly improved, with IC50 values that were 2.94-fold and 2.95-fold lower than those of free FA against HeLa and MCF-7 cells, respectively. BR blockade with biotin reduced FA-SS-Bio cytotoxicity in a concentration-dependent manner, confirming biotin-mediated targeting. Apoptosis assays showed enhanced FA-induced apoptosis due to biotin and disulfide bonds. FA-SS-Bio demonstrated excellent blood compatibility, with a hemolysis rate below 0.5 %, compared to ∼1.5 % for FA. Additionally, FA-SS-Bio exhibited higher cell viability in MCF-10 A cells than in cancer cells, highlighting its favorable safety profile. These findings provide a novel perspective on the design of prodrug conjugates for improved cancer therapy.
Collapse
Affiliation(s)
- Xiaoshuang Dai
- College of Ecology and Environment, Chengdu University of Technology, Chengdu 610059, China
| | - Ke Mei
- College of Ecology and Environment, Chengdu University of Technology, Chengdu 610059, China
| | - Jianpeng Liu
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Bin Sun
- College of Ecology and Environment, Chengdu University of Technology, Chengdu 610059, China
| | - Neng Qiu
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China.
| |
Collapse
|
2
|
Tan K, Zhang H, Yang J, Wang H, Li Y, Ding G, Gu P, Yang S, Li J, Fan X. Organelle-oriented nanomedicines in tumor therapy: Targeting, escaping, or collaborating? Bioact Mater 2025; 49:291-339. [PMID: 40161442 PMCID: PMC11953998 DOI: 10.1016/j.bioactmat.2025.02.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Precise tumor therapy is essential for improving treatment specificity, enhancing efficacy, and minimizing side effects. Targeting organelles is a key strategy for achieving this goal and is a frontier research area attracting a considerable amount of attention. The concept of organelle targeting has a significant effect on the structural design of the nanodrugs employed. Most notably, the intricate interactions among different organelles in a tumor cell essentially create a unified system. Unfortunately, this aspect might have been somewhat overlooked when existing organelle-targeting nanodrugs were designed. In this review, we underscore the synergistic relationship among the various organelles and advocate for a holistic view of organelle-targeting design. Through the integration of biology and material science, recent advancements in organelle targeting, escaping, and collaborating are consolidated to offer fresh perspectives for the development of antitumor nanomedicines.
Collapse
Affiliation(s)
- Kexin Tan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Haiyang Zhang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Jianyuan Yang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Hang Wang
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Yongqiang Li
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Guqiao Ding
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Ping Gu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Siwei Yang
- National Key Laboratory of Materials for Integrated Circuits, Joint Laboratory of Graphene Materials and Applications, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Jipeng Li
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, and Center for Basic Medical Research and Innovation in Visual System Diseases of Ministry of Education, Shanghai, 200011, PR China
| |
Collapse
|
3
|
Nguyen CTG, Meng F. Unleashing the power of nucleic acid therapeutics through efficient cytosolic delivery. J Control Release 2025; 383:113774. [PMID: 40280238 DOI: 10.1016/j.jconrel.2025.113774] [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: 12/11/2024] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
The approval of siRNA-based therapy for liver disease in 2018 and the subsequent success of mRNA-based SARS-CoV-2 vaccines have inaugurated a new era in nucleic acid-based therapeutics. These breakthroughs underscore the transformative potential of nucleic acid-based therapeutics, which modulate gene function, correct genetic defects, or disrupt pathological molecular processes. Such advances represent a paradigm shift in modern medicine. Despite their immense promise, the clinical realization of nucleic acid-based therapies is fundamentally constrained by endosomal entrapment, a critical barrier that significantly limits therapeutic efficacy. Overcoming this obstacle is imperative to fully unlock the potential of these therapies. Designing effective strategies to facilitate the escape of nucleic acids from endosomes-or bypassing endosomal pathways altogether-remains a central challenge in the field. In this review, we provide a comprehensive and critical analysis of current approaches aimed at enhancing endosomal escape or circumventing endosomal entrapment. By highlighting both the successes and limitations of these strategies, we aim to offer valuable insights to inform the development of more efficient and clinically viable nucleic acid delivery systems, advancing the future of molecular medicine.
Collapse
Affiliation(s)
- Cao Thuy Giang Nguyen
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, MA 01854, USA
| | - Fanfei Meng
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, MA 01854, USA.
| |
Collapse
|
4
|
Zhang J, Yang H, Li L, Peng C, Li J. Noninvasive Transdermal Delivery of STING Agonists Reshapes the Immune Microenvironment of Melanoma and Potentiates Checkpoint Blockade Therapy Efficacy. ACS APPLIED BIO MATERIALS 2025; 8:3156-3166. [PMID: 40191891 DOI: 10.1021/acsabm.4c02004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
The emergence of immunotherapy as a revolutionary therapeutic modality has fostered confidence and underscored its potent efficacy in tumor therapy. However, enhancing the therapeutic efficacy of immunotherapy by precise and judicious administration poses a significant challenge. In this context, we have developed a disulfide-bearing transdermal nanovaccine by integrating a thiol-reactive agent lipoic acid (LA) into a metal-coordinated cyclic dinucleotide nanoassembly, designated as LA-Mn-cGAMP (LMC) nanovaccines. Upon topical application to the skin with melanoma, the dithiolane moiety of LA enables thiol-disulfide dynamic exchange in the skin, hence facilitating penetration into both the skin and subcutaneous tumor tissues via the thiol-mediated uptake (TMU) mechanism. Our findings demonstrate that transdermal administration of LMC significantly enhances STING activation, mitigates the immunosuppressive tumor microenvironment (TME), and retards melanoma progression. Moreover, the remodeled TME amplifies the efficacy of immune checkpoint inhibitors. This advancement offers an administration strategy for existing STING agonist therapy, potentially improving the biosafety of immunotherapy.
Collapse
Affiliation(s)
- Junjie Zhang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
| | - Hui Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
| | - Liang Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, China
| | - Changkun Peng
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
| | - Jingying Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, China
| |
Collapse
|
5
|
Li Z, Xie X, Lu Z, Zhang Y, Kong Y, You J, Zhu JJ. Albumin Modulated Homodimer as an Efficient Photosensitizer for Long-Term Imaging-Guided Tumor Therapy Directed with Sunlight Irradiation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411736. [PMID: 40095450 DOI: 10.1002/smll.202411736] [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: 12/04/2024] [Revised: 03/05/2025] [Indexed: 03/19/2025]
Abstract
The reactive oxygen species (ROS) amplification caused by inevitable plasma albumin encapsulation is still a challenge to circumvent the systemic adverse effects in the photodynamic therapy (PDT) process. Herein, a disulfide bond linked homodimer, Cy1280, which is modulated by albumin to accurately balance the fluorescence and ROS generation and exhibit a weak fluorescence and sealed PDT effect during blood circulation, is exploited. Cy1280 can be specifically internalized and dispersed at the tumor site via Organic Anion Transporter Proteins (OATPs) and thiol-disulfide exchange mediated synergistic uptake and activated after mild sunlight irradiation (100 ± 5 Klx) to sensitize neighboring oxygen in cellular mitochondria to execute direct protein dysfunction effect. The dynamic covalent chemistry (DCC) facilitates prolonged and sustained retention in tumors (>336 h) and demonstrates the efficacy of imaging-guided solid-tumor therapy in tumor-bearing BALB/C mice. This study resolves the inevitable stubborn impotent tumor penetration caused by bulky-sized nanoparticles and high interstitial pressure of tumor with synergistic uptake manner, the long-term circulation and sealed PDT manipulated with albumin also improve the whole body phototoxic symptom. The advantageous feature of Cy1280 provides a promising candidate for overcoming the off-target phototoxicity and inadequate accumulation challenges in clinical translation with photosensitizers (PSs).
