1
|
Wang K, Jiang S, Wang W, Chen W, Kai T. Dual-miRNA guided in-vivo imaging and multimodal nanomedicine approaches for precise hepatocellular carcinoma differentiation and synergistic cancer theranostics using DNA hairpins and dual-ligand functionalized zirconium-MOF nanohybrids. Biomaterials 2025; 321:123330. [PMID: 40222259 DOI: 10.1016/j.biomaterials.2025.123330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
As one of the most common and heterogeneous liver malignancies, hepatocellular carcinoma (HCC) remains a significant clinical challenge due to the lack of biomarkers for early diagnosis, challenges in accurate subtyping, and limitations of current therapeutic strategies with poor efficacy. Herein, based on DNA hairpin probes and dual-ligand zirconium (Zr)-based metal-organic frameworks (DMOFs), the multifunctional nanohybrids (DMOF@MnCO@CuS@Hairpin probe, DMCH) were developed to overcome these diagnostic and therapeutic obstacles. Two improved DNA molecular beacons and APE1 enzyme within HCC cells were utilized for sensitive miRNA imaging in vivo with high accuracy to differentiate HCC subtypes precisely. Furthermore, this "all-in-one" theranostic platform not only facilitates the generation of active oxygen species and conversion of near-infrared light into heat, but also releases carbon monoxide to inhibit the expression of HSP70 protein to improve photothermal (PTT) therapy efficiency during laser radiation, which enables PTT, photodynamic (PDT), chemodynamic (CDT), and gas therapy (GAT) for HCC treatment simultaneously. The developed nano-theranostics platform provides a novel way for efficient early screening, diagnosis, and intervention of HCC, and paves the path for future "bench-to-bedside" design of theranostics.
Collapse
Affiliation(s)
- Kun Wang
- Xiangya School of Public Health, Central South University, Changsha, Hunan, 410013, China
| | - Siyu Jiang
- Xiangya School of Public Health, Central South University, Changsha, Hunan, 410013, China
| | - Wanli Wang
- Xiangya School of Public Health, Central South University, Changsha, Hunan, 410013, China
| | - Wansong Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Tianhan Kai
- Xiangya School of Public Health, Central South University, Changsha, Hunan, 410013, China; Furong Laboratory, Changsha, Hunan, 410008, 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
|
Tu T, Huan S, Feng X, Ke G, Li L, Zhang XB. Spatial Confinement of a Dual Activatable DNAzyme Sensor in the Cavity of a DNA Nanocage for Logic-Gated Molecular Imaging. Angew Chem Int Ed Engl 2025; 64:e202424684. [PMID: 39902629 DOI: 10.1002/anie.202424684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/05/2025]
Abstract
Despite intense interest in design of DNAzyme sensors for molecular detection and imaging in living cells, their intracellular applications are still hampered by limited spatial control and poor bio-stability. Here we present controlled spatial confinement of a rationally designed, microRNA (miRNA)-activatable DNAzyme sensor probe (mDz) within the cavity of DNA nanocage, enabling efficient intracellular delivery with improved bio-stability for AND-gate molecular imaging. The mDz that possesses inactive DNAzyme activity is designed by the introduction of a blocking DNA strand, while miR-21 mediated strand displacement reaction allows for the formation of an intact DNAzyme structure for metal-ion-mediated catalytic reaction. Furthermore, the DNA nanocage serves as a nanocarrier for intracellular delivery of mDz, in which the cavity is accessible to the dual targets for logic-gated molecular imaging, while the confinement effect can provide steric protection of mDz by obstructing nuclease from entering the cavity of the DNA nanocage, resulting in enhanced bio-stability and improved molecular imaging precision. This strategy paves a way for the engineering of activatable DNA nanosensors with both self-delivery and self-protection capabilities to detect diverse intracellular targets.
Collapse
Affiliation(s)
- Tingting Tu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha
| | - Shuangyan Huan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha
| | - Xueyan Feng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190
| | - Guoliang Ke
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha
| | - Lele Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha
| |
Collapse
|
4
|
Kansara K, Laha A, Kumar A, Bhatia D. Programmable DNA-Based Nanodevices for Next-Generation Clinical and Healthcare Applications. Adv Biol (Weinh) 2025; 9:e2400593. [PMID: 39913110 DOI: 10.1002/adbi.202400593] [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: 10/06/2024] [Revised: 12/17/2024] [Indexed: 02/07/2025]
Abstract
Deoxyribonucleic acid (DNA) nanotechnology has brought an unparalleled set of possibilities for self-assembled structures emerging as an independent branch of synthetic biology. The field of science uses the molecular properties of DNA to build nanoparticles and nanodevices that have the potential to bring breakthroughs in medical science. On the one hand, their biocompatibility, precision, synthetic ease, and programmability make them an ideal choice in drug delivery and healthcare. On the other, the lack of proper biodistribution profiles, stability inside the system, enzymatic cleavage, immune recognition, and translational barriers are some of the hurdles it faces. Many recent technological advancements are in progress to tackle these challenges, while some already have been used. These tools and technologies need to be understood and studied for the successful transition of these intelligent DNA nanostructures (DNs) to healthcare applications. This review thus, highlights some of the challenges being faced by the DNs in healthcare. Additionally, it provides an overview of the recent trends in using these devices in disease detection and remission and finally talks about the future scope and opportunities for an effective transition from bench to bedside.
Collapse
Affiliation(s)
- Krupa Kansara
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat, 382055, India
| | - Anwesha Laha
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat, 382055, India
| |
Collapse
|
5
|
Li L, Xing Z, Wang J, Guo Y, Wu X, Ma Y, Xu Z, Kuang Y, Liao T, Li C. Hyaluronic acid-mediated targeted nano-modulators for activation of pyroptosis for cancer therapy through multichannel regulation of Ca 2+ overload. Int J Biol Macromol 2025; 299:140116. [PMID: 39842602 DOI: 10.1016/j.ijbiomac.2025.140116] [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: 11/17/2024] [Revised: 01/05/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
Calcium-based nanomaterials-mediated Ca2+ overload-induced pyroptosis and its application in tumor therapy have received considerable attention. However, the calcium buffering capacity of tumor cells can maintain mitochondrial calcium homeostasis, so it is important to effectively disrupt this homeostasis to activate pyroptosis. Here, a nano-modulator CUR@CaCO3-PArg@HA (CCAH) was developed to regulate calcium overload in multiple channels and activate pyroptosis. Hyaluronic acid (HA)-coated nano-modulators achieve tumor targeting, and under the weakly acidic conditions of the tumor microenvironment (TME), CaCO3 nanoparticles rapidly release curcumin (CUR), inhibit the outflow of intracellular Ca2+, and release exogenous Ca2+. Meanwhile, poly-L-arginine (PArg) reacts with reactive oxygen species (ROS) generated by mitochondrial imbalance, releasing nitric oxide (NO) and stimulating the endoplasmic reticulum to release endogenous Ca2+. The combined action of endogenous and exogenous Ca2+ effectively activates caspase-1, which cleaves gasdermin-D (GSDMD) to produce the active N-terminus (GSDMD-N), effectively activating pyroptosis. Notably, the generated ROS and NO can also generate more oxidizing ONOO-, further exacerbating the imbalance in mitochondrial homeostasis. This work demonstrates that simultaneous modulation of exogenous and endogenous Ca2+ can disrupt mitochondrial Ca2+ homeostasis and effectively activate pyroptosis to treat tumors, which is expected to promote the progression of cancer treatment in the future.
Collapse
Affiliation(s)
- Linwei Li
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Glyn O. Phillips Hydrocolloid Research Centre at HBUT, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China; Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Zihan Xing
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Jinyu Wang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Glyn O. Phillips Hydrocolloid Research Centre at HBUT, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Yuhao Guo
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Xiaomei Wu
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Yiming Ma
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Ziqiang Xu
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Ying Kuang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Glyn O. Phillips Hydrocolloid Research Centre at HBUT, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China.
| | - Tao Liao
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Glyn O. Phillips Hydrocolloid Research Centre at HBUT, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China.
| | - Cao Li
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Glyn O. Phillips Hydrocolloid Research Centre at HBUT, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China; Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China.
| |
Collapse
|
6
|
Kim HK, Cheong H, Kim MY, Jin HE. Therapeutic Targeting in Ovarian Cancer: Nano-Enhanced CRISPR/Cas9 Gene Editing and Drug Combination Therapy. Int J Nanomedicine 2025; 20:3907-3931. [PMID: 40191042 PMCID: PMC11970428 DOI: 10.2147/ijn.s507688] [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: 11/21/2024] [Accepted: 02/26/2025] [Indexed: 04/09/2025] Open
Abstract
Ovarian cancer is the third most common gynecological cancer worldwide. Due to the high recurrence rate of advanced-stage ovarian cancer, often resulting from drug-resistant and refractory disease, various treatment strategies are under investigation. Genome editing of therapeutic target genes holds promise in enhancing cancer treatment efficacy by elucidating gene functions and mechanisms involved in cancer progression. The CRISPR/Cas9 system, in particular, shows great potential in ovarian cancer gene therapy and drug development. Targeting therapeutic genes such as BRCA1/2, P53, Snai1 etc, could improve the therapeutic strategy in ovarian cancer. CRISPR/Cas9 is a powerful gene-editing tool that there are many on-going clinical trials to treat various diseases including cancer. Nano-based delivery systems for CRISPR/Cas9 offer further therapeutic benefits, leveraging the unique properties of nanoparticles to improve delivery efficiency. Nano-based delivery systems could enhance the stability of CRISPR/Cas9 delivery formats (such as plasmid, mRNA, etc) and improve the delivery precision of delivery to target tumors. Additionally, combining CRISPR/Cas9 with targeted drug treatments, especially those aimed at genes associated with drug resistance, may significantly improve therapeutic outcomes in ovarian cancer. In this review, we discuss therapeutic target genes and their mechanisms in ovarian cancer, advances in nano-based CRISPR/Cas9 delivery, and the therapeutic potential of combining CRISPR/Cas9 with drug treatments for ovarian cancer.
Collapse
Affiliation(s)
- Hong-Kook Kim
- AI-Super Convergence KIURI Translational Research Center, Ajou University, Suwon, 16499, Republic of Korea
- Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Heedon Cheong
- Department of Pharmacy, Ajou University, Suwon, 16499, Republic of Korea
| | - Moo-Yeon Kim
- Department of Pharmacy, Ajou University, Suwon, 16499, Republic of Korea
| | - Hyo-Eon Jin
- Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
- Department of Pharmacy, Ajou University, Suwon, 16499, Republic of Korea
| |
Collapse
|
7
|
Tan LQ, Zeng WJ, Chen QL, Tao XL, Yuan R, Lei YM, Zhuo Y. Programmable Split DNAzyme Modulators via Allosteric Cooperative Activation for mRNA Electrochemiluminescence Biosensing. Anal Chem 2025; 97:5789-5797. [PMID: 40032605 DOI: 10.1021/acs.analchem.4c07071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
DNAzymes, known for their programmability, stability, and cost-effectiveness, are powerful tools for signal transduction in complex biological systems. However, their application in responding to target effectors is often hindered by limited catalytic efficiency and susceptibility to unintended activation. Here we propose an allosteric cooperative activation strategy to program a split DNAzyme modulator (STATER) that enables sensitive and accurate electrochemiluminescence (ECL) biosensing of interleukin-6 (IL-6) mRNA. Our design features a STATER that leverages a DNA tetrahedron as a central scaffold, equipped with two pairs of T-shaped hairpin probes (TP) and helper hairpin probes (HP). Specifically, the TP contains two apurinic/apyrimidinic endonuclease 1 (APE1) recognition sites, an IL-6 mRNA recognition region, and a partzyme fragment, while the HP contains a corresponding paired partzyme fragment. Unlike conventional DNAzyme modulators that rely on single effector activation, the STATER integrates an allosteric cooperative activation mechanism, which ensures that all preblocked components are synergistically activated and assembled within a confined space, facilitating rapid and specific reconstruction of the DNAzyme's catalytic active domain. Furthermore, upon cooperative recognition by APE1 and IL-6 mRNA, two inactive partzymes undergo an allosteric assembly via a toehold exchange displacement reaction, switching on the cleavage reactivity of STATER. This mechanism enables the establishment of an activation threshold for IL-6 mRNA, thereby minimizing nonspecific activation in complex scenarios. Our studies demonstrate that the STATER exhibits outstanding sensitivity and selectivity for IL-6 mRNA detection using the supramolecular gold nanoclusters network-based ECL platform. The biosensor provides a linear detection span from 1 × 10-13 to 1 × 10-7 M, with a limit of detection as low as 3.26 × 10-14 M, highlighting STATER's potential for detecting various analytes in complex biological systems.
