1
|
Liu L, Che B, Zhang W, Du D, Zhang D, Li J, Chen Z, Yu X, Ye M, Wang W, Li Z, Xie F, Wang Q, Chen L, Shao J. Mechanistic insights into the role of FAT10 in modulating NCOA4-mediated ferroptosis in pancreatic acinar cells during acute pancreatitis. Cell Death Dis 2025; 16:385. [PMID: 40374601 DOI: 10.1038/s41419-025-07715-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 04/25/2025] [Accepted: 05/02/2025] [Indexed: 05/17/2025]
Abstract
Acute pancreatitis (AP) is characterised by inflammation and cell death in pancreatic tissue, with ferroptosis playing a critical role in its pathophysiology by mediating cellular damage and exacerbating inflammation. This study investigated the role of human leukocyte antigen (HLA)-F adjacent transcript 10 (FAT10) in AP, specifically its involvement in ferroptosis within pancreatic acinar cells. We observed that FAT10 expression was significantly elevated in AP tissues, which correlated with increased ferroptosis. Overexpression of FAT10 in pancreatic acinar cells enhances ferroptosis, whereas its knockdown reduced levels of ferroptosis markers. Furthermore, we confirmed that FAT10 enhanced ferroptosis in pancreatic acinar cells primarily by upregulating nuclear receptor coactivator 4 (NCOA4) expression. Mechanistic investigations revealed that FAT10 regulates NCOA4 expression to promote ferroptosis in a complex manner. FAT10 inhibits NCOA4 ubiquitination by reducing ubiquitin-NCOA4 complexes. Meanwhile, NCOA4 expression increased alongside the increase in FAT10-NCOA4 complexes, which are resistant to proteasomal degradation. Notably, we identified silibinin, a natural compound, as an effective inhibitor of the FAT10-NCOA4 axis, leading to reduced ferroptosis and alleviation of pancreatic damage in vivo. Silibinin treatment decreased the levels of ferroptosis-related proteins and inflammatory markers in both cell and animal models. Our findings highlight the FAT10-NCOA4 axis as a crucial regulator of ferroptosis in pancreatic acinar cells and suggest that targeting this pathway could offer a therapeutic strategy for mitigating AP. This study provides new insights into the regulatory mechanisms of ferroptosis in pancreatic acinar cells, identifying FAT10 as a potential therapeutic target for AP management.
Collapse
Affiliation(s)
- Lingpeng Liu
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ben Che
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenming Zhang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dongnian Du
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dandan Zhang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiajuan Li
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zehao Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuzhe Yu
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Miao Ye
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Wang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zijing Li
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fei Xie
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qing Wang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Leifeng Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China.
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China.
- Precision Oncology Medicine Center, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China.
| | - Jianghua Shao
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China.
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China.
- Liver Cancer Institute, Nanchang University, Nanchang, China.
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Nanchang University, Nanchang, China.
| |
Collapse
|
2
|
Zhao M, Cui M, Fan M, Huang C, Wang J, Zeng Y, Wang X, Lu Y. Octreotide attenuates experimental severe acute pancreatitis through inhibiting pyroptosis and modulating intestinal homeostasis. Eur J Pharmacol 2025; 994:177314. [PMID: 39922420 DOI: 10.1016/j.ejphar.2025.177314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/10/2025]
Abstract
Severe acute pancreatitis (SAP) is a common clinical condition characterized by acute abdominal symptoms. Octreotide (OCT) is a commonly prescribed treatment for acute pancreatitis (AP). Recent research shows that pyroptosis and intestinal homeostasis significantly contribute to the progression of AP. However, it remains unclear whether OCT treats SAP through modulating pyroptosis and intestinal microbiota. Our study aimed to investigate and validate the potential therapeutic effects of OCT on SAP and underlying mechanisms. The inhibition of pyroptosis in mice using disulfiram was investigated to elucidate the role of pyroptosis in AP. Molecular biology experiments confirmed that OCT effectively inhibited the expression of pyroptosis-related markers. Additionally, the composition, abundance, and functionality of the intestinal microbiota were analyzed using 16S rRNA sequencing, while short-chain fatty acids (SCFAs) were quantified by targeted metabolomics. Our study demonstrated that the administration of OCT significantly mitigated the severity of SAP in a dose-dependent manner. Furthermore, the inhibition of pyroptosis in mice attenuated SAP, thereby highlighting the critical role of pyroptosis in this condition. OCT administration was observed to suppress the expression of key pyroptosis markers. Additionally, there was a notable reduction in intestinal permeability and bacterial translocation. OCT reverses gut dysbiosis caused by SAP, increasing beneficial bacteria while inhibiting pathogenic strains. Furthermore, OCT administration enhanced the levels of SCFAs, including propanoic acid, acetic acid, and butyric acid. Our findings indicate OCT has the potential to alleviate SAP by suppressing pyroptosis and restoring intestinal homeostasis.
