1
|
Zhang J, Gai T, Wang J, Wu Y, Zeng SM, Zhao D, Li W. Nucleic Acid-Locked Smart Carrier for Photothermal/Chemotherapy-Amplified Immunogenic Cell Death to Enhance Systemic Antitumor Efficacy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2503299. [PMID: 40184610 DOI: 10.1002/advs.202503299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/25/2025] [Indexed: 04/06/2025]
Abstract
Immunotherapy holds great promise in the fight against cancer; however, it often encounters poor immunogenicity with limited therapeutic efficacy. Combining multiple treatment modalities provides a trustworthy strategy for achieving a robust antitumor effect. In this study, a nucleic acid-locked smart carrier (NASC) is developed to amplify immunogenic cell death (ICD) through the synergistic integration of photothermal therapy (PTT) and chemotherapy for high-performance monotherapy. Mesoporous silica-coated gold nanorod (MSGNR) serves as the reservoir for anticancer drug doxorubicin (DOX) and is capped with a sequence-specific duplex unit containing a tumor-specific targeting AS1411 fragment, resulting in the formation of NASC. With AS1411 targeting, the NASC can specifically target and be internalized into tumor cells with high expression of nucleolin, where the duplex capping can be unlocked by the intracellularly overexpressed adenosine triphosphate. Subsequently, the released DOX synergized with MSGNR-mediated PTT following laser irradiation induces direct cell killing, which, concurrently, triggers ICD to activate antineoplastic immunity with an increased number of T lymphocytes. This triple-collaborative strategy, further combined with anti-programmed death-1 antibody (αPD-1)-mediated immune checkpoint blockade (ICB) therapy, shows a robust therapeutic efficacy in both unilateral and bilateral tumor models.
Collapse
Affiliation(s)
- Jiayang Zhang
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Tianyu Gai
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Jiwei Wang
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Yucai Wu
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Si-Ming Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Dongyuan Zhao
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Wei Li
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
2
|
Li S, Zhao Y, Lyu X, Chen Y, Zhang T, Lin S, Liu Z, Cai X, Tian T, Lin Y. Enzyme-Responsive Nanoparachute for Targeted miRNA Delivery: A Protective Strategy Against Acute Liver and Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411210. [PMID: 39717886 PMCID: PMC11905073 DOI: 10.1002/advs.202411210] [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: 09/12/2024] [Revised: 11/10/2024] [Indexed: 12/25/2024]
Abstract
MicroRNA (miRNA)-based therapy holds significant potential; however, its structural limitations pose a challenge to the full exploitation of its biomedical functionality. Framework nucleic acids are promising owing to their transportability, biocompatibility, and functional editability. MiRNA-125 is embedded into a nucleic acid framework to create an enzyme-responsive nanoparachute (NP), enhancing the miRNA loading capacity while preserving the attributes of small-scale framework nucleic acids and circumventing the uncertainty related to RNA exposure in conventional loading methods. An enzyme-sensitive sequence is designed in NP as a bioswitchable apparatus for cargo miRNAs release. NP is compared with conventional delivery modes and delivery vehicles, confirming its excellent transportability and sustained release properties. Moreover, NP confers good enzyme and serum resistance to the cargo miRNAs. Simultaneously, it can easily deliver miRNA-125 to liver and kidney lesions owing to its passive targeting properties. This allows for Keap1/Nrf2 pathway regulation and p53 protein targeting in the affected tissues. Additionally, NP negatively regulates the expression of Bax and Caspase-3. These combined actions help to inhibit oxidation, prevent cell cycle arrest, and reduce the apoptosis of liver and kidney cells. Consequently, this strategy offers a potential treatment for acute liver and kidney injury.
Collapse
Affiliation(s)
- Songhang Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Yuxuan Zhao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Xiaoying Lyu
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Ye Chen
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Tao Zhang
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Shiyu Lin
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Zhiqiang Liu
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Taoran Tian
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
- College of Biomedical EngineeringSichuan UniversityChengdu610041P. R. China
| |
Collapse
|
3
|
Long Q, Rabi K, Cai Y, Li L, Huang S, Qian B, Zhong Y, Qi Z, Zhang Y, Huang K, Wang X, Chang L, Xie W, Jiang H, Zhang H, Zhang J, Ren T, Wang Z, Teesalu T, Wu C, Lu L, Zhu Z, Chu Y, Santos HA, Liu Z, Zhao Q, Ye X. Identification of splenic IRF7 as a nanotherapy target for tele-conditioning myocardial reperfusion injury. Nat Commun 2025; 16:1909. [PMID: 39994192 PMCID: PMC11850716 DOI: 10.1038/s41467-025-57048-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
The sequestration of nanoparticles by mononuclear phagocyte system is a challenge for the use of nanotherapy for treating cardiovascular diseases due to the conventionally perceived loss of therapeutic potency. Here, we revitalize cardiovascular nanotherapy by unlocking an alternative route in which nanomedicines are redirected to the spleen, leveraging its potential as a highly efficient and targeted site for remote conditioning, or tele-conditioning myocardial reperfusion injury. The theoretical foundation underpinning is the splenogenic nature of recruited monocytes upon myocardial reperfusion in the acute stage, which is confirmed through murine heterotopic spleen transplantation. Single-cell RNA-seq analysis identifies IRF7 as a pivotal mediator in the spleen-heart communication network that is initially induced in the spleen and orchestrates functional changes in myocardial macrophages. Spleen-related induction of IRF7 is also valid in human myocardial reperfusion scenarios. In addition, in a murine preclinical model of male mice, temporal inhibition of splenic IRF7 through the designed spleen-targeting erythrosome engineered with the targeting peptide RP182, termed as STEER nanoparticles, mitigates the acute-stage innate immune responses and improves the cardiac function in the long term. In contrast, systemic inhibition, genetic knockout of IRF7 or absolute depletion of splenic monocytes does not have therapeutic benefits, indicating the superiority of nanoparticle-based targeted treatment. These findings establish the spleen as a naturally favored site for nanoparticle-based treatments, offering promising avenues for managing myocardial reperfusion injury.
Collapse
Affiliation(s)
- Qiang Long
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kristina Rabi
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Yu Cai
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Lihui Li
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shixing Huang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei Qian
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiming Zhong
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaoxi Qi
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yecen Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaichen Huang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinming Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lan Chang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weichang Xie
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huaiyu Jiang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haonan Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjie Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Ren
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zichen Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Lin Lu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengbin Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Biotherapy Research Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen, University of Groningen (UMCG), AV, Groningen, Netherlands
| | - Zehua Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen, University of Groningen (UMCG), AV, Groningen, Netherlands.
| | - Qiang Zhao
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaofeng Ye
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Li S, Gao H, Wang H, Zhao X, Pan D, Pacheco-Fernández I, Ma M, Liu J, Hirvonen J, Liu Z, Santos HA. Tailored polysaccharide entrapping metal-organic framework for RNAi therapeutics and diagnostics in atherosclerosis. Bioact Mater 2025; 43:376-391. [PMID: 39399834 PMCID: PMC11470791 DOI: 10.1016/j.bioactmat.2024.08.041] [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: 05/30/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024] Open
Abstract
Metal-organic frameworks (MOFs) hold promise as theranostic carriers for atherosclerosis. However, to further advance their therapeutic effects with higher complexity and functionality, integrating multiple components with complex synthesis procedures are usually involved. Here, we reported a facile and general strategy to prepare multifunctional anti-atherosclerosis theranostic platform in a single-step manner. A custom-designed multifunctional polymer, poly(butyl methacrylate-co-methacrylic acid) branched phosphorylated β-glucan (PBMMA-PG), can effectively entrap different MOFs via coordination, simultaneously endow the MOF with enhanced stability, lesional macrophages selectivity and enhanced endosome escape. Sequential ex situ characterization and computational studies elaborated the potential mechanism. This facile post-synthetic modification granted the administered nanoparticles atherosclerotic tropism by targeting Dectin-1+ macrophages, enhancing in situ MR signal intensity by 72 %. Delivery of siNLRP3 effectively mitigated NLRP3 inflammasomes activation, resulting a 43 % reduction of plaque area. Overall, the current study highlights a simple and general approach for fabricating a MOF-based theranostic platform towards atherosclerosis conditioning, which may also expand to other indications targeting the lesional macrophages.
Collapse
Affiliation(s)
- Sen Li
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Han Gao
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Haoji Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Xiaolin Zhao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Da Pan
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, 210009, PR China
| | - Idaira Pacheco-Fernández
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Ming Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Jianjun Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Zehua Liu
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Hélder A. Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
5
|
Nazli A, Irshad Khan MZ, Rácz Á, Béni S. Acid-sensitive prodrugs; a promising approach for site-specific and targeted drug release. Eur J Med Chem 2024; 276:116699. [PMID: 39089000 DOI: 10.1016/j.ejmech.2024.116699] [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: 02/26/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 08/03/2024]
Abstract
Drugs administered through conventional formulations are devoid of targeting and often spread to various undesired sites, leading to sub-lethal concentrations at the site of action and the emergence of undesired effects. Hence, therapeutic agents should be delivered in a controlled manner at target sites. Currently, stimuli-based drug delivery systems have demonstrated a remarkable potential for the site-specific delivery of therapeutic moieties. pH is one of the widely exploited stimuli for drug delivery as several pathogenic conditions such as tumor cells, infectious and inflammatory sites are characterized by a low pH environment. This review article aims to demonstrate various strategies employed in the design of acid-sensitive prodrugs, providing an overview of commercially available acid-sensitive prodrugs. Furthermore, we have compiled the progress made for the development of new acid-sensitive prodrugs currently undergoing clinical trials. These prodrugs include albumin-binding prodrugs (Aldoxorubicin and DK049), polymeric micelle (NC-6300), polymer conjugates (ProLindac™), and an immunoconjugate (IMMU-110). The article encompasses a broad spectrum of studies focused on the development of acid-sensitive prodrugs for anticancer, antibacterial, and anti-inflammatory agents. Finally, the challenges associated with the acid-sensitive prodrug strategy are discussed, along with future directions.
Collapse
Affiliation(s)
- Adila Nazli
- Department of Pharmacognosy, Semmelweis University, 1085, Budapest, Hungary.
| | | | - Ákos Rácz
- Department of Pharmacognosy, Semmelweis University, 1085, Budapest, Hungary.
| | - Szabolcs Béni
- Integrative Health and Environmental Analysis Research Laboratory, Department of Analytical Chemistry, Institute of Chemistry, Eötvös Loránd University, 1117, Budapest, Hungary.
| |
Collapse
|
6
|
Wei H, Yi K, Li F, Li D, Yang J, Shi R, Jin Y, Wang H, Ding J, Tao Y, Li M. Multimodal Tetrahedral DNA Nanoplatform for Surprisingly Rapid and Significant Treatment of Acute Liver Failure. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305826. [PMID: 37801371 DOI: 10.1002/adma.202305826] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/07/2023] [Indexed: 10/08/2023]
Abstract
Acute liver failure (ALF) is a life-threatening disease associated with the rapid development of inflammatory storms, level elevation of reactive oxygen species (ROS), and hepatocyte necrosis, which results in high short-term mortality. Except for liver transplantation, no effective strategies are available for ALF therapy due to the rapid disease progression and narrow window of therapeutic time. Therefore, there is an urgent demand to explore the fast and effective modalities for ALF treatment. Herein, a multifunctional tetrahedral DNA nanoplatform (TDN) is constructed by incorporating tumor necrosis factor-α siRNA (siTNF-α) through DNA hybridization and antioxidant manganese porphyrin (MnP4) via π-π stacking interaction with G-quadruplex (G4) for surprisingly rapid and significant ALF therapy. TDN-siTNF-α/-G4-MnP4 silences TNF-α of macrophages by siTNF-α and polarizes them to the anti-inflammatory M2 phenotype, providing appropriate microenvironments for hepatocyte viability. Additionally, TDN-siTNF-α/-G4-MnP4 scavenges intracellular ROS by MnP4, protecting hepatocytes from oxidative-stress-associated cell death. Furthermore, TDN itself promotes hepatocyte proliferation by modulating the cell cycle. TDN-siTNF-α/-G4-MnP4 shows almost complete liver accumulation after intravenous injection and exhibits excellent therapeutic efficacy of ALF within 2 h. The multifunctional DNA nanoformulation provides an effective strategy for rapid ALF therapy, expanding its application for innovative treatments of liver diseases.
