1
|
Li T, Fu W, Li X, Huo Y, Ji H, Liang T, Zhang R. Quercetin-Loaded Melanin Nanoparticles Decorated with Collagenase Mediates Synergistic Immunomodulation and Restores Extracellular Matrix Homeostasis in Liver Fibrosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:386-397. [PMID: 39692444 DOI: 10.1021/acsami.4c15494] [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: 12/19/2024]
Abstract
Liver fibrosis is a chronic disease that lacks effective drug treatment. Chronic damage and inflammation lead to the formation of collagen and fibrous scars. However, the excessive accumulation of collagen I significantly hinders the delivery of drugs into liver tissue. Therefore, this study developed a quercetin-loaded melanin nanoparticle codecorated collagenase (MNP-QUE-COL) for the treatment of liver fibrosis. These results showed that MNP-QUE-COL degraded excessive collagen I, thereby efficiently delivering melanin and quercetin into the liver tissue. MNP-QUE-COL exhibited optimal PA/MRI dual-mode imaging ability. In addition, the synergistic anti-inflammatory and reactive oxygen species scavenging function of quercetin and melanin was achieved by regulation of M1-M2 macrophage polarization and inhibition of pro-inflammatory cytokine release, reshaping the imbalanced extracellular interstitial inflammatory environment. The results of this research suggest that MNP-QUE-COL is a safe and efficient therapeutic nanoplatform for liver fibrosis, showing promise as a potential therapeutic strategy for liver fibrosis and associated diseases.
Collapse
Affiliation(s)
- Tingting Li
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Weihua Fu
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Xueqi Li
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Yuanqing Huo
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Huifang Ji
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Taigang Liang
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
- Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan 030012, China
| |
Collapse
|
2
|
Dong Z, Wang Y, Jin W. Liver cirrhosis: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2024; 5:e721. [PMID: 39290252 PMCID: PMC11406049 DOI: 10.1002/mco2.721] [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: 04/26/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Liver cirrhosis is the end-stage of chronic liver disease, characterized by inflammation, necrosis, advanced fibrosis, and regenerative nodule formation. Long-term inflammation can cause continuous damage to liver tissues and hepatocytes, along with increased vascular tone and portal hypertension. Among them, fibrosis is the necessary stage and essential feature of liver cirrhosis, and effective antifibrosis strategies are commonly considered the key to treating liver cirrhosis. Although different therapeutic strategies aimed at reversing or preventing fibrosis have been developed, the effects have not be more satisfactory. In this review, we discussed abnormal changes in the liver microenvironment that contribute to the progression of liver cirrhosis and highlighted the importance of recent therapeutic strategies, including lifestyle improvement, small molecular agents, traditional Chinese medicine, stem cells, extracellular vesicles, and gut remediation, that regulate liver fibrosis and liver cirrhosis. Meanwhile, therapeutic strategies for nanoparticles are discussed, as are their possible underlying broad application and prospects for ameliorating liver cirrhosis. Finally, we also reviewed the major challenges and opportunities of nanomedicine‒biological environment interactions. We hope this review will provide insights into the pathogenesis and molecular mechanisms of liver cirrhosis, thus facilitating new methods, drug discovery, and better treatment of liver cirrhosis.
Collapse
Affiliation(s)
- Zihe Dong
- The First School of Clinical Medicine Lanzhou University Lanzhou People's Republic of China
- Institute of Cancer Neuroscience Medical Frontier Innovation Research Center The First Hospital of Lanzhou University Lanzhou People's Republic of China
| | - Yeying Wang
- The First School of Clinical Medicine Lanzhou University Lanzhou People's Republic of China
- Institute of Cancer Neuroscience Medical Frontier Innovation Research Center The First Hospital of Lanzhou University Lanzhou People's Republic of China
| | - Weilin Jin
- The First School of Clinical Medicine Lanzhou University Lanzhou People's Republic of China
- Institute of Cancer Neuroscience Medical Frontier Innovation Research Center The First Hospital of Lanzhou University Lanzhou People's Republic of China
| |
Collapse
|
3
|
Bakrania A, Mo Y, Zheng G, Bhat M. RNA nanomedicine in liver diseases. Hepatology 2024:01515467-990000000-00569. [PMID: 37725757 DOI: 10.1097/hep.0000000000000606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023]
Abstract
The remarkable impact of RNA nanomedicine during the COVID-19 pandemic has demonstrated the expansive therapeutic potential of this field in diverse disease contexts. In recent years, RNA nanomedicine targeting the liver has been paradigm-shifting in the management of metabolic diseases such as hyperoxaluria and amyloidosis. RNA nanomedicine has significant potential in the management of liver diseases, where optimal management would benefit from targeted delivery, doses titrated to liver metabolism, and personalized therapy based on the specific site of interest. In this review, we discuss in-depth the different types of RNA and nanocarriers used for liver targeting along with their specific applications in metabolic dysfunction-associated steatotic liver disease, liver fibrosis, and liver cancers. We further highlight the strategies for cell-specific delivery and future perspectives in this field of research with the emergence of small activating RNA, circular RNA, and RNA base editing approaches.