Collapse
Affiliation(s)
- Zan Li
- Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, P. R. China
| | - Xiunan Xie
- Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, P. R. China
| | - Zhihao Lu
- Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Yifan Zhang
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, P. R. China
| | - Yuerui Kong
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, P. R. China
| | - Jinmao You
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, P. R. China
| | - Jun-Jie Zhu
- Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
6
|
Zhao X, Zhang Y, Wang X, Fu Z, Zhong Z, Deng C. Multivalent ionizable lipid-polypeptides for tumor-confined mRNA transfection. Bioact Mater 2025; 46:423-433. [PMID: 39850023 PMCID: PMC11754973 DOI: 10.1016/j.bioactmat.2024.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/25/2024] [Accepted: 12/28/2024] [Indexed: 01/25/2025] Open
Abstract
mRNA therapeutics is revolutionizing the treatment concepts toward many diseases including cancer. The potential of mRNA is, however, frequently limited by modest control over site of transfection. Here, we have explored a library of multivalent ionizable lipid-polypeptides (MILP) to achieve robust mRNA complexation and tumor-confined transfection. Leveraging the multivalent electrostatic, hydrophobic, and H-bond interactions, MILP efficiently packs both mRNA and plasmid DNA into sub-80 nm nanoparticles that are stable against lyophilization and long-term storage. The best MILP@mRNA complexes afford 8-fold more cellular uptake than SM-102 lipid nanoparticle formulation (SM-102 LNP), efficient endosomal disruption, and high transfection in different cells. Interestingly, MILP@mLuc displays exclusive tumor residence and distribution via multivalency-directed strong affinity and transcytosis, and affords specific protein expression in tumor cells and macrophages at tumor sites following intratumoral injection, in sharp contrast to the indiscriminate distribution and transfection in main organs of SM-102 LNP. Notably, MILP@mIL-12 with specific and efficient cytokine expression generates significant remodeling of tumor immunoenvironments and remarkable antitumor response in subcutaneous Lewis lung carcinoma and 4T1 tumor xenografts. MILP provides a unique strategy to site-specific transfection that may greatly broaden the applications of mRNA.
Collapse
Affiliation(s)
- Xiaofei Zhao
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymers, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Yueyue Zhang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymers, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Xin Wang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymers, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Ziming Fu
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymers, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymers, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Chao Deng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymers, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| |
Collapse
|
7
|
Deng S, Hu L, Chen G, Ye J, Xiao Z, Guan T, Guo S, Xia W, Cheng D, Wan X, Cheng K, Ou C. A PD-L1 siRNA-Loaded Boron Nanoparticle for Targeted Cancer Radiotherapy and Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2419418. [PMID: 39955653 DOI: 10.1002/adma.202419418] [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: 12/11/2024] [Revised: 01/25/2025] [Indexed: 02/17/2025]
Abstract
Although the combination of radiotherapy and immunotherapy is regarded as a promising clinical treatment strategy, numerous clinical trials have failed to demonstrate synergistic effects. One of the key reasons is that conventional radiotherapies inevitably damage intratumoral effector immune cells. Boron Neutron Capture Therapy (BNCT) is a precise radiotherapy that selectively kills tumor cells while sparing adjacent normal cells, by utilizing 10B agents and neutron irradiation. Therefore, combinational BNCT-immunotherapy holds promise for achieving more effective synergistic effects. Here it develops a 10B-containing polymer that self-assembled with PD-L1 siRNA to form 10B/siPD-L1 nanoparticles for combinational BNCT-immunotherapy. Unlike antibodies, PD-L1 siRNA can inhibit intracellular PD-L1 upregulated by BNCT, activating T-cell immunity while also suppressing DNA repair. This can enhance BNCT-induced DNA damage, promoting immunogenic cell death (ICD) and further amplifying the antitumor immune effect. The results demonstrated that BNCT using 10B/siPD-L1 nanoparticles precisely killed tumor cells while sparing adjacent T cells and induced a potent antitumor immune response, inhibiting distal and metastatic tumors.
Collapse
Affiliation(s)
- Shaohui Deng
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Lijun Hu
- The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Guo Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jujian Ye
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zecong Xiao
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Tianwang Guan
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Shuai Guo
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Wei Xia
- Neuboron Medtech Ltd, Nanjing, 211112, China
| | - Du Cheng
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Xiaochun Wan
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York, 10032, USA
| | - Caiwen Ou
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| |
Collapse
|
8
|
Bresinsky M, Goepferich A. Control of biomedical nanoparticle distribution and drug release in vivo by complex particle design strategies. Eur J Pharm Biopharm 2025; 208:114634. [PMID: 39826847 DOI: 10.1016/j.ejpb.2025.114634] [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: 10/16/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
The utilization of targeted nanoparticles as a selective drug delivery system is a powerful tool to increase the amount of active substance reaching the target site. This can increase therapeutic efficacy while reducing adverse drug effects. However, nanoparticles face several challenges: upon injection, the immediate adhesion of plasma proteins may mask targeting ligands, thereby diminishing the target cell selectivity. In addition, opsonization can lead to premature clearance and the widespread presence of receptors or enzymes limits the accuracy of target cell recognition. Nanoparticles may also suffer from endosomal entrapment, and controlled drug release can be hindered by premature burst release or insufficient particle retention at the target site. Various strategies have been developed to address these adverse events, such as the implementation of switchable particle properties, regulating the composition of the formed protein corona, or using click-chemistry based targeting approaches. This has resulted in increasingly complex particle designs, raising the question of whether this development actually improves the therapeutic efficacy in vivo. This review provides an overview of the challenges in targeted drug delivery and explores potential solutions described in the literature. Subsequently, appropriate strategies for the development of nanoparticular drug delivery concepts are discussed.
Collapse
Affiliation(s)
- Melanie Bresinsky
- Department of Pharmaceutical Technology, University of Regensburg 93053 Regensburg, Bavaria, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg 93053 Regensburg, Bavaria, Germany.
| |
Collapse
|
9
|
Sun K, Kang Z, Yan C, Yu Y, Han L, Shi Y, Chen L, Shi J, Chen Y, Sun J. Rescuing ACE2-Deficiency-Mediated Nucleus Pulposus Senescence and Intervertebral Disc Degeneration by a Nanotopology-Enhanced RNAi System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412908. [PMID: 39804951 PMCID: PMC11884558 DOI: 10.1002/advs.202412908] [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: 10/14/2024] [Revised: 12/03/2024] [Indexed: 01/16/2025]
Abstract
Nucleus pulposus cell (NPC) senescence contributes to intervertebral disc degeneration (IVDD). However, the underlying molecular mechanisms are not fully understood. In this study, it is demonstrated that angiotensin-converting enzyme 2 (ACE2) counteracted the aging of NPCs and IVDD at the cellular and physiological levels. The expression of ACE2 correlates negatively with the degree of NPC senescence and IVDD. Using both loss- and gain-of-function mouse models, it is revealed that ACE2 deficiency increased the senescence of NPCs and exacerbated injury- or instability-induced IVDD, whereas ACE2 overexpression counteracted these detrimental effects. Mechanistically, integrated analysis of single-cell and bulk transcriptomics shows that ACE2 deficiency results in the activation of TGFβ2/Smads signaling pathway and the transcription of Serpine1, ultimately triggering NPC senescence and IVDD. A nanomedical delivery system (virus-like nanovectors, VNs) composed of nanovectors with a virus-like surface topology and small interfering RNA targeting Serpine1 (VN-siSer) is developed. With nanotopology-enhanced transfection efficiency, RNA-interfering treatment by VN-siSer effectively alleviated NPC senescence and IVDD at both the cellular and animal levels. Overall, the data reveal the underlying mechanisms of ACE2 in NPC senescence and IVDD pathogenesis and propose a distinct paradigm of precise nanomedical senescence-blockade RNAi for IVDD treatment.