Collapse
Affiliation(s)
- Hongyan Wei
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease, 600 Tianhe Road, Guangzhou, 510630, P. R. China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, 120 Longshan Road, Chongqing, 401147, P. R. China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| | - Fenfang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| | - Di Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Jiazhen Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, P. R. China
| | - Yuanyuan Jin
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 220 Handan Road, Shanghai, 200433, P. R. China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease, 600 Tianhe Road, Guangzhou, 510630, P. R. China
| |
Collapse
|
7
|
Luo R, Le H, Wu Q, Gong C. Nanoplatform-Based In Vivo Gene Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312153. [PMID: 38441386 DOI: 10.1002/smll.202312153] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Indexed: 07/26/2024]
Abstract
Gene therapy uses modern molecular biology methods to repair disease-causing genes. As a burgeoning therapeutic, it has been widely applied for cancer therapy. Since 1989, there have been numerous clinical gene therapy cases worldwide. However, a few are successful. The main challenge of clinical gene therapy is the lack of efficient and safe vectors. Although viral vectors show high transfection efficiency, their application is still limited by immune rejection and packaging capacity. Therefore, the development of non-viral vectors is overwhelming. Nanoplatform-based non-viral vectors become a hotspot in gene therapy. The reasons are mainly as follows. 1) Non-viral vectors can be engineered to be uptaken by specific types of cells or tissues, providing effective targeting capability. 2) Non-viral vectors can protect goods that need to be delivered from degradation. 3) Nanoparticles can transport large-sized cargo such as CRISPR/Cas9 plasmids and nucleoprotein complexes. 4) Nanoparticles are highly biosafe, and they are not mutagenic in themselves compared to viral vectors. 5) Nanoparticles are easy to scale preparation, which is conducive to clinical conversion and application. Here, an overview of the categories of nanoplatform-based non-viral gene vectors, the limitations on their development, and their applications in cancer therapy.
Collapse
Affiliation(s)
- Rui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Le
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
8
|
Rodimova S, Kozlov D, Krylov D, Mikhailova L, Kozlova V, Gavrina A, Mozherov A, Elagin V, Kuznetsova D. Nanoparticles for Creating a Strategy to Stimulate Liver Regeneration. Sovrem Tekhnologii Med 2024; 16:31-41. [PMID: 39650276 PMCID: PMC11618528 DOI: 10.17691/stm2024.16.3.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Indexed: 12/11/2024] Open
Abstract
Presently, there is a need in the developing new approaches to stimulate liver regeneration, which would make its recovery more effective after resection. Application of nanoparticles, loaded with small bioactive molecules, with their targeted delivery into the liver is a promising approach. The aim of the investigation is to study the interaction of nanoparticles with various types of hepatic cells on the models of liver slices and primary hepatic cell cultures using the methods of multiphoton microscopy with fluorescence lifetime imaging. Materials and Methods Nanoparticles have been synthetized from polylactide (PLA), gold (Au), and silicon (SiO2), and characterized using scanning and transmission electron microscopy. These types of particles were labeled with a fluorescent Cy5 dye for their visualization. Liver slices and a primary hepatocyte culture were used as models for biological testing of nanoparticles. Biodistribution of the nanoparticles in the tissue and cells, their cytotoxicity, and the effect on the cell metabolism were assessed using optical bioimaging methods. Results The silicon nanoparticles are accumulated mainly by macrophages, which generate reactive oxygen species in a large amount and impair the native metabolic state of hepatocytes. The gold nanoparticles accumulate in all types of the liver cells but possess a marked toxic effect, which is indicated by the appearance of necrotic and apoptotic cells and a sharp change in the hepatocyte metabolic state. The polylactide nanoparticles accumulate most effectively in the liver cells, preferably in hepatocytes, do not change their native metabolic state, making this type of nanoparticles most promising for creating the bioactive molecule delivery systems to stimulate liver regeneration.
Collapse
Affiliation(s)
- S.A. Rodimova
- Junior Researcher, Research Laboratory of Regenerative Medicine; Research Laboratory of Molecular Biotechnologies, Institute of Experimental Oncology and Biomedical Technologies; The Institute of Experimental Medicine, 12 Akademika Pavlova St., Saint Petersburg, 197376, Russia
| | - D.S. Kozlov
- Laboratory Assistant, Research Laboratory of Molecular Biotechnologies, Institute of Experimental Oncology and Biomedical Technologies; The Institute of Experimental Medicine, 12 Akademika Pavlova St., Saint Petersburg, 197376, Russia; Student; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| | - D.P. Krylov
- Laboratory Assistant, Research Laboratory of Molecular Biotechnologies, Institute of Experimental Oncology and Biomedical Technologies; The Institute of Experimental Medicine, 12 Akademika Pavlova St., Saint Petersburg, 197376, Russia Student; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| | - L.V. Mikhailova
- Engineer, Department of Physics; ITMO University (Saint Petersburg National Research University of Information Technologies, Mechanics and Optics), 49 Kronverksky Pr., Saint Petersburg, 197101, Russia
| | - V.A. Kozlova
- Student; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| | - A.I. Gavrina
- Junior Researcher, Research Laboratory of Molecular Biotechnologies, Institute of Experimental Oncology and Biomedical Technologies; The Institute of Experimental Medicine, 12 Akademika Pavlova St., Saint Petersburg, 197376, Russia
| | - A.M. Mozherov
- Junior Researcher, Research Laboratory of Optical Spectroscopy and Microscopy, Institute of Experimental Oncology and Biomedical Technologies; The Institute of Experimental Medicine, 12 Akademika Pavlova St., Saint Petersburg, 197376, Russia
| | - V.V. Elagin
- PhD, Researcher, Research Laboratory of Optical Spectroscopy and Microscopy; Research Laboratory of Molecular Biotechnologies, Institute of Experimental Oncology and Biomedical Technologies; The Institute of Experimental Medicine, 12 Akademika Pavlova St., Saint Petersburg, 197376, Russia
| | - D.S. Kuznetsova
- PhD, Head of the Research Laboratory of Molecular Biotechnologies, Institute of Experimental Oncology and Biomedical Technologies; The Institute of Experimental Medicine, 12 Akademika Pavlova St., Saint Petersburg, 197376, Russia
| |
Collapse
|
9
|
Gao H, Li S, Lan Z, Pan D, Naidu GS, Peer D, Ye C, Chen H, Ma M, Liu Z, Santos HA. Comparative optimization of polysaccharide-based nanoformulations for cardiac RNAi therapy. Nat Commun 2024; 15:5398. [PMID: 38926348 PMCID: PMC11208445 DOI: 10.1038/s41467-024-49804-x] [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: 10/25/2023] [Accepted: 06/19/2024] [Indexed: 06/28/2024] Open
Abstract
Ionotropic gelation is widely used to fabricate targeting nanoparticles (NPs) with polysaccharides, leveraging their recognition by specific lectins. Despite the fabrication scheme simply involves self-assembly of differently charged components in a straightforward manner, the identification of a potent combinatory formulation is usually limited by structural diversity in compound collections and trivial screen process, imposing crucial challenges for efficient formulation design and optimization. Herein, we report a diversity-oriented combinatory formulation screen scheme to identify potent gene delivery cargo in the context of precision cardiac therapy. Distinct categories of cationic compounds are tested to construct RNA delivery system with an ionic polysaccharide framework, utilizing a high-throughput microfluidics workstation coupled with streamlined NPs characterization system in an automatic, step-wise manner. Sequential computational aided interpretation provides insights in formulation optimization in a broader scenario, highlighting the usefulness of compound library diversity. As a result, the out-of-bag NPs, termed as GluCARDIA NPs, are utilized for loading therapeutic RNA to ameliorate cardiac reperfusion damages and promote the long-term prognosis. Overall, this work presents a generalizable formulation design strategy for polysaccharides, offering design principles for combinatory formulation screen and insights for efficient formulation identification and optimization.
Collapse
Affiliation(s)
- Han Gao
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), The Personalized Medicine Research Institute (PRECISION), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Sen Li
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Zhengyi Lan
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Da Pan
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Gonna Somu Naidu
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Chenyi Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Hangrong Chen
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Ming Ma
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China.
| | - Zehua Liu
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), The Personalized Medicine Research Institute (PRECISION), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands.
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland.
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), The Personalized Medicine Research Institute (PRECISION), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands.
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland.
| |
Collapse
|
10
|
Shen Y, Gwak H, Han B. Advanced manufacturing of nanoparticle formulations of drugs and biologics using microfluidics. Analyst 2024; 149:614-637. [PMID: 38083968 PMCID: PMC10842755 DOI: 10.1039/d3an01739g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Numerous innovative nanoparticle formulations of drugs and biologics, named nano-formulations, have been developed in the last two decades. However, methods for their scaled-up production are still lagging, as the amount needed for large animal tests and clinical trials is typically orders of magnitude larger. This manufacturing challenge poses a critical barrier to successfully translating various nano-formulations. This review focuses on how microfluidics technology has become a powerful tool to overcome this challenge by synthesizing various nano-formulations with improved particle properties and product purity in large quantities. This microfluidic-based manufacturing is enabled by microfluidic mixing, which is capable of the precise and continuous control of the synthesis of nano-formulations. We further discuss the specific applications of hydrodynamic flow focusing, a staggered herringbone micromixer, a T-junction mixer, a micro-droplet generator, and a glass capillary on various types of nano-formulations of polymeric, lipid, inorganic, and nanocrystals. Various separation and purification microfluidic methods to enhance the product purity are reviewed, including acoustofluidics, hydrodynamics, and dielectrophoresis. We further discuss the challenges of microfluidics being used by broader research and industrial communities. We also provide future outlooks of its enormous potential as a decentralized approach for manufacturing nano-formulations.
Collapse
Affiliation(s)
- Yingnan Shen
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Hogyeong Gwak
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA.
- Purdue University Institute for Cancer Research, West Lafayette, IN, 47907, USA
| |
Collapse
|
11
|
Prasad R, Selvaraj K. Effective Distribution of Gold Nanorods in Ordered Thick Mesoporous Silica: A Choice of Noninvasive Theranostics. ACS APPLIED MATERIALS & INTERFACES 2023; 15:47615-47627. [PMID: 37782885 DOI: 10.1021/acsami.3c06108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Porous silica coated gold nanorod core-shell structures demonstrate a multifunctional role in bioimaging, drug delivery, and cancer therapeutics applications. Here, we address a new approach for effective distribution of gold nanorods (GNRs) in a mesoporous silica (MS) shell, viz., one nanorod in one silica particle (GMS). We have studied that silica coating presents major advantages for the better biocompatibility and stability of GNRs. In this study, two different thicknesses of silica shell over GNRs have been discussed as per the application's need; GNRs in thin silica (11 nm) are fit for phototherapy and bioimaging, whereas thick and porous silica (51 nm) coated gold nanorods are suitable for triggered drug delivery and theranostics. However, effective distribution of GNRs in ordered architecture of thick mesoporous silica (MS, more than 50 nm thickness) with high surface area (more than 1000 m2/g) is not well understood so far. Here, we present methodical investigations for uniform and highly ordered mesoporous silica coating over GNRs with tunable thickness (6 to 51 nm). Judicious identification and optimization of different reaction parameters like concentrations of silica precursor (TEOS, 1.85-43.9 mM), template (CTAB, 0.9-5.7 mM), effect of temperature, pH (8.6-10.8), stirring speed (100-400 rpm), and, most importantly, the mode of addition of TEOS with GNRs have been discussed. Studies with thick, porous silica coated GNRs simplify the highest ever reported surface area (1100 m2/g) and cargo capacity (57%) with better product yield (g/batch). First and foremost, we report a highly scalable (more than 500 mL) and rapid direct deposition of an ordered MS shell around GNRs. These engineered core-shell nanoparticles demonstrate X-ray contrast property, synergistic photothermal-chemotherapeutics, and imaging of tumor cell (96% cell death) due to released fluorescent anticancer drug molecules and photothermal effect (52 °C) of embedded GNRs. A deeper insight into their influence on the architectural features and superior theranostics performances has been illustrated in detail. Hence, these findings indicate the potential impact of individual GMS for image guided combination therapeutics of cancer.
