1
|
Hong J, Kim YH. Cutting-edge biotherapeutics and advanced delivery strategies for the treatment of metabolic dysfunction-associated steatotic liver disease spectrum. J Control Release 2025; 380:433-456. [PMID: 39923856 DOI: 10.1016/j.jconrel.2025.02.008] [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: 09/30/2024] [Revised: 12/22/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), a condition with the potential to progress into liver cirrhosis or hepatocellular carcinoma, has become a significant global health concern due to its increasing prevalence alongside obesity and metabolic syndrome. Despite the promise of existing therapies such as thyroid hormone receptor-β (THR-β) agonists, PPAR agonists, FXR agonists, and GLP-1 receptor agonists, their effectiveness is limited by the complexity of the metabolic, inflammatory, and fibrotic pathways that drive MASLD progression, encompassing steatosis, metabolic dysfunction-associated steatohepatitis (MASH), and reversible liver fibrosis. Recent advances in targeted therapeutics, including RNA interference (RNAi), mRNA-based gene therapies, monoclonal antibodies, proteolysis-targeting chimeras (PROTAC), peptide-based strategies, cell-based therapies such as CAR-modified immune cells and stem cells, and extracellular vesicle-based approaches, have emerged as promising interventions. Alongside these developments, innovative drug delivery systems are being actively researched to enhance the stability, precision, and therapeutic efficacy of these biotherapeutics. These delivery strategies aim to optimize biodistribution, improve target-specific action, and reduce systemic exposure, thus addressing critical limitations of existing treatment modalities. This review provides a comprehensive exploration of the underlying biological mechanisms of MASLD and evaluates the potential of these cutting-edge biotherapeutics in synergy with advanced delivery approaches to address unmet clinical needs. By integrating fundamental disease biology with translational advancements, it aims to highlight future directions for the development of effective, targeted treatments for MASLD and its associated complications.
Collapse
Affiliation(s)
- Juhyeong Hong
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research Hanyang University, 04763 Seoul, South Korea; Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 04763 Seoul, South Korea
| | - Yong-Hee Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research Hanyang University, 04763 Seoul, South Korea; Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 04763 Seoul, South Korea; Cursus Bio Inc., Icure Tower, Gangnam-gu, Seoul 06170, Republic of Korea.
| |
Collapse
|
2
|
Xiang L, Xiang X, Jiao Q, Luo Y, Zeng G, Zhang W, Qin Y, Chen Y. Inhibition of HSC proliferation and hepatic fibrogenesis with Erythrocyte membrane coated Doxorubicin/Black phosphorus nanosheets. Int J Pharm 2025; 673:125403. [PMID: 40015579 DOI: 10.1016/j.ijpharm.2025.125403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Rapid proliferation underlies the abnormal expansion of activated hepatic stellate cells (aHSCs) and thereby contributes to the development and progression of liver fibrosis, so inhibition of HSC proliferation serves as a good antifibrotic strategy. As a potent topoisomerase II inhibitor, doxorubicin (DOX), an antineoplastic drug, exhibits a significant antifibrotic activity in vitro via retarding the growth of aHSCs and reversing their myofibroblastic phenotype, but its severe hepatotoxicity, cardiotoxicity, and renal toxicity limit its wide clinical application. Therefore, enhancing the specificity and efficacy of DOX in targeting aHSCs to improve its therapeutic index and minimize its adverse effects has become a key point for the success of DOX in antifibrotic treatment. In this study aimed at liver fibrosis treatment, we combined the excellent drug-loading capability and good biocompatibility of black phosphorus nanosheets (BPNSs), the protective and camouflaging properties of red blood cell membrane encapsulation, and the HSCs-targetability provided by the surface modification with vitamin A derivatives, into the construction of HSCs-targeted BP/DOX nanovesicles (BP/DOX@RMV-VA). The obtained DOX nanovesicles exhibited a uniform particle size and spheroid morphology, excellent diffusion property and stability, and high DOX loading. Specifically, they demonstrated outstanding biosafety, effective HSCs-targetability both in vivo and in vitro, and markedly improved pharmacokinetic profile of DOX. BP/DOX@RMV-VA produced strong antiproliferative and MF-phenotype reverting activity both in cultured aHSCs and in mice chronically injured by CCl4. And accordingly, the administration of BP/DOX@RMV-VA to CCl4-injured mice effectively suppressed the expansion of aHSCs and fibrogenesis, and significantly improved liver structure and function without causing detectable cardiotoxicity. These results highly suggest the therapeutic potential of BP/DOX@RMV-VA in treating liver fibrosis and other fibrosis-associated liver diseases.
Collapse
Affiliation(s)
- Li Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China; The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Xianjing Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Qiangqiang Jiao
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Yu Luo
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Guodong Zeng
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Wenhui Zhang
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Yuting Qin
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Yuping Chen
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China; The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China; MOE Key Laboratory of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China.
| |
Collapse
|
3
|
Vosoughi P, Naghib SM, Kangarshahi BM, Mozafari MR. A review of RNA nanoparticles for drug/gene/protein delivery in advanced therapies: Current state and future prospects. Int J Biol Macromol 2025; 295:139532. [PMID: 39765293 DOI: 10.1016/j.ijbiomac.2025.139532] [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: 09/22/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Nanotechnology involves the utilization of materials with exceptional properties at the nanoscale. Over the past few years, nanotechnologies have demonstrated significant potential in improving human health, particularly in medical treatments. The self-assembly characteristic of RNA is a highly effective method for designing and constructing nanostructures using a combination of biological, chemical, and physical techniques from different fields. There is great potential for the application of RNA nanotechnology in therapeutics. This review explores various nano-based drug delivery systems and their unique features through the impressive progress of the RNA field and their significant therapeutic promises due to their unique performance in the COVID-19 pandemic. However, a significant hurdle in fully harnessing the power of RNA drugs lies in effectively delivering RNA to precise organs and tissues, a critical factor for achieving therapeutic effectiveness, minimizing side effects, and optimizing treatment outcomes. There have been many efforts to pursue targeting, but the clinical translation of RNA drugs has been hindered by the lack of clear guidelines and shared understanding. A comprehensive understanding of various principles is essential to develop vaccines using nucleic acids and nanomedicine successfully. These include mechanisms of immune responses, functions of nucleic acids, nanotechnology, and vaccinations. Regarding this matter, the aim of this review is to revisit the fundamental principles of the immune system's function, vaccination, nanotechnology, and drug delivery in relation to the creation and manufacturing of vaccines utilizing nanotechnology and nucleic acids. RNA drugs have demonstrated significant potential in treating a wide range of diseases in both clinical and preclinical research. One of the reasons is their capacity to regulate gene expression and manage protein production efficiently. Different methods, like modifying chemicals, connecting ligands, and utilizing nanotechnology, have been essential in enabling the effective use of RNA-based treatments in medical environments. The article reviews stimuli-responsive nanotechnologies for RNA delivery and their potential in RNA medicines. It emphasizes the notable benefits of these technologies in improving the effectiveness of RNA and targeting specific cells and organs. This review offers a comprehensive analysis of different RNA drugs and how they work to produce therapeutic benefits. Recent progress in using RNA-based drugs, especially mRNA treatments, has shown that targeted delivery methods work well in medical treatments.
Collapse
Affiliation(s)
- Pegah Vosoughi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran.
| | - Babak Mikaeeli Kangarshahi
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
4
|
Wu Z, Zhao C, Ai H, Wang Z, Chen M, Lyu Y, Tong Q, Liu L, Sun H, Pu J, Zhang R, Hu X, Liu J, Ma X, Sun Y. A Susceptible Cell-Selective Delivery (SCSD) of mRNA-Encoded Cas13d Against Influenza Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414651. [PMID: 39792803 PMCID: PMC11884569 DOI: 10.1002/advs.202414651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/17/2024] [Indexed: 01/12/2025]
Abstract
To bolster the capacity for managing potential infectious diseases in the future, it is critical to develop specific antiviral drugs that can be rapidly designed and delivered precisely. Herein, a CRISPR/Cas13d system for broad-spectrum targeting of influenza A virus (IAV) from human, avian, and swine sources is designed, incorporating Cas13d mRNA and a tandem CRISPR RNA (crRNA) specific for the highly conserved regions of viral polymerase acidic (PA), nucleoprotein (NP), and matrix (M) gene segments, respectively. Given that the virus targets cells with specific receptors but is not limited to a single organ, a Susceptible Cell Selective Delivery (SCSD) system is developed by modifying a lipid nanoparticle with a peptide mimicking the function of the hemagglutinin of influenza virus to target sialic acid receptors. The SCSD system can precisely deliver an all-RNA-based CRISPR/Cas13d system into potentially infected cells. This drug is shown to reduce the viral load in the lungs by 2.37 log10 TCID50 mL-1 and protect 100% of mice from lethal influenza infection. The SCSD-based CRISPR/Cas13d system shows promise for the flexible and efficient therapy of infections caused by rapidly evolving and novel viruses.
Collapse
Affiliation(s)
- Zhuanli Wu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
| | - Chengcheng Zhao
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Hui Ai
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
| | - Zhen Wang
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
| | - Mingyue Chen
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
- Sanya Institute of China Agricultural UniversityHainan572025China
| | - Yanli Lyu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
- Veterinary Teaching HospitalChina Agricultural UniversityBeijing100193China
| | - Qi Tong
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
| | - Litao Liu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
| | - Honglei Sun
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
| | - Juan Pu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
| | - Ran Zhang
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Xiaoxiang Hu
- State Key Laboratory of Animal Biotech BreedingCollege of Biological SciencesChina Agricultural UniversityBeijing100193China
| | - Jinhua Liu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
| | - Xiaowei Ma
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Sanya Institute of China Agricultural UniversityHainan572025China
- Beijing Key Laboratory of Detection Technology for Animal‐Derived Food SafetyBeijing100193China
| | - Yipeng Sun
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural AffairsKey Laboratory for Prevention and Control of Avian Influenza and Other Major Poultry Diseases of the Ministry of Agriculture and Rural AffairsBeijing100193China
- Sanya Institute of China Agricultural UniversityHainan572025China
| |
Collapse
|
5
|
Cui X, Zhang R, Li Y, Li P, Liu Y, Yu X, Zhou J, Wang L, Tian X, Li H, Zhang S, Lan T, Li X, Zhang G, Li J, Liu Z. Bie Jia Jian pill ameliorates BDL-induced cholestatic hepatic fibrosis in rats by regulating intestinal microbial composition and TMAO-mediated PI3K/AKT signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118910. [PMID: 39369915 DOI: 10.1016/j.jep.2024.118910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/16/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a compound of traditional Chinese medicine (TCM), Bie Jia Jian pill (BJJP) is extensively used to treat the clinical chronic liver disease. Nevertheless, the specific mechanism through which BJJP affects hepatic fibrosis (HF) remains unknown. AIM OF THE STUDY To explore the role and potential mechanism of BJJP involved in treating HF. MATERIALS AND METHODS HF model of Sprague-Dawley (SD) rats was induced by a bile duct ligation (BDL). The function of BJJP involved in the intestinal microbiota (IM) and its metabolites in BDL-induced HF rats were explored through the 16S rRNA sequencing and untargeted metabolomics technologies. Network pharmacology was used to forecast mechanism underlying BJJP's anti-HF effects, which were validated in BDL-induced rats and trimethylamine N-oxide (TMAO)-induced LX-2 and HSC-T6 cells. RESULTS BJJP effectively ameliorated pathological liver damage, inflammation, and fibrosis of the BDL-induced HF rats. BJJP regulated IM diversity and composition and interfered with trimethylamine (TMA)-flavin monooxygenase 3 (FMO3)-TMAO process. In vitro, BJJP significantly inhibited the TMAO-induced activation of hepatic stellate cells (HSCs) (rat HSC cell line, HSC-T6; human HSC cell line, LX-2). Network pharmacology results demonstrated that PI3K/AKT signal pathway is crucially involved in BJJP treatment of HF. Further research revealed that BJJP inhibited the PI3K/AKT signal pathway in BDL-induced HF rats. Moreover, TMAO activated the PI3K/AKT pathway, whereas BJJP suppressed TMAO-induced activation. Subsequent intervention with 740Y-P (the PI3K agonist) successfully neutralized the repression effect on PI3K/AKT signal pathway by BJJP. CONCLUSION These results clearly show that BJJP attenuates HF by regulating the IM, as well as inhibiting PI3K/AKT pathway mediated by TMAO.