Collapse
Affiliation(s)
- Kaiqiang Sun
- Department of Orthopedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
- Department of OrthopedicsNaval Medical Center of PLAShanghai200052P. R. China
| | - Zijian Kang
- Department of Rheumatology and ImmunologyShanghai Sixth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200233P. R. China
| | - Chen Yan
- Department of Orthopedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Yan Yu
- State Key Laboratory of Molecular Engineering of PolymersFudan UniversityShanghai200433P.R. China
| | - Linhui Han
- Department of Orthopedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Yangyang Shi
- Department of Orthopedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Liang Chen
- State Key Laboratory of Molecular Engineering of PolymersFudan UniversityShanghai200433P.R. China
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Jiangang Shi
- Department of Orthopedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Jingchuan Sun
- Department of Orthopedic SurgeryChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| |
Collapse
|
10
|
Liu S, Li H, Xi S, Zhang Y, Sun T. Advancing CNS Therapeutics: Enhancing Neurological Disorders with Nanoparticle-Based Gene and Enzyme Replacement Therapies. Int J Nanomedicine 2025; 20:1443-1490. [PMID: 39925682 PMCID: PMC11806685 DOI: 10.2147/ijn.s457393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 12/12/2024] [Indexed: 02/11/2025] Open
Abstract
Given the complexity of the central nervous system (CNS) and the diversity of neurological conditions, the increasing prevalence of neurological disorders poses a significant challenge to modern medicine. These disorders, ranging from neurodegenerative diseases to psychiatric conditions, not only impact individuals but also place a substantial burden on healthcare systems and society. A major obstacle in treating these conditions is the blood-brain barrier (BBB), which restricts the passage of therapeutic agents to the brain. Nanotechnology, particularly the use of nanoparticles (NPs), offers a promising solution to this challenge. NPs possess unique properties such as small size, large surface area, and modifiable surface characteristics, enabling them to cross the BBB and deliver drugs directly to the affected brain regions. This review focuses on the application of NPs in gene therapy and enzyme replacement therapy (ERT) for neurological disorders. Gene therapy involves altering or manipulating gene expression and can be enhanced by NPs designed to carry various genetic materials. Similarly, NPs can improve the efficacy of ERT for lysosomal storage disorders (LSDs) by facilitating enzyme delivery to the brain, overcoming issues like immunogenicity and instability. Taken together, this review explores the potential of NPs in revolutionizing treatment options for neurological disorders, highlighting their advantages and the future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Haisong Li
- Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Shiwen Xi
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, People’s Republic of China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, People’s Republic of China
- International Center of Future Science, Jilin University, Changchun, People’s Republic of China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
11
|
Dabas R, Navaratnam N, Iino H, Saidjalolov S, Matile S, Carling D, Rueda DS, Kamaly N. Precise intracellular uptake and endosomal release of diverse functional mRNA payloads via glutathione-responsive nanogels. Mater Today Bio 2025; 30:101425. [PMID: 39839495 PMCID: PMC11745970 DOI: 10.1016/j.mtbio.2024.101425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/23/2025] Open
Abstract
We present a novel, highly customizable glutathione-responsive nanogel (NG) platform for efficient mRNA delivery with precise mRNA payload release control. Optimization of various cationic monomers, including newly synthesized cationic polyarginine, polyhistidine, and acrylated guanidine monomers, allowed fine-tuning of NG properties for mRNA binding. By incorporating a poly(ethylene) glycol-based disulphide crosslinker, we achieved glutathione-triggered mRNA release, enabling targeted intracellular delivery. Our NGs demonstrated superior encapsulation (up to 89.3 %) and loading (10.7 %) efficiencies, with controlled mRNA release kinetics at intracellular glutathione concentrations. NGs outperformed commercial transfection reagents across multiple cell lines, including traditionally difficult-to-transfect lines. We demonstrate the platform's versatility by successfully delivering GFP mRNA, Mango II RNA aptamers, and functionally relevant β2-AMPK mRNA. Furthermore, we used TIRF microscopy to measure exact RNA copy number within the NGs. Notably, mechanistic cellular uptake studies revealed that disulphide-containing NGs exhibit enhanced cellular uptake and endosomal escape, potentially due to interactions with cell surface thiols. This work represents a highly tuneable, efficient, and biocompatible platform for mRNA delivery with relevance for gene therapy and vaccine development.
Collapse
Affiliation(s)
- Rupali Dabas
- Cellular Stress Research Group, MRC Laboratory of Medical Sciences, Imperial College London, W12 0HS, London, UK
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, W12 0BZ, London, UK
| | - Naveenan Navaratnam
- Cellular Stress Research Group, MRC Laboratory of Medical Sciences, Imperial College London, W12 0HS, London, UK
| | - Haruki Iino
- Single Molecule Imaging Group, MRC Laboratory of Medical Sciences, Imperial College London, W12 0HS, London, UK
- Section of Virology, Department of Infectious Disease, Imperial College London, W12 0HS, London, UK
| | | | - Stefan Matile
- Department of Organic Chemistry, University of Geneva, Geneva, Switzerland
| | - David Carling
- Cellular Stress Research Group, MRC Laboratory of Medical Sciences, Imperial College London, W12 0HS, London, UK
| | - David S. Rueda
- Single Molecule Imaging Group, MRC Laboratory of Medical Sciences, Imperial College London, W12 0HS, London, UK
- Section of Virology, Department of Infectious Disease, Imperial College London, W12 0HS, London, UK
| | - Nazila Kamaly
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, W12 0BZ, London, UK
| |
Collapse
|
12
|
Luo Y, Wang Y, Chen Y, Li B, Yang T, Zhao X, Ding P. Development and evaluation of a novel biodegradable Poly(amidoamine) with Bis(guanidinium) and benzene ring structures for enhanced gene delivery. J Drug Deliv Sci Technol 2025; 104:106452. [DOI: 10.1016/j.jddst.2024.106452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
13
|
Wang Y, Yin S, He D, Zhang Y, Dong Z, Tian Z, Li J, Chen F, Wang Y, Li M, He Q. Dual Strategies Based on Golgi Apparatus/Endoplasmic Reticulum Targeting and Anchoring for High-Efficiency siRNA Delivery and Tumor RNAi Therapy. ACS NANO 2025; 19:3791-3806. [PMID: 39801087 DOI: 10.1021/acsnano.4c14778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Endolysosomal degradation of small interfering RNA (siRNA) significantly reduces the efficacy of RNA interference (RNAi) delivered by nonviral systems. Leveraging Golgi apparatus/endoplasmic reticulum (Golgi/ER) transport can help siRNA bypass the endolysosomal degradation pathway, but this approach may also result in insufficient siRNA release and an increased risk of Golgi/ER-mediated exocytosis. To address these challenges, we developed two distinct strategies using a nanocomplex of cell-penetrating poly(disulfide)s and chondroitin sulfate, which enhances targeted internalization, Golgi transport, and rapid cytoplasmic release of loaded siRNA. In the first strategy, monensin synergy was found to enhance RNAi by inhibiting both exocytosis and autophagic degradation. In the second strategy, a "directed sorting" approach based on KDEL peptide-mediated retrograde transport was introduced. By conjugation of the KDEL peptide to chondroitin sulfate, Golgi-to-ER transport was promoted, reducing "random" Golgi/ER-related exocytosis. These two strategies operate alternatively to achieve high-efficiency RNAi with a significant therapeutic potential. Notably, in a mouse melanoma model using anti-Bcl-2 siRNA, the strategies achieved tumor inhibition rates of 87.1 and 90.1%, respectively. These two strategies, based on "targeting" and "anchoring" Golgi/ER, provide potent solutions to overcome the challenges of cellular internalization, intracellular release, and exocytosis in efficient siRNA delivery.