Collapse
Affiliation(s)
- Rajendra Prasad
- Nano and Computational Materials Lab, Catalysis and Inorganic Chemistry Division, CSIR National Chemical Laboratory, Pune 411008, India
- Interventional Theranostics & Multimode Imaging Lab, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Kaliaperumal Selvaraj
- Nano and Computational Materials Lab, Catalysis and Inorganic Chemistry Division, CSIR National Chemical Laboratory, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), (CSIR-HRDG) Campus, Postal Staff College area, Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
12
|
Lv L, Zhou X. Targeting Hippo signaling in cancer: novel perspectives and therapeutic potential. MedComm (Beijing) 2023; 4:e375. [PMID: 37799806 PMCID: PMC10547939 DOI: 10.1002/mco2.375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023] Open
Abstract
As highly conserved among diverse species, Hippo signaling pathway regulates various biological processes, including development, cell proliferation, stem cell function, tissue regeneration, homeostasis, and organ size. Studies in the last two decades have provided a good framework for how these fundamental functions of Hippo signaling are tightly regulated by a network with numerous intracellular and extracellular factors. The Hippo signaling pathway, when dysregulated, may lead to a wide variety of diseases, especially cancer. There is growing evidence demonstrating that dysregulated Hippo signaling is closely associated with tumorigenesis, cancer cell invasion, and migration, as well as drug resistance. Therefore, the Hippo pathway is considered an appealing therapeutic target for the treatment of cancer. Promising novel agents targeting the Hippo signaling pathway for cancers have recently emerged. These novel agents have shown antitumor activity in multiple cancer models and demonstrated therapeutic potential for cancer treatment. However, the detailed molecular basis of the Hippo signaling-driven tumor biology remains undefined. Our review summarizes current advances in understanding the mechanisms by which Hippo signaling drives tumorigenesis and confers drug resistance. We also propose strategies for future preclinical and clinical development to target this pathway.
Collapse
Affiliation(s)
- Liemei Lv
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongChina
| | - Xiangxiang Zhou
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongChina
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongChina
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
13
|
Wen X, Ou L, Cutshaw G, Uthaman S, Ou YC, Zhu T, Szakas S, Carney B, Houghton J, Gundlach-Graham A, Rafat M, Yang K, Bardhan R. Physicochemical Properties and Route of Systemic Delivery Control the In Vivo Dynamics and Breakdown of Radiolabeled Gold Nanostars. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204293. [PMID: 36965074 PMCID: PMC10518372 DOI: 10.1002/smll.202204293] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 02/22/2023] [Indexed: 06/18/2023]
Abstract
The in vivo dynamics of nanoparticles requires a mechanistic understanding of multiple factors. Here, for the first time, the surprising breakdown of functionalized gold nanostars (F-AuNSs) conjugated with antibodies and 64 Cu radiolabels in vivo and in artificial lysosomal fluid ex vivo, is shown. The short-term biodistribution of F-AuNSs is driven by the route of systemic delivery (intravenous vs intraperitoneal) and long-term fate is controlled by the tissue type in vivo. In vitro studies including endocytosis pathways, intracellular trafficking, and opsonization, are combined with in vivo studies integrating a milieu of spectroscopy and microcopy techniques that show F-AuNSs dynamics is driven by their physicochemical properties and route of delivery. F-AuNSs break down into sub-20 nm broken nanoparticles as early as 7 days postinjection. Martini coarse-grained simulations are performed to support the in vivo findings. Simulations suggest that shape, size, and charge of the broken nanoparticles, and composition of the lipid membrane depicting various tissues govern the interaction of the nanoparticles with the membrane, and the rate of translocation across the membrane to ultimately enable tissue clearance. The fundamental study addresses critical gaps in the knowledge regarding the fate of nanoparticles in vivo that remain a bottleneck in their clinical translation.
Collapse
Affiliation(s)
- Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, 37235, USA
- Nanovaccine Institute, Iowa State University, Ames, IA, 50012, USA
| | - Luping Ou
- Center for Soft Condensed Matter Physics and Interdisciplinary Research and School of Physical Science and Technology, Soochow University, Suzhou, 215006, China
| | - Gabriel Cutshaw
- Nanovaccine Institute, Iowa State University, Ames, IA, 50012, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50012, USA
| | - Saji Uthaman
- Nanovaccine Institute, Iowa State University, Ames, IA, 50012, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50012, USA
| | - Yu-Chuan Ou
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Tian Zhu
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Sarah Szakas
- Department of Chemistry, Iowa State University, Ames, IA, 50011, USA
| | - Brandon Carney
- Department of Radiology, Stony Brook University, Stony Brook, New York, NY, 11794, USA
| | - Jacob Houghton
- Department of Radiology, Stony Brook University, Stony Brook, New York, NY, 11794, USA
| | | | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research and School of Physical Science and Technology, Soochow University, Suzhou, 215006, China
| | - Rizia Bardhan
- Nanovaccine Institute, Iowa State University, Ames, IA, 50012, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50012, USA
| |
Collapse
|
14
|
Huang Z, Wang H, Chun C, Li X, Xu S, Zhao Y. Self-assembled FGF21 nanoparticles alleviate drug-induced acute liver injury. Front Pharmacol 2023; 13:1084799. [PMID: 36703750 PMCID: PMC9871310 DOI: 10.3389/fphar.2022.1084799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/23/2022] [Indexed: 01/12/2023] Open
Abstract
Acetaminophen (N-acetyl-p-aminophenol, APAP) is a common antipyretic agent and analgesic. An overdose of APAP can result in acute liver injury (ALI). Oxidative stress and inflammation are central to liver injury. N-acetylcysteine (NAC), a precursor of glutathione, is used commonly in clinical settings. However, the window of NAC treatment is limited, and more efficacious alternatives must be found. Endogenous cytokines such as fibroblast growth factor (FGF) 21 can improve mitochondrial function while decreasing intracellular oxidative stress and inflammatory responses, thereby exhibiting antioxidant-like effects. In this study, self-assembled nanoparticles comprising chitosan and heparin (CH) were developed to deliver FGF21 (CH-FGF21) to achieve the sustained release of FGF21 and optimize the in vivo distribution of FGF21. CH-FGF21 attenuated the oxidative damage and intracellular inflammation caused by APAP to hepatocytes effectively. In a murine model of APAP-induced hepatotoxicity, CH-FGF21 could alleviate ALI progression and promote the recovery of liver function. These findings demonstrated that a simple assembly of CH nanoparticles carrying FGF21 could be applied for the treatment of liver diseases.
Collapse
Affiliation(s)
- Zhiwei Huang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China,College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju, South Korea,*Correspondence: Zhiwei Huang, ; Shihao Xu, ; Yingzheng Zhao,
| | - Hengcai Wang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Changju Chun
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju, South Korea
| | - Xinze Li
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shihao Xu
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,*Correspondence: Zhiwei Huang, ; Shihao Xu, ; Yingzheng Zhao,
| | - Yingzheng Zhao
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China,*Correspondence: Zhiwei Huang, ; Shihao Xu, ; Yingzheng Zhao,
| |
Collapse
|
15
|
Wang J, Qin X, Kong B, Ren H. Celery-derived scaffolds with liver lobule-mimicking structures for tissue engineering transplantation. SMART MEDICINE 2022; 1:e20220002. [PMID: 39188745 PMCID: PMC11236025 DOI: 10.1002/smmd.20220002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/25/2022] [Indexed: 08/28/2024]
Abstract
Decellularized scaffolds have a demonstrated value in liver tissue engineering. Challenges in this area are focused on effectively eliminating the biological rejection of scaffolds and finding a suitable liver cell source. Here, inspired by the natural microstructure of hepatic lobules, we present a novel decellularized celery-derived scaffold cultured with human-induced pluripotent stem cell-derived hepatocytes (hiPSC-Heps) bioengineering liver tissue construction. Because of the natural hollow channels, interconnected porous structures, and excellent physicochemical characterization of the decellularized celery-derived scaffold, the resultant bioengineering liver tissue can maintain the hiPSC-Heps viability and the hepatic functions in the in vitro cultures. Based on this bioengineering liver tissue, we have demonstrated its good biocompatibility and the significantly higher expressions of albumin (ALB) and periodic acid-schiff stain (PAS) when it was implanted in nude mice. These remarkable properties endow the hiPSC-Heps integrated decellularized celery scaffolds system with promising prospects in the field of liver transplantation and other regeneration medicine.
Collapse
Affiliation(s)
- Jinglin Wang
- Department of Hepatobiliary SurgeryNanjing Drum Tower HospitalClinical College of Traditional Chinese and Western MedicineSchool of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Xueqian Qin
- Department of Hepatobiliary SurgeryNanjing Drum Tower HospitalClinical College of Traditional Chinese and Western MedicineSchool of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Bin Kong
- Department of Hepatobiliary SurgeryNanjing Drum Tower HospitalClinical College of Traditional Chinese and Western MedicineSchool of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Haozhen Ren
- Department of Hepatobiliary SurgeryNanjing Drum Tower HospitalClinical College of Traditional Chinese and Western MedicineSchool of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| |
Collapse
|
16
|
Fu M, Hu Y, Lan T, Guan KL, Luo T, Luo M. The Hippo signalling pathway and its implications in human health and diseases. Signal Transduct Target Ther 2022; 7:376. [PMID: 36347846 PMCID: PMC9643504 DOI: 10.1038/s41392-022-01191-9] [Citation(s) in RCA: 257] [Impact Index Per Article: 85.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 11/11/2022] Open
Abstract
As an evolutionarily conserved signalling network, the Hippo pathway plays a crucial role in the regulation of numerous biological processes. Thus, substantial efforts have been made to understand the upstream signals that influence the activity of the Hippo pathway, as well as its physiological functions, such as cell proliferation and differentiation, organ growth, embryogenesis, and tissue regeneration/wound healing. However, dysregulation of the Hippo pathway can cause a variety of diseases, including cancer, eye diseases, cardiac diseases, pulmonary diseases, renal diseases, hepatic diseases, and immune dysfunction. Therefore, therapeutic strategies that target dysregulated Hippo components might be promising approaches for the treatment of a wide spectrum of diseases. Here, we review the key components and upstream signals of the Hippo pathway, as well as the critical physiological functions controlled by the Hippo pathway. Additionally, diseases associated with alterations in the Hippo pathway and potential therapies targeting Hippo components will be discussed.
Collapse
Affiliation(s)
- Minyang Fu
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China
| | - Yuan Hu
- Department of Pediatric Nephrology Nursing, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, China
| | - Tianxia Lan
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Ting Luo
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China.
| | - Min Luo
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China.
| |
Collapse
|
17
|
Gonçalves AI, Gomes ME. Outlook in Tissue Engineered Magnetic Systems and Biomagnetic Control. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
18
|
Liu C, Yan P, Xu X, Zhou W, Prakash DR, Wang S, Zhou J, Wang R, Huang H, Chen J, Zhang H, Shen J. In Vivo Kidney Allograft Endothelial Specific Scavengers for On-Site Inflammation Reduction under Antibody-Mediated Rejection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106746. [PMID: 35235710 DOI: 10.1002/smll.202106746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/21/2022] [Indexed: 06/14/2023]
Abstract
Kidney transplantation is the most effective therapy for patients with end-stage renal disease. However, antibody-mediated rejection (ABMR) threatens long-term survival of renal grafts. Although ABMR can be controlled by donor-specific antibody clearance and B- or (and) plasma-cells inhibition, the treatment often causes severe side effects in patients. Therefore, there is need to explore site-specific scavengers. In this study, a nanovehicle carrying an anti-inflammatory drug is developed with complement component 4d targeting, a specific biomarker expressed on allograft endothelium under ABMR. Moreover, the nanovehicle is endowed with photothermal properties to control drug release. Analysis through systematic in vitro and in vivo toxicity, non-invasive targeted imaging, and in situ remote controlled drug release show the nanovehicle specifically targets allograft kidney endothelium, releases an anti-inflammatory drug, methylprednisolone, locally upon laser irradiation, and promotes recovery of injured endothelium, without affecting systemic inflammation or innate immune responses. This strategy has the potential for future clinical application in ABMR treatment.