Collapse
Affiliation(s)
- Xiaoyan Cui
- Hebei Provincial Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Ronghua Zhang
- Hebei Provincial Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Yufeng Li
- The Cancer Institute, Hebei Key Laboratory of Molecular Oncology, Tangshan People's Hospital, Tangshan, 063001, China
| | - Ping Li
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan, 063000, China
| | - Yankun Liu
- The Cancer Institute, Hebei Key Laboratory of Molecular Oncology, Tangshan People's Hospital, Tangshan, 063001, China
| | - Xiaohan Yu
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan, 063000, China
| | - Jing Zhou
- Hebei Provincial Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Luyao Wang
- Hebei Provincial Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Xuetao Tian
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan, 063000, China
| | - Hongjie Li
- Hebei Provincial Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Shukun Zhang
- Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, Nankai Clinical College, Tianjin Medical University, Tianjin, 300100, China
| | - Tao Lan
- Hepatobiliary Pancreatic Surgery Department, Cangzhou People's Hospital, Cangzhou, 061000, China
| | - Xin Li
- Hepatobiliary Pancreatic Surgery Department, Cangzhou People's Hospital, Cangzhou, 061000, China
| | - Guangling Zhang
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan, 063000, China.
| | - Jingwu Li
- The Cancer Institute, Hebei Key Laboratory of Molecular Oncology, Tangshan People's Hospital, Tangshan, 063001, China.
| | - Zhiyong Liu
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan, 063000, China.
| |
Collapse
|
6
|
Gao F, Feng X, Li X. Recent advances in polymeric nanoparticles for the treatment of hepatic diseases. Front Pharmacol 2025; 16:1528752. [PMID: 39925843 PMCID: PMC11802823 DOI: 10.3389/fphar.2025.1528752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025] Open
Abstract
The liver performs crucial roles in energy metabolism, detoxification, and immune regulation. Hepatic diseases, including hepatitis, liver fibrosis, and liver cancer, have posed a significant threat to global health, emphasizing the critical need for the development of novel and effective treatment approaches. Nanotechnology, an emerging technology, has been extensively researched in medicine. Among the many types of nanomaterials, polymeric nanoparticles (NPs) are widely used in drug delivery systems. Compared to traditional therapies, they offer significant advantages in the treatment of liver disease by improving outcomes and reducing side effects. This review introduced the development of liver disease and discussed the application of natural polymers and synthetic polymers in their management. Furthermore, this paper reviewed the application of polymeric nanoparticles -mainly chitosan (CS), hyaluronic acid (HA), polyethylene glycol (PEG) and poly (lactic-co-glycolic acid) (PLGA)-in liver disease treatment, focusing on their use in various delivery systems for pure bioactive compounds of natural origin, drugs, nucleic acids, peptides, and others. Finally, the challenges and future perspectives of the NPs were discussed to provide guidance for further research directions, with the aim of promoting the clinical application of nanotherapeutics in treating hepatic diseases.
Collapse
Affiliation(s)
| | | | - Xinyu Li
- Clinical Laboratory of China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
7
|
Zou Y, Sun Z, Wang Q, Ju Y, Sun N, Yue Q, Deng Y, Liu S, Yang S, Wang Z, Li F, Hou Y, Deng C, Ling D, Deng Y. Core-Shell Magnetic Particles: Tailored Synthesis and Applications. Chem Rev 2025; 125:972-1048. [PMID: 39729245 DOI: 10.1021/acs.chemrev.4c00710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Core-shell magnetic particles consisting of magnetic core and functional shells have aroused widespread attention in multidisciplinary fields spanning chemistry, materials science, physics, biomedicine, and bioengineering due to their distinctive magnetic properties, tunable interface features, and elaborately designed compositions. In recent decades, various surface engineering strategies have been developed to endow them desired properties (e.g., surface hydrophilicity, roughness, acidity, target recognition) for efficient applications in catalysis, optical modulation, environmental remediation, biomedicine, etc. Moreover, precise control over the shell structure features like thickness, porosity, crystallinity and compositions including metal oxides, carbon, silica, polymers, and metal-organic frameworks (MOFs) has been developed as the major method to exploit new functional materials. In this review, we highlight the synthesis methods, regulating strategies, interface engineering, and applications of core-shell magnetic particles over the past half-century. The fundamental methodologies for controllable synthesis of core-shell magnetic materials with diverse organic, inorganic, or hybrid compositions, surface morphology, and interface property are thoroughly elucidated and summarized. In addition, the influences of the synthesis conditions on the physicochemical properties (e.g., dispersibility, stability, stimulus-responsiveness, and surface functionality) are also discussed to provide constructive insight and guidelines for designing core-shell magnetic particles in specific applications. The brand-new concept of "core-shell assembly chemistry" holds great application potential in bioimaging, diagnosis, micro/nanorobots, and smart catalysis. Finally, the remaining challenges, future research directions and new applications for the core-shell magnetic particles are predicted and proposed.
Collapse
Affiliation(s)
- Yidong Zou
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and State Key Laboratory of Molecular Engineering of Polymers, iChEM, Fudan University, Shanghai 200433, P. R. China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai 201804, P. R. China
| | - Zhenkun Sun
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and State Key Laboratory of Molecular Engineering of Polymers, iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Qiyue Wang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, School of Biomedical Engineering, National Center for Translational Medicine,, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
- Department of Clinical Laboratory, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, P. R. China
| | - Yanmin Ju
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, P. R. China
| | - Nianrong Sun
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and State Key Laboratory of Molecular Engineering of Polymers, iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Qin Yue
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and State Key Laboratory of Molecular Engineering of Polymers, iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Yu Deng
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and State Key Laboratory of Molecular Engineering of Polymers, iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Shanbiao Liu
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Shengfei Yang
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Zhiyi Wang
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, P. R. China
- School of Materials, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Fangyuan Li
- Department of Clinical Laboratory, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, P. R. China
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yanglong Hou
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, P. R. China
- School of Materials, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Chunhui Deng
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and State Key Laboratory of Molecular Engineering of Polymers, iChEM, Fudan University, Shanghai 200433, P. R. China
| | - Daishun Ling
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, School of Biomedical Engineering, National Center for Translational Medicine,, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Yonghui Deng
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, and State Key Laboratory of Molecular Engineering of Polymers, iChEM, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
8
|
Liu Y, Chen S, Huang H, Midgley AC, Han Z, Han ZC, Li Q, Li Z. Ligand-Tethered Extracellular Vesicles Mediated RNA Therapy for Liver Fibrosis. Adv Healthc Mater 2025; 14:e2403068. [PMID: 39520385 DOI: 10.1002/adhm.202403068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Liver fibrosis poses a significant global health burden, in which hepatic stellate cells (HSCs) play a crucial role. Targeted nanomedicine delivery systems directed at HSCs have shown immense potential in the treatment of liver fibrosis. Herein, a bioinspired material, engineered therapeutic miR-181a-5p (a miRNA known to inhibit fibrotic signaling pathways) and targeted moiety hyaluronic acid (HA) co-functionalized extracellular vesicles (EVs) are developed. HA is incorporated onto the surface of EVs using DSPE-PEG as a linker, allowing preferential binding to CD44 receptors, which are overexpressed on activated HSCs. Our results confirmed enhanced cellular uptake and improved payload delivery, as evidenced by the increased intracellular abundance of miR-181a-5p in activated HSCs and fibrotic livers. HA-equipped EVs loaded with miR-181a-5p (DPH-EVs@miR) significantly reduce HSC activation and extracellular matrix (ECM) deposition by inhibiting the TGF-β/Smad signaling pathway, thus alleviating the progression of liver fibrosis. Additionally, DPH-EVs@miR improves liver function, ameliorates inflammatory infiltration, and mitigates hepatocyte apoptosis, demonstrating superior hepatic protective effects. Collectively, this study reports a prospective nanovesicle therapeutic platform loaded with therapeutic miRNA and targeting motifs for liver fibrosis. The biomarker-guided EV-engineering technology utilized in this study provides a promising tool for nanomedicine and precision medicine.
Collapse
Affiliation(s)
- Yue Liu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Shang Chen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Haoyan Huang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Adam C Midgley
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Zhibo Han
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co. Ltd., 80 4th Street, TEDA, Tianjin, 300457, China
- Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health Biotech Co. Ltd., No. 1 Kangding Road, Daxing, Beijing, 100176, China
| | - Zhong-Chao Han
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co. Ltd., 80 4th Street, TEDA, Tianjin, 300457, China
- Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health Biotech Co. Ltd., No. 1 Kangding Road, Daxing, Beijing, 100176, China
| | - Qiong Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, China
| | - Zongjin Li
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, 17 Jingnan 5th Road, Zhengzhou, 450016, China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100039, China
| |
Collapse
|
9
|
Wang J, Chen Z, Zhao P, Wang Y, Chen J, Lin Q. PDGFR-α shRNA-polyplex for uveal melanoma treatment via EMT mediated vasculogenic mimicry interfering. J Nanobiotechnology 2024; 22:797. [PMID: 39726008 DOI: 10.1186/s12951-024-03077-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Up to 50% of individuals with uveal melanoma (UM), a frequent cancer of the eye, pass away from metastases. One of the major challenges in treating UM is the role of receptor tyrosine kinases (RTKs), which mediate the epithelial-mesenchymal transition (EMT) of tumors. RTKs are involved in binding multiple growth factors, leading to angiogenesis and vasculogenic mimicry (VM) phenomena. Currently, most anti-angiogenic drugs have shown a tendency to increase the VM of tumors in clinical trials, resulting in limited efficacy. The existing gap in UM treatment lies in the lack of effective strategies to target RTK-mediated EMT and VM. While some approaches have been attempted, there is still a need for novel therapeutic interventions that can specifically interfere with these processes. This research employed the gene vector PEI-g-PEG to interfere with the platelet derived growth factor-alpha receptor (PDGFR-α)-mediated EMT process, thereby retarding the growth of UM. The cell experiments demonstrated that the gene polyplex exhibited favorable cell uptake and lysosome escape properties, effectively suppressing the expression of PDGFR-α protein and EMT marker proteins and the occurrence of VM phenomenon. In vivo animal studies also inhibited the growth of UM, and PAS assays showed that the treatment reduced the generation of VM in tumor tissue. This study broadens the application of PEI-g-PEG while interfering with the RTK-mediated tumor EMT process with the help of RNAi technology, providing a new idea for tumor reduction research.
Collapse
Affiliation(s)
- Jiahao Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhirong Chen
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Peiyi Zhao
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yajia Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiang Chen
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Quankui Lin
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
10
|
Cui J, Wang G, Yip LX, Dong M, Mu M, Tian L, Gao Y, Fan Q, Zhu Q, Zhao X, Xu X, Leong DT, Sun X. Enhanced Ferritin‐Manganese Interaction by Nanoplatinum Growth Enabling Liver Fibrosis 3D Magnetic Resonance Visualization and Synergistic Therapy with Real‐Time Monitoring. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202410748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Indexed: 04/09/2025]
Abstract
AbstractEarly detection and timely intervention are essential to prevent liver fibrosis from progressing to cirrhosis or hepatocellular carcinoma. Herein, utilizing the enhanced ferritin‐manganese interaction by nanoplatinum growth, a novel ferritin‐platinum‐manganese magnetic resonance nanoplatform with RGD grafting and metformin loading (FNMMR) is developed. RGD can enhance the targeting ability of the nanoplatform toward integrin αVβ3 on activated hepatic stellate cells (aHSCs) in liver fibrosis. Systemic delivery of FNMMR shows clear degree‐dependent magnetic resonance contrast enhancement in liver fibrosis. 3D reconstruction techniques and histogram‐based features are achieved to qualitatively and quantitatively analyze the inhomogeneous liver fibrosis areas. FNMMR with catalase‐like activity can catalyze the generation of O2 to alleviate the liver fibrosis hypoxia and inhibit the expression of HIF‐1α, blocking the TGF‐β1/Smad signaling pathway. In addition, metformin shows synergy with HIF‐1α reduction in blocking the TGF‐β1/Smad pathway, effectively inhibiting the activation of HSCs and reducing collagen formation. Furthermore, FNMMR can achieve real‐time anti‐fibrotic therapy monitoring by magnetic resonance imaging. Importantly, no obvious side effects can be observed in both histological and hematology examinations. Therefore, this work presents a novel nanoplatform for accurate liver fibrosis diagnosis and synergistic anti‐fibrotic therapy with real‐time monitoring.