Collapse
Affiliation(s)
- Liu-Qing Tan
- MOE Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Chemistry and Chemical Engineering, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Chongqing 400715, P. R. China
| | - Wei-Jia Zeng
- MOE Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Chemistry and Chemical Engineering, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Chongqing 400715, P. R. China
| | - Qiao-Lin Chen
- MOE Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Chemistry and Chemical Engineering, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Chongqing 400715, P. R. China
| | - Xiu-Li Tao
- MOE Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Chemistry and Chemical Engineering, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Chongqing 400715, P. R. China
| | - Ruo Yuan
- MOE Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Chemistry and Chemical Engineering, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Chongqing 400715, P. R. China
| | - Yan-Mei Lei
- MOE Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Chemistry and Chemical Engineering, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Chongqing 400715, P. R. China
| | - Ying Zhuo
- MOE Key Laboratory of Luminescence Analysis and Molecular Sensing, College of Chemistry and Chemical Engineering, Institute of Developmental Biology and Regenerative Medicine, Southwest University, Chongqing 400715, P. R. China
| |
Collapse
|
8
|
Gao S, Ge H, Gao L, Gao Y, Tang S, Li Y, Yuan Z, Chen W. Silk Fibroin Nanoparticles for Enhanced Cuproptosis and Immunotherapy in Pancreatic Cancer Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417676. [PMID: 40091480 DOI: 10.1002/advs.202417676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/16/2025] [Indexed: 03/19/2025]
Abstract
Cuproptosis is a newly discovered copper ion-dependent programmed cell death. Elesclomol (ES) is a Cu2+ transporter that delivers Cu2+ into tumor cells, causing cell death at toxic doses. However, ES has a short blood half-life, limiting its accumulation in tumors. This study introduces Tussah silk fibroin nanoparticles (TSF@ES-Cu NPs) to protect ES and Cu2+. TSF, with a stable structure, resists metabolism in circulation. Targeting tumors with natural RGD peptides and TSF's unique secondary structure, enhances drug enrichment and special release in pancreatic tumors, improving treatment efficacy. In vitro, TSF@ES-Cu induces tumor cell cuproptosis, releases DAMPs, promotes dendritic cells (DCs) maturation, and macrophage M1 polarization. In vivo, TSF@ES-Cu reshapes the tumor microenvironment (TME), increasing mature DCs from 22.7% to 43.3%, CD8+ T cells from 5.08% to 17.1%, and reducing M2 macrophages from 50.7% to 18.4%. Additionally, the combined anti-tumor efficacy of TSF@ES-Cu and αPDL-1 is 1.6 times higher than TSF@ES-Cu alone and 2.5 times higher than αPDL-1 alone. In summary, this study reports that the combination of TSF@ES-Cu and αPDL-1 effectively induces cuproptosis and reshapes the TME, offering a new approach for copper nanomaterial-based tumor immunotherapy.
Collapse
Affiliation(s)
- Si Gao
- Department of Biliary-pancreatic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Haodong Ge
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200020, China
| | - Lili Gao
- Department of Pathology, Xinhua Hospital, Affiliated to Medicine School of Shanghai Jiaotong University, Shanghai, 200092, China
| | - Ying Gao
- School of Stomatology, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, 010030, China
| | - Shuibin Tang
- Department of Biliary-pancreatic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Yiming Li
- Department of Biliary-pancreatic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
- Department of Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Zhiqing Yuan
- Department of Biliary-pancreatic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Wei Chen
- Department of Biliary-pancreatic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| |
Collapse
|
9
|
Zhang D, Zhang N, Zhao J, Li X, Bian F, Zhang Y, Ge Y, Li Z. Label-free multiplexed detection based on core-shell photonic barcodes integrated RCA. Biosens Bioelectron 2025; 271:117037. [PMID: 39657551 DOI: 10.1016/j.bios.2024.117037] [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: 08/07/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
Multiplexed, rapid, and accurate virus quantification is of great value in biomedical detection. Herein, we proposed a label-free multiplexed virus screening quantitative biosensor based on color core-shell hydrogel photonic crystal (PhC) barcode integrated rolling circle amplification (RCA). The composite hydrogel shell was formed by acrylic acid and polyethylene glycol diacrylate, and the core silica photonic crystal was used as a detector. In addition, by adjusting the internal periodic structure, the PhC microcarrier was able to perform various color barcodes for the detection of different targets. Based on these excellent properties of the nanocomposite barcode, the biosensor not only demonstrated the ability to rapidly and accurately detect SARS-COV-2-N, SARS-COV-2-S, and H1N1 simultaneously in one tube, but also converting the signal of target protein to nucleic acid signal based on DNA decorated antibody complex combine with the blocked primer and RCA strategy. As a result, the platform achieved highly sensitive multiplexed quantitative detection with a detection limit in the range of 0.30 pg/mL. In addition, the platform we developed was validated by clinical sample analysis with acceptable accuracy and high specificity, demonstrating the good potential applicability of the proposed detection method in clinical screening and diagnosis.
Collapse
Affiliation(s)
- Dagan Zhang
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China.
| | - Nan Zhang
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Junqi Zhao
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Xueqin Li
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Feika Bian
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Yi Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China.
| | - Yizhi Ge
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China.
| | - Zhiyang Li
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China.
| |
Collapse
|
10
|
Zhang M, Yue W, Ma W, Wang X, Xu Y, Li A. Heterostructure Nanozyme with Hyperthermia-Amplified Enzyme-Like Activity and Controlled Silver Release for Synergistic Antibacterial Therapy. Adv Healthc Mater 2025; 14:e2401602. [PMID: 38900390 DOI: 10.1002/adhm.202401602] [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: 04/30/2024] [Revised: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Heterostructure nanozymes as antibiotic-free antimicrobial agents exhibit great potential for multidrug-resistant (MDR) bacterial strains elimination. However, realization of heterostructure antimicrobials with enhanced interfacial interaction for synergistically amplified antibacterial therapy is still a great challenge. Herein, oxygen-vacancy-enriched glucose modified MoOx (G-MoOx) is exploited as a reducing agent to spontaneously reduce Ag (I) into Ag (0) that in situ grows onto the surface of G-MoOx. The resultant Ag doped G-MoOx (Ag/G-MoOx) heterostructure displays augmenting photothermal effect and NIR-enhanced oxidase-like activity after introducing Ag nanoparticles. What's more, NIR hyperthermia accelerate Ag+ ions release from Ag nanoparticles. Introduction of Ag greatly enhances antimicrobial activities of Ag/G-MoOx against MDR bacteria, especially the hybrid loading with 1 wt% Ag NPs exhibiting antibacterial efficacy up to 99.99% against Methicillin-resistant Staphylococcus aureus (MRSA, 1×106 CFU mL-1).
Collapse
Affiliation(s)
- Meng Zhang
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao, 266071, P. R. China
| | - Wenhui Yue
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao, 266071, P. R. China
| | - Weishuai Ma
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao, 266071, P. R. China
| | - Xiaoning Wang
- College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Yuanhong Xu
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao, 266071, P. R. China
| | - Aihua Li
- College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, P. R. China
| |
Collapse
|
11
|
Wu R, Zhang Y, Teng J, Zhang Q, Zhang C. Molecular Circuit-Controlled Nanoparticle Folders for Programmable DNA Information Access. ACS NANO 2025; 19:6918-6928. [PMID: 39945285 DOI: 10.1021/acsnano.4c13882] [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: 02/26/2025]
Abstract
DNA storage has become an attractive alternative to long-term, stable digital data storage because of its high storage density and strong stability. Recently, numerous efforts have been made to develop DNA data access methods to improve the efficiency and accuracy of molecular data reading. However, most current data access methods were achieved by well-developed polymerase chain reaction (PCR) and DNA hybridization, which lack the exploration of dynamic and programmable operations for data access. Here, we propose a programmable DNA data access strategy in which the nanoparticle folders are controlled by DNAzyme circuits to achieve specific information manipulation. We experimentally demonstrate three kinds of circuit programs that access specific information in YES, AND, and OR logic manner. In addition, the selective information access was performed by using a DNAzyme circuit to obtain the target information from a DNA data pool. Importantly, we have extended the circuit-controlled framework to multiple manipulation modes, demonstrating four manipulations on two AuNP folders to access different information on demand. The programmable access strategy provides a paradigm for integrated DNA computing and storage systems and has more applications in the fields of molecular computation and DNA data storage.
Collapse
Affiliation(s)
- Ranfeng Wu
- School of Computer Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Yongpeng Zhang
- School of Control and Computer Engineering, North China Electric Power University, Beijing 100096, China
| | - Jiongjiong Teng
- School of Control and Computer Engineering, North China Electric Power University, Beijing 100096, China
| | - Qiang Zhang
- School of Computer Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Cheng Zhang
- School of Computer Science, Key Laboratory of High Confidence Software Technologies, Peking University, Beijing 100871, China
| |
Collapse
|
12
|
Wang J, Cai L, Li N, Luo Z, Ren H, Zhang B, Zhao Y. Developing mRNA Nanomedicines with Advanced Targeting Functions. NANO-MICRO LETTERS 2025; 17:155. [PMID: 39979495 PMCID: PMC11842722 DOI: 10.1007/s40820-025-01665-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 02/22/2025]
Abstract
The emerging messenger RNA (mRNA) nanomedicines have sprung up for disease treatment. Developing targeted mRNA nanomedicines has become a thrilling research hotspot in recent years, as they can be precisely delivered to specific organs or tissues to enhance efficiency and avoid side effects. Herein, we give a comprehensive review on the latest research progress of mRNA nanomedicines with targeting functions. mRNA and its carriers are first described in detail. Then, mechanisms of passive targeting, endogenous targeting, and active targeting are outlined, with a focus on various biological barriers that mRNA may encounter during in vivo delivery. Next, emphasis is placed on summarizing mRNA-based organ-targeting strategies. Lastly, the advantages and challenges of mRNA nanomedicines in clinical translation are mentioned. This review is expected to inspire researchers in this field and drive further development of mRNA targeting technology.
Collapse
Affiliation(s)
- Ji Wang
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Lijun Cai
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Ning Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Zhiqiang Luo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China
| | - Haozhen Ren
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
| | - Yuanjin Zhao
- Department of Radiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, People's Republic of China.
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, People's Republic of China.
| |
Collapse
|
13
|
Wang J, Chen Q, Wang Y, Gan Z, Wu M, Shang L, Duan P. Multiresponsive Microcapsules for Prevention of Intrauterine Adhesion. ACS NANO 2025; 19:6499-6510. [PMID: 39915116 DOI: 10.1021/acsnano.4c17645] [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: 02/19/2025]
Abstract
Intrauterine adhesion (IUA) has a significant negative impact on women's reproductive health. One of the development trends of biomaterials for prevention of IUA is improving the stability and multifaceted functions. Here, we propose a multiresponsive microcapsule (A/G-Fe3O4-Se) with an alginate (ALG) and gelatin methacryloyl (GelMA) dual-network hydrogel shell loaded with magnetic nanoparticles (Fe3O4-Se) and an ultrasound-responsive decafluoropentane core for the prevention of IUA using a microfluidic technique. The microcapsules inherited the biofriendly advantages of ALG and GelMA. The encapsulated magneto-responsive Fe3O4-Se made the microcapsule flexibly change the distribution to adapt to the irregular shape of the uterus and better exert the therapeutic effect. Besides, the A/G-Fe3O4-Se microcapsules demonstrated antioxidant, antibacterial, and pro-healing properties in vitro. Moreover, in the IUA rat models, we also observed a reduction in oxidative stress, better endometrial regeneration, and improved endometrial receptivity and pregnancy rates after treatment of the microcapsules. Consequently, the A/G-Fe3O4-Se microcapsules can be used as a promising strategy for the treatment of damaged endometrium as well as prevention of IUA.