Collapse
Affiliation(s)
- Mengqi Zhao
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China; Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China
| | - Mengyan Cui
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China; Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China
| | - Miaoyan Fan
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China; Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China
| | - Chunlan Huang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China
| | - Jingjing Wang
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China
| | - Yue Zeng
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China
| | - Xingpeng Wang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China.
| | - Yingying Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China.
| |
Collapse
|
3
|
Ma J, Li X, Wang C. The Application of Nanomaterials in the Treatment of Pancreatic-Related Diseases. Int J Mol Sci 2024; 25:13158. [PMID: 39684868 DOI: 10.3390/ijms252313158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/16/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic diseases, typically including pancreatic cancer, pancreatitis, and diabetes, pose enormous threats to people's lives and health. To date, therapeutics with high therapeutic efficacy and low side effects are still challenging. With the development of nanotechnology, nanomaterials have successfully been applied in pancretic disease treatment. Here, we first introduce the diversity of nanomaterials and the effects of their different physicochemical properties on pancreatic function. Following this, we analyze the potential of nanomaterials to enhance pancreatic targeting by overcoming the challenges of traditional delivery methods through surface modifications, structural adjustments, and optimized drug loading. Then, we introduce the application of structurally optimized nanomaterials to pancreatic-related diseases. For instance, on pancreatic cancer (as drug delivery platforms, for the promotion of radiation therapy, and as multifunctional tools), pancreatitis (as drug delivery systems, anti-inflammatory and anti-fibrotic agents), and diabetes (as insulin delivery carriers, for protecting pancreatic β cells, and for improving insulin resistance). Through analysis of the progress of current research, we summarize how nanomaterials can enhance treatment efficacy while minimizing side effects. Finally, we look forward to the prospects of nanomaterials in pancreatic disease treatment.
Collapse
Affiliation(s)
- Jing Ma
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue Li
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunru Wang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
4
|
Su J, Lin C, Lin X, Hu S, Deng X, Xie L, Ye H, Zhou F, Wu S. Combining ulinastatin with TIENAM improves the outcome of sepsis induced by cecal ligation and puncture in mice by reducing inflammation and regulating immune responses. Int Immunopharmacol 2024; 141:112927. [PMID: 39163689 DOI: 10.1016/j.intimp.2024.112927] [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: 06/19/2024] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Despite the high mortality associated with sepsis, effective and targeted treatments remain scarce. The use of conventional antibiotics such as TIENAM (imipenem and cilastatin sodium for injection, TIE) is challenging because of the increasing bacterial resistance, which diminishes their efficacy and leads to adverse effects. Our previous studies demonstrated that ulinastatin (UTI) exerts a therapeutic impact on sepsis by reducing systemic inflammation and modulating immune responses. In this study, we examined the possibility of administering UTI and TIE after inducing sepsis in a mouse model using cecal ligation and puncture (CLP). We assessed the rates of survival, levels of inflammatory cytokines, the extent of tissue damage, populations of immune cells, microbiota in ascites, and important signaling pathways. The combination of UTI and TIE significantly improved survival rates and reduced inflammation and bacterial load in septic mice, indicating potent antimicrobial properties. Notably, the survival rates of UTI+TIE-treated mice increased from 10 % to 75 % within 168 h compared to those of mice that were subjected to CLP. The dual treatment successfully regulated the levels of inflammatory indicators (interleukin [IL]-6, IL-1β, and tumor necrosis factor [TNF]-α) and immune cell numbers by reducing B cells, natural killer cells, and TNFR2+ Treg cells and increasing CD8+ T cells. Additionally, the combination of UTI and TIE alleviated tissue damage, reduced bacterial load in the peritoneal cavity, and suppressed the NF-κB signaling pathway. Our findings indicate that UTI and TIE combination therapy can significantly enhance sepsis outcomes by reducing inflammation and boosting the immune system. The results offer a promising therapeutic approach for future sepsis treatment.