Collapse
Affiliation(s)
- Jin Cui
- Department of Radiology Shandong Provincial Hospital affiliated to Shandong First Medical University China 250021 China
| | - Gongzheng Wang
- Department of Radiology Shandong Provincial Hospital affiliated to Shandong First Medical University China 250021 China
| | - Li Xian Yip
- Department of Chemical and Biomolecular Engineering National University of Singapore Singapore 117585 Singapore
| | - Mengzhen Dong
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| | - Mengyao Mu
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| | - Liya Tian
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| | - Yuan Gao
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| | - Qing Fan
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| | - Qiang Zhu
- Department of Radiology Shandong Provincial Hospital affiliated to Shandong First Medical University China 250021 China
| | - Xinya Zhao
- Department of Radiology Shandong Provincial Hospital affiliated to Shandong First Medical University China 250021 China
| | - Xueli Xu
- School of Science Shandong Jianzhu University Jinan 250101 China
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering National University of Singapore Singapore 117585 Singapore
| | - Xiao Sun
- Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan 250117 China
| |
Collapse
|
11
|
Yuan Y, Li J, Chen M, Zhao Y, Zhang B, Chen X, Zhao J, Liang H, Chen Q. Nano-encapsulation of drugs to target hepatic stellate cells: Toward precision treatments of liver fibrosis. J Control Release 2024; 376:318-336. [PMID: 39413846 DOI: 10.1016/j.jconrel.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Liver fibrosis is characterized by excessive extracellular matrix (ECM) deposition triggered by hepatic stellate cells (HSCs). As central players in fibrosis progression, HSCs are the most important therapeutic targets for antifibrotic therapy. However, owing to the limitations of systemic drug administration, there is still no suitable and effective clinical treatment. In recent years, nanosystems have demonstrated expansive therapeutic potential and evolved into a clinical modality. In liver fibrosis, nanosystems have undergone a paradigm shift from targeting the whole liver to locally targeted modifying processes. Nanomedicine delivered to HSCs has significant potential in managing liver fibrosis, where optimal management would benefit from targeted delivery, personalized therapy based on the specific site of interest, and minor side effects. In this review, we present a brief overview of the role of HSCs in the pathogenesis of liver fibrosis, summarize the different types of nanocarriers and their specific delivery applications in liver fibrosis, and highlight the biological barriers associated with the use of nanosystems to target HSCs and approaches available to solve this issue. We further discuss in-depth all the molecular target receptors overexpressed during HSC activation in liver fibrosis and their corresponding ligands that have been used for drug or gene delivery targeting HSCs.
Collapse
Affiliation(s)
- Yue Yuan
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Jiaxuan Li
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Min Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Ying Zhao
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Jianping Zhao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| | - Qian Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China.
| |
Collapse
|
12
|
Cao Z, Zuo X, Liu X, Xu G, Yong KT. Recent progress in stimuli-responsive polymeric micelles for targeted delivery of functional nanoparticles. Adv Colloid Interface Sci 2024; 330:103206. [PMID: 38823215 DOI: 10.1016/j.cis.2024.103206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Stimuli-responsive polymeric micelles have emerged as a revolutionary approach for enhancing the in vivo stability, biocompatibility, and targeted delivery of functional nanoparticles (FNPs) in biomedicine. This article comprehensively reviews the preparation methods of these polymer micelles, detailing the innovative strategies employed to introduce stimulus responsiveness and surface modifications essential for precise targeting. We delve into the breakthroughs in utilizing these micelles to selectively deliver various FNPs including magnetic nanoparticles, upconversion nanoparticles, gold nanoparticles, and quantum dots, highlighting their transformative impact in the biomedical realm. Concluding, we present an insight into the current research landscape, addressing the challenges at hand, and envisioning the future trajectory in this burgeoning domain. Join us as we navigate the exciting confluence of polymer science and nanotechnology in reshaping biomedical solutions.
Collapse
Affiliation(s)
- Zhonglin Cao
- College of Materials Science and Engineering, Guizhou Minzu University, Guiyang 550025, China
| | - Xiaoling Zuo
- College of Materials Science and Engineering, Guizhou Minzu University, Guiyang 550025, China
| | - Xiaochen Liu
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia; The Biophotonics and Mechano-Bioengineering Lab, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia; The Biophotonics and Mechano-Bioengineering Lab, The University of Sydney, Sydney, New South Wales 2006, Australia.
| |
Collapse
|
13
|
Chen S, Zhuang D, Jia Q, Guo B, Hu G. Advances in Noninvasive Molecular Imaging Probes for Liver Fibrosis Diagnosis. Biomater Res 2024; 28:0042. [PMID: 38952717 PMCID: PMC11214848 DOI: 10.34133/bmr.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/08/2024] [Indexed: 07/03/2024] Open
Abstract
Liver fibrosis is a wound-healing response to chronic liver injury, which may lead to cirrhosis and cancer. Early-stage fibrosis is reversible, and it is difficult to precisely diagnose with conventional imaging modalities such as magnetic resonance imaging, positron emission tomography, single-photon emission computed tomography, and ultrasound imaging. In contrast, probe-assisted molecular imaging offers a promising noninvasive approach to visualize early fibrosis changes in vivo, thus facilitating early diagnosis and staging liver fibrosis, and even monitoring of the treatment response. Here, the most recent progress in molecular imaging technologies for liver fibrosis is updated. We start by illustrating pathogenesis for liver fibrosis, which includes capillarization of liver sinusoidal endothelial cells, cellular and molecular processes involved in inflammation and fibrogenesis, as well as processes of collagen synthesis, oxidation, and cross-linking. Furthermore, the biological targets used in molecular imaging of liver fibrosis are summarized, which are composed of receptors on hepatic stellate cells, macrophages, and even liver collagen. Notably, the focus is on insights into the advances in imaging modalities developed for liver fibrosis diagnosis and the update in the corresponding contrast agents. In addition, challenges and opportunities for future research and clinical translation of the molecular imaging modalities and the contrast agents are pointed out. We hope that this review would serve as a guide for scientists and students who are interested in liver fibrosis imaging and treatment, and as well expedite the translation of molecular imaging technologies from bench to bedside.
Collapse
Affiliation(s)
- Shaofang Chen
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Danping Zhuang
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Qingyun Jia
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application,
Harbin Institute of Technology, Shenzhen 518055, China
| | - Genwen Hu
- Department of Radiology, Shenzhen People’s Hospital (The Second Clinical Medical College,
Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| |
Collapse
|
14
|
Chen L, Guo W, Mao C, Shen J, Wan M. Liver fibrosis: pathological features, clinical treatment and application of therapeutic nanoagents. J Mater Chem B 2024; 12:1446-1466. [PMID: 38265305 DOI: 10.1039/d3tb02790b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Liver fibrosis is a reversible damage-repair response, the pathological features of which mainly include damage to hepatocytes, sinusoid capillarization, hepatic stellate cells activation, excessive accumulation of extracellular matrix and inflammatory response. Although some treatments (including drugs and stem cell therapy) for these pathological features have been shown to be effective, more clinical trials are needed to confirm their effectiveness. In recent years, nanomaterials-based therapies have emerged as an innovative and promising alternative to traditional drugs, being explored for the treatment of liver fibrosis diseases. Natural nanomaterials (including extracellular vesicles) and synthetic nanomaterials (including inorganic nanomaterials and organic nanomaterials) are developed to facilitate drug targeting delivery and combination therapy. In this review, the pathological features of liver fibrosis and the current anti-fibrosis drugs in clinical trials are briefly introduced, followed by a detailed introduction of the therapeutic nanoagents for the precise delivery of anti-fibrosis drugs. Finally, the future development trend in this field is discussed.
Collapse
Affiliation(s)
- Lin Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Wenyan Guo
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
15
|
Zhang JB, Zhang QR, Jin Q, Yang J, Lin SZ, Fan JG. Sestrin2 maintains hepatic immune homeostasis and redox balance partially via inhibiting RIPK3-mediated necroptosis in metabolic dysfunction-associated steatohepatitis. Mol Metab 2024; 80:101865. [PMID: 38163459 PMCID: PMC10825057 DOI: 10.1016/j.molmet.2023.101865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND & AIMS Necroptosis, a novel type of programmed cell death, is intricately associated with inflammatory response. Currently, most studies focus on the activation of necroptosis, while the mechanisms underlying the negative regulation of necroptosis remain poorly understood. METHODS The effects of sestrin2 (SESN2) overexpression or knockdown on the regulation of necroptosis were assessed in the TNFα/Smac-mimetic/Z-VAD-FMK (T/S/Z)-induced necroptosis model and palmitic acid (PA)-induced lipotoxicity model. Western-blot, co-Immunoprecipitation, Glutathione S-transferase pull-down, and confocal assays were employed to explore the regulatory mechanisms including protein-protein interactions and post-translational modification. Furthermore, we used GSK'872, a specific inhibitor of receptor-interacting serine/threonine-protein kinase (RIPK) 3, to evaluate the relationship between SESN2-related alterations and RIPK3-mediated necroptosis in T/S/Z-induced necroptosis model, PA-induced lipotoxicity model, and high-fat high-cholesterol diet (HFHCD)-induced non-alcoholic steatohepatitis model. RESULTS Our findings revealed that SESN2 was upregulated under conditions that induce necroptosis and functioned as a negative regulator of necroptosis. High levels of SESN2 could equipped hepatocytes with the ability to defend against necroptotic inflammation and oxidative stress. Mechanistically, SESN2 interacted with RIPK3 and tuned down necroptosis by inhibiting the phosphorylation of RIPK3, promoting the ubiquitination of RIPK3, and preventing the formation of the RIPK1/RIPK3 necrosome. The depletion of SESN2 resulted in excessive necroptosis, accompanied by increased fat accumulation, inflammation, and oxidative stress in the experimental steatohepatitis model. Blocking necroptosis by GSK'872 reduced the liberation of pro-inflammatory cytokines and reactive oxygen species generation, but not hepatocyte fat deposition, in both PA-treated SESN2 knockout cells and HFHCD-fed SESN2 knockout mice, suggesting that the activation of RIPK3-mediated necroptosis may partially account for the hyperinflammation and excessive oxidative stress induced by SESN2 deficiency. CONCLUSION Our results suggested that SESN2 inhibited RIPK3-mediated necroptosis; this regulation is an important for the immune homeostasis and the redox balance in the liver.
Collapse
Affiliation(s)
- Jian-Bin Zhang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qian-Ren Zhang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qian Jin
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jing Yang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Shuang-Zhe Lin
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
16
|
Niu X, Meng Y, Cui J, Li R, Ding X, Niu B, Chang G, Xu N, Li G, Wang Y, Wang L. Hepatic Stellate Cell- and Liver Microbiome-Specific Delivery System for Dihydrotanshinone I to Ameliorate Liver Fibrosis. ACS NANO 2023; 17:23608-23625. [PMID: 37995097 DOI: 10.1021/acsnano.3c06626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Liver fibrosis is a major contributor to the morbidity and mortality associated with liver diseases, yet effective treatment options remain limited. Hepatic stellate cells (HSCs) are a promising target for hepatic fibrogenesis due to their pivotal role in disease progression. Our previous research has demonstrated the potential of Dihydrotanshinone I (DHI), a lipophilic component derived from the natural herb Salvia miltiorrhiza Bunge, in treating liver fibrosis by inhibiting the YAP/TEAD2 interaction in HSCs. However, the clinical application of DHI faces challenges due to its poor aqueous solubility and lack of specificity for HSCs. Additionally, recent studies have implicated the impact of liver microbiota, distinct from gut microbiota, on the pathogenesis of liver diseases. In this study, we have developed an HSC- and microbiome-specific delivery system for DHI by conjugating prebiotic-like cyclodextrin (CD) with vitamin A, utilizing PEG2000 as a linker (VAP2000@CD). Our results demonstrate that VAP2000@CD markedly enhances the cellular uptake in human HSC line LX-2 and enhances the deposition of DHI in the fibrotic liver in vivo. Subsequently, intervention with DHI-VAP2000@CD has shown a notable reduction in bile duct-like structure proliferation, collagen accumulation, and the expression of fibrogenesis-associated genes in rats subjected to bile duct ligation. These effects may be attributed to the regulation of the YAP/TEAD2 interaction. Importantly, the DHI-VAP2000@CD intervention has also restored microbial homeostasis in the liver, promoting the amelioration of liver inflammation. Overall, our findings indicate that DHI-VAP2000@CD represents a promising therapeutic approach for liver fibrosis by specifically targeting HSCs and restoring the liver microbial balance.