Collapse
Affiliation(s)
- Anita Bakrania
- Department of Medicine, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Department of Medicine, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yulin Mo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Mamatha Bhat
- Department of Medicine, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Department of Medicine, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Division of Gastroenterology, University Health Network and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Vu HT, Nguyen VD, Ikenaga H, Matsubara T. Application of PPAR Ligands and Nanoparticle Technology in Metabolic Steatohepatitis Treatment. Biomedicines 2024; 12:1876. [PMID: 39200340 PMCID: PMC11351628 DOI: 10.3390/biomedicines12081876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD/MASH) is a major disease worldwide whose effective treatment is challenging. Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear receptor superfamily and function as ligand-activated transcription factors. To date, three distinct subtypes of PPARs have been characterized: PPARα, PPARβ/δ, and PPARγ. PPARα and PPARγ are crucial regulators of lipid metabolism that modulate the transcription of genes involved in fatty acid (FA), bile acid, and cholesterol metabolism. Many PPAR agonists, including natural (FAs, eicosanoids, and phospholipids) and synthetic (fibrate, thiazolidinedione, glitazar, and elafibranor) agonists, have been developed. Furthermore, recent advancements in nanoparticles (NPs) have led to the development of new strategies for MASLD/MASH therapy. This review discusses the applications of specific cell-targeted NPs and highlights the potential of PPARα- and PPARγ-targeted NP drug delivery systems for MASLD/MASH treatment.
Collapse
Affiliation(s)
- Hung Thai Vu
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Osaka, Japan; (H.T.V.); (V.D.N.)
| | - Vien Duc Nguyen
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Osaka, Japan; (H.T.V.); (V.D.N.)
| | - Hiroko Ikenaga
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Osaka, Japan
| | - Tsutomu Matsubara
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Osaka, Japan; (H.T.V.); (V.D.N.)
- Research Institute for Light-induced Acceleration System (RILACS), Osaka Metropolitan University, Sakai 599-8570, Osaka, Japan
| |
Collapse
|
5
|
Liu J, Liu J, Mu W, Ma Q, Zhai X, Jin B, Liu Y, Zhang N. Delivery Strategy to Enhance the Therapeutic Efficacy of Liver Fibrosis via Nanoparticle Drug Delivery Systems. ACS NANO 2024; 18:20861-20885. [DOI: 10.1021/acsnano.4c02380] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Jie Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jinhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiangyu Zhai
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Bin Jin
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
6
|
Peng Y, Qu R, Xu S, Bi H, Guo D. Regulatory mechanism and therapeutic potentials of naringin against inflammatory disorders. Heliyon 2024; 10:e24619. [PMID: 38317884 PMCID: PMC10839891 DOI: 10.1016/j.heliyon.2024.e24619] [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: 08/04/2023] [Revised: 12/02/2023] [Accepted: 01/11/2024] [Indexed: 02/07/2024] Open
Abstract
Naringin is a natural flavonoid with therapeutic properties found in citrus fruits and an active natural product from herbal plants. Naringin has become a focus of attention in recent years because of its ability to actively participate in the body's immune response and maintain the integrity of the immune barrier. This review aims to elucidate the mechanism of action and therapeutic efficacy of naringin in various inflammatory diseases and to provide a valuable reference for further research in this field. The review provided the chemical structure, bioavailability, pharmacological properties, and pharmacokinetics of naringin and found that naringin has good therapeutic potential for inflammatory diseases, exerting anti-inflammatory, anti-apoptotic, anti-oxidative stress, anti-ulcerative and detoxifying effects in the disease. Moreover, we found that the great advantage of naringin treatment is that it is safe and can even alleviate the toxic side effects associated with some of the other drugs, which may become a highlight of naringin research. Naringin, an active natural product, plays a significant role in systemic diseases' anti-inflammatory and antioxidant regulation through various signaling pathways and molecular mechanisms.