Collapse
Affiliation(s)
- Chang Liu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | - Pengpeng Yan
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xiaoyu Xu
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200031, China
| | - Wenhui Zhou
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | | | - Shuqi Wang
- Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Junnian Zhou
- Experimental Hematology and Biochemistry Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Rending Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Hongfeng Huang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
- 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, Shanghai, 200025, China
| | - Jia Shen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
19
|
Formulation attributes, acid tunable degradability and cellular interaction of acetalated maltodextrin nanoparticles. Carbohydr Polym 2022; 288:119378. [DOI: 10.1016/j.carbpol.2022.119378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 01/06/2023]
|
20
|
Gupta A, Sood A, Fuhrer E, Djanashvili K, Agrawal G. Polysaccharide-Based Theranostic Systems for Combined Imaging and Cancer Therapy: Recent Advances and Challenges. ACS Biomater Sci Eng 2022; 8:2281-2306. [PMID: 35513349 DOI: 10.1021/acsbiomaterials.1c01631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Designing novel systems for efficient cancer treatment and improving the quality of life for patients is a prime requirement in the healthcare sector. In this regard, theranostics have recently emerged as a unique platform, which combines the benefits of both diagnosis and therapeutics delivery. Theranostics have the desired contrast agent and the drugs combined in a single carrier, thus providing the opportunity for real-time imaging to monitor the therapy results. This helps in reducing the hazards related to treatment overdose or underdose and gives the possibility of personalized therapy. Polysaccharides, as natural biomolecules, have been widely explored to develop theranostics, as they act as a matrix for simultaneously loading both contrast agents and drugs for their utility in drug delivery and imaging. Additionally, their remarkable physicochemical attributes (biodegradability, satisfactory safety profile, abundance, and diversity in functionality and charge) can be tuned via postmodification, which offers numerous possibilities to develop theranostics with desired characteristics. Hence, we provide an overview of recent advances in polysaccharide matrix-based theranostics for drug delivery combined with magnetic resonance imaging, computed tomography, positron emission tomography, single photon emission computed tomography, and ultrasound imaging. Herein, we also summarize the toxicity assessment of polysaccharides, associated contrast agents, and nanotoxicity along with the challenges and future research directions.
Collapse
Affiliation(s)
- Aastha Gupta
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh-175075, India
| | - Ankur Sood
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh-175075, India
| | - Erwin Fuhrer
- School of Computing and Electrical Engineering, Indian Institute of Technology Mandi, Himachal Pradesh-175075, India
| | - Kristina Djanashvili
- Department of Biotechnology, Delft University of Technology, Van der Maasweg 9, 2629 HZ Delft, The Netherlands
| | - Garima Agrawal
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh-175075, India
| |
Collapse
|
21
|
Ju Y, Liao H, Richardson JJ, Guo J, Caruso F. Nanostructured particles assembled from natural building blocks for advanced therapies. Chem Soc Rev 2022; 51:4287-4336. [PMID: 35471996 DOI: 10.1039/d1cs00343g] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Advanced treatments based on immune system manipulation, gene transcription and regulation, specific organ and cell targeting, and/or photon energy conversion have emerged as promising therapeutic strategies against a range of challenging diseases. Naturally derived macromolecules (e.g., proteins, lipids, polysaccharides, and polyphenols) have increasingly found use as fundamental building blocks for nanostructured particles as their advantageous properties, including biocompatibility, biodegradability, inherent bioactivity, and diverse chemical properties make them suitable for advanced therapeutic applications. This review provides a timely and comprehensive summary of the use of a broad range of natural building blocks in the rapidly developing field of advanced therapeutics with insights specific to nanostructured particles. We focus on an up-to-date overview of the assembly of nanostructured particles using natural building blocks and summarize their key scientific and preclinical milestones for advanced therapies, including adoptive cell therapy, immunotherapy, gene therapy, active targeted drug delivery, photoacoustic therapy and imaging, photothermal therapy, and combinational therapy. A cross-comparison of the advantages and disadvantages of different natural building blocks are highlighted to elucidate the key design principles for such bio-derived nanoparticles toward improving their performance and adoption. Current challenges and future research directions are also discussed, which will accelerate our understanding of designing, engineering, and applying nanostructured particles for advanced therapies.
Collapse
Affiliation(s)
- Yi Ju
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia. .,School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Haotian Liao
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China. .,Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Sichuan 610065, China
| | - Joseph J Richardson
- Department of Materials Engineering, University of Tokyo, 7-3-1 Bunkyo-ku, Tokyo 113-8656, Japan
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China. .,State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan 610065, China. .,Bioproducts Institute, Departments of Chemical and Biological Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
22
|
Tran VA, Vo GV, Tan MA, Park JS, An SSA, Lee SW. Dual Stimuli-Responsive Multifunctional Silicon Nanocarriers for Specifically Targeting Mitochondria in Human Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14040858. [PMID: 35456692 PMCID: PMC9028052 DOI: 10.3390/pharmaceutics14040858] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 03/30/2022] [Accepted: 04/11/2022] [Indexed: 01/16/2023] Open
Abstract
Specific targeting, selective stimuli-responsiveness, and controlled release of anticancer agents are requested for high therapeutic efficiency with a minimal adverse effect. Herein, we report the sophisticated synthesis and functionalization of fluorescent mesoporous silicon (FMPSi) nanoparticles decorated with graphene oxide (GO) nanosheets. GO-wrapped FMPSi (FMPSi@GO) was loaded with a cisplatin (Cis) anticancer agent, and Cis-loaded FMPSi@GO (FMPSi-Cis@GO) exhibited the dual stimuli (pH and NIR)-responsiveness of controlled drug release, i.e., the drug release rate was distinctly enhanced at acidic pH 5.5 than at neutral pH 7.0 and further enhanced under NIR irradiation at acidic pH condition. Notably, dequalinium-conjugated FMPSi-Cis@GO (FMPSi-Cis@GO@DQA) demonstrated an excellent specificity for mitochondrial targeting in cancer cells without noticeable toxicity to normal human cells. Our novel silicon nanocarriers demonstrated not only stimuli (pH and NIR)-responsive controlled drug release, but also selective accumulation in the mitochondria of cancer cells and destroying them.
Collapse
Affiliation(s)
- Vy Anh Tran
- Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnam-daero, Sujung-gu, Seongnam-si 461-701, Gyeonggi-do, Korea;
| | - Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City 700000, Vietnam;
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City 700000, Vietnam
| | - Mario A. Tan
- College of Science and Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila 1015, Philippines;
| | - Joon-Seo Park
- Department of Chemistry, Eastern University, 1300 Eagle Road, St. Davids, PA 19087, USA;
| | - Seong Soo A. An
- Department of Bionano Technology, Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujung-gu, Seongnam-si 461-701, Gyeonggi-do, Korea
- Correspondence: (S.S.A.A.); (S.-W.L.); Tel.: +82-31-750-8755 (S.S.A.A.); +82-31-750-5360 (S.-W.L.)
| | - Sang-Wha Lee
- Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnam-daero, Sujung-gu, Seongnam-si 461-701, Gyeonggi-do, Korea;
- Correspondence: (S.S.A.A.); (S.-W.L.); Tel.: +82-31-750-8755 (S.S.A.A.); +82-31-750-5360 (S.-W.L.)
| |
Collapse
|
23
|
Chen Y, Shi S, Li B, Lan T, Yuan K, Yuan J, Zhou Y, Song J, Lv T, Shi Y, Xiang B, Tian T, Zhang T, Yang J, Lin Y. Therapeutic Effects of Self-Assembled Tetrahedral Framework Nucleic Acids on Liver Regeneration in Acute Liver Failure. ACS APPLIED MATERIALS & INTERFACES 2022; 14:13136-13146. [PMID: 35285610 DOI: 10.1021/acsami.2c02523] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Liver failure is a serious disease that is characterized by global hepatocyte necrosis. Hepatocyte proliferation and liver regeneration are critically important for the success of treatments for liver disease. Tetrahedral framework nucleic acids (TFNAs), which are widely used antioxidants and anti-inflammatory nanomaterials, activate multiple proliferation and prosurvival pathways. Therefore, the effects of a TFNA on hepatocyte proliferation and liver regeneration in mouse livers injured by 70% partial hepatectomy (PHx), acetaminophen overdose, and carbon tetrachloride were explored in this study. The TFNA, which was successfully self-assembled from four specifically designed ssDNAs, entered the body quickly and was taken up effectively by hepatocytes in the liver and could eventually be cleared by the kidneys. The TFNA promoted hepatocyte proliferation in vitro by activating the Notch and Wnt signaling pathways. In the three in vivo mouse models of liver injury, the TFNA attenuated the injuries and enhanced liver regeneration by regulating the cell cycle and the P53 signaling pathway. Therefore, by promoting hepatocyte proliferation and enhancing liver regeneration, the TFNA shows potential as an effective therapeutic agent for treating acute liver injury induced by 70% PHx and other factors, thereby preventing the progression to acute liver failure and reducing the associated mortality rate.
Collapse
Affiliation(s)
- Yang Chen
- Department of Liver Surgery& Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Department of Pediatric Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bo Li
- Department of Liver Surgery& Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Tian Lan
- Department of Liver Surgery& Liver Transplantation Center, Laboratory of Liver Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Kefei Yuan
- Department of Liver Surgery& Liver Transplantation Center, Laboratory of Liver Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingsheng Yuan
- Department of Liver Surgery& Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Yongjie Zhou
- Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Laboratory of Pathology, Key Laboratory of Transplant Engineering, and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiulin Song
- Department of Liver Surgery& Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Department of Pediatric Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Tao Lv
- Department of Liver Surgery& Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Department of Pediatric Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Yujun Shi
- Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- Laboratory of Pathology, Key Laboratory of Transplant Engineering, and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bo Xiang
- Department of Pediatric Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiayin Yang
- Department of Liver Surgery& Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
24
|
Lee J, Byun J, Shim G, Oh YK. Fibroblast activation protein activated antifibrotic peptide delivery attenuates fibrosis in mouse models of liver fibrosis. Nat Commun 2022; 13:1516. [PMID: 35314685 PMCID: PMC8938482 DOI: 10.1038/s41467-022-29186-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/04/2022] [Indexed: 02/07/2023] Open
Abstract
In liver fibrosis, activated hepatic stellate cells are known to overexpress fibroblast activation protein. Here we report a targeted antifibrotic peptide-delivery system in which fibroblast activation protein, which is overexpressed in fibrotic regions of the liver, liberates the antifibrotic peptide melittin by cleaving a fibroblast activation protein-specific site in the peptide. The promelittin peptide is linked to pegylated and maleimide-functionalized liposomes, resulting in promelittin-modified liposomes. The promelittin-modified liposomes were effective in reducing the viability of activated hepatic stellate cells but not that of control cells. In three types of liver fibrosis mouse models, intravenously administered promelittin-modified liposomes significantly reduces fibrotic regions. In addition, in the bile duct ligation mouse model promelittin-modified liposome-treatment increases overall survival. Although this peptide-delivery concept was tested for liver fibrosis, it can potentially be adapted to other fibrotic diseases. Activated hepatic stellate cells contribute towards the pathogenesis of liver fibrosis, and overexpress fibroblast activation protein. Here the authors report a targeted peptide-delivery system in which fibroblast activation protein liberates the antifibrotic peptide melittin, and demonstrate the approach attenuates fibrosis in mouse models of liver fibrosis.
Collapse
|
25
|
Li F, Zhang J, Yi K, Wang H, Wei H, Chan HF, Tao Y, Li M. Delivery of Stem Cell Secretome for Therapeutic Applications. ACS APPLIED BIO MATERIALS 2022; 5:2009-2030. [PMID: 35285638 DOI: 10.1021/acsabm.1c01312] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Intensive studies on stem cell therapy reveal that benefits of stem cells attribute to the paracrine effects. Hence, direct delivery of stem cell secretome to the injured site shows the comparative therapeutic efficacy of living cells while avoiding the potential limitations. However, conventional systemic administration of stem cell secretome often leads to rapid clearance in vivo. Therefore, a variety of different biomaterials are developed for sustained and controllable delivery of stem cell secretome to improve therapeutic efficiency. In this review, we first introduce current approaches for the preparation and characterization of stem cell secretome as well as strategies to improve their therapeutic efficacy and production. The up-to-date delivery platforms are also summarized, including nanoparticles, injectable hydrogels, microneedles, and scaffold patches. Meanwhile, we discuss the underlying therapeutic mechanism of stem cell secretome for the treatment of various diseases. In the end, future opportunities and challenges are proposed.