Collapse
Affiliation(s)
- Jing Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Oncology Discipline Group, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Qiong Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yueyue Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhouyi Gan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Meiling Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Luoran Shang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Ping Duan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Oncology Discipline Group, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
14
|
Wang R, Wang Z, Zhang M, Zhong D, Zhou M. Application of photosensitive microalgae in targeted tumor therapy. Adv Drug Deliv Rev 2025; 219:115519. [PMID: 39955076 DOI: 10.1016/j.addr.2025.115519] [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: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/18/2025] [Indexed: 02/17/2025]
Abstract
Microalgae present a novel and multifaceted approach to cancer therapy by modulating the tumor-associated microbiome (TAM) and the tumor microenvironment (TME). Through their ability to restore gut microbiota balance, reduce inflammation, and enhance immune responses, microalgae contribute to improved cancer treatment outcomes. As photosynthetic microorganisms, microalgae exhibit inherent anti-tumor, antioxidant, and immune-regulating properties, making them valuable in photodynamic therapy and tumor imaging due to their capacity to generate reactive oxygen species. Additionally, microalgae serve as effective drug delivery vehicles, leveraging their biocompatibility and unique structural properties to target the TME more precisely. Microalgae-based microrobots further expand their therapeutic potential by autonomously navigating complex biological environments, offering a promising future for precision-targeted cancer treatments. We position microalgae as a multifunctional agent capable of modulating TAM, offering novel strategies to enhance TME and improve the efficacy of cancer therapies.
Collapse
Affiliation(s)
- Ruoxi Wang
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310029, China; Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China; Zhejiang University-Ordos City Etuoke Banner Joint Research Center, Haining 314400, China
| | - Zhouyue Wang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| | - Min Zhang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| | - Danni Zhong
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Min Zhou
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310029, China; Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China; Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China; Zhejiang University-Ordos City Etuoke Banner Joint Research Center, Haining 314400, China.
| |
Collapse
|
15
|
Lu J, Huang D, Liu R, Zhu H, Wang D, Zhao Y, Sun L. Extracellular Matrix-Inspired Dendrimer Nanogels Encapsulating Cyclophosphamide for Systemic Sclerosis Treatment. ACS NANO 2025; 19:4672-4683. [PMID: 39834323 DOI: 10.1021/acsnano.4c15164] [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: 01/22/2025]
Abstract
Cyclophosphamide has a certain therapeutic effect on treating systemic sclerosis (SSc), while difficulties exist in controlling severe systematic side effects and enhancing targeting capacity. Here, inspired from the natural extracellular matrix composition, we propose a cyclophosphamide-encapsulated nanogel based on dendritic polymers polyamidoamine (PAMAM) for SSc treatment. We combine bovine serum albumin and generation 5 (G5) PAMAM dendrimers with polyphenol modification to obtain nanogels featured with antioxidant and anti-inflammatory effects. The nanogels can possess excellent biocompatibility and prevent fibroblasts from oxidative stress damage and TGF-β-mediated activation. Furthermore, in the bleomycin-induced SSc mouse model, dendrimer nanogels encapsulating cyclophosphamide also exhibit the ability to attenuate fibrosis by modulating immunity, suppressing inflammation, and reducing collagen synthesis. These findings underscore the value of this dendritic polymer nanogel in the treatment of chronic SSc, indicating its broader potential for clinical applications.
Collapse
Affiliation(s)
- Junjie Lu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Danqing Huang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Rui Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Haofang Zhu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| |
Collapse
|
16
|
Roh J, Park S, Kim H, Kim W, Kim J, Park C, Choi H, Song S, Choi E, Choi Y. Polymorphing Hydrogels Regulated by Photo-reactive DNA Cross-links. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414648. [PMID: 39617981 DOI: 10.1002/adma.202414648] [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: 09/27/2024] [Revised: 11/10/2024] [Indexed: 02/06/2025]
Abstract
Polymorphing hydrogels can morph into another structure on demand with reprogrammable features. This concept extends the degree of morphing beyond that of traditional shape-morphing hydrogels, which predetermine their morphing capabilities at the fabrication stage. However, current polymorphing hydrogels face limitations due to the need for complex, non-sustained responsiveness or additional chemical steps for reconfigurable morphing. Here, photo-reactive DNA-cross-linked polymorphing hydrogels are presented that enable polymorphing under patterned light. The photo-reactive DNA cross-links act as a regulator in changing the shapes of the hydrogels by adjusting the lengths of the cross-links depending on the wavelength of light, which allows precise and dynamic morphing in a programmable way through a one-pot reaction. The high programmability involving spatiotemporal controllability and reprogrammable features offers advanced solutions for multifunctional soft machines and applications requiring complex processes.
Collapse
Affiliation(s)
- Junho Roh
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123, Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Seongjun Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123, Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Hoeseong Kim
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123, Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Woojin Kim
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123, Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Jinho Kim
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123, Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Cheolheon Park
- Bio-MAX Institute, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
- Department of Electrical and Computer Engineering, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Hansol Choi
- Bio-MAX Institute, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
- Department of Electrical and Computer Engineering, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Department of Biological Chemistry and Molecular Pharmacology (BCMP), Boston, MA, 02115, USA
| | - Seowoo Song
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Eunjin Choi
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123, Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| | - Yeongjae Choi
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), 123, Cheomdangwagi-ro, Buk-gu, Gwangju, 61005, Republic of Korea
| |
Collapse
|
17
|
Jia J, Zhang X, Li Y, Wang T, An Y, Yan X, Liu B, Yang C, Ju H. Remotely Sequential Activation of Biofunctional MXenes for Spatiotemporally Controlled Photothermal Cancer Therapy Integrated with Multimodal Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410535. [PMID: 39665387 DOI: 10.1002/smll.202410535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/26/2024] [Indexed: 12/13/2024]
Abstract
Spatiotemporally controlled cancer therapy may offer great advantages in precision medicine, but still remains some challenges in programmed sequential release and co-localization of components at target sites. Herein, a MXene-based nanoprobe (TCC@M) is meticulously designed by engineering of photodynamically activated CRISPR-Cas9 and cancer cell membrane-camouflaged Ti3C2 MXenes for targeting delivery and spatiotemporally controlled gene regulation followed by enhanced photothermal therapy (PTT) via two near-infrared irradiations. The first irradiation can activate the photosensitizer bound in cancer cells internalized TCC@M to release Cas9 ribonucleoprotein (RNP) by photodynamic effect. The released Cas9 RNP then enters the nuclei directed by the fused nuclear localization sequence in Cas9 to cleave the heat shock protein (HSP) 90α gene, which greatly reduces the expression of HSP90α protein and thus effectively sensitizes cancer cells to heat, leading to enhanced PTT at a mild temperature (<45 °C) risen by Ti₃C₂ MXenes under the second irradiation. Simultaneously, TCC@M can produce fluorescence, photoacoustic, and thermal imaging signals to guide the optimal irradiation timing. The in vivo studies have demonstrated the spatiotemporally selective therapeutic efficacy of the designed TCC@M. This innovative approach presents an effective integration of gene regulation and enhanced PTT, exemplifying a precise cancer treatment strategy.
Collapse
Affiliation(s)
- Jing Jia
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Xiaobo Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Tian Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Ying An
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Xinrong Yan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Bin Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Chaoyi Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
18
|
Wang J, Shang J, Yu S, Lin M, Gong X, Liu X, Liu Z, Wang F. Self-Adaptive Activation of DNAzyme Nanoassembly for Synergistically Combined Gene Therapy. Angew Chem Int Ed Engl 2025; 64:e202417363. [PMID: 39415359 DOI: 10.1002/anie.202417363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/12/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
DNAzyme represents a promising gene silencing toolbox yet is obstructed by the poor substrate accessibility in specific cells. Herein, a compact DNA nanoassembly, incorporating multimeric therapeutic DNAzyme, was prepared for selective delivery of gene-silencing DNAzyme with requisite cofactors and auxiliary chemo-drugs. By virtue of the sequence-conservative duplex-specific nuclease, the endogenous miRNA catalyzes the successive and site-specific cleavage of DNA nanoassembly substrate (nominated as the localized RNA walking machine) and thus ensures the liberation/activation of therapeutic agents with high accuracy and efficacy. The miR-10b-stimulated DNAzyme was designed to downregulate the TWIST transcription factor, an upstream promotor of miR-10b, thus acquiring the self-sufficient downregulation of TWIST/miR-10b signaling nodes (self-adaptive negative feedback loop) for abrogating tumor metastasis and chemo-resistance issues.
Collapse
Affiliation(s)
- Jing Wang
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, 430072, Wuhan, P. R. China
- College of Health Science and Engineering, Key Laboratory for the Synthesis and Application of Organic Functional Molecules (Ministry of Education), Hubei University, 430062, Wuhan, P. R. China
| | - Jinhua Shang
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, 430072, Wuhan, P. R. China
| | - Shanshan Yu
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, 430072, Wuhan, P. R. China
| | - Mengru Lin
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, 430072, Wuhan, P. R. China
| | - Xue Gong
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, 430072, Wuhan, P. R. China
| | - Xiaoqing Liu
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, 430072, Wuhan, P. R. China
| | - Zhihong Liu
- College of Health Science and Engineering, Key Laboratory for the Synthesis and Application of Organic Functional Molecules (Ministry of Education), Hubei University, 430062, Wuhan, P. R. China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan University, 430072, Wuhan, P. R. China
| |
Collapse
|
19
|
Zhu H, Huang D, Nie M, Zhao Y, Sun L. Dexamethasone loaded DNA scavenger nanogel for systemic lupus erythematosus treatment. Bioact Mater 2025; 43:330-339. [PMID: 40115883 PMCID: PMC11923376 DOI: 10.1016/j.bioactmat.2024.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/09/2024] [Accepted: 08/27/2024] [Indexed: 03/23/2025] Open
Abstract
Lupus nephritis (LN) poses a severe risk for individuals with systemic lupus erythematosus (SLE), prompting extensive research into targeted delivery systems capable of modulating immune responses and clearing cell-free DNA (cfDNA). Here, we propose a novel renal homing nanogel that acts as a cfDNA scavenger and a dexamethasone (DXM) delivery carrier for LN treatment. Based on the generation 3 polylysine dendrimers, the created cationic nanogels (G3DSP) exhibit minimal toxicity and outstanding DXM loading efficiency. Our studies confirm that these nanogels can competitively bind with anionic cfDNA in vitro, leading to the suppression of toll-like receptor 9 (TLR9) activation. When administered systemically to MRL/lpr mice, the nanogels preferentially localize to and are retained in the inflamed kidneys, releasing their payload in response to reactive oxygen species (ROS), therefore effectively ameliorating SLE symptoms. Consequently, G3DSP nanogels emerge as a promising effective combined therapy for LN, minimizing cfDNA accumulation in vital organs and delivering immunomodulatory benefits through DXM.
Collapse
Affiliation(s)
- Haofang Zhu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Danqing Huang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Min Nie
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| |
Collapse
|
20
|
Liu Y, Bai X, Wang H, Wang J, Li S, Zhang H, Wang F, Hong Z. PEG-modification enhances the targeted photothermal therapy of affibody-conjugated indocyanine green for precision cancer treatment. Biochem Biophys Res Commun 2025; 742:151155. [PMID: 39662453 DOI: 10.1016/j.bbrc.2024.151155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Photothermal therapy (PTT) is an innovative cancer treatment that leverages heat generated from near-infrared light exposure to induce tumor cell death. A major challenge in PTT is achieving precise delivery of the photothermal agent to tumor tissues to maximize efficacy and minimize off-target effects. In this study, we introduce a novel ligand-coupled photothermal reagent that addresses this challenge by leveraging the high-affinity HER2 affibody ZHER2:2891 (referred to as ZHER2), conjugated with indocyanine green (ICG) for targeted delivery. Polyethylene glycol (PEG) was incorporated as a hydrophilic linker to further optimize photothermal conversion efficiency and enhance tumor-specific targeting. Among the conjugates tested, ZHER2-PEG1000-ICG, modified with a PEG chain of 1000 Da molecular weight, demonstrated exceptional performance. In vitro studies revealed that ZHER2-PEG1000-ICG specifically bound to HER2-expressing cells and effectively induced cell death. In vivo experiments using HER2-positive N87 tumor-bearing mice showed that ZHER2-PEG1000-ICG accumulated highly and specifically in tumor tissues over an extended period. Upon light irradiation, this conjugate caused a significant rise in temperature at the tumor site, resulting in complete tumor elimination with a single photothermal treatment. This PEG-modified affibody-ICG conjugate represents a precise and effective approach to PTT, offering a promising new therapeutic strategy for cancer treatment with the potential to significantly impact future cancer therapies.
Collapse
Affiliation(s)
- Yanting Liu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, PR China; State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Xuerui Bai
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Henan Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Jian Wang
- National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, PR China.
| | - Shuang Li
- National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, PR China.
| | - Hongru Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Fengwei Wang
- People's Hospital of Tianjin, School of Medicine, Nankai University, Tianjin, 300071, PR China.