Collapse
Affiliation(s)
- Yuan Peng
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Ruyi Qu
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Shuqin Xu
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| |
Collapse
|
7
|
Shinn J, Park S, Lee S, Park N, Kim S, Hwang S, Moon JJ, Kwon Y, Lee Y. Antioxidative Hyaluronic Acid-Bilirubin Nanomedicine Targeting Activated Hepatic Stellate Cells for Anti-Hepatic-Fibrosis Therapy. ACS NANO 2024; 18:4704-4716. [PMID: 38288705 DOI: 10.1021/acsnano.3c06107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Liver fibrosis is a life-threatening and irreversible disease. The fibrosis process is largely driven by hepatic stellate cells (HSCs), which undergo transdifferentiation from an inactivated state to an activated one during persistent liver damage. This activated state is responsible for collagen deposition in liver tissue and is accompanied by increased CD44 expression on the surfaces of HSCs and amplified intracellular oxidative stress, which contributes to the fibrosis process. To address this problem, we have developed a strategy that combines CD44-targeting of activated HSCs with an antioxidative approach. We developed hyaluronic acid-bilirubin nanoparticles (HABNs), composed of endogenous bilirubin, an antioxidant and anti-inflammatory bile acid, and hyaluronic acid, an endogenous CD44-targeting glycosaminoglycan biopolymer. Our findings demonstrate that intravenously administered HABNs effectively targeted the liver, particularly activated HSCs, in fibrotic mice with choline-deficient l-amino acid-defined high-fat diet (CD-HFD)-induced nonalcoholic steatohepatitis (NASH). HABNs were able to inhibit HSC activation and proliferation and collagen production. Furthermore, in a murine CD-HFD-induced NASH fibrosis model, intravenously administered HABNs showed potent fibrotic modulation activity. Our study suggests that HABNs have the potential to serve as a targeted anti-hepatic-fibrosis therapy by modulating activated HSCs via CD44-targeting and antioxidant strategies. This strategy could also be applied to various ROS-related diseases in which CD44-overexpressing cells play a pivotal role.
Collapse
Affiliation(s)
- Jongyoon Shinn
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul 03760, South Korea
| | - Seojeong Park
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul 03760, South Korea
| | - Seonju Lee
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul 03760, South Korea
| | - Nayoon Park
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul 03760, South Korea
| | - Seojeong Kim
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul 03760, South Korea
| | - Seohui Hwang
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul 03760, South Korea
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Youngjoo Kwon
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul 03760, South Korea
| | - Yonghyun Lee
- Department of Pharmacy, College of Pharmacy, Ewha Womans University, Seoul 03760, South Korea
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Zhang YW, Hou LS, Xing JH, Zhang TR, Zhou SY, Zhang BL. Two-Membrane Hybrid Nanobiomimetic Delivery System for Targeted Autophagy Inhibition of Activated Hepatic Stellate Cells To Synergistically Treat Liver Fibrosis. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37899504 DOI: 10.1021/acsami.3c11046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Liver fibrosis is one of the most common and highly prevalent chronic liver diseases caused by multiple pathogenic factors, and there is still no effective therapeutic drugs up to now. The activated hepatic stellate cells (aHSCs) are the main executor in liver fibrosis, and the autophagy plays a key role in the proliferation and differentiation of aHSCs, which promotes the development of liver fibrosis. However, autophagy has the opposite effect on the different kinds of liver cells in the development of liver fibrosis, and the clinical treatment has been limited by the poor selectivity and inefficient drug delivery to aHSCs. Therefore, in this study, a liposome (Lip) and exosome (Exo) two-membrane hybrid nanobiomimetic delivery system HCQ@VA-Lip-Exo was designed, which was modified by vitamin A (VA) to target the aHSCs and carried the autophagy inhibitor hydroxychloroquine (HCQ). The experimental results in vitro and in vivo revealed that the constructed aHSC-targeted hybrid delivery system HCQ@VA-Lip-Exo combined with the benefits of HCQ and exosomes derived from bone marrow mesenchymal stem cells. HCQ@VA-Lip-Exo had good aHSC-targeted delivery ability, effective autophagy inhibition, and synergistical anti-liver fibrosis performance, thus reducing the production and deposition of the extracellular matrix to inhibit the liver fibrosis. This combined strategy provided a potential idea for the construction and clinical application of a two-membrane hybrid delivery system as an effective targeted therapy of liver fibrosis.