Collapse
Affiliation(s)
- Fenfang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Jiabin Zhang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hongyan Wei
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Liver Disease, Guangzhou 510630, China
| |
Collapse
|
26
|
Zhu L, Shao C, Chen H, Chen Z, Zhao Y. Hierarchical Hydrogels with Ordered Micro-Nano Structures for Cancer-on-a-Chip Construction. RESEARCH (WASHINGTON, D.C.) 2021; 2021:9845679. [PMID: 35028584 PMCID: PMC8724685 DOI: 10.34133/2021/9845679] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/25/2021] [Indexed: 06/01/2023]
Abstract
In the drug therapy of tumor, efficient and stable drug screening platforms are required since the drug efficacy varies individually. Here, inspired by the microstructures of hepatic lobules, in which hepatocytes obtain nutrients from both capillary vessel and the central vein, we present a novel hierarchical hydrogel system with ordered micro-nano structure for liver cancer-on-a-chip construction and drug screening. The hierarchical hydrogel system was fabricated by using pregel to fill and replicate self-assembled colloidal crystal arrays and microcolumn array template. Due to the synergistic effect of its interconnected micro-nano structures, the resultant system could not only precisely control the size of cell spheroids but also realize adequate nutrient supply of cell spheroids. We have demonstrated that by integrating the hierarchical hydrogel system into a multichannel concentration gradients microfluidic chip, a functional liver cancer-on-a-chip could be constructed for high-throughput drug screening with good repeatability and high accuracy. These results indicated that the hierarchical hydrogel system and its derived liver cancer-on-a-chip are ideal platforms for drug screening and have great application potential in the field of personalized medicine.
Collapse
Affiliation(s)
- Luyao Zhu
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Changmin Shao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325001, China
| | - Hanxu Chen
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhuoyue Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yuanjin Zhao
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| |
Collapse
|
27
|
miR122-controlled all-in-one nanoplatform for in situ theranostic of drug-induced liver injury by visualization imaging guided on-demand drug release. Mater Today Bio 2021; 12:100157. [PMID: 34825161 PMCID: PMC8604687 DOI: 10.1016/j.mtbio.2021.100157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 01/10/2023] Open
Abstract
Drug-induced liver injury (DILI) is a challenging clinical problem with respect to both diagnosis and management. As a newly emerging biomarker of liver injury, miR122 shows great potential in early and sensitive in situ detection of DILI. Glycyrrhetinic acid (GA) possesses desirable therapeutic effect on DILI, but its certain dose-dependent side effects after long-term and/or high-dose administration limit its clinical application. In this study, in order to improve the precise diagnosis and effective treatment of DILI, GA loaded all-in-one theranostic nanoplatform was designed by assembling of upconversion nanoparticles and gold nanocages. As a proof of concept, we demonstrated the applicability of this single-wavelength laser-triggered theranostic nanoplatform for the spatiotemporally controllable in situ imaging of DILI and miR122-controlled on-demand drug release in vitro and in vivo. This novel nanoplatform opens a promising avenue for the clinical diagnosis and treatment of DILI.
Collapse
|
28
|
Liu Z, Wang S, Tapeinos C, Torrieri G, Känkänen V, El-Sayed N, Python A, Hirvonen JT, Santos HA. Non-viral nanoparticles for RNA interference: Principles of design and practical guidelines. Adv Drug Deliv Rev 2021; 174:576-612. [PMID: 34019958 DOI: 10.1016/j.addr.2021.05.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/04/2021] [Accepted: 05/15/2021] [Indexed: 02/08/2023]
Abstract
Ribonucleic acid interference (RNAi) is an innovative treatment strategy for a myriad of indications. Non-viral synthetic nanoparticles (NPs) have drawn extensive attention as vectors for RNAi due to their potential advantages, including improved safety, high delivery efficiency and economic feasibility. However, the complex natural process of RNAi and the susceptible nature of oligonucleotides render the NPs subject to particular design principles and requirements for practical fabrication. Here, we summarize the requirements and obstacles for fabricating non-viral nano-vectors for efficient RNAi. To address the delivery challenges, we discuss practical guidelines for materials selection and NP synthesis in order to maximize RNA encapsulation efficiency and protection against degradation, and to facilitate the cytosolic release of oligonucleotides. The current status of clinical translation of RNAi-based therapies and further perspectives for reducing the potential side effects are also reviewed.
Collapse
|
29
|
Kim S, Han G, Hwang D, Won D, Shin Y, Kim C, Kang JM, Park J, Jung H, Park W, Yun J. Design and Usability Evaluations of a 3D-Printed Implantable Drug Delivery Device for Acute Liver Failure in Preclinical Settings. Adv Healthc Mater 2021; 10:e2100497. [PMID: 34160141 DOI: 10.1002/adhm.202100497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/18/2021] [Indexed: 12/11/2022]
Abstract
Acute liver failure (ALF) requiring liver transplantation is a disease that occurs due to rapid hepatocellular dysfunction. As liver transplantation has various limitations, including donor scarcity, high cost, and immuno-incompatibility, continuous local delivery of biopharmaceuticals to the liver tissue can be a promising ALF treatment option. Here, the in vivo safety and usability of a 3D-printed implantable drug delivery device for effective ALF treatment is evaluated. The implantable reservoir consists of a 3D-printed container and a semipermeable membrane for repeated administrations of drugs, specifically to the liver tissue. The physical stability and function of the 3D-printed reservoir are confirmed by the mechanical properties and in vitro drug release test, respectively. In mice implanted with the reservoir system, mortality, weight changes, clinical signs, hematological and serum biochemical changes, and organ weight changes are not observed, suggesting no foreign body reaction. The usability of the reservoir system is further evaluated using an ALF model of 70% hepatectomized mice treated with N-acetylcysteine through the system, showing cell-specific regeneration and significant liver injury alleviation. Overall, the 3D-printed reservoir system is safe for studying the therapeutic potential of ALF treatment, and it can be used for the delivery of various active pharmaceutical ingredients.
Collapse
Affiliation(s)
- Shin‐Young Kim
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Ginam Han
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
- Department of Biomedical‐Chemical Engineering The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Da‐Bin Hwang
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Dong‐Hoon Won
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Yoo‐Sub Shin
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Changuk Kim
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Jeon Min Kang
- Biomedical Engineering Research Center Asan Institute for Life Sciences Asan Medical Center 88 Olympic‐ro 43‐gil Songpa‐gu Seoul 05505 Republic of Korea
| | - Jung‐Hoon Park
- Biomedical Engineering Research Center Asan Institute for Life Sciences Asan Medical Center 88 Olympic‐ro 43‐gil Songpa‐gu Seoul 05505 Republic of Korea
| | - Hyun‐Do Jung
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
- Department of Biomedical‐Chemical Engineering The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Wooram Park
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
- Department of Biomedical‐Chemical Engineering The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| | - Jun‐Won Yun
- Department of Biotechnology The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
- Department of Medical and Biological Sciences The Catholic University of Korea 43 Jibong‐ro Bucheon‐Si Gyeonggi‐do 14662 Republic of Korea
| |
Collapse
|
30
|
Sun L, Fan L, Bian F, Chen G, Wang Y, Zhao Y. MXene-Integrated Microneedle Patches with Innate Molecule Encapsulation for Wound Healing. RESEARCH (WASHINGTON, D.C.) 2021; 2021:9838490. [PMID: 34308359 PMCID: PMC8267825 DOI: 10.34133/2021/9838490] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/14/2021] [Indexed: 04/13/2023]
Abstract
Wound healing is a complex physiological process that involves coordinated phases such as inflammation and neovascularization. Attempts to promote the healing process tend to construct an effective delivery system based on different drugs and materials. In this paper, we propose novel MXene-integrated microneedle patches with adenosine encapsulation for wound healing. Owing to the dynamic covalent bonding capacity of boronate molecules with adenosine, 3-(acrylamido)phenylboronic acid- (PBA-) integrated polyethylene glycol diacrylate (PEGDA) hydrogel is utilized as the host material of microneedle patches. Benefitting from photothermal conversion capacity of MXene, the release of loaded adenosine could be accelerated under NIR irradiation for maintaining the activation signal around injury site. In vitro cell experiments proved the effect of MXene-integrated microneedle patches with adenosine encapsulation in enhancing angiogenesis. When applied for treating animal models, it is demonstrated that the microneedle patches efficiently promote angiogenesis, which is conductive to wound healing. These features make the proposed microneedle patch potential for finding applications in wound healing and other biomedical fields.
Collapse
Affiliation(s)
- Lingyu Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Lu Fan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Feika Bian
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
| | - Guopu Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yuetong Wang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008 Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
31
|
Castro R, Spivak Y, Shevchenko S, Moshnikov V. Low-Frequency Dielectric Relaxation in Structures Based on Macroporous Silicon with Meso-Macroporous Skin-Layer. MATERIALS (BASEL, SWITZERLAND) 2021; 14:2471. [PMID: 34064623 PMCID: PMC8151545 DOI: 10.3390/ma14102471] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 11/24/2022]
Abstract
The spectra of dielectric relaxation of macroporous silicon with a mesoporous skin layer in the frequency range 1-106 Hz during cooling (up to 293-173 K) and heating (293-333 K) are presented. Macroporous silicon (pore diameter ≈ 2.2-2.7 μm) with a meso-macroporous skin layer was obtained by the method of electrochemical anodic dissolution of monocrystalline silicon in a Unno-Imai cell. A mesoporous skin layer with a thickness of about 100-200 nm in the form of cone-shaped nanostructures with pore diameters near 13-25 nm and sizes of skeletal part about 35-40 nm by ion-electron microscopy was observed. The temperature dependence of the relaxation of the most probable relaxation time is characterized by two linear sections with different slope values; the change in the slope character is observed at T ≈ 250 K. The features of the distribution of relaxation times in meso-macroporous silicon at temperatures of 223, 273, and 293 K are revealed. The Havriliak-Negami approach was used for approximation of the relaxation curves ε″ = f(ν). The existence of a symmetric distribution of relaxers for all temperatures was found (Cole-Cole model). A discussion of results is provided, taking into account the structure of the studied object.
Collapse
Affiliation(s)
- Rene Castro
- Department of Physical Electronics, Faculty of Physics, Herzen State Pedagogical University of Russia (Herzen University), 191186 Saint Petersburg, Russia;
| | - Yulia Spivak
- Department of Micro- and Nanoelectronics, Faculty of Electronics, Saint Petersburg Electrotechnical University «LETI», 197376 Saint Petersburg, Russia;
| | - Sergey Shevchenko
- Department of Laser Measuring and Navigation Systems, Faculty of Information Measurement and Biotechnical Systems, Saint Petersburg Electrotechnical University «LETI», 197376 Saint Petersburg, Russia;
| | - Vyacheslav Moshnikov
- Department of Micro- and Nanoelectronics, Faculty of Electronics, Saint Petersburg Electrotechnical University «LETI», 197376 Saint Petersburg, Russia;
| |
Collapse
|
32
|
Wang S, Fontana F, Shahbazi MA, Santos HA. Acetalated dextran based nano- and microparticles: synthesis, fabrication, and therapeutic applications. Chem Commun (Camb) 2021; 57:4212-4229. [PMID: 33913978 DOI: 10.1039/d1cc00811k] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acetalated dextran (Ac-DEX) is a pH-responsive dextran derivative polymer. Prepared by a simple acetalation reaction, Ac-DEX has tunable acid-triggered release profile. Despite its relatively short research history, Ac-DEX has shown great potential in various therapeutic applications. Furthermore, the recent functionalization of Ac-DEX makes versatile derivatives with additional properties. Herein, we summarize the cutting-edge development of Ac-DEX and related polymers. Specifically, we focus on the chemical synthesis, nano- and micro-particle fabrication techniques, the controlled-release mechanisms, and the rational design Ac-DEX-based of drug delivery systems in various biomedical applications. Finally, we briefly discuss the challenges and future perspectives in the field.