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| |
Collapse
|
21
|
Zhu L, Zhang Y, Ding J, Xin W, Fan GC, Song ZL, Zhang P, Luo X. Intelligent DNA Nanosystem for Broad-Spectrum Oncological Typing and Therapy. ACS Sens 2024; 9:6655-6666. [PMID: 39622787 DOI: 10.1021/acssensors.4c02246] [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: 12/28/2024]
Abstract
The occurrence of multiple primary cancers in individual patients underscores the need for diagnostic and therapeutic techniques with augmented cancer-targeting selectivity and broad-spectrum antitumor effects. To address this, we develop a quadruple-input-triggered OR-AND-AND logic gated oncological nanosystem (OAA). This system employs four cancer-related markers (EpCAM, MUC1, APE1, and miR-21) to generate three distinct fluorescence signals, enabling precise differentiation of various cancer cell lines (MCF-7, HepG2, and HeLa) from normal cells (MCF-10A). Additionally, the OAA system integrates photodynamic therapy (PDT) and gene silencing strategies, allowing selective activation of Ce6 release, miR-21 gene silencing, and VEGFR2 mRNA gene silencing through the OR-AND-AND logic gating mechanism in a cancer-specific manner. This synergetic therapeutic approach induces significant apoptosis in multiple cancer cell lines while sparing normal cells, demonstrating improved cancer-targeting specificity and broad-spectrum versatility. This intelligent platform precisely types and treats diverse cancer cells, powering the future exploration of advanced diagnostic and therapeutic strategies to combat highly heterogeneous diseases.
Collapse
Affiliation(s)
- Lina Zhu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yuxi Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jiani Ding
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Wenjuan Xin
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Gao-Chao Fan
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Zhi-Ling Song
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453000, China
| | - Peisen Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiliang Luo
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| |
Collapse
|
22
|
Chen Y, Yang SY, Ou X, Wang H, Kong FC, Chow PCY, Wang Y, Jiang Y, Zhao W, Sun J, Kwok RTK, Zheng DW, Yu W, Wang F, Lam JWY, Tang BZ. Engineering a Near-Infrared Spiro-Based Aggregation-Induced Emission Luminogen for DNAzyme-Sensitized Photothermal Therapy with High Efficiency and Accuracy. J Am Chem Soc 2024; 146:35462-35477. [PMID: 39665333 DOI: 10.1021/jacs.4c14818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Aggregation-induced emission luminogen (AIEgens)-based photothermal therapy (PTT) has grown into a sparkling frontier for tumor ablation. However, challenges remain due to the uncoordinated photoluminescence (PL) and photothermal properties of classical AIEgens, along with hyperthermia-induced antiapoptotic responses in tumor cells, hindering satisfactory therapeutic outcomes. Herein, a near-infrared (NIR) spiro-AIEgen TTQ-SA was designed for boosted PTT by auxiliary DNAzyme-regulated tumor cell sensitization. TTQ-SA with a unique molecular structure and packing mode was initially fabricated, endowing it with a strong AIE effect, favorable PL quantum yield, and good photothermal performance. DNAzyme, as a gene silencing tool, could alleviate antiapoptosis response during PTT. By integrating TTQ-SA and DNAzyme into folate-modified poly(lactic-co-glycolic acid) (PLGA) polymer, the as-fabricated nanosystem could promote cell apoptosis and sensitize tumor cells to PTT, thereby maximizing the therapeutic outcomes. With the combination of spiro-AIEgen-based PTT and DNAzyme-based gene silencing, the as-designed nanosystem showed promising NIR and photothermal imaging abilities for tumor targeting and demonstrated significant cell apoptotic, antitumor, and antimetastasis effects against orthotopic breast cancer. Furthermore, a synergistic antitumor effect was realized in spontaneous MMTV-PyMT transgenic mice. These findings offer new insights into AIEgen-based photothermal theranostics and DNAzyme-regulated tumor cell sensitization, paving the way for synergistic gene silencing-PTT nanoplatforms in clinical research.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Sheng-Yi Yang
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Xinwen Ou
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Hui Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430071, China
| | - Fan-Cheng Kong
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam 999077, Hong Kong SAR, China
| | - Philip C Y Chow
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam 999077, Hong Kong SAR, China
| | - Yifei Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430071, China
| | - Yuqian Jiang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430071, China
| | - Wei Zhao
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou City 510280, Guangdong, China
| | - Jianwei Sun
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Ryan T K Kwok
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Di-Wei Zheng
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Wenqian Yu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430071, China
| | - Fuan Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430071, China
| | - Jacky W Y Lam
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Ben Zhong Tang
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Shenzhen 518172, Guangdong, China
| |
Collapse
|
23
|
Zhang R, Zhou T, Huo X, Du H, Ning G, Weng T, Wang X, Zhang G, Wang F, Zhang Z. A Multifunctional DNA Nanoassembly for Cancer Cell Detection and Combined Gene-Chemotherapy Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:26033-26040. [PMID: 39601798 DOI: 10.1021/acs.langmuir.4c03495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Although DNAzyme is a promising gene therapy agent, low cellular uptake efficiency, poor biological stability, and the unsatisfactory effect of monotherapy limit its development. Herein, a multifunctional DNA nanoassembly (RCA product-aptamer-DNAzyme, RAD) was constructed for cancer cell detection and targeted delivery of doxorubicin (DOX) and DNAzyme. Briefly, the rolling circle amplification (RCA) product was employed as a scaffold, and each repeated sequence was designed to combine with three single-stranded DNA (ssDNA), which carried the aptamer AS1411 sequence, fluorescent group, and DNAzyme sequence, respectively. Up to 40 groups of ssDNA can be assembled into an RCA product, resulting in a high affinity for cancer cells and stronger fluorescent signals. Due to the high binding affinity, RAD displayed high sensitivity for the detection of HepG-2 cells (the limit of detection was 200 cells/mL). In addition, with the formation of the double helix structure, each RAD could load up to 200 DOX molecules. Subsequently, RAD could efficiently and selectively deliver DOX and DNAzyme into cancer cells through the multivalent interaction between the aptamers and membrane nucleolin. Then, the released DNAzyme could recognize and cleave survivin mRNA under the action of Mg2+, leading to the apoptosis of HepG-2 cells for gene therapy, while DOX inserted into intracellular DNA to inhibit cell proliferation, realizing chemotherapy. According to the results, RAD-DOX displayed enhanced therapeutic effects compared with individual gene therapy or chemotherapy, and RAD could protect membrane nucleolin-negative cells from the effects of DOX. Overall, given the enhanced serum stability, high drug-loading capacity, and excellent selective cellular uptake ability of RAD, this strategy shows great potential in the field of cancer therapy.
Collapse
Affiliation(s)
- Ruyan Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Ting Zhou
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Xiaobing Huo
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Huan Du
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Gan Ning
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Tianxin Weng
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Xiufeng Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Guodong Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Fang Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Zhiqing Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
24
|
Xie X, Nan H, Peng J, Zeng K, Wang HH, Huang Y, Nie Z. Hydrogen Sulfide-Triggered Artificial DNAzyme Switches for Precise Manipulation of Cellular Functions. Angew Chem Int Ed Engl 2024; 63:e202410380. [PMID: 39327234 DOI: 10.1002/anie.202410380] [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: 06/02/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 09/28/2024]
Abstract
The development of synthetic molecular tools responsive to biological cues is crucial for advancing targeted cellular regulation. A significant challenge is the regulation of cellular processes in response to gaseous signaling molecules such as hydrogen sulfide (H2S). To address this, we present the design of Gas signaling molecule-Responsive Artificial DNAzyme-based Switches (GRAS) to manipulate cellular functions via H2S-sensitive synthetic DNAzymes. By incorporating stimuli-responsive moieties to the phosphorothioate backbone, DNAzymes are strategically designed with H2S-responsive azide groups at cofactor binding locations within the catalytic core region. These modifications enable their activation through H2S-reducing decaging, thereby initiating substrate cleavage activity. Our approach allows for the flexible customization of various DNAzymes to regulate distinct cellular processes in diverse scenarios. Intracellularly, the enzymatic activity of GRAS promotes H2S-induced cleavage of specific mRNA sequences, enabling targeted gene silencing and inducing apoptosis in cancer cells. Moreover, integrating GRAS with dynamic DNA assembly allows for grafting these functional switches onto cell surface receptors, facilitating H2S-triggered receptor dimerization. This extracellular activation transmits signals intracellularly to regulate cellular behaviors such as migration and proliferation. Collectively, synthetic switches are capable of rewiring cellular functions in response to gaseous cues, offering a promising avenue for advanced targeted cellular engineering.
Collapse
Affiliation(s)
- Xuan Xie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Hexin Nan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Jialong Peng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Kaiqiang Zeng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Yan Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, 410082, Changsha, P. R. China
| |
Collapse
|
25
|
Wang P, Sun X, Tang L, Li N, Wang Q, Gan B, Zhang Y. CaCO 3-encircled hollow CuS nanovehicles to suppress cervical cancer through enhanced calcium overload-triggered mitochondria damage. Asian J Pharm Sci 2024; 19:100989. [PMID: 39640053 PMCID: PMC11616050 DOI: 10.1016/j.ajps.2024.100989] [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: 05/13/2024] [Revised: 06/25/2024] [Accepted: 07/28/2024] [Indexed: 12/07/2024] Open
Abstract
Cervical cancer stands is a formidable malignancy that poses a significant threat to women's health. Calcium overload, a minimally invasive tumor treatment, aims to accumulate an excessive concentration of Ca2+ within mitochondria, triggering apoptosis. Copper sulfide (CuS) represents a photothermal mediator for tumor hyperthermia. However, relying solely on thermotherapy often proves insufficient in controlling tumor growth. Curcumin (CUR), an herbal compound with anti-cancer properties, inhibits the efflux of exogenous Ca2+ while promoting its excretion from the endoplasmic reticulum into the cytoplasm. To harness these therapeutic modalities, we have developed a nanoplatform that incorporates hollow CuS nanoparticles (NPs) adorned with multiple CaCO3 particles and internally loaded with CUR. This nanocomposite exhibits high uptake and easy escape from lysosomes, along with the degradation of surrounding CaCO3, provoking the generation of abundant exogenous Ca2+ in situ, ultimately damaging the mitochondria of diseased cells. Impressively, under laser excitation, the CuS NPs demonstrate a photothermal effect that accelerates the degradation of CaCO3, synergistically enhancing the antitumor effect through photothermal therapy. Additionally, fluorescence imaging reveals the distribution of these nanovehicles in vivo, indicating their effective accumulation at the tumor site. This nanoplatform shows promising outcomes for tumor-targeting and the effective treatment in a murine model of cervical cancer, achieved through cascade enhancement of calcium overload-based dual therapy.
Collapse
Affiliation(s)
- Pengfei Wang
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China
| | - Xichen Sun
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China
| | - Liuyan Tang
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China
| | - Ningning Li
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China
| | - Qing Wang
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China
| | - Bicheng Gan
- College of Petroleum Engineering, Heilongjiang, Northeast Petroleum University, Daqing 163318, China
| | - Yuezhou Zhang
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China
- Ningbo Institute of Northwestern Polytechnical University, Frontiers Science Center for Flexible Electronics (FSCFE), Key laboratory of Flexible Electronics of Zhejiang Province, Ningbo 315103, China
| |
Collapse
|
26
|
Chen Q, Liu Y, Chen Q, Li M, Xu L, Lin B, Tan Y, Liu Z. DNA Nanostructures: Advancing Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2405231. [PMID: 39308253 DOI: 10.1002/smll.202405231] [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: 06/26/2024] [Revised: 08/28/2024] [Indexed: 12/06/2024]
Abstract
Cancer immunotherapy is a groundbreaking medical revolution and a paradigm shift from traditional cancer treatments, harnessing the power of the immune system to target and destroy cancer cells. In recent years, DNA nanostructures have emerged as prominent players in cancer immunotherapy, exhibiting immense potential due to their controllable structure, surface addressability, and biocompatibility. This review provides an overview of the various applications of DNA nanostructures, including scaffolded DNA, DNA hydrogels, tetrahedral DNA nanostructures, DNA origami, spherical nucleic acids, and other DNA-based nanostructures in cancer immunotherapy. These applications explore their roles in vaccine development, immune checkpoint blockade therapies, adoptive cellular therapies, and immune-combination therapies. Through rational design and optimization, DNA nanostructures significantly bolster the immunogenicity of the tumor microenvironment by facilitating antigen presentation, T-cell activation, tumor infiltration, and precise immune-mediated tumor killing. The integration of DNA nanostructures with cancer therapies ushers in a new era of cancer immunotherapy, offering renewed hope and strength in the battle against this formidable foe of human health.