Collapse
Affiliation(s)
- Yao-Wen Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Li-Shuang Hou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Jie-Hua Xing
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Tang-Rui Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Si-Yuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
- Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Bang-Le Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
- Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| |
Collapse
|
11
|
Xiang L, Wang X, Jiao Q, Shao Y, Luo R, Zhang J, Zheng X, Zhou S, Chen Y. Selective inhibition of glycolysis in hepatic stellate cells and suppression of liver fibrogenesis with vitamin A-derivative decorated camptothecin micelles. Acta Biomater 2023; 168:497-514. [PMID: 37507035 DOI: 10.1016/j.actbio.2023.07.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/14/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023]
Abstract
The persistent transformation of quiescent hepatic stellate cells (HSCs) into myofibroblasts (MFs) and the excessive proliferation of MF-HSCs in the liver contribute to the pathogenesis of liver fibrosis, cirrhosis, and liver cancer. Glycolysis inhibition of MF-HSCs can reverse their MF phenotype and suppress their abnormal expansion. Here, we have developed vitamin A-derivative (VA) decorated PEG-PCL polymeric micelles to encapsulate the labile and hydrophobic camptothecin (CPT) and direct its active attack on HSCs, selectively inhibiting of HIF-1α and cellular glycolysis, ultimately repressing hepatic fibrogenesis. The obtained micelles exhibited a good stability, biocompatibility, pH sensitivity, and exceptional HSC-targetability, allowing an efficient accumulation of their carried CPT in acutely and chronically injured livers. On their intracellular release of CPT specifically in MF-HSCs, these CPT micelles nicely inhibited the HIF-1α and a series of glycolytic players in MF-HSCs and prominently suppressed their proliferation and MF phenotypic characteristics. Accordingly, on in vitro administration to the mice challenged by CCl4 or subjected to bile duct ligation, these VA-decorated CPT micelles ameliorated the pathological symptoms of the livers, as evidenced by the significant reduction in serum levels of ALT and AST, infiltration of inflammatory cells, and collagen accumulation, the drastic down-regulation of multiple fibrotic genes, and the good recovery of attenuated hepatocyte CYP2E1 and lipogenesis regulator PPARγ. Overall, the CPT carried by VA-decorated PEG-PCL polymeric micelles can selectively inhibit the glycolysis and expansion of HSCs and thus suppress fibrogenesis, providing an original and effective approach for anti-fibrotic therapy. STATEMENT OF SIGNIFICANCE: Our work introduces an innovative antifibrotic drug system that is developed upon the active targeting of CPT and aims for the fate reversal of HSCs. Through HSC-targeted delivery achieved by PEG-PCL polymeric micelles decorated with vitamin A-derivatives, CPT significantly suppressed the expressions of HIF-1α and glycolytic enzymes in MF-HSCs, as well as their pathologic expansion in mouse livers. It effectively ameliorated chronic liver fibrosis in mice induced by CCl4 injection or BDL and restored the damaged liver structure and function. These compelling findings demonstrate the therapeutic potential of glycolytic HSC-targeting in combating fibrosis and related disorders and thus provide new promise for future clinical management of such prevalent and life-threatening conditions.
Collapse
Affiliation(s)
- Li Xiang
- Hengyang Medical School, University of South China, Hengyang, Hunan, 410001, China
| | - Xin Wang
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Qiangqiang Jiao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yaru Shao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Rui Luo
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Jie Zhang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Xiaotong Zheng
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Shaobing Zhou
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Yuping Chen
- Hengyang Medical School, University of South China, Hengyang, Hunan, 410001, China; School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China.
| |
Collapse
|
12
|
Cha GD, Kim M, Park OK, Sunwoo SH, Kang T, Lee WH, Nam S, Hyeon T, Choi SH, Kim DH. Minimally-Invasive and In-Vivo Hydrogel Patterning Method for In Situ Fabrication of Implantable Hydrogel Devices. SMALL METHODS 2023; 7:e2300032. [PMID: 37199695 DOI: 10.1002/smtd.202300032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/19/2023] [Indexed: 05/19/2023]
Abstract
Despite advances in a wide range of device applications of hydrogels, including implantable ones, a method for deploying patterned hydrogel devices into the body in a minimally-invasive manner is not available yet. However, in situ patterning of the hydrogel in vivo has an obvious advantage, by which incision surgery for implantation of the hydrogel device can be avoided. Here, a minimally-invasive and in vivo hydrogel patterning method for in situ fabrication of implantable hydrogel devices is presented. The sequential application of injectable hydrogels and enzymes, with assistance of minimally-invasive surgical instruments, enables the in vivo and in situ hydrogel patterning. This patterning method can be achieved by adopting an appropriate combination of the sacrificial mold hydrogel and the frame hydrogel, in consideration of unique material properties of the hydrogels such as high softness, facile mass transfer, biocompatibility, and diverse crosslinking mechanisms. In vivo and in situ patterning of the hydrogels functionalized with nanomaterials is also demonstrated to fabricate the wireless heater and tissue scaffold, showcasing broad applicability of the patterning method.
Collapse
Affiliation(s)
- Gi Doo Cha
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Minjeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ok Kyu Park
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Sung-Hyuk Sunwoo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Taegyu Kang
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Wang Hee Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seonghyeon Nam
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Hong Choi
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Dae-Hyeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
13
|
Zheng D, Pisano F, Collard L, Balena A, Pisanello M, Spagnolo B, Mach-Batlle R, Tantussi F, Carbone L, De Angelis F, Valiente M, de la Prida LM, Ciracì C, De Vittorio M, Pisanello F. Toward Plasmonic Neural Probes: SERS Detection of Neurotransmitters through Gold-Nanoislands-Decorated Tapered Optical Fibers with Sub-10 nm Gaps. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2200902. [PMID: 36479741 DOI: 10.1002/adma.202200902] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 10/21/2022] [Indexed: 06/17/2023]
Abstract
Integration of plasmonic nanostructures with fiber-optics-based neural probes enables label-free detection of molecular fingerprints via surface-enhanced Raman spectroscopy (SERS), and it represents a fascinating technological horizon to investigate brain function. However, developing neuroplasmonic probes that can interface with deep brain regions with minimal invasiveness while providing the sensitivity to detect biomolecular signatures in a physiological environment is challenging, in particular because the same waveguide must be employed for both delivering excitation light and collecting the resulting scattered photons. Here, a SERS-active neural probe based on a tapered optical fiber (TF) decorated with gold nanoislands (NIs) that can detect neurotransmitters down to the micromolar range is presented. To do this, a novel, nonplanar repeated dewetting technique to fabricate gold NIs with sub-10 nm gaps, uniformly distributed on the wide (square millimeter scale in surface area), highly curved surface of TF is developed. It is experimentally and numerically shown that the amplified broadband near-field enhancement of the high-density NIs layer allows for achieving a limit of detection in aqueous solution of 10-7 m for rhodamine 6G and 10-5 m for serotonin and dopamine through SERS at near-infrared wavelengths. The NIs-TF technology is envisioned as a first step toward the unexplored frontier of in vivo label-free plasmonic neural interfaces.