Collapse
Affiliation(s)
- Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland. and Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran and Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland. and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
33
|
Wen Y, Liu Y, Chen C, Chi J, Zhong L, Zhao Y, Zhao Y. Metformin loaded porous particles with bio-microenvironment responsiveness for promoting tumor immunotherapy. Biomater Sci 2021; 9:2082-2089. [PMID: 33475656 DOI: 10.1039/d0bm01931c] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PD1/PD-L1 antibody blockade-based immunotherapy has been widely recognized in the field of cancer treatment; however, only a small number of cancer patients have been shown to respond well due to the PD1/PD-L1 antibody hydrolysis induced substandard immunotherapeutic efficacy and the low immunogenicity and immunosuppressive tumor microenvironment of the patients. Here, we present a novel tumor microenvironment (TME) responsive particle delivery system with a metformin-loaded chitosan (CS) inverse opal core and a manganese dioxide (MnO2) shell (denoted as CS-metformin@MnO2 particles) for inhibiting the PD-1/PD-L1 signaling pathway and promoting tumor immunotherapy. Benefiting from the interconnected porous structure of the inverse opal, metformin can be easily extensively loaded into the CS particles. With the coating of the TME responsive MnO2 shells, the particle delivery system was imparted with an intelligent "trigger" to prevent premature leaking of the drug until it reaches the tumor tissue. We have demonstrated that CS-metformin@MnO2 particles were able to promote the apoptosis of tumor cells through immunotherapeutic means both in vivo and in vitro. Specifically, the viability of tumor cells in the drug carrier-treated group was nearly 20% less than in the untreated group. In addition, the CS particles could serve as scaffolds for the regeneration of normal tissues and promote post-surgical wound healing due to their biocompatibility and antibacterial ability. These results make CS-metformin@MnO2 particles an excellent delivery system in tumor immunotherapy and post-surgical wound healing applications.
Collapse
Affiliation(s)
- Yuanyuan Wen
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | | | | | | | | | | | | |
Collapse
|
34
|
Li J, Zhang W, Gao Y, Tong H, Chen Z, Shi J, Santos HA, Xia B. Near-infrared light and magnetic field dual-responsive porous silicon-based nanocarriers to overcome multidrug resistance in breast cancer cells with enhanced efficiency. J Mater Chem B 2021; 8:546-557. [PMID: 31854435 DOI: 10.1039/c9tb02340b] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The development of drug delivery systems based on external stimuli-responsive nanocarriers is important to overcome multidrug resistance in breast cancer cells. Herein, iron oxide/gold (Fe3O4/Au) nanoparticles were first fabricated via a simple hydrothermal reaction, and subsequently loaded into porous silicon nanoparticles (PSiNPs) via electrostatic interactions to construct PSiNPs@(Fe3O4/Au) nanocomposites. The as-prepared PSiNPs@(Fe3O4/Au) nanocomposites exhibited excellent super-paramagnetism, photothermal effect, and T2-weight magnetic resonance imaging capability. In particular, with the help of a magnetic field, the cellular uptake of PSiNPs@(Fe3O4/Au) nanocomposites was significantly enhanced in drug-resistant breast cancer cells. Moreover, PSiNPs@(Fe3O4/Au) nanocomposites as carriers showed a high loading and NIR light-triggered release of anticancer drugs. Based on the synergistic effect of magnetic field-enhanced cellular uptake and NIR light-triggered intracellular release, the amount of anticancer drug carried by PSiNPs@(Fe3O4/Au) nanocarriers into the nuclei of drug-resistant breast cancer cells sharply increased, accompanied by improved chemo-photothermal therapeutic efficacy. Finally, PSiNPs@(Fe3O4/Au) nanocomposites under the combined conditions of magnetic field attraction and NIR light irradiation also showed improved anticancer drug penetration and accumulation in three-dimensional multicellular spheroids composed of drug-resistant breast cancer cells, leading to a better growth inhibition effect. Overall, the fabricated PSiNPs@(Fe3O4/Au) nanocomposites demonstrated great potential for the therapy of multidrug-resistant breast cancer in future.
Collapse
Affiliation(s)
- Jiachen Li
- Key Laboratory of Forest Genetics & Biotechnology (Ministry of Education of China), College of Science, Nanjing Forestry University, Nanjing 210037, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Advanced Multi-Dimensional Cellular Models as Emerging Reality to Reproduce In Vitro the Human Body Complexity. Int J Mol Sci 2021; 22:ijms22031195. [PMID: 33530487 PMCID: PMC7865724 DOI: 10.3390/ijms22031195] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
A hot topic in biomedical science is the implementation of more predictive in vitro models of human tissues to significantly improve the knowledge of physiological or pathological process, drugs discovery and screening. Bidimensional (2D) culture systems still represent good high-throughput options for basic research. Unfortunately, these systems are not able to recapitulate the in vivo three-dimensional (3D) environment of native tissues, resulting in a poor in vitro–in vivo translation. In addition, intra-species differences limited the use of animal data for predicting human responses, increasing in vivo preclinical failures and ethical concerns. Dealing with these challenges, in vitro 3D technological approaches were recently bioengineered as promising platforms able to closely capture the complexity of in vivo normal/pathological tissues. Potentially, such systems could resemble tissue-specific extracellular matrix (ECM), cell–cell and cell–ECM interactions and specific cell biological responses to mechanical and physical/chemical properties of the matrix. In this context, this review presents the state of the art of the most advanced progresses of the last years. A special attention to the emerging technologies for the development of human 3D disease-relevant and physiological models, varying from cell self-assembly (i.e., multicellular spheroids and organoids) to the use of biomaterials and microfluidic devices has been given.
Collapse
|
36
|
Zhan C, Lin G, Huang Y, Wang Z, Zeng F, Wu S. A dopamine-precursor-based nanoprodrug for in-situ drug release and treatment of acute liver failure by inhibiting NLRP3 inflammasome and facilitating liver regeneration. Biomaterials 2020; 268:120573. [PMID: 33260093 DOI: 10.1016/j.biomaterials.2020.120573] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/18/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023]
Abstract
Acute liver failure (ALF) is a severe liver disease with high mortality rate. Inflammasome is a newly-found and promising target for effective treatment of immunity-associated diseases including liver disease, and dopamine has recently been proved as an inhibitor for NLRP3 inflammasome. This work demonstrates a diselenide-based nanodrug for ALF treatment through inhibiting NLRP3 inflammasome activation and enhancing liver regeneration. A diselenide-containing molecule (DSeSeD) has been synthesized via covalently linking two l-Dopa molecules to a diselenide linker, and the resultant molecules form stable nanoparticles in aqueous media and encapsulate SW033291 (an inhibitor of prostaglandin-degrading enzyme that hampers liver regeneration) to produce the nanodrug (SW@DSeSeD). As a nanoscale prodrug, SW@DSeSeD protects its payloads from decomposition in bloodstream upon administration, accumulates in liver of ALF mice, then responds to the overexpressed ROS and thereby releases SW033291 as well as a stable dopamine precursor that can transform into dopamine in hepatic cells, thus achieving significant therapeutic efficacy against ALF through inhibiting NLRP3 inflammasome activation and enhancing hepatic regeneration. Moreover, multiple contrast agents have been loaded onto the nanodrug to achieve fluorescence, optoacoustic and magnetic resonance imaging for nanodrug location and disease evaluation.
Collapse
Affiliation(s)
- Chenyue Zhan
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Guifang Lin
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Yong Huang
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Ziqian Wang
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Fang Zeng
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China.
| | - Shuizhu Wu
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China.
| |
Collapse
|
37
|
Jin Y, Wang H, Yi K, Lv S, Hu H, Li M, Tao Y. Applications of Nanobiomaterials in the Therapy and Imaging of Acute Liver Failure. NANO-MICRO LETTERS 2020; 13:25. [PMID: 34138224 PMCID: PMC8187515 DOI: 10.1007/s40820-020-00550-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/22/2020] [Indexed: 05/02/2023]
Abstract
This review focuses on the therapeutic mechanisms, targeting strategies of various nanomaterials in acute liver failure, and recent advances of diverse nanomaterials for acute liver failure therapy, diagnosis, and imaging. This review provides an outlook on the applications of nanomaterials, especially on the new horizons in acute liver failure therapy, and inspires broader interests across various disciplines. Acute liver failure (ALF), a fatal clinical disease featured with overwhelming hepatocyte necrosis, is a grand challenge in global health. However, a satisfactory therapeutic option for curing ALF is still absent, other than liver transplantation. Nanobiomaterials are currently being developed for the diagnosis and treatment of ALF. The liver can sequester most of nanoparticles from blood circulation, which becomes an intrinsic superiority for nanobiomaterials targeting hepatic diseases. Nanobiomaterials can enhance the bioavailability of free drugs, thereby significantly improving the therapeutic effects in ALF. Nanobiomaterials can also increase the liver accumulation of therapeutic agents and enable more effective targeting of the liver or specific liver cells. In addition, stimuli-responsive, optical, or magnetic nanomaterials exhibit great potential in the therapeutical, diagnostic, and imaging applications in ALF. Therefore, therapeutic agents in combination with nanobiomaterials increase the specificity of ALF therapy, diminish adverse systemic effects, and offer a multifunctional theranostic platform. Nanobiomaterial holds excellent significance and prospects in ALF theranostics. In this review, we summarize the therapeutic mechanisms and targeting strategies of various nanobiomaterials in ALF. We highlight recent developments of diverse nanomedicines for ALF therapy, diagnosis, and imaging. Furthermore, the challenges and future perspectives in the theranostics of ALF are also discussed.
Collapse
Affiliation(s)
- Yuanyuan Jin
- Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, People's Republic of China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, People's Republic of China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, People's Republic of China
| | - Shixian Lv
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Hanze Hu
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, People's Republic of China.
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, People's Republic of China.
| |
Collapse
|
38
|
Dou Y, Li C, Li L, Guo J, Zhang J. Bioresponsive drug delivery systems for the treatment of inflammatory diseases. J Control Release 2020; 327:641-666. [PMID: 32911014 PMCID: PMC7476894 DOI: 10.1016/j.jconrel.2020.09.008] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Inflammation is intimately related to the pathogenesis of numerous acute and chronic diseases like cardiovascular disease, inflammatory bowel disease, rheumatoid arthritis, and neurodegenerative diseases. Therefore anti-inflammatory therapy is a very promising strategy for the prevention and treatment of these inflammatory diseases. To overcome the shortcomings of existing anti-inflammatory agents and their traditional formulations, such as nonspecific tissue distribution and uncontrolled drug release, bioresponsive drug delivery systems have received much attention in recent years. In this review, we first provide a brief introduction of the pathogenesis of inflammation, with an emphasis on representative inflammatory cells and mediators in inflammatory microenvironments that serve as pathological fundamentals for rational design of bioresponsive carriers. Then we discuss different materials and delivery systems responsive to inflammation-associated biochemical signals, such as pH, reactive oxygen species, and specific enzymes. Also, applications of various bioresponsive drug delivery systems in the treatment of typical acute and chronic inflammatory diseases are described. Finally, crucial challenges in the future development and clinical translation of bioresponsive anti-inflammatory drug delivery systems are highlighted.
Collapse
Affiliation(s)
- Yin Dou
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lanlan Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Chemistry, College of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiawei Guo
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Pharmaceutical Analysis, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China; Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
39
|
Huang K, Yang MS, Tang YJ, Ling SY, Pan F, Liu XD, Chen J. Porous shape memory scaffold of dextran and hydroxyapatite for minimum invasive implantation for bone tissue engineering applications. J Biomater Appl 2020; 35:823-837. [PMID: 32842853 DOI: 10.1177/0885328220950062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Minimally invasive implantation of a porous scaffold of large volume into bone defect site remains a challenge. Scaffolds based on shape memory polymer (SMP) show potential to be delivered in the compact form via minimally invasive surgery. The present study chooses poly (ε-caprolactone)-diols (PCL-diols) as the SMP to cross-link carboxyl dextran via ester bonds together with particle leaching method to yield a porous SMP scaffold. The inner surfaces of porous SMP scaffold are then mineralized via in situ precipitation to yield mineralized porous SMP-hydroxyapatite (SMP-HA) scaffold. The porous SMP-HA scaffold possesses pore size of 400-500 μm, with HA particles uniformly distributed and orientationally aligned on the inner surfaces of scaffold. X-ray diffraction (XRD) and differential scanning calorimetry (DSC) are carried out to identify the HA deposition. The phase transition temperature of the scaffold is adjusted to 38°C via changing the dosage of PCL (molecule weight: 2800) to endow the scaffold with shape deformation and fixed properties, as well as well-performed shape recovery property under body temperature. Bone marrow mesenchymal stem cells (BMSCs) adhere on the inner surfaces of SMP-HA scaffold, exhibiting larger spreading area when compared to cells adhered on SMP scaffold without HA, promoting its osteogenesis. In vivo degradation showed that the scaffold degrades completely after 6 months post-implantation. At the same time, significant tissue and capillary invasion indicated that the present porous SMP-HA scaffold hold great promise towards bone tissue engineering applications.