Collapse
Affiliation(s)
- Qianqian Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| | - Bingyu Lin
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| |
Collapse
|
27
|
Huang H, Chen Y, Li L, Zheng L. A DNAzyme-Based Nanohybrid for Ultrasound and Enzyme Dual-Controlled mRNA Regulation and Combined Tumor Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409035. [PMID: 39470099 DOI: 10.1002/adma.202409035] [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: 06/24/2024] [Revised: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Despite the significant potential of RNA-cleaving DNAzymes for gene regulation, their application is limited by low therapeutic efficacy and lack of cell-specific control. Here, a DNAzyme-based nanohybrid designed for ultrasound (US)-controlled, enzyme-activatable mRNA regulation is presented, enabling tumor cell-selective combination therapy. The nanohybrid is constructed by coordination-directed self-assembly of an enzymatically-triggerable therapeutic DNAzyme (En-Dz) and natural sonosensitizer hemoglobin (Hb). Controlled US exposure induces reactive oxygen species generation from Hb units, which not only facilitates efficient endosomal escape of En-Dz, but also promotes the translocation of specific enzyme from the nucleus to the cytoplasm, thereby enhancing gene regulation efficacy. Notably, the enzyme-triggered, spatiotemporally-controlled activation of En-Dz's catalytic activity results in enhanced cancer-cell selectivity in the therapeutic treatment. Furthermore, the combination of enzyme-activated mRNA regulation and sonodynamic therapy significantly enhances anti-tumor efficacy both in vitro and in vivo. This work highlights the potential of integrating a sonodynamic strategy to overcome the current limitations of DNAzyme-based gene regulators, providing a spatiotemporally-controlled approach for precise tumor treatment.
Collapse
Affiliation(s)
- Hanji Huang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
| | - Yaoxuan Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Lele Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
28
|
Cheng Y, Zhang X, Wu X, Ding Y, Zhu L, Pan J, Zhao Y, Zhou M. Multifunctional Microneedle Patches for Perivascular Gene Delivery and Treatment of Vascular Intimal Hyperplasia. ACS NANO 2024; 18:32578-32588. [PMID: 39545708 DOI: 10.1021/acsnano.4c09527] [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: 11/17/2024]
Abstract
Gene therapy has emerged as a promising approach to address challenging cardiovascular diseases. Extensive efforts have been focused on developing highly efficient gene vectors with precise delivery techniques to enhance its effectiveness. In this study, we present multifunctional dopamine-gelatin microneedle patches with gene therapy capabilities to achieve perivascular gene delivery for intimal hyperplasia treatment. These patches that were fabricated through freeze-drying of gelatin are with recombinant adeno-associated virus (rAAVs)-carrying tips and dopamine coating backing layers. The lyophilized gelatin could not only effectively preserve the therapeutic activity of rAAVs but could also demonstrate the capability to penetrate the adventitia for efficient delivery. The incorporation of dopamine facilitated patch adhesion and extended the release duration. Based on these advantages, we have demonstrated that the rAAVs-loaded microneedle patches (AMNPs) behave satisfactorily in perivascular gene delivery to inhibit carotid artery restenosis in rats. These features indicate that the AMNPs are clinically valuable in the treatment of vascular intimal hyperplasia diseases.
Collapse
Affiliation(s)
- Yi Cheng
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xiaoxuan Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiangyi Wu
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Youjun Ding
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Linxi Zhu
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jun Pan
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yuanjin Zhao
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Min Zhou
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
29
|
Hua S, Hu H, Liu J, Lu F, Yu R, Zhang X, Sun H, Wang Z, Li Y, Xia J, Xu F, Zhou M. A Mucous Permeable Local Delivery Strategy Based on Manganese-Enhanced Bacterial Cuproptosis-like Death for Bacterial Pneumonia Treatment. ACS NANO 2024; 18:31923-31940. [PMID: 39506631 DOI: 10.1021/acsnano.4c09695] [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: 11/08/2024]
Abstract
Bacterial pneumonia is one of the most challenging global infectious diseases with high morbidity and mortality. Considering the antibiotic abuse and resistance of bacterial biofilms, a variety of metal-based materials have been developed. However, due to the high oxygen environment of the lungs, some aerobic infection bacteria have high tolerance to oxygen and ROS, and most of the metal-based materials based on ROS may not achieve good therapeutic effects. Inspired by the sensitivity of cuproptosis to aerobic respiratory cells, we designed a copper composite antibacterial nanoparticle and found that it can effectively induce cuproptosis-like death in the aerobic bacteria of the lungs. To address the challenge of in vivo application of cuproptosis, manganese dioxide was first incorporated to deplete protective glutathione, which can interact with copper and thus hinder the interaction of copper with proteins and assist in antibacterial action through immune enhancement. Cuproptosis-like death also requires a large number of copper ions. To meet this demand, we deliver positively hydrophilic modified composite nanoparticles that effectively penetrate the lung mucus layer directly to the lungs through local administration, and the copper ions are further released rapidly by the acidic environment at the infected site, which can further destroy bacterial biofilms in synergy with manganese. This drug-delivery system can effectively treat pneumonia caused by aerobic bacteria and avoid systemic toxicity that can be caused by large doses of copper.
Collapse
Affiliation(s)
- Shiyuan Hua
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310009, China
- ZJU-Etuoke Joint Research Centre, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining 314400, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Huiqun Hu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jin Liu
- ZJU-Etuoke Joint Research Centre, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining 314400, China
| | - Feng Lu
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou213003, China
| | - Runchang Yu
- ZJU-Etuoke Joint Research Centre, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining 314400, China
| | - Xuening Zhang
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310009, China
- ZJU-Etuoke Joint Research Centre, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining 314400, China
| | - Haimeng Sun
- ZJU-Etuoke Joint Research Centre, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining 314400, China
| | - Ziwei Wang
- ZJU-Etuoke Joint Research Centre, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining 314400, China
| | - Yige Li
- ZJU-Etuoke Joint Research Centre, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining 314400, China
| | - Jingyan Xia
- Department of Radiation Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Min Zhou
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310009, China
- ZJU-Etuoke Joint Research Centre, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining 314400, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
30
|
Zhu C, Yu M, Lv J, Sun F, Qin A, Chen Z, Hu X, Yang Z, Fang Z. De novo strategy of organic semiconducting polymer brushes for NIR-II light-triggered carbon monoxide release to boost deep-tissue cancer phototheranostics. J Nanobiotechnology 2024; 22:708. [PMID: 39543646 PMCID: PMC11562092 DOI: 10.1186/s12951-024-02984-6] [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: 07/07/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
The integration of photoacoustic imaging (PAI) and photothermal therapy (PTT) within the second near-infrared (NIR-II) window, offering a combination of high-resolution imaging and precise non-invasive thermal ablation, presents an attractive opportunity for cancer treatment. Despite the significant promise, the development of this noninvasive phototheranostic nanomedicines encounters challenges that stem from tumor thermotolerance and limited therapeutic efficacy. In this contribution, we designed an amphiphilic semiconducting polymer brush (SPB) featuring a thermosensitive carbon monoxide (CO) donor (TDF-CO) for NIR-II PAI-assisted gas-augmented deep-tissue tumor PTT. TDF-CO nanoparticles (NPs) exhibited a powerful photothermal conversion efficiency (43.1%) and the capacity to trigger CO release after NIR-II photoirradiation. Notably, the liberated CO not only acts on mitochondria, leading to mitochondrial dysfunction and promoting cellular apoptosis but also hinders the overexpression of heat shock proteins (HSPs), enhancing the tumor's thermosensitivity to PTT. This dual action accelerates cellular thermal ablation, achieving a gas-augmented synergistic therapeutic effect in cancer treatment. Intravenous administration of TDF-CO NPs in 4T1 tumor-bearing mice demonstrated bright PAI signals and remarkable tumor ablation under 1064 nm laser irradiation, underscoring the potential of CO-mediated photothermal/gas synergistic therapy. We envision this tailor-made multifunctional NIR-II light-triggered SPB provides a feasible approach to amplify the performance of PTT for advancing future cancer phototheranostics.
Collapse
Affiliation(s)
- Caijun Zhu
- Department of Interventional Radiology, Department of Nuclear Medicine, Fuzhou University Affiliated Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, No. 134, Dongjie Road, Fuzhou, 350001, China
- School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, China
| | - Mingdian Yu
- Department of Interventional Radiology, Department of Nuclear Medicine, Fuzhou University Affiliated Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, No. 134, Dongjie Road, Fuzhou, 350001, China
| | - Jingqi Lv
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Fengwei Sun
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Achen Qin
- School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, China
| | - Zejing Chen
- School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, China
| | - Xiaoming Hu
- Department of Interventional Radiology, Department of Nuclear Medicine, Fuzhou University Affiliated Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, No. 134, Dongjie Road, Fuzhou, 350001, China.
- School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, China.
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China.
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China.
| | - Zhuting Fang
- Department of Interventional Radiology, Department of Nuclear Medicine, Fuzhou University Affiliated Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, No. 134, Dongjie Road, Fuzhou, 350001, China.
- Department of Oncology and Vascular Interventional Therapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, 350014, China.
| |
Collapse
|
31
|
Xiao Y, Tang Z, Zhang J, Saiding Q, Li Y, Du J, Tao W. One-Pot Synthesis of Fe-Norepinephrine Nanoparticles for Synergetic Thermal-Enhanced Chemodynamic Therapy. NANO LETTERS 2024; 24:13825-13833. [PMID: 39392201 DOI: 10.1021/acs.nanolett.4c04375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Chemodynamic therapy (CDT) is an innovative and burgeoning strategy that utilizes Fenton-Fenton-like chemistry and specific microenvironments to produce highly toxic hydroxyl radicals (•OH), with numerous methods emerging to refine this approach. Herein, we report a coordination compound, Fe-norepinephrine nanoparticles (Fe-NE NPs), via a one-pot synthesis. The Fe-NE NPs are based on ferrous ions (Fe2+) and norepinephrine, which are capable of efficient Fe2+/Fe3+ delivery. Once internalized by tumor cells, the released Fe2+/Fe3+ exerts the Fenton reaction to specifically produce toxic •OH. Moreover, the internal photothermal conversion ability of Fe-NE NPs allows us to simultaneously introduce light to trigger local heat generation and then largely improve the Fenton reaction efficiency, which enables a synergetic photothermal and chemodynamic therapy to realize satisfactory in vivo antitumor efficiency. This proof-of-concept work offers a promising approach to developing nanomaterials and refining strategies for enhanced CDT against tumors.
Collapse
Affiliation(s)
- Yufen Xiao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Zhongmin Tang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jiamin Zhang
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
32
|
Zheng L, Ding Y, Fang S, Yang W, Chen J, Ma J, Wang M, Wang J, Zhang F, Guo X, Zhang K, Shu GF, Weng Q, Wu F, Zhao Z, Chen M, Jiansong J. Potentiated Calcium Carbonate with Enhanced Calcium Overload Induction and Acid Neutralization Capabilities to Boost Chemoimmunotherapy against Liver Cancer. ACS NANO 2024; 18:27597-27616. [PMID: 39342637 DOI: 10.1021/acsnano.4c08690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Unfavorable phenotypes characterized by low immunogenicity and acidity within the tumor microenvironment (TME) contribute to immunosuppression and therapeutic resistance. Herein, we rationally synthesized a multifunctional nanoregulator by encapsulating DOX and erianin into calcium carbonate (CaCO3)-based nanoparticles using a modified double emulsion method. The DOX and erianin-loaded CaCO3-based nanoparticles, termed DECaNPs, could effectively induce the calcium overload by triggering calcium influx and absorbing CaCO3 nanoparticles. Additionally, DECaNPs also neutralize the acidic TME by interacting with extracellular protons and limiting lactic acid production, a result of metabolic remodeling in cancer cells. As a result, DECaNPs elicit cellular oxidative stress damage, which mediates the activation of ferroptosis/apoptosis hybrid pathways, and profound immunogenic cell death. Treatment with DECaNPs could inhibit the growth of tumors by promoting oxidative stress, acid neutralization, metabolic remodeling, and protective antitumor immunity in vivo. In addition, DECaNPs could synergistically amplify the antitumor effects of αPD-L1 in a bilateral tumor model by eliciting systemic immune responses. In all, our work presents the preparation of CaCO3-based nanoregulators designed to reverse the unfavorable TME and enhance αPD-L1 immunotherapy through multiple mechanisms.