Collapse
Affiliation(s)
- Di Zheng
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Filippo Pisano
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Liam Collard
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Antonio Balena
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Marco Pisanello
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Barbara Spagnolo
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Rosa Mach-Batlle
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Francesco Tantussi
- Istituto Italiano di Tecnologia, Center for Convergent Technologies, Genova, 16163, Italy
| | - Luigi Carbone
- CNR NANOTEC - Institute of Nanotechnology, University of Salento, Lecce, 73100, Italy
| | - Francesco De Angelis
- Istituto Italiano di Tecnologia, Center for Convergent Technologies, Genova, 16163, Italy
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Center (CNIO), Madrid, 28029, Spain
| | | | - Cristian Ciracì
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Massimo De Vittorio
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
- Dipartimento di Ingegneria Dell'Innovazione, Università del Salento, Lecce, 73100, Italy
| | - Ferruccio Pisanello
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| |
Collapse
|
14
|
Wu Y, Zhu K, Zhang X, Du W, Song J, Yang H. Emerging plasmonic nanoparticles and their assemblies for cancer radiotherapy. Adv Drug Deliv Rev 2023; 194:114710. [PMID: 36708774 DOI: 10.1016/j.addr.2023.114710] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/07/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023]
Abstract
Plasmonic nanoparticles and their assemblies have been widely used in biosensing, optical imaging, and biomedicine over the past few decades. Especially in the field of radiotherapy, the physicochemical properties of high-Z plasmonic nanomaterials endow them with the ability to sensitize radiotherapy. Compared with single particles, the assembled structure with tunable properties leads to versatile applications in drug delivery and cancer treatment. In this review, we focus on plasmonic nanoparticles and their assemblies for cancer radiotherapy. First, the sensitization mechanism of plasmonic radiosensitizers is briefly introduced. Subsequently, the recent progress in cancer radiotherapy is systematically discussed according to the structure and shape classification. Finally, the current challenges and future perspectives in this field are also discussed in detail.
Collapse
Affiliation(s)
- Ying Wu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, PR China
| | - Kang Zhu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, PR China
| | - Xuan Zhang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350116, PR China
| | - Wei Du
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350116, PR China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 10010, PR China.
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350116, PR China.
| |
Collapse
|
15
|
Jiang M, Huang C, Wu Q, Su Y, Wang X, Xuan Z, Wang Y, Xu F, Ge C. Sini San ameliorates CCl4-induced liver fibrosis in mice by inhibiting AKT-mediated hepatocyte apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:115965. [PMID: 36460296 DOI: 10.1016/j.jep.2022.115965] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/12/2022] [Accepted: 11/21/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sini San (SNS) is recorded in Zhang Zhongjing's "Treatise on Typhoids" and is used in the treatment of non-alcoholic fatty liver disease, hepatitis, and other liver diseases, with good efficacy in liver fibrosis. However, its anti-liver fibrosis mechanism remains unclear. AIM OF THE STUDY This study aimed to evaluate the ameliorative effect of SNS on carbon tetrachloride (CCl4)-induced liver fibrosis in mice and the underlying mechanisms. MATERIALS AND METHODS The active ingredients in the water extract of SNS were determined using high-performance liquid chromatography (HPLC). CCl4-induced liver fibrosis mice were subsequently treated with different doses of SNS for 3 weeks, and AST, ALT, and T-BIL were detected in the serum. The pathological characteristics of the liver were observed using hematoxylin and eosin (H&E) and Masson's staining. Hepatocyte apoptosis was assessed using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The proteins expression of PI3K, p-PI3K, AKT, p-AKT, FXR, caspase-8, Bax, and Bcl-2 was analyzed using western blotting and immunofluorescence. FXR mRNA expression was measured using quantitative real-time reverse transcription-polymerase chain reaction (RT-qPCR). Using network pharmacology and bioinformatics to search for active ingredients that regulate PI3K/AKT signaling in the SNS. The material basis for regulating PI3K/AKT signaling in SNS was searched using network pharmacology and bioinformatics. Based on the network pharmacology results, isorhamnetin or SNS-containing serum was added to HepG2 cells stimulated with TNF-α. The Cell Counting Kit (CCK)-8 assay was used to analyze cell viability and apoptosis of HepG2 cells was detected using flow cytometry. RESULTS SNS reduced serum levels of AST, ALT and T-BIL, down-regulated caspase-8 protein expression and the ratio of Bcl-2/Bax protein expression, and improved apoptosis in liver fibrosis mice. In addition, SNS downregulated the ratio of p-PI3K/PI3K and p-AKT/AKT protein expression and increased FXR expression. Network pharmacology studies showed that quercetin, kaempferol and isorhamnetin in SNS can bind to AKT. In vitro experiments showed that isorhamnetin inhibited HepG2 cell apoptosis, upregulated FXR expression and suppressed AKT activity, whereas AKT inhibitors blocked the effects of isorhamnetin. The effect of the SNS-containing serum was similar to that of isorhamnetin. CONCLUSION SNS ameliorated the progression of fibrosis and improved hepatocyte apoptosis in liver fibrosis mice. The anti-apoptotic mechanism was related to the inhibition of AKT-mediated down-regulation of FXR expression by its active ingredient, isorhamnetin.