Collapse
Affiliation(s)
- Xia Niu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yanan Meng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jinjin Cui
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Rui Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiao Ding
- State Key Laboratory of Phytochemistry and Plant Resource in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Bingyu Niu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Ge Chang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Ning Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guiling Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yucheng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Lulu Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
17
|
Zhou H, Chen DS, Hu CJ, Hong X, Shi J, Xiao Y. Stimuli-Responsive Nanotechnology for RNA Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303597. [PMID: 37915127 PMCID: PMC10754096 DOI: 10.1002/advs.202303597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/30/2023] [Indexed: 11/03/2023]
Abstract
Ribonucleic acid (RNA) drugs have shown promising therapeutic effects for various diseases in clinical and preclinical studies, owing to their capability to regulate the expression of genes of interest or control protein synthesis. Different strategies, such as chemical modification, ligand conjugation, and nanotechnology, have contributed to the successful clinical translation of RNA medicine, including small interfering RNA (siRNA) for gene silencing and messenger RNA (mRNA) for vaccine development. Among these, nanotechnology can protect RNAs from enzymatic degradation, increase cellular uptake and cytosolic transportation, prolong systemic circulation, and improve tissue/cell targeting. Here, a focused overview of stimuli-responsive nanotechnologies for RNA delivery, which have shown unique benefits in promoting RNA bioactivity and cell/organ selectivity, is provided. Many tissue/cell-specific microenvironmental features, such as pH, enzyme, hypoxia, and redox, are utilized in designing internal stimuli-responsive RNA nanoparticles (NPs). In addition, external stimuli, such as light, magnetic field, and ultrasound, have also been used for controlling RNA release and transportation. This review summarizes a wide range of stimuli-responsive NP systems for RNA delivery, which may facilitate the development of next-generation RNA medicines.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Cardiology, Clinical Trial CenterZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan University430071WuhanChina
- Center for Nanomedicine and Department of AnesthesiologyPerioperative and Pain MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM)Nanjing University of Posts & Telecommunications210023NanjingChina
| | - Dean Shuailin Chen
- Center for Nanomedicine and Department of AnesthesiologyPerioperative and Pain MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Caleb J. Hu
- Center for Nanomedicine and Department of AnesthesiologyPerioperative and Pain MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Xuechuan Hong
- Department of Cardiology, Clinical Trial CenterZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan University430071WuhanChina
| | - Jinjun Shi
- Center for Nanomedicine and Department of AnesthesiologyPerioperative and Pain MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Yuling Xiao
- Center for Nanomedicine and Department of AnesthesiologyPerioperative and Pain MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| |
Collapse
|
18
|
Li F, Zhao Y, Nie G. Nanotechnology-based combinational strategies toward the regulation of myofibroblasts and diseased microenvironment in liver fibrosis and hepatic carcinoma. NANO RESEARCH 2023; 16:13042-13055. [DOI: 10.1007/s12274-023-5809-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/29/2023] [Accepted: 05/05/2023] [Indexed: 01/03/2025]
|
19
|
Jouve M, Carpentier R, Kraiem S, Legrand N, Sobolewski C. MiRNAs in Alcohol-Related Liver Diseases and Hepatocellular Carcinoma: A Step toward New Therapeutic Approaches? Cancers (Basel) 2023; 15:5557. [PMID: 38067261 PMCID: PMC10705678 DOI: 10.3390/cancers15235557] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 06/29/2024] Open
Abstract
Alcohol-related Liver Disease (ALD) is the primary cause of chronic liver disorders and hepatocellular carcinoma (HCC) development in developed countries and thus represents a major public health concern. Unfortunately, few therapeutic options are available for ALD and HCC, except liver transplantation or tumor resection for HCC. Deciphering the molecular mechanisms underlying the development of these diseases is therefore of major importance to identify early biomarkers and to design efficient therapeutic options. Increasing evidence indicate that epigenetic alterations play a central role in the development of ALD and HCC. Among them, microRNA importantly contribute to the development of this disease by controlling the expression of several genes involved in hepatic metabolism, inflammation, fibrosis, and carcinogenesis at the post-transcriptional level. In this review, we discuss the current knowledge about miRNAs' functions in the different stages of ALD and their role in the progression toward carcinogenesis. We highlight that each stage of ALD is associated with deregulated miRNAs involved in hepatic carcinogenesis, and thus represent HCC-priming miRNAs. By using in silico approaches, we have uncovered new miRNAs potentially involved in HCC. Finally, we discuss the therapeutic potential of targeting miRNAs for the treatment of these diseases.
Collapse
Affiliation(s)
- Mickaël Jouve
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Rodolphe Carpentier
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Sarra Kraiem
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Noémie Legrand
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Cyril Sobolewski
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| |
Collapse
|
20
|
Wang K, Chen H, Qin S, Chen S, Zhang Q, Chen J, Di D, Su G, Yuan Y. Co-delivery of pirfenidone and siRNA in ZIF-based nanoparticles for dual inhibition of hepatic stellate cell activation in liver fibrotic therapy. Colloids Surf B Biointerfaces 2023; 231:113567. [PMID: 37797465 DOI: 10.1016/j.colsurfb.2023.113567] [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: 06/20/2023] [Revised: 09/07/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023]
Abstract
Hepatic fibrosis, as a destructive liver disease, occurs due to activated hepatic stellate cells (HSCs) producing excessive extracellular matrix deposition. If left untreated, it could further deteriorate into cirrhosis and hepatoma with high morbidity and mortality. Currently, to break the dilemma of poor targeting efficiency on HSCs and limited effect of monotherapy, it is urgent to explore a precise and efficient treatment against liver fibrosis. In the present work, a novel multifunctional nanoplatform based on vitamin A (VA) modified zeolitic imidazolate framework-8 (ZIF-8) nanoparticles was designed for co-delivery of chemical drug (Pirfenidone) and genetic drug (TGF-β1 siRNA) to achieve HSCs targeting mediated synergistic chemo-gene therapy against liver fibrosis. With the large specific surface area and acid-responsive degradation characteristics, ZIF-8 nanoparticles have great advantages to achieve high loading efficiency of Pirfenidone and enable acid-reactive drug release. After complexing siRNA, the prepared chemo-gene drug co-delivered nanocomplex (GP@ZIF-VL) proved excellent serum stability and effectively protected siRNA from degradation. Importantly, in vitro cell uptake and in vivo biodistribution demonstrated that VA functionalization markedly enhanced the delivery efficiency of GP@ZIF-VL nanocomplex into HSCs. As expected, GP@ZIF-VL significantly reduced extracellular matrix deposition and ameliorated hepatic fibrosis, as evidenced by decreased levels of liver enzymes in serum and a reduction in the hydroxyproline content in liver tissue. Therefore, GP@ZIF-VL nanocomplex displayed a bright future on the treatment of liver fibrosis with HSCs-targeting mediated chemo-gene synergetic therapy.
Collapse
Affiliation(s)
- Kaili Wang
- Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Hao Chen
- Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Si Qin
- Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Shuhui Chen
- Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Qian Zhang
- Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Jiali Chen
- Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Donghua Di
- Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Guangyue Su
- Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China.
| | - Yue Yuan
- Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China.
| |
Collapse
|
21
|
Li X, Yu M, Zhao Q, Yu Y. Prospective therapeutics for intestinal and hepatic fibrosis. Bioeng Transl Med 2023; 8:e10579. [PMID: 38023697 PMCID: PMC10658571 DOI: 10.1002/btm2.10579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/17/2023] [Accepted: 07/12/2023] [Indexed: 12/01/2023] Open
Abstract
Currently, there are no effective therapies for intestinal and hepatic fibrosis representing a considerable unmet need. Breakthroughs in pathogenesis have accelerated the development of anti-fibrotic therapeutics in recent years. Particularly, with the development of nanotechnology, the harsh environment of the gastrointestinal tract and inaccessible microenvironment of fibrotic lesions seem to be no longer considered a great barrier to the use of anti-fibrotic drugs. In this review, we comprehensively summarize recent preclinical and clinical studies on intestinal and hepatic fibrosis. It is found that the targets for preclinical studies on intestinal fibrosis is varied, which could be divided into molecular, cellular, and tissues level, although little clinical trials are ongoing. Liver fibrosis clinical trials have focused on improving metabolic disorders, preventing the activation and proliferation of hepatic stellate cells, promoting the degradation of collagen, and reducing inflammation and cell death. At the preclinical stage, the therapeutic strategies have focused on drug targets and delivery systems. At last, promising remedies to the current challenges are based on multi-modal synergistic and targeted delivery therapies through mesenchymal stem cells, nanotechnology, and gut-liver axis providing useful insights into anti-fibrotic strategies for clinical use.
Collapse
Affiliation(s)
- Xin Li
- Department of Clinical Pharmacy, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Institute of Pharmaceutics, College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Mengli Yu
- Department of Gastroenterology, The Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Qingwei Zhao
- Department of Clinical Pharmacy, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yang Yu
- College of Pharmaceutical SciencesSouthwest UniversityChongqingChina
| |
Collapse
|
22
|
Kozlov D, Rodimova S, Kuznetsova D. The Role of MicroRNAs in Liver Functioning: from Biogenesis to Therapeutic Approaches (Review). Sovrem Tekhnologii Med 2023; 15:54-79. [PMID: 39967915 PMCID: PMC11832066 DOI: 10.17691/stm2023.15.5.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Indexed: 01/03/2025] Open
Abstract
Molecular diagnostics based on small non-coding RNA molecules (in particular microRNA) is a new direction in modern biomedicine and is considered a promising method for identification of a wide range of pathologies at an early stage, clinical phenotype assessment, as well as monitoring the course of the disease, evaluation of therapy efficacy and the risk of the disease recurrence. Currently, the role of microRNAs as the most important epigenetic regulator in cancer development has been proven within the studies of normal and pathogenic processes. However, currently, there are insignificant studies devoted to studying the role of microRNAs in functioning of other organs and tissues, as well as to development of possible therapeutic approaches based on microRNAs. A huge number of metabolic processes in the liver are controlled by microRNAs, which creates enormous potential for the use of microRNAs as a diagnostic marker and makes it a target for therapeutic intervention in metabolic, oncological, and even viral diseases of this organ. This review examines various aspects of biological functions of microRNAs in different types of liver cells. Both canonical and non-canonical pathways of biogenesis, epigenetic regulation mediated by microRNAs, as well as the microRNAs role in intercellular communication and the course of viral diseases are shown. The potential of microRNAs as a diagnostic marker for various liver pathologies is described, as well as therapeutic approaches and medicines based on microRNAs, which are approved for clinical use and currently being developed.
Collapse
Affiliation(s)
- D.S. Kozlov
- Laboratory Assistant, Scientific Laboratory of Molecular Biotechnologies, I Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Student, Institute of Biology and Biomedicine; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| | - S.A. Rodimova
- Junior Researcher, Laboratory of Regenerative Medicine; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Junior Researcher, Scientific Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - D.S. Kuznetsova
- PhD, Head of the Scientific Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Head of the Research Laboratory for Molecular Genetic Researches, Institute of Clinical Medicine; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| |
Collapse
|
23
|
Huang J, Huang H, Wang Y, Xu B, Lin M, Han S, Yuan Y, Wang Y, Shuai X. Retinol-binding protein-hijacking nanopolyplex delivering siRNA to cytoplasm of hepatic stellate cell for liver fibrosis alleviation. Biomaterials 2023; 299:122134. [PMID: 37167895 DOI: 10.1016/j.biomaterials.2023.122134] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/13/2023]
Abstract
Activated hepatic stellate cell (aHSC) is mainly responsible for deposition of extracellular collagen matrix that causes liver fibrosis. Although several siRNAs adequately inhibited HSC activation in vitro, they were demonstrated poor RNAi efficiency in vivo. Developing HSC-targeting and cytoplasmic delivery nanocarrier is highly essential to acquire a desirable siRNA therapeutic index for anti-liver fibrosis. Here, we developed a unique crosslinking nanopolyplex (called T-C-siRNA) modified by vitamin A (VA) with the well-designed natures, including the negative charge, retinol-binding protein (RBP) hijacking, and cytoplasmic siRNA release in response to ROS and cis diol molecules. The nanopolyplex was given a yolk-shell-like shape, camouflage ability in blood, and HSC-targeting capability by hijacking the endogenous ligand RBP via surface VA. PDGFR-β siRNA (siPDGFR-β) supplied via T-C-siPDGFR-β nanopolyplex dramatically reduced HSC activation and its production of pro-fibrogenic proteins in vitro and in vivo. Furthermore, T-C-siPDGFR-β nanopolyplex effectively alleviated CCl4-induced liver injury, decreased hepatic collagen sediment, and recovered liver function in mice. This study provides a sophisticated method for HSC-targeting cytoplasmic RNA delivery using endogenous ligand hijacking and dual sensitivity of ROS and cis diol compounds.