Collapse
Affiliation(s)
- Kai Huang
- Shanghai Zhabei District Library, Shanghai, China
| | - Mo-Song Yang
- Shanghai Zhabei District Library, Shanghai, China
| | - Yu-Jun Tang
- Shanghai Zhabei District Library, Shanghai, China
| | | | - Feng Pan
- Shanghai Zhabei District Library, Shanghai, China
| | | | - Jun Chen
- Department of Orthopedic, Zhabei Central Hospital of Jin'an District, Shanghai, China
| |
Collapse
|
40
|
Li Y, Liu Z, Li L, Lian W, He Y, Khalil E, Mäkilä E, Zhang W, Torrieri G, Liu X, Su J, Xiu Y, Fontana F, Salonen J, Hirvonen J, Liu W, Zhang H, Santos HA, Deng X. Tandem-Mass-Tag Based Proteomic Analysis Facilitates Analyzing Critical Factors of Porous Silicon Nanoparticles in Determining Their Biological Responses under Diseased Condition. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001129. [PMID: 32775170 PMCID: PMC7404168 DOI: 10.1002/advs.202001129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/25/2020] [Indexed: 05/11/2023]
Abstract
The analysis of nanoparticles' biocompatibility and immunogenicity is mostly performed under a healthy condition. However, more clinically relevant evaluation conducted under pathological condition is less known. Here, the immunogenicity and bio-nano interactions of porous silicon nanoparticles (PSi NPs) are evaluated in an acute liver inflammation mice model. Interestingly, a new mechanism in which PSi NPs can remit the hepatocellular damage and inflammation activation in a surface dependent manner through protein corona formation, which perturbs the inflammation by capturing the pro-inflammatory signaling proteins that are inordinately excreted or exposed under pathological condition, is found. This signal sequestration further attenuates the nuclear factor κB pathway activation and cytokines production from macrophages. Hence, the study proposes a potential mechanism for elucidating the altered immunogenicity of nanomaterials under pathological conditions, which might further offer insights to establish harmonized standards for assessing the biosafety of biomaterials in a disease-specific or personalized manner.
Collapse
Affiliation(s)
- Yunzhan Li
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell SignalingNetworkSchool of Life SciencesXiamen UniversityFujian361101China
- State‐Province Joint Engineering Laboratory of Targeted Drugs from Natural ProductsSchool of Life SciencesXiamen UniversityFujian361101China
| | - Zehua Liu
- Drug Research programDivision of Pharmaceutical Chemistry and TechnologyDrug Research ProgramFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Li Li
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell SignalingNetworkSchool of Life SciencesXiamen UniversityFujian361101China
- State‐Province Joint Engineering Laboratory of Targeted Drugs from Natural ProductsSchool of Life SciencesXiamen UniversityFujian361101China
| | - Wenhua Lian
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell SignalingNetworkSchool of Life SciencesXiamen UniversityFujian361101China
- State‐Province Joint Engineering Laboratory of Targeted Drugs from Natural ProductsSchool of Life SciencesXiamen UniversityFujian361101China
| | - Yaohui He
- School of Pharmaceutical SciencesXiamen UniversityFujian361101China
| | - Elbadry Khalil
- Drug Research programDivision of Pharmaceutical Chemistry and TechnologyDrug Research ProgramFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Ermei Mäkilä
- Laboratory of Industrial PhysicsDepartment of PhysicsUniversity of TurkuTurkuFI‐20014Finland
| | - Wenzhong Zhang
- Department of ChemistryUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Giulia Torrieri
- Drug Research programDivision of Pharmaceutical Chemistry and TechnologyDrug Research ProgramFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Xueyan Liu
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell SignalingNetworkSchool of Life SciencesXiamen UniversityFujian361101China
- State‐Province Joint Engineering Laboratory of Targeted Drugs from Natural ProductsSchool of Life SciencesXiamen UniversityFujian361101China
| | - Jingyi Su
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell SignalingNetworkSchool of Life SciencesXiamen UniversityFujian361101China
- State‐Province Joint Engineering Laboratory of Targeted Drugs from Natural ProductsSchool of Life SciencesXiamen UniversityFujian361101China
| | - Yuanming Xiu
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell SignalingNetworkSchool of Life SciencesXiamen UniversityFujian361101China
- State‐Province Joint Engineering Laboratory of Targeted Drugs from Natural ProductsSchool of Life SciencesXiamen UniversityFujian361101China
| | - Flavia Fontana
- Drug Research programDivision of Pharmaceutical Chemistry and TechnologyDrug Research ProgramFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Jarno Salonen
- Laboratory of Industrial PhysicsDepartment of PhysicsUniversity of TurkuTurkuFI‐20014Finland
| | - Jouni Hirvonen
- Drug Research programDivision of Pharmaceutical Chemistry and TechnologyDrug Research ProgramFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Wen Liu
- School of Pharmaceutical SciencesXiamen UniversityFujian361101China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory and Turku Bioscience CentreAbo Akademi UniversityTurkuFI‐20520Finland
| | - Hélder A. Santos
- Drug Research programDivision of Pharmaceutical Chemistry and TechnologyDrug Research ProgramFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFI‐00014Finland
| | - Xianming Deng
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell SignalingNetworkSchool of Life SciencesXiamen UniversityFujian361101China
- State‐Province Joint Engineering Laboratory of Targeted Drugs from Natural ProductsSchool of Life SciencesXiamen UniversityFujian361101China
| |
Collapse
|
41
|
Zhang DX, Esser L, Vasani RB, Thissen H, Voelcker NH. Porous silicon nanomaterials: recent advances in surface engineering for controlled drug-delivery applications. Nanomedicine (Lond) 2020; 14:3213-3230. [PMID: 31855121 DOI: 10.2217/nnm-2019-0167] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Porous silicon (pSi) nanomaterials are increasingly attractive for biomedical applications due to their promising properties such as simple and feasible fabrication procedures, tunable morphology, versatile surface modification routes, biocompatibility and biodegradability. This review focuses on recent advances in surface modification of pSi for controlled drug delivery applications. A range of functionalization strategies and fabrication methods for pSi-polymer hybrids are summarized. Surface engineering solutions such as stimuli-responsive polymer grafting, stealth coatings and active targeting modifications are highlighted as examples to demonstrate what can be achieved. Finally, the current status of engineered pSi nanomaterials for in vivo applications is reviewed and future prospects and challenges in drug-delivery applications are discussed.
Collapse
Affiliation(s)
- De-Xiang Zhang
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.,Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia
| | - Lars Esser
- Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia
| | - Roshan B Vasani
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Helmut Thissen
- Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia
| | - Nicolas H Voelcker
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.,Commonwealth Scientific & Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, 3168, Australia.,Melbourne Centre for Nanofabrication, Victorian Node of Australian National Fabrication Facility, Clayton, Victoria, 3168, Australia
| |
Collapse
|
42
|
Chaudhary M, Verma M, Jena KC, Singh N. Histidine‐Naphthalimide based Organic‐Inorganic Nanohybrid for Electrochemical Detection of Cyanide and Iodide ions. ChemistrySelect 2020. [DOI: 10.1002/slct.202001968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Monika Chaudhary
- Center for Biomedical Engineering Indian institute of Technology Ropar, Roopnagar Punjab 140001 India
| | - Meenakshi Verma
- Department of UCRD Chandigarh University, Ghraun Mohali, 140413 India
| | - Kailash C. Jena
- Center for Biomedical Engineering Indian institute of Technology Ropar, Roopnagar Punjab 140001 India
- Department of Physics Indian institute of Technology Ropar Roopnagar, Punjab 140001 India
| | - Narinder Singh
- Center for Biomedical Engineering Indian institute of Technology Ropar, Roopnagar Punjab 140001 India
- Department of Chemistry Indian institute of Technology Ropar Roopnagar, Punjab 140001 India
| |
Collapse
|
43
|
Shen J, Shafiq M, Ma M, Chen H. Synthesis and Surface Engineering of Inorganic Nanomaterials Based on Microfluidic Technology. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1177. [PMID: 32560284 PMCID: PMC7353232 DOI: 10.3390/nano10061177] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/15/2022]
Abstract
The controlled synthesis and surface engineering of inorganic nanomaterials hold great promise for the design of functional nanoparticles for a variety of applications, such as drug delivery, bioimaging, biosensing, and catalysis. However, owing to the inadequate and unstable mass/heat transfer, conventional bulk synthesis methods often result in the poor uniformity of nanoparticles, in terms of microstructure, morphology, and physicochemical properties. Microfluidic technologies with advantageous features, such as precise fluid control and rapid microscale mixing, have gathered the widespread attention of the research community for the fabrication and engineering of nanomaterials, which effectively overcome the aforementioned shortcomings of conventional bench methods. This review summarizes the latest research progress in the microfluidic fabrication of different types of inorganic nanomaterials, including silica, metal, metal oxides, metal organic frameworks, and quantum dots. In addition, the surface modification strategies of nonporous and porous inorganic nanoparticles based on microfluidic method are also introduced. We also provide the readers with an insight on the red blocks and prospects of microfluidic approaches, for designing the next generation of inorganic nanomaterials.
Collapse
Affiliation(s)
- Jie Shen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; (J.S.); (H.C.)
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Muhammad Shafiq
- Department of Chemistry, Pakistan Institute of Engineering & Applied Sciences (PIEAS), Nilore, Islamabad 45650, Pakistan;
| | - Ming Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; (J.S.); (H.C.)
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hangrong Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; (J.S.); (H.C.)
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
44
|
Liu Z, Fontana F, Python A, Hirvonen JT, Santos HA. Microfluidics for Production of Particles: Mechanism, Methodology, and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1904673. [PMID: 31702878 DOI: 10.1002/smll.201904673] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/27/2019] [Indexed: 06/10/2023]
Abstract
In the past two decades, microfluidics-based particle production is widely applied for multiple biological usages. Compared to conventional bulk methods, microfluidic-assisted particle production shows significant advantages, such as narrower particle size distribution, higher reproducibility, improved encapsulation efficiency, and enhanced scaling-up potency. Herein, an overview of the recent progress of the microfluidics technology for nano-, microparticles or droplet fabrication, and their biological applications is provided. For both nano-, microparticles/droplets, the previously established mechanisms behind particle production via microfluidics and some typical examples during the past five years are discussed. The emerging interdisciplinary technologies based on microfluidics that have produced microparticles or droplets for cellular analysis and artificial cells fabrication are summarized. The potential drawbacks and future perspectives are also briefly discussed.