Collapse
Affiliation(s)
- Meijie Jiang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Chunmei Huang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China
| | - Qiong Wu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China
| | - Yong Su
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China
| | - Xinming Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China
| | - Zihua Xuan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, PR China
| | - Yunlai Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, PR China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, PR China
| | - Fan Xu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, PR China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, PR China.
| | - Chaoliang Ge
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China.
| |
Collapse
|
16
|
Hao Y, Song K, Tan X, Ren L, Guo X, Zhou C, Li H, Wen J, Meng Y, Lin M, Zhang Y, Huang H, Wang L, Zheng W. Reactive Oxygen Species-Responsive Polypeptide Drug Delivery System Targeted Activated Hepatic Stellate Cells to Ameliorate Liver Fibrosis. ACS NANO 2022; 16:20739-20757. [PMID: 36454190 DOI: 10.1021/acsnano.2c07796] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Hepatic fibrosis is a chronic liver disease that lacks effective pharmacotherapeutic treatments. As part of the disease's mechanism, hepatic stellate cells (HSCs) are activated by damage-related stimuli to secrete excessive extracellular matrix, leading to collagen deposition. Currently, the drug delivery system that targets HSCs in the treatment of liver fibrosis remains an urgent challenge due to the poor controllability of drug release. Since the level of reactive oxygen species (ROS) increases sharply in activated HSCs (aHSCs), we designed ROS-responsive micelles for the HSC-specific delivery of a traditional Chinese medicine, resveratrol (RES), for treatment of liver fibrosis. The micelles were prepared by the ROS-responsive amphiphilic block copolymer poly(l-methionine-block-Nε-trifluoro-acetyl-l-lysine) (PMK) and a PEG shell modified with a CRGD peptide insertion. The CRGD-targeted and ROS-responsive micelles (CRGD-PMK-MCs) could target aHSCs and control the release of RES under conditions of high intracellular ROS in aHSCs. The CRGD-PMK-MCs treatment specifically enhanced the targeted delivery of RES to aHSCs both in vitro and in vivo. In vitro experiments show that CRGD-PMK-MCs could significantly promote ROS consumption, reduce collagen accumulation, and avert activation of aHSCs. In vivo results demonstrate that CRGD-PMK-MCs could alleviate inflammatory infiltration, prevent fibrosis, and protect hepatocytes from damage in fibrotic mice. In conclusion, CRGD-PMK-MCs show great potential for targeted and ROS-responsive controlled drug release in the aHSCs of liver fibrosis.
Collapse
Affiliation(s)
- Yumei Hao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing 100050, China
| | - Kaichao Song
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiaochuan Tan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ling Ren
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiuping Guo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Chuchu Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - He Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jin Wen
- Chinese Pharmaceutical Association, Beijing 100022, China
| | - Ya Meng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Mingbao Lin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yujia Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hongdong Huang
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing 100050, China
| | - Lulu Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Wensheng Zheng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines & Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
17
|
Zhou R, Zhang M, Xi J, Li J, Ma R, Ren L, Bai Z, Qi K, Li X. Gold Nanorods-Based Photothermal Therapy: Interactions Between Biostructure, Nanomaterial, and Near-Infrared Irradiation. NANOSCALE RESEARCH LETTERS 2022; 17:68. [PMID: 35882718 PMCID: PMC9325935 DOI: 10.1186/s11671-022-03706-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/21/2022] [Indexed: 05/28/2023]
Abstract
Gold nanorods (AuNRs) are ideal inorganic nanophotothermal agents with unique characteristics, including local surface plasmon resonance effects, easy scale preparation and functional modification, and good biocompatibility. This review summarizes several recent advances in AuNRs-based photothermal therapy (PTT) research. Functionalized AuNRs photothermal agents have optimized biocompatibility and targeting properties. The multifunctional AuNRs nanoplatform composite structure meets the requirements for synergistic effects of PTT, photoacoustic imaging, and other therapeutic methods. Photothermal therapy with AuNRs (AuNRs-PTT) is widely used to treat tumors and inflammatory diseases; its tumor-targeting, tumor metastasis inhibition, and photothermal tumor ablation abilities have remarkable curative effects. An in-depth study of AuNRs in living systems and the interactions between biological structure, nanomaterial, and near-infrared irradiation could lay the foundation for further clinical research and the broad application of AuNRs in PTT.