Collapse
Affiliation(s)
- Jinsheng Huang
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Huiling Huang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Yiyao Wang
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Bin Xu
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Minzhao Lin
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China; PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Shisong Han
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yuanyuan Yuan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yong Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
24
|
Li H, Liu T, Yang Y, Cho WC, Flynn RJ, Harandi MF, Song H, Luo X, Zheng Y. Interplays of liver fibrosis-associated microRNAs: Molecular mechanisms and implications in diagnosis and therapy. Genes Dis 2023; 10:1457-1469. [PMID: 37397560 PMCID: PMC10311052 DOI: 10.1016/j.gendis.2022.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/09/2022] [Accepted: 08/20/2022] [Indexed: 11/22/2022] Open
Abstract
microRNAs (miRNAs) are a class of non-coding functional small RNA composed of 21-23 nucleotides, having multiple associations with liver fibrosis. Fibrosis-associated miRNAs are roughly classified into pro-fibrosis or anti-fibrosis types. The former is capable of activating hepatic stellate cells (HSCs) by modulating pro-fibrotic signaling pathways, mainly including TGF-β/SMAD, WNT/β-catenin, and Hedgehog; the latter is responsible for maintenance of the quiescent phenotype of normal HSCs, phenotypic reversion of activated HSCs (aHSCs), inhibition of HSCs proliferation and suppression of the extracellular matrix-associated gene expression. Moreover, several miRNAs are involved in regulation of liver fibrosis via alternative mechanisms, such as interacting between hepatocytes and other liver cells via exosomes and increasing autophagy of aHSCs. Thus, understanding the role of these miRNAs may provide new avenues for the development of novel interventions against hepatic fibrosis.
Collapse
Affiliation(s)
- Hong Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Tingli Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Yongchun Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang 311300, China
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR 999077, China
| | - Robin J. Flynn
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool L3 5RF, UK
- Graduate Studies Office, Department of Research, Innovation and Graduate Studies, Waterford Institute of Technology, X91 K0EK, Ireland
| | - Majid Fasihi Harandi
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman 7616914115, Iran
| | - Houhui Song
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang 311300, China
| | - Xuenong Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Yadong Zheng
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang 311300, China
| |
Collapse
|
25
|
Slott S, Astakhova K. MicroRNA Pools Synthesized Using Tandem Solid-Phase Oligonucleotide Synthesis. J Org Chem 2023. [PMID: 37389967 DOI: 10.1021/acs.joc.3c00376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2023]
Abstract
Herein, we describe a new approach to make pools of microRNA targeting breast cancer cells. The microRNA pools were synthesized at once on the same solid support using the "Tandem Oligonucleotide Synthesis" strategy. We make up to four consecutive microRNAs (miR129-1-5p, miR31, miR206, and miR27b-3p) using 2'/3'OAc nucleotide phosphoramidites, with the total length of the pool reaching 88 nucleotides. The developed phosphoramidites, when combined, give a cleavable moiety that separates the microRNAs and is cleaved using standard post-RNA synthesis cleavage conditions. Furthermore, we investigate making branched pools (microRNA dendrimers) versus linear pools as a strategy to further improve the product yields. Our approach provides with microRNA pools in high yields, which is of relevance to the growing demand on synthetic RNA oligomers for nucleic acid research and technology.
Collapse
Affiliation(s)
- Sofie Slott
- Department of Chemistry, Technical University of Denmark, 206-207, 2800 Kgs. Lyngby, Denmark
| | - Kira Astakhova
- Department of Chemistry, Technical University of Denmark, 206-207, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
26
|
Zhang F, Jiang J, Qian H, Yan Y, Xu W. Exosomal circRNA: emerging insights into cancer progression and clinical application potential. J Hematol Oncol 2023; 16:67. [PMID: 37365670 DOI: 10.1186/s13045-023-01452-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/10/2023] [Indexed: 06/28/2023] Open
Abstract
Exosomal circRNA serves a novel genetic information molecule, facilitating communication between tumor cells and microenvironmental cells, such as immune cells, fibroblasts, and other components, thereby regulating critical aspects of cancer progression including immune escape, tumor angiogenesis, metabolism, drug resistance, proliferation and metastasis. Interestingly, microenvironment cells have new findings in influencing tumor progression and immune escape mediated by the release of exosomal circRNA. Given the intrinsic stability, abundance, and broad distribution of exosomal circRNAs, they represent excellent diagnostic and prognostic biomarkers for liquid biopsy. Moreover, artificially synthesized circRNAs may open up new possibilities for cancer therapy, potentially bolstered by nanoparticles or plant exosome delivery strategies. In this review, we summarize the functions and underlying mechanisms of tumor cell and non-tumor cell-derived exosomal circRNAs in cancer progression, with a special focus on their roles in tumor immunity and metabolism. Finally, we examine the potential application of exosomal circRNAs as diagnostic biomarkers and therapeutic targets, highlighting their promise for clinical use.
Collapse
Affiliation(s)
- Fan Zhang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, People's Republic of China
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, People's Republic of China
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Hui Qian
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yongmin Yan
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, People's Republic of China.
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, No. 2 North Yongning Road, Changzhou, 213017, Jiangsu, People's Republic of China.
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, People's Republic of China.
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
27
|
Wu T, Qi Y, Xu C, Sui D, Xu FJ. HSC-targeted delivery of shRNA-TGFβ1 by vitamin A-functionalized polyaminoglycoside for hepatic fibrosis therapy. NANO TODAY 2023; 50:101887. [DOI: 10.1016/j.nantod.2023.101887] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
28
|
Gu J, Sun J, Tian K, Bian J, Peng J, Xu S, Zhao L. Reversal of hepatic fibrosis by the co-delivery of drug and ribonucleoprotein-based genome editor. Biomaterials 2023; 298:122133. [PMID: 37146364 DOI: 10.1016/j.biomaterials.2023.122133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/09/2023] [Accepted: 04/25/2023] [Indexed: 05/07/2023]
Abstract
Liver fibrosis is a chronic disease without effective treatment in the clinic. Gene editing systems such as the well-known CRISPR/Cas9 have shown great potential in the biomedical field. However, the delivery of the ribonucleoprotein is challenging due to the unstable RNA probe and the requirement for the entrance to the nucleus. Recently, a structure-guided endonuclease (SGN) has been reported as an effective gene-editing system composed of a nuclease and stable DNA probes, which can regulate the protein expression by targeting specific mRNA outside the nucleus. Here, we conjugated the SGN to a nanomicelle as the delivery system. In the resulting material, the chance of the collision between the endonuclease and the probe was raised due to the confinement of the two components within the 40-nm nanomicelle, thus the mRNA can be cleaved immediately after being captured by the probe, resulting in a space-induced nucleotide identification-cleavage acceleration effect. The delivery system was used to treat liver fibrosis via the co-delivery of SGN and a drug rosiglitazone to the hepatic stellate cells, which separately downregulated the expression of tissue inhibitor of metalloprotease-1 and inactivated the hepatic stellate cells. The system successfully reversed the liver fibrosis in mice through the bidirectional regulatory that simultaneously promoted the degradation and inhibited the production of the collagen, demonstrating the great potency of the SGN system as gene medicine.
Collapse
Affiliation(s)
- Jiayu Gu
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China
| | - Jingfang Sun
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China
| | - Kun Tian
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China
| | - Jinlei Bian
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| | - Juanjuan Peng
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| | - Shu Xu
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| | - Lingzhi Zhao
- State Key Laboratory of Natural Medicine, The School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, PR China.
| |
Collapse
|
29
|
Mosley RJ, Rucci B, Byrne ME. Recent advancements in design of nucleic acid nanocarriers for controlled drug delivery. J Mater Chem B 2023; 11:2078-2094. [PMID: 36806872 DOI: 10.1039/d2tb02325c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Research of nanoscale nucleic acid carriers has garnered attention in recent years due to their distinctive and controllable properties. However, current knowledge is limited in how we can efficiently utilize these systems for clinical applications. Several researchers have pioneered new and innovative nanocarrier drug delivery systems, but understanding physiochemical properties and behavior in vivo is vital to implementing them as clinical drug delivery platforms. In this review, we outline the most significant innovations in the synthesis, physical properties, and utilization of nucleic acid nanocarriers in the past 5 years, addressing the crucial properties which improve nanocarrier characteristics, delivery, and drug release. The challenges of controlling the transport of nucleic acid nanocarriers and therapeutic release for biological applications are outlined. Barriers which inhibit effective transport into tissue are discussed with emphasis on the modifications needed to overcome such obstacles. The novel strategies discussed in this work summarize the pivotal features of modern nucleic nanocarriers and postulate where future developments could revolutionize the translation of these tools into a clinical setting.
Collapse
Affiliation(s)
- Robert J Mosley
- Biomimetic and Biohybrid Materials, Biomedical Devices, and Drug Delivery Laboratories, Department of Biomedical Engineering, 201 Mullica Hill Rd, Rowan University, Glassboro, NJ, 08028, USA.
| | - Brendan Rucci
- Biomimetic and Biohybrid Materials, Biomedical Devices, and Drug Delivery Laboratories, Department of Biomedical Engineering, 201 Mullica Hill Rd, Rowan University, Glassboro, NJ, 08028, USA.
| | - Mark E Byrne
- Biomimetic and Biohybrid Materials, Biomedical Devices, and Drug Delivery Laboratories, Department of Biomedical Engineering, 201 Mullica Hill Rd, Rowan University, Glassboro, NJ, 08028, USA. .,Department of Chemical Engineering, Rowan University, Glassboro, NJ, 08028, USA
| |
Collapse
|
30
|
Vyas K, Patel MM. Insights on drug and gene delivery systems in liver fibrosis. Asian J Pharm Sci 2023; 18:100779. [PMID: 36845840 PMCID: PMC9950450 DOI: 10.1016/j.ajps.2023.100779] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/30/2023] Open
Abstract
Complications of the liver are amongst the world's worst diseases. Liver fibrosis is the first stage of liver problems, while cirrhosis is the last stage, which can lead to death. The creation of effective anti-fibrotic drug delivery methods appears critical due to the liver's metabolic capacity for drugs and the presence of insurmountable physiological impediments in the way of targeting. Recent breakthroughs in anti-fibrotic agents have substantially assisted in fibrosis; nevertheless, the working mechanism of anti-fibrotic medications is not fully understood, and there is a need to design delivery systems that are well-understood and can aid in cirrhosis. Nanotechnology-based delivery systems are regarded to be effective but they have not been adequately researched for liver delivery. As a result, the capability of nanoparticles in hepatic delivery was explored. Another approach is targeted drug delivery, which can considerably improve efficacy if delivery systems are designed to target hepatic stellate cells (HSCs). We have addressed numerous delivery strategies that target HSCs, which can eventually aid in fibrosis. Recently genetics have proved to be useful, and methods for delivering genetic material to the target place have also been investigated where different techniques are depicted. To summarize, this review paper sheds light on the most recent breakthroughs in drug and gene-based nano and targeted delivery systems that have lately shown useful for the treatment of liver fibrosis and cirrhosis.