Collapse
Affiliation(s)
- Liyun Zheng
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Yiming Ding
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Shiji Fang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Wenjing Yang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Jiale Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Ji Ma
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Mengyuan Wang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Jiaoli Wang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Feng Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Xiaoju Guo
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Kun Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Gao-Feng Shu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Qiaoyou Weng
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Fazong Wu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Ji Jiansong
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310000, China
| |
Collapse
|
33
|
Du C, Chen Z, Liu S, Liu J, Zhan J, Zou J, Liao J, Huang W, Lei Y. Lubricin-Inspired Nanozymes Reconstruct Cartilage Lubrication System with an "In-Out" Strategy. SMALL METHODS 2024; 8:e2400757. [PMID: 38962862 DOI: 10.1002/smtd.202400757] [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: 05/22/2024] [Revised: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Lubricin, secreted primarily by chondrocytes, plays a critical role in maintaining the function of the cartilage lubrication system. However, both external factors such as friction and internal factors like oxidative stress can disrupt this system, leading to osteoarthritis. Inspired by lubricin, a lubricating nanozyme, that is, Poly-2-acrylamide-2-methylpropanesulfonic acid sodium salt-grafted aminofullerene, is developed to restore the cartilage lubrication system using an "In-Out" strategy. The "Out" aspect involves reducing friction through a combination of hydration lubrication and ball-bearing lubrication. Simultaneously, the "In" aspect aims to mitigate oxidative stress by reducing free radical, increasing autophagy, and improving the mitochondrial respiratory chain. This results in reduced chondrocyte senescence and increased lubricin production, enhancing the natural lubrication ability of cartilage. Transcriptome sequencing and Western blot results demonstrate that it enhances the functionality of mitochondrial respiratory chain complexes I, III, and V, thereby improving mitochondrial function in chondrocytes. In vitro and in vivo experiments show that the lubricating nanozymes reduce cartilage wear, improve chondrocyte senescence, and mitigate oxidative stress damage, thereby mitigating the progression of osteoarthritis. These findings provide novel insights into treating diseases associated with oxidative stress and frictional damage, such as osteoarthritis, and set the stage for future research and development of therapeutic interventions.
Collapse
Affiliation(s)
- Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhuolin Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Senrui Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jingdi Zhan
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jing Zou
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Junyi Liao
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
34
|
Sun P, Gou H, Che X, Chen G, Feng C. Recent advances in DNAzymes for bioimaging, biosensing and cancer therapy. Chem Commun (Camb) 2024; 60:10805-10821. [PMID: 39248025 DOI: 10.1039/d4cc03774j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
DNAzymes, a class of single-stranded catalytic DNA with good stability, high catalytic activity, and easy synthesis, functionalization and modification properties, have garnered significant interest in the realm of biosensing and bioimaging. Their integration with fluorescent dyes or chemiluminescent moieties has led to remarkable bioimaging outcomes, while DNAzyme-based biosensors have demonstrated robust sensitivity and selectivity in detecting metal ions, nucleic acids, proteins, enzyme activities, exosomes, bacteria and microorganisms. In addition, by delivering DNAzymes into tumor cells, the mRNA therein can be cleaved to regulate the expression of corresponding proteins, which has further propelled the application of DNAzymes in cancer gene therapy and synergistic therapy. This paper reviews the strategies for screening attractive DNAzymes such as SELEX and high-throughput sequencing, and briefly describes the amplification strategies of DNAzymes, which mainly include catalytic hairpin assembly (CHA), DNA walker, hybridization chain reaction (HCR), DNA origami, CRISPR-Cas12a, rolling circle amplification (RCA), and aptamers. In addition, applications of DNAzymes in bioimaging, biosensing, and cancer therapy are also highlighted. Subsequently, the possible challenges of these DNAzymes in practical applications are further pointed out, and future research directions are suggested.
Collapse
Affiliation(s)
- Pei Sun
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Hongquan Gou
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, P. R. China
| | - Xinran Che
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Guifang Chen
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Chang Feng
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| |
Collapse
|
35
|
He Y, Wang Q, Zhang Q, Wang Y, Jiang Y, Zhao Q, Liu X, Wang F. A Methyl-Engineered DNAzyme for Endogenous Alkyltransferase Monitoring and Self-Sufficient Gene Regulation. SMALL METHODS 2024:e2401160. [PMID: 39295467 DOI: 10.1002/smtd.202401160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/30/2024] [Indexed: 09/21/2024]
Abstract
The on-demand gene regulation is crucial for extensively exploring specific gene functions and developing personalized gene therapeutics, which shows great promise in precision medicines. Although some nucleic acid-based gene regulatory tools (antisense oligonucleotides and small interfering RNAs) are devised for achieving on-demand activation, the introduction of chemical modifications may cause undesired side effects, thereby impairing the gene regulatory efficacy. Herein, a methyl-engineered DNAzyme (MeDz) is developed for the visualization of endogenous alkyltransferase (AGT) and the simultaneous self-sufficiently on-demand gene regulation. The catalytic activity of DNAzyme can be efficiently blocked by O6-methylguanine (O6MeG) modification and specifically restored via the AGT-mediated DNA-repairing pathway. This simply designed MeDz is demonstrated to reveal AGT of varying expression levels in different cells, opening the possibility to explore the AGT-related biological processes. Moreover, the AGT-guided MeDz exhibits cell-selective regulation on the human early growth response-1 (EGR-1) gene, with efficient gene repression in breast cancer cells and low effectiveness in normal cells. The proposed MeDz offers an attractive strategy for on-demand gene regulation, displaying great potential in biomedical applications.
Collapse
Affiliation(s)
- Yuqiu He
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Qing Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Qingqing Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yifei Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yuqian Jiang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Xiaoqing Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| |
Collapse
|
36
|
Qi G, Chen K, Guan W, Xie J, Chen X, Zhang G, Yan R, Yang G. One-Pot Synthesis of a pH-Sensitive MOF Integrated with Glucose Oxidase for Amplified Tumor Photodynamic/Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:49083-49091. [PMID: 39228328 DOI: 10.1021/acsami.4c10006] [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: 09/05/2024]
Abstract
Photothermal therapy (PTT) and photodynamic therapy (PDT) provide targeted approaches to cancer treatment, but each therapy has inherent limitations such as insufficient tissue penetration, uneven heat distribution, extreme hypoxia, and overexpressed HSP90 in tumor cells. To address these issues, herein, by encapsulating the IR780 dye and glucose oxidase (GOx) enzyme within ZIF-8 nanoparticles, we created a versatile system capable of combining photodynamic and enhanced photothermal therapy. The integration of the IR780 dye facilitated the generation of reactive oxygen species and hyperthermia upon light activation, enabling dual-mode cancer cell ablation. Moreover, GOx catalyzes the decomposition of glucose into gluconic acid and hydrogen peroxide, leading to the inhibition of ATP production and downregulation of heat shock protein 90 (HSP90) expression, sensitizing cancer cells to heat-induced cytotoxicity. This synergistic combination resulted in significantly improved therapeutic outcomes. Both in vitro and in vivo results validated that the nanoplatform demonstrated superior specificity and favorable therapeutic responses. Our innovative approach represents a promising strategy for overcoming current limitations in cancer treatments and offers the potential for clinical translation in the future.
Collapse
Affiliation(s)
- Guiqiang Qi
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Kang Chen
- Department of Gastroenterology of Southwest Hospital Army Medical University (Third Military Medical University), Chongqing 400038, P.R. China
| | - Wenhua Guan
- Linyi Hospital of Traditional Chinese Medicine, Linyi 276005, P. R. China
| | - Junyu Xie
- Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| | - Xiangyan Chen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, P.R. China
| | - Guanhua Zhang
- Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| | - Ran Yan
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, P.R. China
| | - Geng Yang
- Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, P. R. China
| |
Collapse
|
37
|
Chen M, Tong X, Sun Y, Dong C, Li C, Wang C, Zhang M, Wen Y, Ye P, Li R, Wan J, Liang S, Shi S. A ferroptosis amplifier based on triple-enhanced lipid peroxides accumulation strategy for effective pancreatic cancer therapy. Biomaterials 2024; 309:122574. [PMID: 38670032 DOI: 10.1016/j.biomaterials.2024.122574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/03/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
As an iron dependent regulatory cell death process driven by excessive lipid peroxides (LPO), ferroptosis is recognized as a powerful weapon for pancreatic cancer (PC) therapy. However, the tumor microenvironment (TME) with hypoxia and elevated glutathione (GSH) expression not only inhibits LPO production, but also induces glutathione peroxidase 4 (GPX4) mediated LPO clearance, which greatly compromise the therapeutic outcomes of ferroptosis. To address these issues, herein, a novel triple-enhanced ferroptosis amplifier (denoted as Zal@HM-PTBC) is rationally designed. After intravenous injection, the overexpressed H2O2/GSH in TME induces the collapse of Zal@HM-PTBC and triggers the production of oxygen and reactive oxygen species (ROS), which synergistically amplify the degree of lipid peroxidation (broaden sources). Concurrently, GSH consumption because of the degradation of the hollow manganese dioxide (HM) significantly weakens the activity of GPX4, resulting in a decrease in LPO clearance (reduce expenditure). Moreover, the loading and site-directed release of zalcitabine further promotes autophagy-dependent LPO accumulation (enhance effectiveness). Both in vitro and in vivo results validated that the ferroptosis amplifier demonstrated superior specificity and favorable therapeutic responses. Overall, this triple-enhanced LPO accumulation strategy demonstrates the ability to facilitate the efficacy of ferroptosis, injecting vigorous vitality into the treatment of PC.
Collapse
Affiliation(s)
- Mengyao Chen
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Xiaohan Tong
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Yanting Sun
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Chunyan Dong
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Chen Li
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Chunhui Wang
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Minyi Zhang
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Yixuan Wen
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Pinting Ye
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Ruihao Li
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Jie Wan
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China
| | - Shujing Liang
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China.
| | - Shuo Shi
- School of Chemical Science and Engineering, Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai, 200092, PR China.
| |
Collapse
|
38
|
Zhang J, Yang L, Zeng H, Zhao Z, Han Y, Zhao Y, Qu S, Gong Z, Wang Z, Bai Y, Zhao Q. Targeted Reprogramming of Pathogenic Fibroblast Genes at the 3'-Untranslated Regions by DNA Nanorobots for Periodontitis. ACS NANO 2024; 18:22139-22152. [PMID: 39110572 DOI: 10.1021/acsnano.4c05475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Periodontitis, with its persistent nature, causes significant distress for most sufferers. Current treatments, such as mechanical cleaning and surgery, often fail to fully address the underlying overactivation of fibroblasts that drives this degradation. Targeting the post-transcriptional regulation of fibroblasts, particularly at the 3'-untranslated regions (3'UTR) of pathogenic genes, offers a therapeutic strategy for periodontitis. Herein, we developed a DNA nanorobot for this purpose. This system uses a dynamic DNA nanoframework to incorporate therapeutic microRNAs through molecular recognition and covalent bonds, facilitated by DNA monomers modified with disulfide bonds. The assembled-DNA nanoframework is encapsulated in a cell membrane embedded with a fibroblast-targeting peptide. By analyzing the 3'UTR regions of pathogenic fibroblast genes FOSB and JUND, we identified the therapeutic microRNA as miR-1-3p and integrated it into this system. As expected, the DNA nanorobot delivered the internal components to fibroblasts by the targeting peptide and outer membrane that responsively releases miR-1-3p under intracellular glutathione. It resulted in a precise reduction of mRNA and suppression of protein function in pathogenic genes, effectively reprogramming fibroblast behavior. Our results confirm that this approach not only mitigates the inflammation but also promotes tissue regeneration in periodontal models, offering a promising therapeutic avenue for periodontitis.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Liu Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Hao Zeng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zifan Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yue Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yilong Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Shuyuan Qu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zijian Gong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Ziming Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yi Bai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qin Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
39
|
Zhang M, Shi F, Chen Y, Yang C, Zhang X, Deng C, Sun N. Straightforward Creation of Multishell Hollow Hybrids for an Integrated Metabolic Monitoring System in Disease Management. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400941. [PMID: 38529737 DOI: 10.1002/smll.202400941] [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: 02/05/2024] [Revised: 03/08/2024] [Indexed: 03/27/2024]
Abstract
Multidimensional metabolic analysis has become a new trend in establishing efficient disease monitoring systems, as the constraints associated with relying solely on a single dimension in refined monitoring are increasingly pronounced. Here, coordination polymers are employed as derivative precursors to create multishell hollow hybrids, developing an integrated metabolic monitoring system. Briefly, metabolic fingerprints are extracted from hundreds of serum samples and urine samples, encompassing not only membranous nephropathy but also related diseases, using high-throughput mass spectrometry. With optimized algorithm and initial feature selection, the established combined panel demonstrates enhanced accuracy in both subtype differentiation (over 98.1%) and prognostic monitoring (over 95.6%), even during double blind test. This surpasses the serum biomarker panel (≈90.7% for subtyping, ≈89.7% for prognosis) and urine biomarker panel (≈94.4% for subtyping, ≈76.5% for prognosis). Moreover, after attempting to further refine the marker panel, the blind test maintains equal sensitivity, specificity, and accuracy, showcasing a comprehensive improvement over the single-fluid approach. This underscores the remarkable effectiveness and superiority of the integrated strategy in discriminating between MN and other groups. This work has the potential to significantly advance diagnostic medicine, leading to the establishment of more effective strategies for patient management.