Collapse
Affiliation(s)
- Ruili Zhou
- The First School of Clinical Medicine, Lanzhou University, No. 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
| | - Meigui Zhang
- The First School of Clinical Medicine, Lanzhou University, No. 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
| | - Jiahui Xi
- The First School of Clinical Medicine, Lanzhou University, No. 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
| | - Jing Li
- The First School of Clinical Medicine, Lanzhou University, No. 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
| | - Ruixia Ma
- The First School of Clinical Medicine, Lanzhou University, No. 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
| | - Longfei Ren
- The First School of Clinical Medicine, Lanzhou University, No. 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Zhongtian Bai
- The First School of Clinical Medicine, Lanzhou University, No. 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, China
| | - Kuo Qi
- The First School of Clinical Medicine, Lanzhou University, No. 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China.
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, China.
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, No. 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- Hepatopancreatobiliary Surgery Institute of Gansu Province, Medical College Cancer Center of Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
18
|
Bucharskaya AB, Khlebtsov NG, Khlebtsov BN, Maslyakova GN, Navolokin NA, Genin VD, Genina EA, Tuchin VV. Photothermal and Photodynamic Therapy of Tumors with Plasmonic Nanoparticles: Challenges and Prospects. MATERIALS (BASEL, SWITZERLAND) 2022; 15:1606. [PMID: 35208145 PMCID: PMC8878601 DOI: 10.3390/ma15041606] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 01/27/2023]
Abstract
Cancer remains one of the leading causes of death in the world. For a number of neoplasms, the efficiency of conventional chemo- and radiation therapies is insufficient because of drug resistance and marked toxicity. Plasmonic photothermal therapy (PPT) using local hyperthermia induced by gold nanoparticles (AuNPs) has recently been extensively explored in tumor treatment. However, despite attractive promises, the current PPT status is limited by laboratory experiments, academic papers, and only a few preclinical studies. Unfortunately, most nanoformulations still share a similar fate: great laboratory promises and fair preclinical trials. This review discusses the current challenges and prospects of plasmonic nanomedicine based on PPT and photodynamic therapy (PDT). We start with consideration of the fundamental principles underlying plasmonic properties of AuNPs to tune their plasmon resonance for the desired NIR-I, NIR-2, and SWIR optical windows. The basic principles for simulation of optical cross-sections and plasmonic heating under CW and pulsed irradiation are discussed. Then, we consider the state-of-the-art methods for wet chemical synthesis of the most popular PPPT AuNPs such as silica/gold nanoshells, Au nanostars, nanorods, and nanocages. The photothermal efficiencies of these nanoparticles are compared, and their applications to current nanomedicine are shortly discussed. In a separate section, we discuss the fabrication of gold and other nanoparticles by the pulsed laser ablation in liquid method. The second part of the review is devoted to our recent experimental results on laser-activated interaction of AuNPs with tumor and healthy tissues and current achievements of other research groups in this application area. The unresolved issues of PPT are the significant accumulation of AuNPs in the organs of the mononuclear phagocyte system, causing potential toxic effects of nanoparticles, and the possibility of tumor recurrence due to the presence of survived tumor cells. The prospective ways of solving these problems are discussed, including developing combined antitumor therapy based on combined PPT and PDT. In the conclusion section, we summarize the most urgent needs of current PPT-based nanomedicine.
Collapse
Affiliation(s)
- Alla B. Bucharskaya
- Core Facility Center, Saratov State Medical University, 112 Bol′shaya Kazachya Str., 410012 Saratov, Russia; (G.N.M.); (N.A.N.)
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia; (V.D.G.); (E.A.G.); (V.V.T.)
- Laser Molecular Imaging and Machine Learning Laboratory, Tomsk State University, 36 Lenin′s Av., 634050 Tomsk, Russia
| | - Nikolai G. Khlebtsov
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia; (V.D.G.); (E.A.G.); (V.V.T.)
- Nanobiotechnology Laboratory, Institute of Biochemistry and Physiology of Plants and Microorganisms RAS, FRC “Saratov Scientific Centre of the Russian Academy of Sciences”, 13 Prospekt Entuziastov, 410049 Saratov, Russia;
| | - Boris N. Khlebtsov
- Nanobiotechnology Laboratory, Institute of Biochemistry and Physiology of Plants and Microorganisms RAS, FRC “Saratov Scientific Centre of the Russian Academy of Sciences”, 13 Prospekt Entuziastov, 410049 Saratov, Russia;
| | - Galina N. Maslyakova
- Core Facility Center, Saratov State Medical University, 112 Bol′shaya Kazachya Str., 410012 Saratov, Russia; (G.N.M.); (N.A.N.)