Collapse
Affiliation(s)
- Kunj Vyas
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University SG Highway, Gujarat 382481, India
| | - Mayur M Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University SG Highway, Gujarat 382481, India
| |
Collapse
|
31
|
Yuan Y, Wang Y, Huang H, Tao S, Huang J. Fine-Tuned Polymer Nanoassembly for Codelivery of Chemotherapeutic Drug and siRNA. Macromol Biosci 2023; 23:e2200529. [PMID: 36640140 DOI: 10.1002/mabi.202200529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Successful clinical application of siRNA to liver-associated diseases reinvigorates the RNAi therapeutics and delivery vectors, especially for anticancer combination therapy. Fine tuning of copolymer-based assembly configuration is highly important for a desirable synergistic cancer cell-killing effect via the codelivery of chemotherapeutic drug and siRNA. Herein, an amphiphilic triblock copolymer methoxyl poly(ethylene glycol)-block-poly(L-lysine)-block-poly(2-(diisopropyl amino)ethyl methacrylate) (abbreviated as mPEG-PLys-PDPA or PLD) consisting of a hydrophilic diblock mPEG-PLys and a hydrophobic block PDPA is synthesized. Three distinct assemblies (i.e., nanosized micelle, nanosized polymersome, and microparticle) are acquired, along with the increase in PDPA block length. Furthermore, the as-obtained polymersome can efficiently codeliver doxorubicin hydrochloride (DOX) as a hydrophilic chemotherapeutic model and siRNA against ADP-ribosylation factor 6 (siArf6) as an siRNA model into cancer cell via lysosomal pH-triggered payload release. PC-3 prostate cell is synergistically killed by the DOX- and siArf6-coloading polymersome (namely PLD@DOX/siArf6). PLD@DOX/siArf6 may serve as a robust nanomedicine for anticancer therapy.
Collapse
Affiliation(s)
- Yuanyuan Yuan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yiyao Wang
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Huiling Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shuiliang Tao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jinsheng Huang
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
32
|
Polyethyleneimine-Based Drug Delivery Systems for Cancer Theranostics. J Funct Biomater 2022; 14:jfb14010012. [PMID: 36662059 PMCID: PMC9862060 DOI: 10.3390/jfb14010012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
With the development of nanotechnology, various types of polymer-based drug delivery systems have been designed for biomedical applications. Polymer-based drug delivery systems with desirable biocompatibility can be efficiently delivered to tumor sites with passive or targeted effects and combined with other therapeutic and imaging agents for cancer theranostics. As an effective vehicle for drug and gene delivery, polyethyleneimine (PEI) has been extensively studied due to its rich surface amines and excellent water solubility. In this work, we summarize the surface modifications of PEI to enhance biocompatibility and functionalization. Additionally, the synthesis of PEI-based nanoparticles is discussed. We further review the applications of PEI-based drug delivery systems in cancer treatment, cancer imaging, and cancer theranostics. Finally, we thoroughly consider the outlook and challenges relating to PEI-based drug delivery systems.
Collapse
|
33
|
Chen G, Deng S, Zuo M, Wang J, Cheng D, Chen B. Non-viral CRISPR activation system targeting VEGF-A and TGF-β1 for enhanced osteogenesis of pre-osteoblasts implanted with dual-crosslinked hydrogel. Mater Today Bio 2022; 16:100356. [PMID: 35898441 PMCID: PMC9309523 DOI: 10.1016/j.mtbio.2022.100356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
Healing of large calvarial bone defects remains challenge but may be improved by stimulating bone regeneration of implanted cells. The aim of this study is to specially co-activate transforming growth factor β1 (TGF-β1) and vascular endothelial growth factor (VEGF-A) genes expressions in pre-osteoblast MC3T3-E1 cells through the non-viral CRISPR activation (CRISPRa) system to promote osteogenesis. A cationic copolymer carrying nucleus localizing peptides and proton sponge groups dimethyl-histidine was synthesized to deliver CRISPRa system into MC3T3-E1 cells with high cellular uptake, lysosomal escape, and nuclear translocation, which activated VEGF-A and TGF-β1 genes expressions and thereby additively or synergistically induced several osteogenic genes expressions. A tunable dual-crosslinked hydrogel was developed to implant the above engineered cells into mice calvaria bone defect site to promote bone healing in vivo. The combination of multi-genes activation through non-viral CRISPRa system and tunable dual-crosslinked hydrogel provides a versatile strategy for promoting bone healing with synergistic effect.
Collapse
Affiliation(s)
- Guo Chen
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, PR China
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Shaohui Deng
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Mingxiang Zuo
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Jin Wang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
| | - Du Cheng
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, PR China
- Corresponding author.
| | - Bin Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Corresponding author.
| |
Collapse
|
34
|
Luo F, Yu Y, Li M, Chen Y, Zhang P, Xiao C, Lv G. Polymeric nanomedicines for the treatment of hepatic diseases. J Nanobiotechnology 2022; 20:488. [PMCID: PMC9675156 DOI: 10.1186/s12951-022-01708-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022] Open
Abstract
The liver is an important organ in the human body and performs many functions, such as digestion, detoxification, metabolism, immune responses, and vitamin and mineral storage. Therefore, disorders of liver functions triggered by various hepatic diseases, including hepatitis B virus infection, nonalcoholic steatohepatitis, hepatic fibrosis, hepatocellular carcinoma, and transplant rejection, significantly threaten human health worldwide. Polymer-based nanomedicines, which can be easily engineered with ideal physicochemical characteristics and functions, have considerable merits, including contributions to improved therapeutic outcomes and reduced adverse effects of drugs, in the treatment of hepatic diseases compared to traditional therapeutic agents. This review describes liver anatomy and function, and liver targeting strategies, hepatic disease treatment applications and intrahepatic fates of polymeric nanomedicines. The challenges and outlooks of hepatic disease treatment with polymeric nanomedicines are also discussed.
Collapse
Affiliation(s)
- Feixiang Luo
- grid.430605.40000 0004 1758 4110Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021 People’s Republic of China
| | - Ying Yu
- grid.430605.40000 0004 1758 4110Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021 People’s Republic of China
| | - Mingqian Li
- grid.430605.40000 0004 1758 4110Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021 People’s Republic of China
| | - Yuguo Chen
- grid.430605.40000 0004 1758 4110Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021 People’s Republic of China
| | - Peng Zhang
- grid.9227.e0000000119573309Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 People’s Republic of China
| | - Chunsheng Xiao
- grid.9227.e0000000119573309Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 People’s Republic of China
| | - Guoyue Lv
- grid.430605.40000 0004 1758 4110Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021 People’s Republic of China
| |
Collapse
|
35
|
Deng S, Wang S, Xiao Z, Cheng D. Unprotonatable and ROS-Sensitive Nanocarrier for NIR Spatially Activated siRNA Therapy with Synergistic Drug Effect. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203823. [PMID: 36094800 DOI: 10.1002/smll.202203823] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/13/2022] [Indexed: 06/15/2023]
Abstract
Although small interfering RNA (siRNA) therapy has achieved great progress, unwanted gene inhibition in normal tissues severely limits its extensive clinical applications due to uncontrolled siRNA biodistribution. Herein, a spatially controlled siRNA activation strategy is developed to achieve tumor-specific siRNA therapy without gene inhibition in the normal tissues. The quaternary ammonium moieties are conjugated to amphiphilic copolymers via reactive oxygen species (ROS)-sensitive thioketal (TK) linkers for co-delivery of siRNA and photosensitizer chlorin e6 (Ce6), showing excellent siRNA complexation capacity and near infrared (NIR)-controlled siRNA release. In the normal tissue, siRNAs are trapped and degraded in the endo-lysosomes due to the unprotonatable property of quaternary ammonium moiety, showing the siRNA activity "off" state. When NIR irradiation is spatially applied to the tumor tissue, the NIR irradiation/Ce6-induced ROS trigger siRNA endo-lysosomal escape and cytosolic release through the photochemical internalization effect and cleavage of TK bonds, respectively, showing the siRNA activity "on" state. The siRNA-mediated glutathione peroxidase 4 gene inhibition enhances ROS accumulation. The synergistic antitumor activity of Ce6 photodynamic therapy and gene inhibition is confirmed in vivo. Spatially controlled tumor-specific siRNA activation and co-delivery with Ce6 using unprotonatable and ROS-sensitive cationic nanocarriers provide a feasible strategy for tumor-specific siRNA therapy with synergistic drug effects.
Collapse
Affiliation(s)
- Shaohui Deng
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Shiyin Wang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| | - Zecong Xiao
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Du Cheng
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China
| |
Collapse
|
36
|
He H, Zhang X, Du L, Ye M, Lu Y, Xue J, Wu J, Shuai X. Molecular imaging nanoprobes for theranostic applications. Adv Drug Deliv Rev 2022; 186:114320. [PMID: 35526664 DOI: 10.1016/j.addr.2022.114320] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/11/2022] [Accepted: 04/30/2022] [Indexed: 12/13/2022]
Abstract
As a non-invasive imaging monitoring method, molecular imaging can provide the location and expression level of disease signature biomolecules in vivo, leading to early diagnosis of relevant diseases, improved treatment strategies, and accurate assessment of treating efficacy. In recent years, a variety of nanosized imaging probes have been developed and intensively investigated in fundamental/translational research and clinical practice. Meanwhile, as an interdisciplinary discipline, this field combines many subjects of chemistry, medicine, biology, radiology, and material science, etc. The successful molecular imaging not only requires advanced imaging equipment, but also the synthesis of efficient imaging probes. However, limited summary has been reported for recent advances of nanoprobes. In this paper, we summarized the recent progress of three common and main types of nanosized molecular imaging probes, including ultrasound (US) imaging nanoprobes, magnetic resonance imaging (MRI) nanoprobes, and computed tomography (CT) imaging nanoprobes. The applications of molecular imaging nanoprobes were discussed in details. Finally, we provided an outlook on the development of next generation molecular imaging nanoprobes.
Collapse
Affiliation(s)
- Haozhe He
- Nanomedicine Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Xindan Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lihua Du
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510260, China
| | - Minwen Ye
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yonglai Lu
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiajia Xue
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Jun Wu
- PCFM Lab of Ministry of Education, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510260, China.
| |
Collapse
|
37
|
Li X, Xu X, Huang K, Wu Y, Lin Z, Yin L. Hypoxia-Reinforced Antitumor RNA Interference Mediated by Micelleplexes with Programmed Disintegration. Acta Biomater 2022; 148:194-205. [PMID: 35662669 DOI: 10.1016/j.actbio.2022.05.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 12/17/2022]
Abstract
The performance of polycation-mediated siRNA delivery is often hurdled by the multiple systemic and cellular barriers that pose conflicting requirements for materials properties. Herein, micelleplexes (MPs) capable of programmed disintegration were developed to mediate efficient delivery of siRNA against XIAP (siXIAP) in a hypoxia-reinforced manner. MPs were assembled from azobenzene-crosslinked oligoethylenimine (AO), acid-transformable diblock copolymer PPDHP with conjugated photosensitizer, and siXIAP. AO efficiently condensed siXIAP via electrostatic interaction, and PPDHP rendered additional hydrophobic interaction with AO to stabilize the MPs against salt. The hydrophilic PEG corona enhanced the serum stability of MPs to prolong blood circulation and promote tumor accumulation. After internalization into cancer cells, the endolysosomal acidity triggered shedding of PPDHP, exposing AO to induce endolysosomal escape. Then, light irradiation generated lethal amount of ROS, and concurrently aggravated intracellular hypoxia level to degrade AO into low-molecular weight segments, release siXIAP, and potentiate the XIAP silencing efficiency. Thus, siXIAP-mediated pro-apoptosis cooperated with generated ROS to provoke pronounced anti-cancer efficacy against Skov-3 tumors in vitro and in vivo. This study provides a hypoxia-instructed strategy to overcome the multiple barriers against anti-cancer siRNA delivery in a programmed manner. STATEMENT OF SIGNIFICANCE: : The success of RNA interference (RNAi) heavily depends on delivery systems that can enable spatiotemporal control over siRNA delivery. Herein, we developed micelleplexes (MPs) constructed from hypoxia-degradable, azobenzene-crosslinked oligoethylenimine (AO) and acid-responsive, photosensitizer-conjugated diblock copolymer PPDHP, to mediate efficient anti-tumor siRNA (siXIAP) delivery via programmed disintegration. MPs possessed high salt/serum stability and underwent acid-triggered PPDHP detachment to promote endolysosomal escape. Then, light irradiation aggravated hypoxia to trigger AO degradation and intracellular siXIAP release, which cooperated with photodynamic therapy to eradicate tumor cells. This study presents a new example of hypoxia-degradable polycation to mediate hypoxia-reinforced RNAi, and it also renders an effective strategy to overcome the complicated extracellular/intracellular barriers against systemic siRNA delivery.