Collapse
Affiliation(s)
- Yashi Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Sheng Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Dan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yujia Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Ziyan Dong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zhipeng Tian
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jiayu Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Fang Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yang Wang
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm 17177, Sweden
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Immunology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
14
|
Wang S, Zou Y, Hu L, Lv Y. Manganese-doped liquid metal nanoplatforms for cellular uptake and glutathione depletion-enhanced photothermal and chemodynamic combination tumor therapy. Acta Biomater 2025; 191:369-385. [PMID: 39522626 DOI: 10.1016/j.actbio.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/02/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Chemodynamic therapy (CDT) involves the catalysis of in situ overexpressed hydrogen peroxide (H2O2) into highly toxic reactive oxygen species (ROS) to treat tumors. However, the efficacy of CDT is greatly hampered by limited cellular internalization efficiency, ROS scavenging by glutathione (GSH), and slow reaction rate. To overcome the current limitations of CDT, a manganese-doped and polyethylene glycol (PEG)-modified liquid metal (LM)-silica nanoplatform (labeled as Mn-LMOP) with varying stiffness is constructed to achieve synergistic photothermal therapy (PTT) and CDT, which can further induce immunogenic cell death (ICD) in tumors to enhance the anti-tumour effects. Significantly, benefiting from the increased stiffness, the Mn-LMOP nanoparticles (NPs) can enhance cellular uptake and lysosomal escape, and gradually accumulate in tumor sites. Moreover, manganese-doped NPs exhibite good photothermal effects and can rapidly reacte with intratumoral GSH to produce Mn2+, inhibiting GSH-mediated ROS clearance and promoting the efficiency of CDT. This combined treatment strategy can activate the immune response of the tumors, which holds the promise of photothermal/chemodynamic/immune multimodal therapeutic effects. This LM-based nanosystem will provide a paradigm for enhanced CDT/PTT combination anti-tumour efficacy. STATEMENT OF SIGNIFICANCE: Chemodynamic therapy (CDT) is a promising drug-free treatment approach characterized by its low invasiveness and minimal side effect. However, CDT encounters challenges such as high levels of glutathione (GSH), low Fenton-like reaction rate, and inefficient cellular uptake in tumor tissues. Here, a manganese-doped liquid metal (LM) nanomaterial was designed to achieve synergistic photothermal therapy (PTT) and CDT. This innovative strategy enhanced cellular uptake by adjusting the mechanical property of nanoparticles (NPs) and facilitated the consumption of GSH, while simultaneously accelerating the Fenton-like reaction rate with the assistance of PTT-mediated hyperthermia. This combined CDT/PTT strategy also activated the immune response within the tumor, demonstrating significant therapeutic potential.
Collapse
Affiliation(s)
- Shuai Wang
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Yang Zou
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, PR China
| | - Liefeng Hu
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, PR China; School of Materials Science and Engineering, Wuhan Textile University, Wuhan 430200, PR China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, PR China.
| |
Collapse
|
15
|
Gong S, Liu B, Qiu J, Huang F, Thayumanavan S. Antibody-Directing Antibody Conjugates (ADACs) Enabled by Orthogonal Click Chemistry for Targeted Intracellular Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402874. [PMID: 39162119 PMCID: PMC11581923 DOI: 10.1002/smll.202402874] [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: 04/10/2024] [Revised: 08/03/2024] [Indexed: 08/21/2024]
Abstract
Using orthogonal click chemistries for efficient nanoscale self-assembly, a new antibody-directing antibody conjugate (ADAC) nanogel is generated. In this system, one of the antibodies is displayed on the nanogel surface to specifically recognize cell-surface epitopes while the other antibody is encapsulated inside the nanogel core. The system is programmed to release the latter antibody in its functional form in the cytosolic environment of a specific cell to engage intracellular targets. ADACs offer a potential solution to harness the advantages seen with antibody-drug conjugates (ADCs) to deliver therapeutic cargos to specific tissues, but with the added capability of carrying biologics as the cargo. In this manuscript, this potential is demonstrated through delivery of antibodies against intracellular targets in specific cells. This platform offers new avenues for precise therapeutic interventions and the potential to address previously "undruggable" cellular targets.
Collapse
Affiliation(s)
- Shuai Gong
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Bin Liu
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Jingyi Qiu
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Fangying Huang
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| |
Collapse
|
16
|
Bernal-Martínez AM, Bedrina B, Angulo-Pachón CA, Galindo F, Miravet JF, Castelletto V, Hamley IW. pH-Induced conversion of bolaamphiphilic vesicles to reduction-responsive nanogels for enhanced Nile Red and Rose Bengal delivery. Colloids Surf B Biointerfaces 2024; 242:114072. [PMID: 39024718 DOI: 10.1016/j.colsurfb.2024.114072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024]
Abstract
This study details the preparation and investigation of molecular nanogels formed by the self-assembly of bolaamphiphilic dipeptide derivatives containing a reduction-sensitive disulfide unit. The described bolaamphiphiles, featuring amino acid terminal groups, generate cationic vesicles at pH 4, which evolve into gel-like nanoparticles at pH 7. The critical aggregation concentration has been determined, and the nanogels' size and morphology have been characterized through Dynamic Light Scattering (DLS) and Transmission Electron Microscopy (TEM). Circular Dichroism (CD) spectroscopy reveals substantial molecular reconfigurations accompanying the pH shift. These nanogels enhance the in vitro cellular uptake of the lipophilic dye Nile Red and the ionic photosensitizer Rose Bengal into Human colon adenocarcinoma (HT-29) cells, eliminating the need for organic co-solvents in the former case. Fluorescence measurements with Nile Red as a probe indicate the reduction-sensitive disassembly of the nanogels. In photodynamic therapy (PDT) applications, Rose Bengal-loaded nanogels demonstrate notable improvements, with flow cytometry analysis evidencing increased apoptotic activity in the study with HT-29 cells.
Collapse
Affiliation(s)
- Ana M Bernal-Martínez
- Department of Inorganic and Organic Chemistry, Universitat Jaume I, Avda. Sos Baynat s/n, Castelló 12071, Spain
| | - Begoña Bedrina
- Department of Inorganic and Organic Chemistry, Universitat Jaume I, Avda. Sos Baynat s/n, Castelló 12071, Spain
| | - César A Angulo-Pachón
- Department of Inorganic and Organic Chemistry, Universitat Jaume I, Avda. Sos Baynat s/n, Castelló 12071, Spain; Departamento de Química Orgánica y Bio-orgánica, Facultad de Ciencias, Universidad Nacional de Educación a Distancia, UNED, Las Rozas, Madrid 28232, Spain
| | - Francisco Galindo
- Department of Inorganic and Organic Chemistry, Universitat Jaume I, Avda. Sos Baynat s/n, Castelló 12071, Spain
| | - Juan F Miravet
- Department of Inorganic and Organic Chemistry, Universitat Jaume I, Avda. Sos Baynat s/n, Castelló 12071, Spain.
| | - Valeria Castelletto
- School of Chemistry, Pharmacy and Food Biosciences, University of Reading, Reading RG6 6AD, UK
| | - Ian W Hamley
- School of Chemistry, Pharmacy and Food Biosciences, University of Reading, Reading RG6 6AD, UK
| |
Collapse
|
17
|
Wang S, Lv Y. Silica-coated liquid metal nanoparticles with different stiffness for cellular uptake-enhanced tumor photothermal therapy. BIOMATERIALS ADVANCES 2024; 161:213872. [PMID: 38733802 DOI: 10.1016/j.bioadv.2024.213872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/08/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024]
Abstract
Cells can sense the mechanical stimulation of nanoparticles (NPs) and then regulate the cellular uptake process. The enhanced endocytosis efficiency can improve the concentration of NPs in tumor cells significantly, which is the key prerequisite for achieving efficient biological performance. However, the preparation methods of NPs with flexible and tunable stiffness are relatively limited, and the impact of stiffness property on their interaction with tumor cells remains unclear. In this study, soft liquid metal (LM) core was coated with hard silica layer, the obtained core-shell NPs with a wide range of Young's modulus (130.5 ± 25.6 MPa - 1729.2 ± 146.7 MPa) were prepared by adjusting the amount of silica. It was found that the NPs with higher stiffness exhibited superior cellular uptake efficiency and lysosomal escape ability compared to the NPs with lower stiffness. The silica layer not only affected the stiffness, but also improved the photothermal stability of the LM NPs. Both in vitro and in vivo results demonstrated that the NPs with higher stiffness displayed significantly enhanced tumor hyperthermia capability. This work may provide a paradigm for the preparation of NPs with varying stiffness and offer insights into the role of the mechanical property of NPs in their delivery.