Collapse
Affiliation(s)
- Man Zhang
- Department of Chemistry, Department of Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Fangying Shi
- Department of Chemistry, Department of Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Yijie Chen
- Department of Chemistry, Department of Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Chenyu Yang
- Department of Chemistry, Department of Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Xiangmin Zhang
- Department of Chemistry, Department of Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Chunhui Deng
- Department of Chemistry, Department of Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
40
|
Cao Y, Xu R, Liang Y, Tan J, Guo X, Fang J, Wang S, Xu L. Nature-inspired protein mineralization strategies for nanoparticle construction: advancing effective cancer therapy. NANOSCALE 2024; 16:13718-13754. [PMID: 38954406 DOI: 10.1039/d4nr01536c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Recently, nanotechnology has shown great potential in the field of cancer therapy due to its ability to improve the stability and solubility and reduce side effects of drugs. The biomimetic mineralization strategy based on natural proteins and metal ions provides an innovative approach for the synthesis of nanoparticles. This strategy utilizes the unique properties of natural proteins and the mineralization ability of metal ions to combine nanoparticles through biomimetic mineralization processes, achieving the effective treatment of tumors. The precise control of the mineralization process between proteins and metal ions makes it possible to obtain nanoparticles with the ideal size, shape, and surface characteristics, thereby enhancing their stability and targeting ability in vivo. Herein, initially, we analyze the role of protein molecules in biomineralization and comprehensively review the functions, properties, and applications of various common proteins and metal particles. Subsequently, we systematically review and summarize the application directions of nanoparticles synthesized based on protein biomineralization in tumor treatment. Specifically, we discuss their use as efficient drug delivery carriers and role in mediating monotherapy and synergistic therapy using multiple modes. Also, we specifically review the application of nanomedicine constructed through biomimetic mineralization strategies using natural proteins and metal ions in improving the efficiency of tumor immunotherapy.
Collapse
Affiliation(s)
- Yuan Cao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Rui Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Yixia Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Jiabao Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Xiaotang Guo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Junyue Fang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Shibo Wang
- Institute of Smart Biomaterials, School of Materials Science and Engineering and Zhejiang Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Lei Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| |
Collapse
|
41
|
Xiang L, An Z, Wu X, Wang J, Cai S, Lu Y, Li L, Huang W, Wu D, Lu L, Shi S, Bi H, Kou X. Carbon Dot-Loaded Apoptotic Vesicles Improve the Liver Kupffer Cell-Mediated Antibacterial Effect to Synergistically Alleviate Sepsis. ACS NANO 2024; 18:16726-16742. [PMID: 38888383 DOI: 10.1021/acsnano.4c01780] [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: 06/20/2024]
Abstract
Sepsis is a lethal systemic inflammatory disease against infection that lacks effective therapeutic approaches. Liver resident macrophage Kupffer cell (KC)-initiated bacterial clearance is crucial for the host to defend against infection. However, it remains unclear whether this process also governs the antibacterial therapy of sepsis that would be used to improve therapeutic outcomes. Here, we found that copper-doped carbon dots (Cu-CDs) exhibited superior antibacterial capabilities in vitro but displayed limited therapeutic effects in septic mice due to their limited ability to target the liver and restore KC antimicrobial capacity. Thus, we developed a composite nanodrug of copper-doped carbon dot-loaded apoVs (CC-apoVs) that combined the antibacterial ability of Cu-CDs and liver KC targeting features of apoV. Moreover, intravenous injection of CC-apoVs markedly alleviated the systemic infection and decreased the mortality of septic mice compared to Cu-CD and apoV infusion alone. Mechanistically, CC-apoV injection rescued impaired liver KCs during sepsis and enhanced their ability to capture and kill bloodborne bacteria. In addition, apoV-promoted macrophage killing of bacteria could be blocked by the inhibition of small GTPase Rab5. This study reveals a liver KC-targeted therapeutic strategy for sepsis and provides a nanodrug CC-apoV to improve the host antibacterial defense and amplify the therapeutic effect of the nanodrug.
Collapse
Affiliation(s)
- Lei Xiang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Zhe An
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xiaoyan Wu
- School of Materials Science and Engineering, Anhui University, Hefei 230601, China
| | - Jinyang Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Simin Cai
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Yongxi Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Longchuang Li
- School of Materials Science and Engineering, Anhui University, Hefei 230601, China
| | - Weiying Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Di Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Lu Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Songtao Shi
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Hong Bi
- School of Materials Science and Engineering, Anhui University, Hefei 230601, China
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| |
Collapse
|
42
|
Li F, Yan J, Wei C, Zhao Y, Tang X, Xu L, He B, Sun Y, Chang J, Liang Y. "Cicada Out of the Shell" Deep Penetration and Blockage of the HSP90 Pathway by ROS-Responsive Supramolecular Gels to Augment Trimodal Synergistic Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401214. [PMID: 38647420 PMCID: PMC11220648 DOI: 10.1002/advs.202401214] [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: 02/01/2024] [Revised: 03/18/2024] [Indexed: 04/25/2024]
Abstract
Deep penetration and downregulation of heat shock protein (HSP) expression in multimodal synergistic therapy are promising approaches for curing cancer in clinical trials. However, free small-molecule drugs and most drug vehicles have a low delivery efficiency deep into the tumor owing to poor drug penetration and hypoxic conditions at the tumor site. In this study, the objective is to use reactive oxygen species (ROS)-responsive supramolecular gels co-loaded with the photosensitizer Zn(II) phthalocyanine tetrasulfonic acid (ZnPCS4) and functionalized tetrahedral DNA (TGSAs) (G@P/TGSAs) to enhance deep tissue and cell penetration and block the HSP90 pathway for chemo- photodynamic therapy (PDT) - photothermal therapy (PTT) trimodal synergistic therapy. The (G@P/TGSAs) are injected in situ into the tumor to release ZnPCS4 and TGSAs under high ROS concentrations originating from both the tumor and PDT. TGSAs penetrate deeply into tumor tissues and augment photothermal therapy by inhibiting the HSP90 pathway. Proteomics show that HSP-related proteins and molecular chaperones are inhibited/activated, inhibiting the HSP90 pathway. Simultaneously, the TGSA-regulated apoptotic pathway is activated. In vivo study demonstrates efficient tumor penetration and excellent trimodal synergistic therapy (45% tumor growth inhibition).
Collapse
Affiliation(s)
- Fashun Li
- Department of PharmaceuticsSchool of PharmacyQingdao UniversityQingdao266073China
| | - Jianqin Yan
- Department of PharmaceuticsSchool of PharmacyQingdao UniversityQingdao266073China
| | - Chen Wei
- Department of PharmacyQingdao Women and Children's HospitalQingdao266034China
| | - Yi Zhao
- Department of Recuperation MedicineQingdao Special Service Sanatorium of PLA NavyQingdao266071China
| | - Xiaowen Tang
- Department of Medicinal ChemistrySchool of PharmacyQingdao UniversityQingdao266073China
| | - Long Xu
- School of Materials Science and Chemical EngineeringNingbo UniversityNingbo315211China
| | - Bin He
- National Engineering Research Center for BiomaterialsSichuan UniversityChengdu610064China
| | - Yong Sun
- Department of PharmaceuticsSchool of PharmacyQingdao UniversityQingdao266073China
| | - Jing Chang
- College of Marine Life ScienceOcean University of ChinaQingdao266003China
| | - Yan Liang
- Department of PharmaceuticsSchool of PharmacyQingdao UniversityQingdao266073China
| |
Collapse
|
43
|
Nedelcu A, Mocan T, Sabau LI, Matea CT, Tabaran F, Pop T, Delcea C, Mosteanu O, Mocan L. In vitro photothermal therapy of pancreatic cancer mediated by immunoglobulin G-functionalized silver nanoparticles. Sci Rep 2024; 14:14417. [PMID: 38909066 PMCID: PMC11193743 DOI: 10.1038/s41598-024-63142-4] [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: 12/20/2023] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
Pancreatic cancer is one of the most aggressive forms of cancer, and treatment options are limited. One therapeutic approach is to use nanoparticles to deliver the active agent directly to pancreatic cancer cells. Nanoparticles can be designed to specifically target cancer cells, minimizing damage to healthy tissues. Silver nanoparticles have the unique ability to absorb light, especially in the near-infrared (NIR) region. In this study, silver nanoparticles functionalized with IgG molecules were synthesized and administered to pancreatic cancer cell lines. Subsequently, the cells were photo-excited using a 2 W 808 nm laser and further examined in PANC-1 pancreatic cancer cell lines. Flow cytometry and confocal microscopy combined with immunochemical staining were used to examine the interaction between photo-excited silver nanoparticles and pancreatic cancer cells. The photothermal therapy based on IgG-functionalized silver nanoparticles in pancreatic cancer induces dysfunction in the Golgi apparatus, leading to the activation of the caspase-3 apoptotic pathway and ultimately resulting in cellular apoptosis. These findings suggest that our proposed IgG nanoparticle laser treatment could emerge as a novel approach for the therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Andreea Nedelcu
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology, ClujNapoca, Romania
| | - Teodora Mocan
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology, ClujNapoca, Romania.
- Physiology Department, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania.
| | - Lavinia Ioana Sabau
- Physiology Department, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
| | - Cristian Tudor Matea
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology, ClujNapoca, Romania
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße 34, 5020, Salzburg, Austria
| | - Flaviu Tabaran
- Department of Pathology, Sciences and Veterinary Medicine, University of Agricultural, ClujNapoca, Romania
| | - Teodora Pop
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
- Department of Gastroenterology, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
| | - Cristian Delcea
- Department of Forensic Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania.
| | - Ofelia Mosteanu
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
- Department of Gastroenterology, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
| | - Lucian Mocan
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania.
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology, ClujNapoca, Romania.
| |
Collapse
|
44
|
Chang M, Zhang L, Zhang T, Duan Y, Feng W, Yang S, Chen Y, Wang Z. Ultrasound-augmented enzyodynamic-Ca 2+ overload synergetic tumor nanotherapy. Biomaterials 2024; 307:122513. [PMID: 38432005 DOI: 10.1016/j.biomaterials.2024.122513] [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: 11/25/2023] [Revised: 02/06/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
The excessive intracellular Ca2+ can induce oxidative stress, mitochondrial damage and cell apoptosis, which has been extensively explored for tumor therapy. However, the low Ca2+ accumulation originated from Ca2+-based nanosystems substantially weakens the therapeutic effect. Herein, a functional plant polyphenol-appended enzyodynamic nanozyme system CaFe2O4@BSA-curcumin (abbreviation as CFO-CUR) has been rationally designed and engineered to achieve magnified Ca2+ accumulation process, deleterious reactive oxygen species (ROS) production, as well as mitochondrial dysfunction through enzyodynamic-Ca2+ overload synergistic effect. The exogenous Ca2+ released by CaFe2O4 nanozymes under the weakly acidic tumor microenvironment and Ca2+ efflux inhibition by curcumin boost mitochondria-dominant antineoplastic efficiency. The presence of Fe components with multivalent characteristic depletes endogenous glutathione and outputs the incremental ROS due to the oxidase-, peroxidase-, glutathione peroxidase-mimicking activities. The ROS burst-triggered regulation of Ca2+ channels and pumps strengthens the intracellular Ca2+ accumulation. Especially, the exogenous ultrasound stimulation further amplifies mitochondrial damage. Both in vitro and in vivo experimental results affirm the ultrasound-augmented enzyodynamic-Ca2+ overload synergetic tumor inhibition outcomes. This study highlights the role of ultrasound coupled with functional nanozyme in the homeostasis imbalance and function disorder of mitochondria for highly efficient tumor treatment.
Collapse
Affiliation(s)
- Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, PR China
| | - Lu Zhang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071000, PR China
| | - Tingting Zhang
- Department of Ultrasound, The 985th Hospital of the Joint Logistics Support Force of the Chinese People's Liberation Army, Taiyuan, 030001, PR China; Department of Diving and Hyperbarie Medicine, Naval Medical Center (Naval Medical University), Shanghai, 200433, PR China.