Collapse
|
38
|
Chen WH, Chen QW, Chen Q, Cui C, Duan S, Kang Y, Liu Y, Liu Y, Muhammad W, Shao S, Tang C, Wang J, Wang L, Xiong MH, Yin L, Zhang K, Zhang Z, Zhen X, Feng J, Gao C, Gu Z, He C, Ji J, Jiang X, Liu W, Liu Z, Peng H, Shen Y, Shi L, Sun X, Wang H, Wang J, Xiao H, Xu FJ, Zhong Z, Zhang XZ, Chen X. Biomedical polymers: synthesis, properties, and applications. Sci China Chem 2022; 65:1010-1075. [PMID: 35505924 PMCID: PMC9050484 DOI: 10.1007/s11426-022-1243-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/01/2022] [Indexed: 02/07/2023]
Abstract
Biomedical polymers have been extensively developed for promising applications in a lot of biomedical fields, such as therapeutic medicine delivery, disease detection and diagnosis, biosensing, regenerative medicine, and disease treatment. In this review, we summarize the most recent advances in the synthesis and application of biomedical polymers, and discuss the comprehensive understanding of their property-function relationship for corresponding biomedical applications. In particular, a few burgeoning bioactive polymers, such as peptide/biomembrane/microorganism/cell-based biomedical polymers, are also introduced and highlighted as the emerging biomaterials for cancer precision therapy. Furthermore, the foreseeable challenges and outlook of the development of more efficient, healthier and safer biomedical polymers are discussed. We wish this systemic and comprehensive review on highlighting frontier progress of biomedical polymers could inspire and promote new breakthrough in fundamental research and clinical translation.
Collapse
Affiliation(s)
- Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Chunyan Cui
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350 China
| | - Shun Duan
- Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029 China
| | - Yongyuan Kang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - Yun Liu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
| | - Wali Muhammad
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027 China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215 China
| | - Chengqiang Tang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438 China
| | - Jinqiang Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
| | - Lei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nano-science, National Center for Nanoscience and Technology (NCNST), Beijing, 100190 China
| | - Meng-Hua Xiong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006 China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, 215123 China
| | - Kuo Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nano-science, National Center for Nanoscience and Technology (NCNST), Beijing, 100190 China
| | - Zhanzhan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - Xu Zhen
- Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093 China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
| | - Chaoliang He
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Xiqun Jiang
- Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093 China
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350 China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Huisheng Peng
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438 China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027 China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215 China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - Xuemei Sun
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438 China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nano-science, National Center for Nanoscience and Technology (NCNST), Beijing, 100190 China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006 China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190 China
| | - Fu-Jian Xu
- Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029 China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123 China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123 China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
| |
Collapse
|
39
|
Wang S, Huang Z, Lei Y, Han X, Tian D, Gong J, Liu M. Celastrol Alleviates Autoimmune Hepatitis Through the PI3K/AKT Signaling Pathway Based on Network Pharmacology and Experiments. Front Pharmacol 2022; 13:816350. [PMID: 35359864 PMCID: PMC8960436 DOI: 10.3389/fphar.2022.816350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/24/2022] [Indexed: 12/02/2022] Open
Abstract
Objective: This work aims to explore the potential targets and underlying therapeutic mechanisms of celastrol in autoimmune hepatitis (AIH) through network pharmacology and experiments on Laboratory Animals. Methods: A drug-target interaction network was constructed to predict the possible targets of celastrol and their potential relationship with the drug; docking studies were also performed for validation. This study used both acute and chronic rodent models of autoimmune hepatitis. Gross appearance of liver and spleen were obtained from murine models, hematoxylin-eosin staining and Sirius red staining were performed to examine hepatic inflammation and fibrosis respectively. By combining molecular docking and enrichment analysis results, the most prominent signaling pathway was selected and further confirmed by Western blot in AIH models administered with celastrol. Results: In total, 82 common targets of celastrol and AIH were obtained from databases, identified by network pharmacology, and adequately enriched. Among them, PIK3R1, SRC, MAPK1, AKT1, and HRAS were selected as the top 5 closely related targets to celastrol. They all performed effectively in molecular docking, with AKT1 and PIK3R1 exhibiting more-prominent binding energy. Subsequently, celastrol administration significantly ameliorated hepatitis and liver fibrosis by reducing AKT1 and PI3K phosphorylation in both acute liver injury and chronic models of autoimmune hepatitis. Conclusion: In summary, celastrol significantly attenuates autoimmune hepatitis by suppressing the PI3K/AKT signaling pathway, confirmed by validated animal models. These findings may help identify the mechanism involved in the anti-inflammatory action of celastrol in autoimmune hepatitis and provide ideas for future comprehensive studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Jin Gong
- *Correspondence: Jin Gong, ; Mei Liu,
| | - Mei Liu
- *Correspondence: Jin Gong, ; Mei Liu,
| |
Collapse
|
40
|
Zhou L, Li Y, Liang Q, Liu J, Liu Y. Combination therapy based on targeted nano drug co-delivery systems for liver fibrosis treatment: A review. J Drug Target 2022; 30:577-588. [PMID: 35179094 DOI: 10.1080/1061186x.2022.2044485] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Liver fibrosis is the hallmark of liver disease and occurs prior to the stages of cirrhosis and hepatocellular carcinoma. Any type of liver damage or inflammation can result in fibrosis. Fibrosis does not develop overnight, but rather as a result of the long-term action of injury factors. At present, however, there are no good treatment methods or specific drugs other than removing the pathogenic factors. Drug application is still limited, which means that drugs with good performance in vitro cannot achieve good therapeutic effects in vivo, owing to various factors such as poor drug targeting, large side effects, and strong hydrophobicity. Hepatic stellate cells (HSC) are the primary effector cells in liver fibrosis. The nano-drug delivery system is a new and safe drug delivery system that has many advantages which are widely used in the field of liver fibrosis. Drug resistance and side effects can be reduced when two or more drugs are used in combination drug delivery. Combination therapy of drugs with different targets has emerged as a novel approach to treating liver fibrosis, and the nano co-delivery system enhances the benefits of combination therapy. While nano co-delivery systems can maximize benefits while avoiding drug side effects, this is precisely the advantage of the nano co-delivery system. This review briefly described the pathogenesis and current treatment strategies, the different co-delivery systems of combination drugs in the nano delivery system, and targeting strategies for nano delivery systems on liver fibrosis therapy. Because of their superior performance, nano delivery systems and targeting drug delivery systems have received a lot of attention in the new drug delivery system. The new delivery systems offer a new pathway in the treatment of liver fibrosis, and it is believed that it can be a new treatment for fibrosis in the future. Nano co-delivery system of combination drugs and targeting strategies has proven the effectiveness of anti-fibrosis at the experimental level.
Collapse
Affiliation(s)
- Liyue Zhou
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yifan Li
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Qiangwei Liang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jinxia Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
41
|
Xu Y, Chen J, Jiang W, Zhao Y, Yang C, Wu Y, Li Q, Zhu C. Multiplexing Nanodrug Ameliorates Liver Fibrosis via ROS Elimination and Inflammation Suppression. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2102848. [PMID: 34758098 DOI: 10.1002/smll.202102848] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/31/2021] [Indexed: 06/13/2023]
Abstract
Liver fibrosis is the leading risk factor for hepatocellular carcinoma. Both oxidative stress and inflammation promote the progression of liver fibrosis, but existing therapeutic strategies tend to focus solely on one issue. Additionally, targeting of pathological microstructures is often neglected. Herein, an esterase-responsive carbon quantum dot-dexamethasone (CD-Dex) is developed for liver fibrosis therapy to simultaneously target pathological microstructures, scavenge reactive oxygen species (ROS), and suppress inflammation. Hepatocyte-targeting CD-Dex can efficiently eliminate the intrahepatic ROS, thereby inhibiting the activation of Kupffer cells, preventing further inflammation progression. Moreover, released dexamethasone (Dex) also suppresses inflammatory response by inhibiting the infiltration of inflammatory cells. Antifibrotic experiments demonstrate that CD-Dex significantly alleviates liver injury and collagen deposition, consequently preventing the progression of liver fibrosis. Taken together, these findings suggest that via ROS elimination and inflammation suppression, the newly developed multiplexing nanodrug exhibits great potential in liver fibrosis therapy.
Collapse
Affiliation(s)
- Youcui Xu
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, Intelligent Nanomedicine Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, P. R. China
| | - Jing Chen
- School of Life Sciences, Hefei Normal University, Hefei, 230601, P. R. China
| | - Wei Jiang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Yangyang Zhao
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Chen Yang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, P. R. China
| | - Yi Wu
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, Intelligent Nanomedicine Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, P. R. China
| | - Qianming Li
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, Intelligent Nanomedicine Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, P. R. China
| | - Chen Zhu
- Department of Orthopaedics, The First Affiliated Hospital of University of Science and Technology of China, Intelligent Nanomedicine Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, P. R. China
| |
Collapse
|
42
|
Bholakant R, Dong B, Zhou X, Huang X, Zhao C, Huang D, Zhong Y, Qian H, Chen W, Feijen J. Multi-functional polymeric micelles for chemotherapy-based combined cancer therapy. J Mater Chem B 2021; 9:8718-8738. [PMID: 34635905 DOI: 10.1039/d1tb01771c] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Currently, the therapeutic performance of traditional mono-chemotherapy on cancers remains unsatisfactory because of the tumor heterogeneity and multidrug resistance. In light of intricate tumor structures and distinct tumor microenvironments (TMEs), combinational therapeutic strategies with multiple anticancer drugs from different mechanisms can synergistically optimize the outcomes and concomitantly minimize the adverse effects during the therapy process. Extensive research on polymeric micelles (PMs) for biomedical applications has revealed the growing importance of nanomedicines for cancer therapy in the recent decade. Starting from traditional simple delivery systems, PMs have been extended to multi-faceted therapeutic strategies. Here we review and summarize the most recent advances in combinational therapy based on multifunctional PMs including a combination of multiple anticancer drugs, chemo-gene therapy, chemo-phototherapy and chemo-immunotherapy. The design approaches, action mechanisms and therapeutic applications of these nanodrugs are summarized. In addition, we highlight the opportunities and potential challenges associated with this promising field, which will provide new guidelines for advanced combinational cancer chemotherapy.
Collapse
Affiliation(s)
- Raut Bholakant
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Bin Dong
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiang Zhou
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Xin Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Changshun Zhao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Dechun Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Yinan Zhong
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Hongliang Qian
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Jan Feijen
- Department of Polymer Chemistry and Biomaterials, Faculty of Science and Technology, TECHMED Centre, University of Twente, P. O. Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
43
|
Huang J, Zheng C, Xiao H, Huang H, Wang Y, Lin M, Pang J, Wang Y, Yuan Y, Shuai X. A polymer‑calcium phosphate nanocapsule for RNAi-induced oxidative stress and cascaded chemotherapy. J Control Release 2021; 340:259-270. [PMID: 34740724 DOI: 10.1016/j.jconrel.2021.10.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/25/2021] [Accepted: 10/31/2021] [Indexed: 12/25/2022]
Abstract
As most of intracellular reactive oxygen species (ROS) is produced in the mitochondria, mitochondrial modulation of cancer cell is a promising strategy for maximizing the in situ-activable combination therapy of oxidative catastrophe and cascaded chemotherapy. Herein, a serum-stable polymer‑calcium phosphate (CaP) hybrid nanocapsule carrying siRNA against ADP-ribosylation factor 6 (Arf6) overexpressed in cancer cells and parent drug camptothecin (CPT), designated as PTkCPT/siRNA, was developed for the RNAi-induced oxidative catastrophe and cascaded chemotherapy. A copolymer of mPEG-P(Asp-co-TkCPT), covalently tethered with chemotherapeutic CPT via a ROS-labile dithioketal (Tk) linker, was synthesized and self-assembled into a PTkCPT micelle as a nanotemplate for the CaP mineralization. The as-prepared PTkCPT/siRNA nanoparticle showed a core-shell-distinct nanocapsule which was consisted of a spherical polymeric core enclosed within a CaP shell capable of releasing siRNA in response to lysosomal acidity. Blocking Arf6 signal pathway of cancer cells led to their mitochondrial aggregation and subsequently induced a burst of ROS for oxidative catastrophe, which further triggered the cascaded CPT chemotherapy via the breakage of ROS-labile dithioketal linker. This strategy of RNAi-induced oxidative catastrophe and cascaded chemotherapy resulted in a significant combination effect on cancer cell killing and tumor growth inhibition in mice with low side effects, and provided a promising paradigm for precise cancer therapy.