Collapse
Affiliation(s)
- Shuai Wang
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, PR China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, PR China.
| |
Collapse
|
18
|
Saidjalolov S, Coelho F, Mercier V, Moreau D, Matile S. Inclusive Pattern Generation Protocols to Decode Thiol-Mediated Uptake. ACS CENTRAL SCIENCE 2024; 10:1033-1043. [PMID: 38799667 PMCID: PMC11117725 DOI: 10.1021/acscentsci.3c01601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 05/29/2024]
Abstract
Thiol-mediated uptake (TMU) is an intriguing enigma in current chemistry and biology. While the appearance of cell-penetrating activity upon attachment of cascade exchangers (CAXs) has been observed by many and is increasingly being used in practice, the molecular basis of TMU is essentially unknown. The objective of this study was to develop a general protocol to decode the dynamic covalent networks that presumably account for TMU. Uptake inhibition patterns obtained from the removal of exchange partners by either protein knockdown or alternative inhibitors are aligned with original patterns generated by CAX transporters and inhibitors and patterns from alternative functions (here cell motility). These inclusive TMU patterns reveal that the four most significant CAXs known today enter cells along three almost orthogonal pathways. Epidithiodiketopiperazines (ETP) exchange preferably with integrins and protein disulfide isomerases (PDIs), benzopolysulfanes (BPS) with different PDIs, presumably PDIA3, and asparagusic acid (AspA), and antisense oligonucleotide phosphorothioates (OPS) exchange with the transferrin receptor and can be activated by the removal of PDIs with their respective inhibitors. These findings provide a solid basis to understand and use TMU to enable and prevent entry into cells.
Collapse
Affiliation(s)
| | - Filipe Coelho
- Department
of Organic Chemistry, University of Geneva, CH-1211 Geneva, Switzerland
| | - Vincent Mercier
- Department
of Biochemistry, University of Geneva, CH-1211 Geneva, Switzerland
| | - Dimitri Moreau
- Department
of Biochemistry, University of Geneva, CH-1211 Geneva, Switzerland
| | - Stefan Matile
- Department
of Organic Chemistry, University of Geneva, CH-1211 Geneva, Switzerland
| |
Collapse
|
19
|
Chen P, Yang W, Mochida Y, Li S, Hong T, Kinoh H, Kataoka K, Cabral H. Selective Intracellular Delivery of Antibodies in Cancer Cells with Nanocarriers Sensing Endo/Lysosomal Enzymatic Activity. Angew Chem Int Ed Engl 2024; 63:e202317817. [PMID: 38342757 DOI: 10.1002/anie.202317817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
The differential enzymatic activity in the endo/lysosomes of particular cells could trigger targeted endosomal escape functions, enabling selective intracellular protein delivery. However, this strategy may be jeopardized due to protein degradation during endosomal trafficking. Herein, using custom made fluorescent probes to assess the endosomal activity of cathepsin B (CTSB) and protein degradation, we found that certain cancer cells with hyperacidified endosomes grant a spatiotemporal window where CTSB activity surpass protein digestion. This inspired the engineering of antibody-loaded polymeric nanocarriers having CTSB-activatable endosomal escape ability. The nanocarriers selectively escaped from the endo/lysosomes in the cells with high endosomal CTSB activity and delivered active antibodies to intracellular targets. This study provides a viable strategy for cell-specific protein delivery using stimuli-responsive nanocarriers with controlled endosomal escape.
Collapse
Affiliation(s)
- Pengwen Chen
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Wenqian Yang
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuki Mochida
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki, 210-0821, Japan
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Shangwei Li
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Taehun Hong
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroaki Kinoh
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki, 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki, 210-0821, Japan
| | - Horacio Cabral
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
20
|
Shao Y, Du G, Luo B, Liu T, Zhao J, Zhang S, Wang J, Chi M, Cai C, Liu Y, Meng X, Liu Z, Wang S, Nie S. A Tough Monolithic-Integrated Triboelectric Bioplastic Enabled by Dynamic Covalent Chemistry. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311993. [PMID: 38183330 DOI: 10.1002/adma.202311993] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/30/2023] [Indexed: 01/08/2024]
Abstract
Electronic waste is a growing threat to the global environment and human health, raising particular concerns. Triboelectric devices synthesized from sustainable and degradable materials are a promising electronic alternative, but the mechanical mismatch at the interface between the polymer substrate and the electrodes remains unresolved in practical applications. This study uses the sulfhydryl silanization reaction and the chemical selectivity and site specificity of the thiol-disulfide exchange reaction in dynamic covalent chemistry to prepare a tough monolithic-integrated triboelectric bioplastic. The stress is dissipated by covalent bond adaptation to the interface interaction, which makes the polymer dielectric layer to the conductive layer have a good interface adhesion effect (220.55 kPa). The interfacial interlocking of the polymer substrate with the conductive layer gives the triboelectric bioplastic excellent tensile strength (87.4 MPa) and fracture toughness (33.3 MJ m-3). Even when subjected to a tension force of 10 000 times its weight, it still maintains a stable triboelectric output with no visible cracks. This study provides new insights into the design of reliable and environmentally friendly self-powered devices, which is significant for the development of flexible wearable electronics.
Collapse
Affiliation(s)
- Yuzheng Shao
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Guoli Du
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Bin Luo
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Tao Liu
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Jiamin Zhao
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Song Zhang
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Jinlong Wang
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Mingchao Chi
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Chenchen Cai
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Yanhua Liu
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Xiangjiang Meng
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Zhaomeng Liu
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Shuangfei Wang
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Shuangxi Nie
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| |
Collapse
|
21
|
Veider F, Haddadzadegan S, Sanchez Armengol E, Laffleur F, Kali G, Bernkop-Schnürch A. Inhibition of P-glycoprotein-mediated efflux by thiolated cyclodextrins. Carbohydr Polym 2024; 327:121648. [PMID: 38171673 DOI: 10.1016/j.carbpol.2023.121648] [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: 08/28/2023] [Revised: 11/20/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
Overcoming P-glycoprotein (P-gp)-mediated efflux poses a significant challenge for the pharmaceutical industry. This study investigates the potential of thiolated β-cyclodextrins (β-CD-SHs) as inhibitors of P-gp-mediated efflux in Caco-2 cells. Through a series of transport assays, intracellular accumulation, and efflux of the P-gp substrates Rhodamine 123 (Rh123) and Calcein-AM with and without co-administration of β-CD-SHs were assessed. The results revealed that the cellular uptake of Rh123 and Calcein-AM were enhanced up to 7- and 3-fold, compared to the control, respectively. In efflux studies an up to 2.5-fold reduction of the Rh123 efflux was reached compared the control, indicating a substantial decrease of Rh123 efflux by β-CD-SHs. Furthermore, it was observed that β-CD-SHs led to a decrease in the reactivity of fluorescence-labeled anti-P-gp, suggesting additional effects on the conformation of P-gp. Overall, this study demonstrates the potential of β-CD-SHs as effective modulator of P-gp-mediated drug efflux in Caco-2 cells.