Collapse
Affiliation(s)
- Zehua Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Andre Python
- Nuffield Department of Medicine, Li Ka Shing Centre for Health Information and Discovery, Big Data Institute, University of Oxford, OX3 7LF, Oxford, UK
| | - Jouni T Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014, Helsinki, Finland
| |
Collapse
|
45
|
Tang Z, Luo C, Jun Y, Yao M, Zhang M, He K, Jin L, Ma J, Chen S, Sun S, Tao M, Ding L, Sun X, Chen X, Zhang L, Gao Y, Wang QL. Nanovector Assembled from Natural Egg Yolk Lipids for Tumor-Targeted Delivery of Therapeutics. ACS APPLIED MATERIALS & INTERFACES 2020; 12:7984-7994. [PMID: 31971362 DOI: 10.1021/acsami.9b22293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Nanomedicine uses nanotechnology-based strategies for precision tumor therapy, including passive and ligand-mediated active tumor targeting by nanocarriers. However, the possible biotoxicity of chemosynthetic nanovectors limits their clinical applications. A novel natural egg yolk lipid nanovector (EYLN) was developed for effective loading and delivery of therapeutic agents. Lipids were extracted from egg yolks and reassembled into nanosized particles. EYLNs' stability, cellular uptake, toxicity, and delivery capacity for therapeutic agents were evaluated in vitro. The systemic toxicity and biodistribution of EYLNs were analyzed in normal mice, and the therapeutic effects of doxorubicin (Dox)-loaded EYLNs were evaluated in mouse breast cancer and hepatoma models. EYLNs had a particle size of ∼40 nm and a surface ζ-potential of -45 mV and were effectively internalized by tumor cells, without showing toxicity and side effects in vitro and in vivo. Importantly, their excellent permeability and retention effect significantly enhanced the distribution of EYLNs at tumor sites, and EYLN-Dox effectively inhibited the tumor growth in both mouse models. Targeted modification with folic acid further promoted vector-mediated drug distribution in tumors. This study demonstrates that lipids with specific proportions in the egg yolk can be used to construct natural drug vectors, providing a new strategy for nano-oncology research.
Collapse
Affiliation(s)
- Zhuang Tang
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Chao Luo
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Yali Jun
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Mengchu Yao
- Department of Clinical Oncology, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Mengyan Zhang
- Department of Clinical Oncology, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Kang He
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Luhao Jin
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Jianshe Ma
- School of Basic Medicine , Wenzhou Medical University , Wenzhou 325035 , China
| | - Song Chen
- Institute of Medicinal Biotechnology , Jiangsu College of Nursing , Huai'an 223300 , China
| | - SuAn Sun
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Mingyue Tao
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Lianshu Ding
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Xiaoyang Sun
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Xiaofei Chen
- Department of Clinical Oncology, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Li Zhang
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Yong Gao
- Department of Clinical Oncology, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Qi-Long Wang
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| |
Collapse
|
46
|
Zhang Y, Wang Y, Wen Y, Zhong Q, Zhao Y. Self-Healable Magnetic Structural Color Hydrogels. ACS APPLIED MATERIALS & INTERFACES 2020; 12:7486-7493. [PMID: 31961647 DOI: 10.1021/acsami.9b22579] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Biologically inspired structural color hydrogels with magnetic- and photothermal-controlled self-healable abilities were fabricated by integrating magnetic-responsive photonic crystals into gelatin hydrogels. The self-healable ability of the hydrogel systems was derived from the magnetic response and light-absorbing abilities of the magnetic nanoparticles. When the hydrogels deteriorate or get damaged, magnetic nanoparticles could absorb heat under near-infrared irradiation and external magnetic fields, which stimulates phase transformation in the hydrogels to fill or heal the hydrogels. In addition, the hydrogel systems were demonstrated with high biocompatibility and plasticity. These features of magnetic self-healable structural color hydrogels make them have broad application prospects in the fields of biological engineering and cell engineering.
Collapse
Affiliation(s)
- Yalan Zhang
- Department of Pharmaceutical Engineering, School of Engineering , China Pharmaceutical University , Nanjing 211198 , China
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital , Clinical College of Xuzhou Medical University , Nanjing 210008 , China
- Department of Clinical Laboratory , The Affiliated Drum Tower Hospital of Nanjing University Medical School , Nanjing 210008 , China
| | - Yu Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering , Southeast University , Nanjing 210096 , China
| | - Yuanyuan Wen
- Department of Pharmaceutical Engineering, School of Engineering , China Pharmaceutical University , Nanjing 211198 , China
| | - Qifeng Zhong
- Department of Pharmaceutical Engineering, School of Engineering , China Pharmaceutical University , Nanjing 211198 , China
| | - Yuanjin Zhao
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital , Clinical College of Xuzhou Medical University , Nanjing 210008 , China
- Department of Clinical Laboratory , 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
| |
Collapse
|
47
|
De Stefano L. Porous Silicon Optical Biosensors: Still a Promise or a Failure? SENSORS 2019; 19:s19214776. [PMID: 31684128 PMCID: PMC6864673 DOI: 10.3390/s19214776] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/23/2019] [Accepted: 11/01/2019] [Indexed: 12/21/2022]
Abstract
Even if the first published article on a porous silicon (PSi)-based biosensor dates back to more than twenty years ago, this technology still attracts great attention from many research groups around the world. In this brief review, the pros and cons of porous silicon-based optical biosensors will be highlighted on the basis of some recent results and published papers on this subject. The aim of the paper is to give a straightforward introduction to PhD students and young researchers on this subject, which is particularly full of educative content, since it is highly multidisciplinary. Fabrication of PSi-based optical biosensors requires competencies related to many different scientific topics ranging from material science, physics and optics to healthcare and environmental monitoring through surface chemistry and more.
Collapse
Affiliation(s)
- Luca De Stefano
- Institute for Microelectronics and Microsystems, National Research Council, Via P. Castellino 111, 80131 Napoli, Italy.
| |
Collapse
|
48
|
Bao H, Xia Y, Yu C, Ning X, Liu X, Fu H, Chen Z, Huang J, Zhang Z. CT/Bioluminescence Dual-Modal Imaging Tracking of Mesenchymal Stem Cells in Pulmonary Fibrosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1904314. [PMID: 31565866 DOI: 10.1002/smll.201904314] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/06/2019] [Indexed: 06/10/2023]
Abstract
Human mesenchymal stem cells (hMSCs), due to their immune regulation and collateral secretion effects, are currently explored for potential therapy of idiopathic pulmonary fibrosis (IPF). Understanding the migration, homing, functions, and survival of transplanted hMSCs in vivo is critical to successful IPF treatment. Therefore, it is highly desired to develop noninvasive and effective imaging technologies to track the transplanted hMSCs, providing experimental basis for improving the efficacy of hMSCs in the treatment of IPF. The rational design and development of a dual-labeling strategy are reported by integrating gold nanoparticle (AuNP)-based computed tomography (CT) nanotracers and red-emitting firefly luciferase (RfLuc)-based bioluminescence (BL) tags for CT/BL multimodal imaging tracking of the transplanted hMSCs in a murine model of IPF. In this approach, the CT nanotracer is prepared by sequential coupling of AuNPs with polyethylene glycol and trans-activator of transcription (TAT) peptide (Au@TAT), and employed it to monitor the location and distribution of the transplanted hMSCs in vivo by CT imaging, while RfLuc is used to monitor hMSCs viability by BLI. This facile strategy allows for visualization of the transplanted hMSCs in vivo, thereby enabling profound understanding of the role of hMSCs in the IPF treatment, and advancing stem cell-based regenerative medicine.
Collapse
Affiliation(s)
- Hongying Bao
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- Nano Science and Technology Institute, University of Science and Technology of China, Suzhou, 215123, China
| | - Yuyang Xia
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Chenggong Yu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xinyu Ning
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xiaoyun Liu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Han Fu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Zhongjin Chen
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jie Huang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Zhijun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| |
Collapse
|
49
|
Zou Y, Wei Y, Sun Y, Bao J, Yao F, Li Z, Meng F, Hu C, Storm G, Zhong Z. Cyclic RGD-Functionalized and Disulfide-Crosslinked Iodine-Rich Polymersomes as a Robust and Smart Theranostic Agent for Targeted CT Imaging and Chemotherapy of Tumor. Theranostics 2019; 9:8061-8072. [PMID: 31754381 PMCID: PMC6857068 DOI: 10.7150/thno.37184] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/28/2019] [Indexed: 02/07/2023] Open
Abstract
There is tremendous interest in integrating CT imaging with chemotherapy; however, reported iodine-based nanosystems such as nanogels and nano-emulsions display typically reduced contrast coefficient, low drug loading and stability, and poor targetability. Here, cRGD-functionalized disulfide-crosslinked iodine-rich polymersomes (cRGD-XIPs) were designed as a novel, robust and smart theranostic agent and investigated for targeted CT imaging and chemotherapy of malignant tumors. Methods: cRGD-XIPs were prepared from co-self-assembly of poly(ethylene glycol)-b-poly(dithiolane trimethylene carbonate-co-iodinated trimethylene carbonate) (PEG-P(DTC-IC)) and cRGD-PEG-P(DTC-IC) block copolymers. In vitro and in vivo CT contrast effect of cRGD-XIPs was studied using αvβ3-overexpressing B16 melanoma as a tumor model in comparison with clinical agent iohexol. The therapeutic efficacy of doxorubicin-loaded cRGD-XIPs (cRGD-XIPs-Dox) to B16 melanoma was investigated and compared with XIPs-Dox (non-targeted), cRGD-IPs-Dox (non-crosslinked) and free Dox. Results: cRGD-XIPs were formed with 55.5 wt.% iodine and ca. 90 nm in diameter. cRGD-XIPs-Dox with a Dox loading of 15.3 wt.% bared superior colloidal stability and reduction-responsive drug release. Notably, blank cRGD-XIPs showed a maximum-tolerated dose (MTD) > 400 mg iodine equiv./kg while cRGD-XIPs-Dox had an MTD > 150 mg Dox equiv./kg, ca. 15-fold improvement over free Dox. cRGD-XIPs revealed superior CT contrast effect and achieved 46.5- and 24.0-fold better enhancement of CT imaging of B16 melanoma than iohexol at 4 h following intratumoral and intravenous injection, respectively. cRGD-XIPs-Dox displayed an elimination half-life of 6.5 h and an elevated accumulation of 6.68% ID/g in the tumors. Furthermore, cRGD-XIPs-Dox was significantly more effective than XIPs-Dox and cRGD-XPs-Dox in inhibiting growth of B16 melanoma model. Conclusion: This proof-of-concept study demonstrates that cRGD-XIPs are a robust, non-toxic and smart polymeric theranostic agent that can not only significantly enhance CT imaging of tumors but also mediate efficient tumor-targeted chemotherapy. XIPs offer a unique and safe platform for theranostic polymersomes that pre-select patients using CT imaging prior to targeted chemotherapy with the same system.
Collapse
|
50
|
Zhao X, Liu Y, Shao C, Nie M, Huang Q, Li J, Sun L, Zhao Y. Photoresponsive Delivery Microcarriers for Tissue Defects Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901280. [PMID: 31637165 PMCID: PMC6794614 DOI: 10.1002/advs.201901280] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/03/2019] [Indexed: 05/16/2023]
Abstract
Intelligent responsive microcarriers have emerged as a promising class of biomaterials for therapeutic delivery and tissue regeneration, since they can respond to external stimuli and release the loaded drugs in an active manner. Among various available stimuli, near-infrared (NIR) light is particularly attractive because it can penetrate biotic tissues with sufficient intensity and minimal damage. In this work, a kind of photoresponsive delivery microcarriers (PDMs) is developed using microfluidics. The microcarriers consist of NIR-absorbing graphene oxide, thermosensitive poly(N-isopropylacrylamide), and biocompatible gelatin methacrylate. Under NIR light, the PDMs exhibit an evident volume shrinkage and effectively trigger the drug release. After the NIR light is switched off, the shrunken microcarriers return to their original size. This reversible process can be stably repeated for many cycles. An in vitro experiment demonstrates that the NIR-radiated PDMs can actively release vascular endothelial growth factors and improve the tube formation of human umbilical vein endothelial cells. The results from the in vivo experiment also show an obvious photothermal effect and superior therapeutic efficacy of these PDMs in a rat model of tissue defects. These features make the PDMs an excellent drug delivery system and represent a great potential for clinical applications in tissue repair.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Rheumatology and ImmunologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
- Research Institute of General SurgeryJinling HospitalMedical School of Nanjing UniversityNanjing210002China
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhou215006China
| | - Yuxiao Liu
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Changmin Shao
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Min Nie
- Department of Rheumatology and ImmunologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Qian Huang
- Research Institute of General SurgeryJinling HospitalMedical School of Nanjing UniversityNanjing210002China
| | - Jieshou Li
- Research Institute of General SurgeryJinling HospitalMedical School of Nanjing UniversityNanjing210002China
| | - Lingyun Sun
- Department of Rheumatology and ImmunologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| |
Collapse
|