Collapse
Affiliation(s)
- Jinsheng Huang
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Chujie Zheng
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Hong Xiao
- Department of Medical Ultrasonic, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Huiling Huang
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| | - Yiyao Wang
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Minzhao Lin
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Jun Pang
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Yong Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China.
| | - Yuanyuan Yuan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
44
|
Ezhilararasan D, Lakshmi T, Raut B. Novel Nano-Based Drug Delivery Systems Targeting Hepatic Stellate Cells in the Fibrotic Liver. JOURNAL OF NANOMATERIALS 2021; 2021:1-9. [DOI: 10.1155/2021/4674046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Hepatic stellate cells (HSCs) exist in the liver’s perisinusoidal space, are phenotypically activated, and acquire myofibroblast-like phenotype. This phenotypic transformation is accountable for the accumulation and production of various extracellular matrix (ECM) proteins, involving different fibril-forming collagens in the perisinusoidal space, producing altered hepatic function and portal hypertension and increased vascular resistance, fibrosis, cirrhosis, and hepatocellular carcinoma. The activated HSCs/myofibroblasts are principal collagen-producing cells in the damaged liver. Therefore, fibrosis treatments are often targeting HSCs. HSCs store most of the total body’s retinol in their cytoplasm, and hence, antifibrotic nanomedicines are often targeted with vitamin A decoration. Vitamin A-decorated nanomedicines with siRNAs for transforming growth factor-beta, collagen, and connective tissue growth factors target to inhibit fibrogenesis and the ECM-associated gene expressions, leading to fibrosis regression. Similarly, a variety of miRNAs play pro- and antifibrotic function. In the fibrotic liver, the profibrotic miRNAs are targeted with their respective antagomir and the antifibrotic miRNAs are targeted with their respective agomirs along with HSC-specific nanodecoration. These miRNA treatments reduce fibrogenesis by downregulation of ECM-related gene expressions. However, liver fibrosis is caused by the upregulation of a different type of profibrotic signaling pathways associated with ECM accumulation in the fibrotic liver. Therefore, specific gene silencing by siRNAs or targeting particularly miRNA may also not effectively reduce fibrosis to a greater extent. However, nanodecoration of a drug is useful to deliver drugs into activated HSCs in the injured liver. Therefore, the aim of this review is to focus on targeted drug delivery towards activated HSCs in the persistently damaged liver.
Collapse
Affiliation(s)
- Devaraj Ezhilararasan
- Department of Pharmacology, The Blue Lab, Molecular Medicine and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 600077, India
| | - Thangavelu Lakshmi
- Department of Pharmacology, The Blue Lab, Molecular Medicine and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 600077, India
| | - Biond Raut
- Department of Pharmacology, Kathmandu Medical College Bhaktapur Duwakot, Kathmandu University, Dhulikhel, Kavre, Nepal
| |
Collapse
|
45
|
Liu Y, Wu W, Wang Y, Han S, Yuan Y, Huang J, Shuai X, Peng Z. Recent development of gene therapy for pancreatic cancer using non-viral nanovectors. Biomater Sci 2021; 9:6673-6690. [PMID: 34378568 DOI: 10.1039/d1bm00748c] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Pancreatic cancer (PC), characterized by its dense desmoplastic stroma and hypovascularity, is one of the most lethal cancers with a poor prognosis in the world. Traditional treatments such as chemotherapy, radiotherapy, and targeted therapy show little benefit in the survival rate in patients with advanced PC due to the poor penetration and resistance of drugs, low radiosensitivity, or severe side effects. Gene therapy can modify the morbific and drug-resistant genes as well as insert the tumor-suppressing genes, which has been shown to have great potential in PC treatment. The development of safe non-viral vectors for the highly efficient delivery of nucleic acids is essential for effective gene therapy, and has been attracting much attention. In this review, we first summarized the PC-promoting genes and gene therapies using plasmid DNA, mRNA, miRNA/siRNA-based RNA interference technology, and genome editing technology. Second, the commonly used non-viral nanovector and theranostic gene delivery nanosystem, especially the tumor microenvironment-sensitive delivery nanosystem and the cell/tumor-penetrating delivery nanosystem, were introduced. Third, a combination of non-viral nanovector-based gene therapy and other therapies, such as immunotherapy, chemotherapy, photothermal therapy (PTT), and photodynamic therapy (PDT), for PDAC treatment was discussed. Finally, a number of clinical trials have demonstrated the proof-of-principle that gene therapy or the combination of gene therapy and chemotherapy using non-viral vectors can inhibit the progression of PC. Although most of the non-viral vector-based gene therapies and their combination therapy are still under preclinical research, the development of genetics, molecular biology, and novel vectors would promote the clinical transformation of gene therapy.
Collapse
Affiliation(s)
- Yu Liu
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Wei Wu
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Yiyao Wang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Shisong Han
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yuanyuan Yuan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jinsheng Huang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Zhao Peng
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
46
|
Yu C, Li L, Hu P, Yang Y, Wei W, Deng X, Wang L, Tay FR, Ma J. Recent Advances in Stimulus-Responsive Nanocarriers for Gene Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2100540. [PMID: 34306980 PMCID: PMC8292848 DOI: 10.1002/advs.202100540] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/07/2021] [Indexed: 05/29/2023]
Abstract
Gene therapy provides a promising strategy for curing monogenetic disorders and complex diseases. However, there are challenges associated with the use of viral delivery vectors. The advent of nanomedicine represents a quantum leap in the application of gene therapy. Recent advances in stimulus-responsive nonviral nanocarriers indicate that they are efficient delivery systems for loading and unloading of therapeutic nucleic acids. Some nanocarriers are responsive to cues derived from the internal environment, such as changes in pH, redox potential, enzyme activity, reactive oxygen species, adenosine triphosphate, and hypoxia. Others are responsive to external stimulations, including temperature gradients, light irradiation, ultrasonic energy, and magnetic field. Multiple stimuli-responsive strategies have also been investigated recently for experimental gene therapy.
Collapse
Affiliation(s)
- Cheng Yu
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Long Li
- Department of OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Pei Hu
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Yan Yang
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Wei Wei
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Xin Deng
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Lu Wang
- Department of OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | | | - Jingzhi Ma
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| |
Collapse
|
47
|
Dai X, Zeng Y, Zhang H, Gu Z, Gong Q, Luo K. Advances on Nanomedicines for Diagnosis and Theranostics of Hepatic Fibrosis. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Xinghang Dai
- Huaxi MR Research Center (HMRRC) Department of Radiology Functional and molecular imaging Key Laboratory of Sichuan Province West China Hospital Sichuan University Chengdu 610041 China
- West China School of Medicine Sichuan University Chengdu 610041 China
| | - Yujun Zeng
- Huaxi MR Research Center (HMRRC) Department of Radiology Functional and molecular imaging Key Laboratory of Sichuan Province West China Hospital Sichuan University Chengdu 610041 China
| | - Hu Zhang
- Huaxi MR Research Center (HMRRC) Department of Radiology Functional and molecular imaging Key Laboratory of Sichuan Province West China Hospital Sichuan University Chengdu 610041 China
- Amgen Bioprocessing Centre Keck Graduate Institute CA 91711 USA
| | - Zhongwei Gu
- Research Unit of Psychoradiology Chinese Academy of Medical Sciences Chengdu 610041 China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC) Department of Radiology Functional and molecular imaging Key Laboratory of Sichuan Province West China Hospital Sichuan University Chengdu 610041 China
- Research Unit of Psychoradiology Chinese Academy of Medical Sciences Chengdu 610041 China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC) Department of Radiology Functional and molecular imaging Key Laboratory of Sichuan Province West China Hospital Sichuan University Chengdu 610041 China
- Research Unit of Psychoradiology Chinese Academy of Medical Sciences Chengdu 610041 China
| |
Collapse
|
48
|
Liu S, Deng S, Li X, Cheng D. Size- and Surface- Dual Engineered Small Polyplexes for Efficiently Targeting Delivery of siRNA. Molecules 2021; 26:3238. [PMID: 34072265 PMCID: PMC8199253 DOI: 10.3390/molecules26113238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 01/15/2023] Open
Abstract
Though siRNA-based therapy has achieved great progress, efficient siRNA delivery remains a challenge. Here, we synthesized a copolymer PAsp(-N=C-PEG)-PCys-PAsp(DETA) consisting of a poly(aspartate) block grafted with comb-like PEG side chains via a pH-sensitive imine bond (PAsp(-N=C-PEG) block), a poly(l-cysteine) block with a thiol group (PCys block), and a cationic poly(aspartate) block grafted with diethylenetriamine (PAsp(DETA) block). The cationic polymers efficiently complexed siRNA into polyplexes, showing a sandwich-like structure with a PAsp(-N=C-PEG) out-layer, a crosslinked PCys interlayer, and a complexing core of siRNA and PAsp(DETA). Low pH-triggered breakage of pH-sensitive imine bonds caused PEG shedding. The disulfide bond-crosslinking and pH-triggered PEG shedding synergistically decreased the polyplexes' size from 75 nm to 26 nm. To neutralize excessive positive charges and introduce the targeting ligand, the polyplexes without a PEG layer were coated with an anionic copolymer modified with the targeting ligand lauric acid. The resulting polyplexes exhibited high transfection efficiency and lysosomal escape capacity. This study provides a promising strategy to engineer the size and surface of polyplexes, allowing long blood circulation and targeted delivery of siRNA.
Collapse
Affiliation(s)
- Shuang Liu
- PCFM Lab of Ministry of Education & Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (X.L.)
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Shaohui Deng
- PCFM Lab of Ministry of Education & Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (X.L.)
| | - Xiaoxia Li
- PCFM Lab of Ministry of Education & Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (X.L.)
| | - Du Cheng
- PCFM Lab of Ministry of Education & Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (X.L.)
| |
Collapse
|
49
|
Khurana A, Sayed N, Allawadhi P, Weiskirchen R. It's all about the spaces between cells: role of extracellular matrix in liver fibrosis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:728. [PMID: 33987426 PMCID: PMC8106070 DOI: 10.21037/atm-20-2948] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 07/22/2020] [Indexed: 12/19/2022]
Abstract
Liver fibrosis is one of the leading complications of a variety of chronic liver disorders, including the nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, liver cirrhosis and liver failure. The progression of liver fibrosis is driven by chronic inflammation, which activates the secretory fibroblasts to the myofibroblast phenotype. These specialized liver cells are called as hepatic stellate cells (HSCs). The excessive extracellular matrix (ECM) secretion creates a large number of complications. Fibrosis is the result of imbalance between the matrix synthesizing and matrix degrading factors. The major ECM proteins include the matrix metalloproteinases (MMPs), tissue inhibitor of metalloproteinases (TIMPs), lysyl oxidases (LOX), lysyl oxidase-like (LOXLs) enzymes, tenascins and others. These ECM proteins present novel avenues for the therapeutics of liver fibrosis. The current review highlights the major role played by these critical matrix proteins in liver fibrosis. Further, some of the targeted formulations used against these proteins are discussed and suggestions are provided to select the course of research for successful clinical translation of basic research findings for the amelioration of liver fibrosis.
Collapse
Affiliation(s)
- Amit Khurana
- Center for Biomedical Engineering (CBME), Indian Institute of Technology (IIT), Hauz Khas, New Delhi, India
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Nilofer Sayed
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science, Rajendranagar, Hyderabad, Telangana, India
| | - Prince Allawadhi
- Department of Biotechnology, Indian Institute of Technology (IIT), Roorkee, Uttarakhand, India
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
50
|
Abstract
In this review, we summarized recent advances in the development and biological applications of polymeric nanoparticles embedded with superparamagnetic iron oxide nanoparticles (SPIONs). Superparamagnetic polymeric nanoparticles include core-shell nanoparticles, superparamagnetic polymeric micelles and superparamagnetic polymersomes. They have potential for various biomedical applications, including magnetic resonance imaging (MRI) contrast agents, drug delivery, detection of bacteria, viruses and proteins, etc. Finally, the challenges in the design and preparation of superparamagnetic nanoparticles towards clinical applications are explored and the prospects in this field are proposed.
Collapse
Affiliation(s)
- Yufen Xiao
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China.
| | | |
Collapse
|