Collapse
Affiliation(s)
- Florina Veider
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Soheil Haddadzadegan
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Eva Sanchez Armengol
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Flavia Laffleur
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Gergely Kali
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Andreas Bernkop-Schnürch
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| |
Collapse
|
22
|
Zhang Y, Yu Y, Yang Y, Wang Y, Yu C. Engineered Silica Nanoparticles for Nucleic Acid Delivery. SMALL METHODS 2024; 8:e2300812. [PMID: 37906035 DOI: 10.1002/smtd.202300812] [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: 06/29/2023] [Revised: 09/14/2023] [Indexed: 11/02/2023]
Abstract
The development of nucleic acid-based drugs holds great promise for therapeutic applications, but their effective delivery into cells is hindered by poor cellular membrane permeability and inherent instability. To overcome these challenges, delivery vehicles are required to protect and deliver nucleic acids efficiently. Silica nanoparticles (SiNPs) have emerged as promising nanovectors and recently bioregulators for gene delivery due to their unique advantages. In this review, a summary of recent advancements in the design of SiNPs for nucleic acid delivery and their applications is provided, mainly according to the specific type of nucleic acids. First, the structural characteristics and working mechanisms of various types of nucleic acids are introduced and classified according to their functions. Subsequently, for each nucleic acid type, the use of SiNPs for enhancing delivery performance and their biomedical applications are summarized. The tailored design of SiNPs for selected type of nucleic acid delivery will be highlighted considering the characteristics of nucleic acids. Lastly, the limitations in current research and personal perspectives on future directions in this field are presented. It is expected this opportune review will provide insights into a burgeoning research area for the development of next-generation SiNP-based nucleic acid delivery systems.
Collapse
Affiliation(s)
- Yue Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Yingjie Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
- Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, Institute of Optoelectronics, Fudan University, Shanghai, 200433, P. R. China
| | - Yue Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, P. R. China
| |
Collapse
|
23
|
Raj G, Vasudev DS, Christopher S, Babulal A, Harsha P, Ram S, Tiwari M, Sauer M, Varghese R. Multifunctional siRNA/ferrocene/cyclodextrin nanoparticles for enhanced chemodynamic cancer therapy. NANOSCALE 2024; 16:3755-3763. [PMID: 38299362 DOI: 10.1039/d3nr06071c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
The therapeutic outcome of chemodynamic therapy (CDT) is greatly hindered by the presence of oxidative damage repair proteins (MTH1) inside cancer cells. These oxidative damage repair proteins detoxify the action of radicals generated by Fenton or Fenton-like reactions. Hence, it is extremely important to develop a simple strategy for the downregulation of MTH1 protein inside cancer cells along with the delivery of metal ions into cancer cells. A one-pot host-guest supramolecular approach for the codelivery of MTH1 siRNA and metal ions into a cancer cell is reported. Our approach involves the fabrication of an inclusion complex between cationic β-cyclodextrin and a ferrocene prodrug, which spontaneously undergoes amphiphilicity-driven self-assembly to form spherical nanoparticles (NPs) having a positively charged surface. The cationic surface of the NPs was then explored for the loading of MTH1 siRNA through electrostatic interactions. Using HeLa cells as a representative example, efficient uptake of the NPs, delivery of MTH1 siRNA and the enhanced CDT of the nanoformulation are demonstrated. This work highlights the potential of the supramolecular approach as a simple yet efficient method for the delivery of siRNA across the cell membrane for enhanced chemodynamic therapy.
Collapse
Affiliation(s)
- Gowtham Raj
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - D S Vasudev
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - Sarah Christopher
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - Anupama Babulal
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - P Harsha
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - Soumakanya Ram
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - Mehul Tiwari
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Reji Varghese
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| |
Collapse
|
24
|
Gong S, Qiu J, Thayumanavan S. Self-Assembly of Epitope-Tagged Proteins and Antibodies for Delivering Biologics to Antigen Presenting Cells. J Am Chem Soc 2024; 146:33-38. [PMID: 38147631 PMCID: PMC11131140 DOI: 10.1021/jacs.3c09334] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Inspired by the immune system's own strategy for macrophage activation, we describe here a simple self-assembly strategy for generating artificial immune complexes. The built-in recognition domains in the antibody, viz. the Fab and Fc domains, are judiciously leveraged for cargo conjugation to generate the nanoassembly and macrophage targeting, respectively. A responsive linker is engineered into the nanoassembly for releasing the protein cargo inside the macrophages, while ensuring stability during delivery. The design principles are simple and versatile to be applicable to a range of biologics, from small protein toxins to large enzymes, with high loading capacity. This self-assembly platform has the potential for delivering biologics to immune cells with implications in immunotherapy.
Collapse
Affiliation(s)
- Shuai Gong
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Jingyi Qiu
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
25
|
Yu L, Xu Y, Al-Amin M, Jiang S, Sample M, Prasad A, Stephanopoulos N, Šulc P, Yan H. CytoDirect: A Nucleic Acid Nanodevice for Specific and Efficient Delivery of Functional Payloads to the Cytoplasm. J Am Chem Soc 2023; 145:27336-27347. [PMID: 38055928 PMCID: PMC10789493 DOI: 10.1021/jacs.3c07491] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Direct and efficient delivery of functional payloads such as chemotherapy drugs, siRNA, or small-molecule inhibitors into the cytoplasm, bypassing the endo/lysosomal trapping, is a challenging task for intracellular medicine. Here, we take advantage of the programmability of DNA nanotechnology to develop a DNA nanodevice called CytoDirect, which incorporates disulfide units and human epidermal growth factor receptor 2 (HER2) affibodies into a DNA origami nanostructure, enabling rapid cytosolic uptake into targeted cancer cells and deep tissue penetration. We further demonstrated that therapeutic oligonucleotides and small-molecule chemotherapy drugs can be easily delivered by CytoDirect and showed notable effects on gene knockdown and cell apoptosis, respectively. This study demonstrates the synergistic effect of disulfide and HER2 affibody modifications on the rapid cytosolic delivery of DNA origami and its payloads to targeted cells and deep tissues, thereby expanding the delivery capabilities of DNA nanostructures in a new direction for disease treatment.
Collapse
Affiliation(s)
- Lu Yu
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Yang Xu
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Md Al-Amin
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Shuoxing Jiang
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Matthew Sample
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Abhay Prasad
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Nicholas Stephanopoulos
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Petr Šulc
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Hao Yan
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| |
Collapse
|
26
|
Chowdhry R, Lu SZ, Lee S, Godhulayyagari S, Ebrahimi SB, Samanta D. Enhancing CRISPR/Cas systems with nanotechnology. Trends Biotechnol 2023; 41:1549-1564. [PMID: 37451945 DOI: 10.1016/j.tibtech.2023.06.005] [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: 04/18/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
CRISPR/Cas systems have revolutionized biology and medicine, and have led to new paradigms in disease diagnostics and therapeutics. However, these complexes suffer from key limitations regarding barriers to cellular entry, stability in biological environments, and off-target effects. Integrating nanotechnology with CRISPR/Cas systems has emerged as a promising strategy to overcome these challenges and has further unlocked structures that accumulate preferentially in tissues of interest, have tunable pharmacological properties, and are activated in response to desired stimuli. Nanomaterials can also enhance CRISPR/Cas-mediated detection platforms by enabling faster, more sensitive, and convenient readouts. We highlight recent advances in this rapidly growing field. We also outline areas that need further development to fully realize the potential of CRISPR technologies.
Collapse
Affiliation(s)
- Rupali Chowdhry
- Department of Public Health, The University of Texas at Austin, Austin, TX 78712, USA
| | - Steven Z Lu
- Department of Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Seungheon Lee
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA
| | | | - Sasha B Ebrahimi
- Drug Product Development - Steriles, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Devleena Samanta
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
27
|
Stengel D, Jörgensen AM, Polidori I, Kapitza P, Ricci F, Bernkop-Schnürch A. The power of sulfhydryl groups: Thiolated lipid-based nanoparticles enhance cellular uptake of nucleic acids. J Colloid Interface Sci 2023; 654:1136-1145. [PMID: 39491903 DOI: 10.1016/j.jcis.2023.10.039] [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: 07/11/2023] [Revised: 09/29/2023] [Accepted: 10/09/2023] [Indexed: 11/05/2024]
Abstract
AIM The aim of the study was to evaluate the effect of thiolation of lipid-based nanoparticles (LNPs) on cellular uptake of nucleic acids. METHODS A thiolated surfactant was synthesized by binding palmitic acid covalently to cysteine. Green fluorescent protein (GFP) encoding plasmid DNA (pDNA) was used as model nucleic acid and incorporated via hydrophobic ion-pairing with a cationic cholesterol derivate (DcCholesterol) in LNPs that were prepared by solvent injection method using the thiolated surfactant for surface decoration. LNPs were characterized regarding size, polydispersity index, zeta potential and stability in biorelevant media. The endosomal escape properties of LNPs were evaluated by erythrocytes interaction studies. Cell viability and transfection efficiency on HEK293 cells were investigated. RESULTS The structure of the thiolated surfactant was confirmed by 1H NMR and FT-IR. LNPs containing the nucleic acid-DcCholesterol complex with and without the thiolated surfactant were developed and displayed sizes in the range from 173 nm to 233 nm with a narrow size distribution (PDI < 0.3) and a negative zeta potential. LNPs showed no significant increase in size after 4 h of incubation in artificial body fluids. Erythrocytes interaction studies revealed enhanced endosomal escape properties for thiolated LNPs compared to non-thiolated LNPs. LNPs showed in concentrations lower than 0.74 mg/mL cell viability ≥ 80 %. Transfection studies on HEK cells with thiolated LNPs compared to non-thiolated LNP showed a 4.6-fold higher expression of GFP. CONCLUSION Surface thiolation of LNPs represents a promising tool for enhancing intracellular nucleic acid delivery of LNPs.