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, PR China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Shaoling Yang
- Department of Ultrasound Medicine, Shanghai Eighth People's Hospital, Shanghai, 200235, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| | - Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
45
|
Jia J, Wang X, Lin X, Zhao Y. Engineered Microorganisms for Advancing Tumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313389. [PMID: 38485221 DOI: 10.1002/adma.202313389] [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: 12/09/2023] [Revised: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Engineered microorganisms have attracted significant interest as a unique therapeutic platform in tumor treatment. Compared with conventional cancer treatment strategies, engineering microorganism-based systems provide various distinct advantages, such as the intrinsic capability in targeting tumors, their inherent immunogenicity, in situ production of antitumor agents, and multiple synergistic functions to fight against tumors. Herein, the design, preparation, and application of the engineered microorganisms for advanced tumor therapy are thoroughly reviewed. This review presents a comprehensive survey of innovative tumor therapeutic strategies based on a series of representative engineered microorganisms, including bacteria, viruses, microalgae, and fungi. Specifically, it offers extensive analyses of the design principles, engineering strategies, and tumor therapeutic mechanisms, as well as the advantages and limitations of different engineered microorganism-based systems. Finally, the current challenges and future research prospects in this field, which can inspire new ideas for the design of creative tumor therapy paradigms utilizing engineered microorganisms and facilitate their clinical applications, are discussed.
Collapse
Affiliation(s)
- Jinxuan Jia
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiaocheng Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Xiang Lin
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Yuanjin Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| |
Collapse
|
46
|
Liu Y, Li J, Zhang Y, Wang F, Su J, Ma C, Zhang S, Du Y, Fan C, Zhang H, Liu K. Robotic Actuation-Mediated Quantitative Mechanogenetics for Noninvasive and On-Demand Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401611. [PMID: 38509850 PMCID: PMC11186056 DOI: 10.1002/advs.202401611] [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: 02/14/2024] [Revised: 02/29/2024] [Indexed: 03/22/2024]
Abstract
Cell mechanotransduction signals are important targets for physical therapy. However, current physiotherapy heavily relies on ultrasound, which is generated by high-power equipment or amplified by auxiliary drugs, potentially causing undesired side effects. To address current limitations, a robotic actuation-mediated therapy is developed that utilizes gentle mechanical loads to activate mechanosensitive ion channels. The resulting calcium influx precisely regulated the expression of recombinant tumor suppressor protein and death-associated protein kinase, leading to programmed apoptosis of cancer cell line through caspase-dependent pathway. In stark contrast to traditional gene therapy, the complete elimination of early- and middle-stage tumors (volume ≤ 100 mm3) and significant growth inhibition of late-stage tumor (500 mm3) are realized in tumor-bearing mice by transfecting mechanogenetic circuits and treating daily with quantitative robotic actuation in a form of 5 min treatment over the course of 14 days. Thus, this massage-derived therapy represents a quantitative strategy for cancer treatment.
Collapse
Affiliation(s)
- Yangyi Liu
- Center of Materials Science and Optoelectronics EngineeringCollege of Materials Science and Optoelectronic TechnologyUniversity of Chinese Academy of SciencesBeijing100049China
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education)Department of ChemistryTsinghua UniversityBeijing100084China
| | - Jingjing Li
- State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022China
| | - Yi Zhang
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education)Department of ChemistryTsinghua UniversityBeijing100084China
- State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022China
| | - Fan Wang
- State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022China
| | - Juanjuan Su
- Center of Materials Science and Optoelectronics EngineeringCollege of Materials Science and Optoelectronic TechnologyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Chao Ma
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education)Department of ChemistryTsinghua UniversityBeijing100084China
| | - Shuyi Zhang
- School of Pharmaceutical SciencesTsinghua UniversityBeijing100084China
| | - Yanan Du
- Department of Biomedical EngineeringSchool of MedicineTsinghua‐Peking Center for Life SciencesTsinghua UniversityBeijing100084China
| | - Chunhai Fan
- Xiangfu LaboratoryJiaxing314102China
- School of Chemistry and Chemical EngineeringNew Cornerstone Science LaboratoryFrontiers Science Center for Transformative MoleculesZhangjiang Institute for Advanced Study and National Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
| | - Hongjie Zhang
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education)Department of ChemistryTsinghua UniversityBeijing100084China
- State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022China
- Xiangfu LaboratoryJiaxing314102China
| | - Kai Liu
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education)Department of ChemistryTsinghua UniversityBeijing100084China
- State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchun130022China
- Xiangfu LaboratoryJiaxing314102China
| |
Collapse
|
47
|
Yan J, Bhadane R, Ran M, Ma X, Li Y, Zheng D, Salo-Ahen OMH, Zhang H. Development of Aptamer-DNAzyme based metal-nucleic acid frameworks for gastric cancer therapy. Nat Commun 2024; 15:3684. [PMID: 38693181 PMCID: PMC11063048 DOI: 10.1038/s41467-024-48149-9] [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: 08/22/2023] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
The metal-nucleic acid nanocomposites, first termed metal-nucleic acid frameworks (MNFs) in this work, show extraordinary potential as functional nanomaterials. However, thus far, realized MNFs face limitations including harsh synthesis conditions, instability, and non-targeting. Herein, we discover that longer oligonucleotides can enhance the synthesis efficiency and stability of MNFs by increasing oligonucleotide folding and entanglement probabilities during the reaction. Besides, longer oligonucleotides provide upgraded metal ions binding conditions, facilitating MNFs to load macromolecular protein drugs at room temperature. Furthermore, longer oligonucleotides facilitate functional expansion of nucleotide sequences, enabling disease-targeted MNFs. As a proof-of-concept, we build an interferon regulatory factor-1(IRF-1) loaded Ca2+/(aptamer-deoxyribozyme) MNF to target regulate glucose transporter (GLUT-1) expression in human epidermal growth factor receptor-2 (HER-2) positive gastric cancer cells. This MNF nanodevice disrupts GSH/ROS homeostasis, suppresses DNA repair, and augments ROS-mediated DNA damage therapy, with tumor inhibition rate up to 90%. Our work signifies a significant advancement towards an era of universal MNF application.
Collapse
Affiliation(s)
- Jiaqi Yan
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Rajendra Bhadane
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
- Structural Bioinformatics Laboratory, Biochemistry, Åbo Akademi University, 20520, Turku, Finland
| | - Meixin Ran
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Xiaodong Ma
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Yuanqiang Li
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Dongdong Zheng
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China
| | - Outi M H Salo-Ahen
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Structural Bioinformatics Laboratory, Biochemistry, Åbo Akademi University, 20520, Turku, Finland
| | - Hongbo Zhang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
| |
Collapse
|
48
|
Deng Y, Zhou T, Hu K, Peng Y, Jia X, Yang J, Li G. An electrochemical biosensor designed with entropy-driven autocatalytic DNA circuits for sensitive detection of ovarian cancer-derived exosomes. Biosens Bioelectron 2024; 250:116060. [PMID: 38278121 DOI: 10.1016/j.bios.2024.116060] [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: 12/03/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 01/28/2024]
Abstract
Intelligent artificial DNA circuits have emerged as a promising approach for modulating signaling pathways and signal transduction through rational design, which may contribute to comprehensively realizing biomolecular sensing of organisms. In this work, we have fabricated an electrochemical biosensor for the sensitive and accurate detection of ovarian cancer-derived exosomes by constructing an entropy-driven autocatalytic DNA circuit (EADC). Specifically, the robust EADC is prepared by the self-assembly of well-designed DNA probes, and upon stimulation of the presence of ovarian cancer cells-derived exosomes, numerous inputs can be produced to feedback and accelerate the reaction. The catalytic abilities of the generated input sequences play a pivotal role in EADC and dramatically enhance the signal amplification capability. Through the combination of the autocatalytic circuit and circular cleavage reactions, significantly changed electrochemical signals can be recorded for sensitive analysis of the exosomes with a remarkably low detection limit of 30 particles/μL. Moreover, the proposed enzyme-free biosensor shows exceptional performance in distinguishing patient samples from healthy samples, which exhibits promising prospects for the clinical diagnosis of ovarian cancer.
Collapse
Affiliation(s)
- Ying Deng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Tianci Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Kai Hu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, PR China
| | - Ying Peng
- School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang, 550025, PR China
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, PR China.
| | - Jie Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Genxi Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China; Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
49
|
Wang Y, Chen K, Liu G, Du C, Cheng Z, Wei D, Li F, Li C, Yang Y, Zhao Y, Nie G. Disruption of Super-Enhancers in Activated Pancreatic Stellate Cells Facilitates Chemotherapy and Immunotherapy in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308637. [PMID: 38417121 PMCID: PMC11040371 DOI: 10.1002/advs.202308637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/27/2024] [Indexed: 03/01/2024]
Abstract
One major obstacle in the drug treatment of pancreatic ductal adenocarcinoma (PDAC) is its highly fibrotic tumor microenvironment, which is replete with activated pancreatic stellate cells (a-PSCs). These a-PSCs generate abundant extracellular matrix and secrete various cytokines to form biophysical and biochemical barriers, impeding drug access to tumor tissues. Therefore, it is imperative to develop a strategy for reversing PSC activation and thereby removing the barriers to facilitate PDAC drug treatment. Herein, by integrating chromatin immunoprecipitation (ChIP)-seq, Assays for Transposase-Accessible Chromatin (ATAC)-seq, and RNA-seq techniques, this work reveals that super-enhancers (SEs) promote the expression of various genes involved in PSC activation. Disruption of SE-associated transcription with JQ1 reverses the activated phenotype of a-PSCs and decreases stromal fibrosis in both orthotopic and patient-derived xenograft (PDX) models. More importantly, disruption of SEs by JQ1 treatments promotes vascularization, facilitates drug delivery, and alters the immune landscape in PDAC, thereby improving the efficacies of both chemotherapy (with gemcitabine) and immunotherapy (with IL-12). In summary, this study not only elucidates the contribution of SEs of a-PSCs in shaping the PDAC tumor microenvironment but also highlights that targeting SEs in a-PSCs may become a gate-opening strategy that benefits PDAC drug therapy by removing stromal barriers.
Collapse
Affiliation(s)
- Yazhou Wang
- Pancreas CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210000China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Kai Chen
- Department of General SurgeryPeking University First HospitalBeijing100034China
| | - Gang Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of EducationNortheast Normal UniversityChangchun130024China
| | - Chong Du
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710061China
| | - Zhaoxia Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Dan Wei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Fenfen Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Chen Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Yinmo Yang
- Department of General SurgeryPeking University First HospitalBeijing100034China
| | - Ying Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| |
Collapse
|
50
|
Yu Y, Zhang F, Xiao W, Cheng Q, Li T, Tang J, Tao W, Mei L. Adaptive Design of Nanovesicles Overcoming Immunotherapeutic Limitations of Chemotherapeutic Drugs through Poliovirus Receptor Blockade. ACS NANO 2024. [PMID: 38324591 DOI: 10.1021/acsnano.3c13056] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Chemotherapy is currently a widely used treatment for cancer in clinical settings. Some chemotherapeutic drugs such as oxaliplatin (OXA) can cause tumor immunogenic cell death (ICD), activate immunity, and realize chemoimmunotherapy for tumors. However, the low degree of accumulation and immunosuppressive microenvironment in tumors limit the immunotherapeutic efficacy of these drugs. T cell immunoreceptor with Ig and ITIM domains (TIGIT)/poliovirus receptor (PVR) is an inhibitory immune checkpoint pathway involved in mediating natural killer (NK) cell and T cell exhaustion in tumors. TIGIT expression is up-regulated in NK cells and CD8+ T cells during tumor development. Moreover, we first found that tumors upregulated PVR expression after OXA treatment in previous work. Here, we systematically analyzed the effects of OXA on the TIGIT/PVR pathway, further proving the effectiveness of the combination of OXA and TIGIT/PVR blocking combination. We developed engineered TIGIT-expressing cell membrane nanovesicles loaded with OXA (OXA@TIGIT MVs) for synergistic cancer therapy. OXA@TIGIT showed good efficacy in several cancer models, leading to tumor regression, effectively inhibiting tumor growth and prolonging mouse survival. Furthermore, the OXA@TIGIT MVs activate a strong tumor-specific immune response in the body, providing long-term (more than 2 months) protection from tumor reactivation in the B16F10 melanoma rechallenge mouse model.
Collapse
Affiliation(s)
- Yongkang Yu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, PR China
| | - Fan Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Wenqing Xiao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Qinzhen Cheng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, PR China
| | - Tingxuan Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Jing Tang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, PR China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| |
Collapse
|