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia; (V.D.G.); (E.A.G.); (V.V.T.)
| | - Nikita A. Navolokin
- Core Facility Center, Saratov State Medical University, 112 Bol′shaya Kazachya Str., 410012 Saratov, Russia; (G.N.M.); (N.A.N.)
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia; (V.D.G.); (E.A.G.); (V.V.T.)
| | - Vadim D. Genin
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia; (V.D.G.); (E.A.G.); (V.V.T.)
- Laser Molecular Imaging and Machine Learning Laboratory, Tomsk State University, 36 Lenin′s Av., 634050 Tomsk, Russia
| | - Elina A. Genina
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia; (V.D.G.); (E.A.G.); (V.V.T.)
- Laser Molecular Imaging and Machine Learning Laboratory, Tomsk State University, 36 Lenin′s Av., 634050 Tomsk, Russia
| | - Valery V. Tuchin
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia; (V.D.G.); (E.A.G.); (V.V.T.)
- Laser Molecular Imaging and Machine Learning Laboratory, Tomsk State University, 36 Lenin′s Av., 634050 Tomsk, Russia
- Institute of Precision Mechanics and Control, FRC “Saratov Scientific Centre of the Russian Academy of Sciences”, 24 Rabochaya Str., 410028 Saratov, Russia
| |
Collapse
|
19
|
Gu L, Zhang F, Wu J, Zhuge Y. Nanotechnology in Drug Delivery for Liver Fibrosis. Front Mol Biosci 2022; 8:804396. [PMID: 35087870 PMCID: PMC8787125 DOI: 10.3389/fmolb.2021.804396] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/17/2021] [Indexed: 12/15/2022] Open
Abstract
Liver fibrosis is a reversible disease course caused by various liver injury etiologies, and it can lead to severe complications, such as liver cirrhosis, liver failure, and even liver cancer. Traditional pharmacotherapy has several limitations, such as inadequate therapeutic effect and side effects. Nanotechnology in drug delivery for liver fibrosis has exhibited great potential. Nanomedicine improves the internalization and penetration, which facilitates targeted drug delivery, combination therapy, and theranostics. Here, we focus on new targets and new mechanisms in liver fibrosis, as well as recent designs and development work of nanotechnology in delivery systems for liver fibrosis treatment.
Collapse
Affiliation(s)
- Lihong Gu
- Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Feng Zhang
- Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, China
| | - Yuzheng Zhuge
- Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
20
|
Shu G, Yusuf A, Dai C, Sun H, Deng X. Piperine inhibits AML-12 hepatocyte EMT and LX-2 HSC activation and alleviates mouse liver fibrosis provoked by CCl 4: roles in the activation of the Nrf2 cascade and subsequent suppression of the TGF-β1/Smad axis. Food Funct 2021; 12:11686-11703. [PMID: 34730139 DOI: 10.1039/d1fo02657g] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Piperine (PIP) is an alkaloid derived from peppercorns. Herein, we assessed its effects on hepatocyte EMT and HSC activation in vitro and CCl4-elicited liver fibrosis in mice. Further experiments were performed to unveil the molecular mechanisms underlying the hepatoprotective activity of PIP. We found that PIP inhibited TGF-β1-provoked AML-12 hepatocyte EMT and LX-2 HSC activation. Mechanistically, in AML-12 and LX-2 cells, PIP evoked Nrf2 nuclear translocation and increased transcriptions of Nrf2-responsive antioxidative genes. These events decreased TGF-β1-induced production of ROS. Moreover, PIP increased the expression of Smad7, suppressed phosphorylation and nuclear translocation of Smad2/3, and decreased the transcriptions of Smad2/3-downstream genes. Knockdown of Nrf2 abrogated the protective activity of PIP against TGF-β1. Modulatory effects of PIP on the TGF-β1/Smad cascade were also crippled, which suggested that activation of Nrf2 played critical roles in the regulatory effects of PIP on TGF-β1/Smad signaling. Experiments in vivo unveiled that PIP ameliorated mouse liver fibrosis provoked by CCl4. PIP modulated the intrahepatic contents of the markers of EMT and HSC activation. In mouse livers, PIP activated Nrf2 signaling and reduced Smad2/3-dependent gene transcriptions. Our findings collectively suggested PIP as a new chemical entity with the capacity of alleviating liver fibrosis. The activation of the Nrf2 cascade and subsequent suppression of the TGF-β1/Smad axis are implicated in the hepatoprotective activity of PIP.
Collapse
Affiliation(s)
- Guangwen Shu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, Hubei, China.
| | - Arslan Yusuf
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, Hubei, China.
| | - Chenxi Dai
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, Hubei, China.
| | - Hui Sun
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, Hubei, China.
| | - Xukun Deng
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, Hubei, China.
| |
Collapse
|