Collapse
Affiliation(s)
- Daniel Stengel
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Arne M Jörgensen
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Ilaria Polidori
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Paul Kapitza
- Department of Pharmaceutical Chemistry, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Fabrizio Ricci
- Thiomatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, 6020 Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria.
| |
Collapse
|
28
|
Kaplan Ö, Truszkowska M, Kali G, Knoll P, Blanco Massani M, Braun DE, Bernkop-Schnürch A. Thiolated α-cyclodextrin: The likely smallest drug carrier providing enhanced cellular uptake and endosomal escape. Carbohydr Polym 2023; 316:121070. [PMID: 37321712 DOI: 10.1016/j.carbpol.2023.121070] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023]
Abstract
This study aimed to evaluate the effect of thiolated α-cyclodextrin (α-CD-SH) on the cellular uptake of its payload. For this purpose, α-CD was thiolated using phosphorous pentasulfide. Thiolated α-CD was characterized by FT-IR and 1H NMR spectroscopy, differential scanning calorimetry (DSC), and powder X-ray diffractometry (PXRD). Cytotoxicity of α-CD-SH was evaluated on Caco-2, HEK 293, and MC3T3 cells. Dilauryl fluorescein (DLF) and coumarin-6 (Cou) serving as surrogates for a pharmaceutical payload were incorporated in α-CD-SH, and cellular uptake was analyzed by flow cytometry and confocal microscopy. Endosomal escape was investigated by confocal microscopy and hemolysis assay. Results showed no cytotoxic effect within 3 h, while dose-dependent cytotoxicity was observed within 24 h. The cellular uptake of DLF and Cou was up to 20- and 11-fold enhanced by α-CD-SH compared to native α-CD, respectively. Furthermore, α-CD-SH provided an endosomal escape. According to these results, α-CD-SH is a promising carrier to shuttle drugs into the cytoplasm of target cells.
Collapse
Affiliation(s)
- Özlem Kaplan
- Department of Genetics and Bioengineering, Rafet Kayış Faculty of Engineering, Alanya Alaaddin Keykubat University, 07400 Antalya, Turkey; Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, 34134 Istanbul, Turkey
| | - Martyna Truszkowska
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Gergely Kali
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Patrick Knoll
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Mariana Blanco Massani
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Doris Elfriede Braun
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria.
| |
Collapse
|
29
|
Kanjilal P, Singh K, Das R, Matte J, Thayumanavan S. Antibody Polymer Conjugates (APCs) for Active Targeted Therapeutic Delivery. Biomacromolecules 2023; 24:3638-3646. [PMID: 37478281 PMCID: PMC11145409 DOI: 10.1021/acs.biomac.3c00385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Antibody drug conjugates (ADCs) are poised to have an enormous impact on targeted nanomedicine, especially in many cancer pathologies. The reach of the current format of ADCs is limited by their low drug-to-antibody ratio (DAR) because of the associated physiochemical instabilities. Here, we design antibody polymer conjugates (APCs) as a modular strategy to utilize polymers to address ADC's shortcomings. We show here that conjugation of polymer-based therapeutic molecules to antibodies helps increase the DAR, owing to the hydrophilic comonomer in the polymer that helps in masking the increased hydrophobicity caused by high drug loading. We show that the platform exhibits cell targetability and selective cell killing in multiple cell lines expressing disease-relevant antigens, viz., HER2 and EGFR. The ability to use different functionalities in the drug as the handle for polymer attachment further demonstrates the platform nature of APCs. The findings here could serve as an alternative design strategy for the next generation of active targeted nanomedicine.
Collapse
|
30
|
Wan Y, Wang W, Lai Q, Wu M, Feng S. Advances in cell-penetrating poly(disulfide)s for intracellular delivery of therapeutics. Drug Discov Today 2023:103668. [PMID: 37321318 DOI: 10.1016/j.drudis.2023.103668] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/25/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023]
Abstract
Efficient intracellular delivery is essential for most therapeutic agents; however, existing delivery vectors face a dilemma between efficiency and toxicity, and always encounter the challenge of endolysosomal trapping. The cell-penetrating poly(disulfide) (CPD) is an effective tool for intracellular delivery, as it is taken up through thiol-mediated cellular uptake, thus avoiding endolysosomal entrapment and ensuring efficient cytosolic availability. Upon cellular uptake, CPD undergoes reductive depolymerization by glutathione inside cells and has minimal cytotoxicity. This review summarizes CPD's chemical synthesis approaches, cellular uptake mechanism, and recent advances in the intracellular delivery of proteins, antibodies, nucleic acids, and other nanoparticles. Overall, CPD is a promising candidate carrier for efficient intracellular delivery.
Collapse
Affiliation(s)
- Yu Wan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Wangxia Wang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qiuyue Lai
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Mingyu Wu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Shun Feng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
31
|
Coelho F, Saidjalolov S, Moreau D, Thorn-Seshold O, Matile S. Inhibition of Cell Motility by Cell-Penetrating Dynamic Covalent Cascade Exchangers: Integrins Participate in Thiol-Mediated Uptake. JACS AU 2023; 3:1010-1016. [PMID: 37124287 PMCID: PMC10131202 DOI: 10.1021/jacsau.3c00113] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/05/2023] [Accepted: 04/05/2023] [Indexed: 05/03/2023]
Abstract
Integrins are cell surface proteins responsible for cell motility. Inspired by the rich disulfide exchange chemistry of integrins, we show here the inhibition of cell migration by cascade exchangers (CAXs), which also enable and inhibit cell penetration by thiol-mediated uptake. Fast-moving CAXs such as reversible Michael acceptor dimers, dithiabismepanes, and bioinspired epidithiodiketopiperazines are best, much better than Ellman's reagent. The implication that integrins participate in thiol-mediated uptake is confirmed by reduced uptake in integrin-knockdown cells. Although thiol-mediated uptake is increasingly emerging as a unifying pathway to bring matter into cells, its molecular basis is essentially unknown. These results identify the integrin superfamily as experimentally validated general cellular partners in the dynamic covalent exchange cascades that are likely to account for thiol-mediated uptake. The patterns identified testify to the complexity of the dynamic covalent networks involved. This work also provides chemistry tools to explore cell motility and expands the drug discovery potential of CAXs from antiviral toward antithrombotic and antitumor perspectives.
Collapse
Affiliation(s)
- Filipe Coelho
- Department
of Organic Chemistry, University of Geneva, 1211 Geneva, Switzerland
| | | | - Dimitri Moreau
- Department
of Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Oliver Thorn-Seshold
- Department
of Pharmacy, Ludwig-Maximilians University
of Munich, 81377 Munich, Germany
| | - Stefan Matile
- Department
of Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
32
|
Huang X, Li J, Li G, Ni B, Liang Z, Chen H, Xu C, Zhou J, Huang J, Deng S. Cation-free siRNA-cored nanocapsules for tumor-targeted RNAi therapy. Acta Biomater 2023; 161:226-237. [PMID: 36898473 DOI: 10.1016/j.actbio.2023.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/03/2023] [Accepted: 03/01/2023] [Indexed: 03/10/2023]
Abstract
Cation-associated cytotoxicity limits the systemic administration of RNA delivery in vivo, demanding the development of non-cationic nanosystems. In this study, cation-free polymer-siRNA nanocapsules with disulfide-crosslinked interlayer, namely T-SS(-), were prepared via the following steps: 1) complexation of siRNA with a cationic block polymer cRGD-poly(ethylene glycol)-b-poly[(2-aminoethanethiol)aspartamide]-b-poly{N'-[N-(2-aminoethyl)-2-ethylimino-1-aminomethyl]aspartamide}, abbreviated as cRGD-PEG-PAsp(MEA)-PAsp(C=N-DETA), 2) interlayer crosslinking via disulfide bond in pH 7.4 solution, and 3) removal of cationic DETA pendant at pH 5.0 via breakage of imide bond. The cationic-free nanocapsules with siRNA cores not only showed great performance (such as efficient siRNA encapsulation, high stability in serum, cancer cell targeting via cRGD modification, and GSH-triggered siRNA release), but also achieved tumor-targeted gene silencing in vivo. Moreover, the nanocapsules loaded with siRNA against polo-like kinase 1 (siRNA-PLK1) significantly inhibited tumor growth without showing cation-associated toxicity side effects and remarkably improved the survival rate of PC-3 tumor-bearing mice. The cation-free nanocapsules could potentially serve as a safe and effective platform for siRNA delivery. STATEMENT OF SIGNIFICANCE: Cation-associated toxicity limits the clinical translation of cationic carriers for siRNA delivery. Recently, several non-cationic carriers, such as siRNA micelles, DNA-based nanogels, and bottlebrush-architectured poly(ethylene glycol), have been developed to deliver siRNA. However, in these designs, siRNA as a hydrophilic macromolecule was attached to the nanoparticle surface instead of being encapsulated. Thus, it was easily degraded by serum nuclease and often induced immunogenicity. Herein, we demonstrate a new type of cation-free siRNA-cored polymeric nanocapsules. The developed nanocapsules not only showed capacities including efficient siRNA encapsulation, high stability in serum, and cancer cell targeting via cRGD modification, but also achieved an efficient tumor-targeted gene silencing in vivo. Importantly, unlike cationic carriers, the nanocapsules exhibited no cation-associated side effects.
Collapse
Affiliation(s)
- Xinghua Huang
- Department of Urology, Longgang District People's Hospital of Shenzhen, Shenzhen 518000, China
| | - Jianwei Li
- Department of Urology, Longgang District People's Hospital of Shenzhen, Shenzhen 518000, China
| | - Guanyi Li
- Department of Urology, Shenzhen Samii Medical Center, Shenzhen 518000, China
| | - Binyu Ni
- Department of Paediatrics, Longgang District People's Hospital of Shenzhen, Shenzhen 518000, China
| | - Ziji Liang
- Department of Urology, Longgang District People's Hospital of Shenzhen, Shenzhen 518000, China
| | - Haodong Chen
- Department of Urology, Longgang District People's Hospital of Shenzhen, Shenzhen 518000, China
| | - Chaozhang Xu
- Department of Urology, Longgang District People's Hospital of Shenzhen, Shenzhen 518000, China
| | - Jianhua Zhou
- Department of Urology, Longgang District People's Hospital of Shenzhen, Shenzhen 518000, China.
| | - Jinsheng Huang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Shaohui Deng
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China; Affiliated Dongguan Hospital, Southern Medical University, Dongguan 523059, China.
| |
Collapse
|
33
|
Yao SY, Yue YX, Ying AK, Hu XY, Li HB, Cai K, Guo DS. An Antitumor Dual-Responsive Host-Guest Supramolecular Polymer Based on Hypoxia-Cleavable Azocalix[4]arene. Angew Chem Int Ed Engl 2023; 62:e202213578. [PMID: 36353747 DOI: 10.1002/anie.202213578] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Indexed: 11/11/2022]
Abstract
The exploitation of specific guests which can respond to external stimuli is the main approach for the construction of stimuli-responsive supramolecular polymers (SPs) based on host-guest interactions. Most functional guests, however, fail to manifest stimuli-responses. Herein, a hypoxia-responsive dimeric azocalixarene (D-SAC4A) with outstanding hosting properties was used as the macrocyclic building block for the preparation of host stimuli-responsive SPs. Since azocalixarenes can also be compatible with stimuli-responsive guests, an antitumor drug, camptothecin (CPT), was chosen and linked via a disulfide-containing linker to afford a glutathione (GSH)-responsive ditropic guest (D-CPT). A unique dual-responsive SP was obtained by 1 : 1 mixing of D-SAC4A and D-CPT in water, which further assembled into SP nanoparticles (DSPNs). DSPNs displayed outstanding stability against dilution and biological interferants, as well as precise CPT-release under GSH and hypoxia conditions. In vitro and in vivo experiments demonstrated the good biosafety and tumor-suppressive effects of DSPNs.
Collapse
Affiliation(s)
- Shun-Yu Yao
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Yu-Xin Yue
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, 300071, China
| | - An-Kang Ying
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Xin-Yue Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Hua-Bin Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Kang Cai
- College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, 300071, China
| |
Collapse
|
34
|
Hemmati S, Rasekhi Kazerooni H. Polypharmacological Cell-Penetrating Peptides from Venomous Marine Animals Based on Immunomodulating, Antimicrobial, and Anticancer Properties. Mar Drugs 2022; 20:md20120763. [PMID: 36547910 PMCID: PMC9787916 DOI: 10.3390/md20120763] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/09/2022] Open
Abstract
Complex pathological diseases, such as cancer, infection, and Alzheimer's, need to be targeted by multipronged curative. Various omics technologies, with a high rate of data generation, demand artificial intelligence to translate these data into druggable targets. In this study, 82 marine venomous animal species were retrieved, and 3505 cryptic cell-penetrating peptides (CPPs) were identified in their toxins. A total of 279 safe peptides were further analyzed for antimicrobial, anticancer, and immunomodulatory characteristics. Protease-resistant CPPs with endosomal-escape ability in Hydrophis hardwickii, nuclear-localizing peptides in Scorpaena plumieri, and mitochondrial-targeting peptides from Synanceia horrida were suitable for compartmental drug delivery. A broad-spectrum S. horrida-derived antimicrobial peptide with a high binding-affinity to bacterial membranes was an antigen-presenting cell (APC) stimulator that primes cytokine release and naïve T-cell maturation simultaneously. While antibiofilm and wound-healing peptides were detected in Synanceia verrucosa, APC epitopes as universal adjuvants for antiviral vaccination were in Pterois volitans and Conus monile. Conus pennaceus-derived anticancer peptides showed antiangiogenic and IL-2-inducing properties with moderate BBB-permeation and were defined to be a tumor-homing peptide (THP) with the ability to inhibit programmed death ligand-1 (PDL-1). Isoforms of RGD-containing peptides with innate antiangiogenic characteristics were in Conus tessulatus for tumor targeting. Inhibitors of neuropilin-1 in C. pennaceus are proposed for imaging probes or therapeutic delivery. A Conus betulinus cryptic peptide, with BBB-permeation, mitochondrial-targeting, and antioxidant capacity, was a stimulator of anti-inflammatory cytokines and non-inducer of proinflammation proposed for Alzheimer's. Conclusively, we have considered the dynamic interaction of cells, their microenvironment, and proportional-orchestrating-host- immune pathways by multi-target-directed CPPs resembling single-molecule polypharmacology. This strategy might fill the therapeutic gap in complex resistant disorders and increase the candidates' clinical-translation chance.
Collapse
Affiliation(s)
- Shiva Hemmati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Department of Pharmaceutical Biology, Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Correspondence: ; Tel.: +98-7132-424-128
| | | |
Collapse
|