Collapse
Affiliation(s)
- Jingqian Su
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China.
| | - Congfan Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Xinrui Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Shan Hu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Xiaohui Deng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Lian Xie
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Hui Ye
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Fen Zhou
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| | - Shun Wu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian Province 350117, PR China
| |
Collapse
|
5
|
Pan Y, Chen J, Zhang Y, Ren Y, Wu Z, Xue Q, Zeng S, Fang C, Zhang H, Zhang L, Liu C, Zeng J. Second Near-Infrared Macrophage-Biomimetic Nanoprobes for Photoacoustic Imaging of Neuroinflammation. Mol Pharm 2024; 21:1804-1816. [PMID: 38466359 DOI: 10.1021/acs.molpharmaceut.3c01115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Neuroinflammation is a significant pathological event involving the neurodegenerative process associated with many neurological disorders. Diagnosis and treatment of neuroinflammation in its early stage are essential for the prevention and management of neurological diseases. Herein, we designed macrophage membrane-coated photoacoustic (PA) probes (MSINPs), with targeting specificities based on naturally existing target-ligand interactions for the early diagnosis of neuroinflammation. The second near-infrared dye, IR1061, was doped into silica as the core and was encapsulated with a macrophage membrane. In vitro as well as in vivo, the MSINPs could target inflammatory cells via the inflammation chemotactic effect. PA imaging was used to trace the MSINPs in a neuroinflammation mouse model and showed a great targeted effect of MSINPs in the prefrontal cortex. Therefore, the biomimetic nanoprobe prepared in this study offers a new strategy for PA molecular imaging of neuroinflammation, which can enhance our understanding of the evolution of neuroinflammation in specific brain regions.
Collapse
Affiliation(s)
- Yingying Pan
- Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Jingqin Chen
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuling Zhang
- Shenzhen Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518116, China
| | - Yaguang Ren
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhifeng Wu
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qiang Xue
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Ultrasound, Shenzhen People's Hospital, The Second Clinical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Silue Zeng
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chihua Fang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Hai Zhang
- Department of Ultrasound, Shenzhen People's Hospital, The Second Clinical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Lingyan Zhang
- Lab of Molecular Imaging and Medical Intelligence, Department of Radiology, Longgang Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Chengbo Liu
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jie Zeng
- Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| |
Collapse
|
6
|
Shang J, Liang T, Wei D, Qin F, Yang J, Ye Y, Zhou M. Quercetin-loaded PLGA nanoparticles coating with macrophage membranes for targeted delivery in acute liver injury. NANOTECHNOLOGY 2023; 35:115102. [PMID: 38156649 DOI: 10.1088/1361-6528/ad1440] [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: 10/18/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Quercetin (QU), a natural flavonoid with potent anti-inflammatory and antioxidant properties, holds promise in treating acute liver injury (ALI). Nonetheless, its limited solubility hampers its efficacy, and its systemic distribution lacks targeting, leading to off-target effects. To address these challenges, we developed macrophage membrane-coated quercetin-loaded PLGA nanoparticles (MVs-QU-NPs) for active ALI targeting. The resulting MVs-QU-NPs exhibited a spherical morphology with a clear core-shell structure. The average size and zeta potential were assessed as 141.70 ± 0.89 nm and -31.83 ± 0.76 mV, respectively. Further studies revealed sustained drug release characteristics from MVs-QU-NPs over a continuous period of 24 h. Moreover, these MVs-QU-NPs demonstrated excellent biocompatibility when tested on normal liver cells. The results of biodistribution analysis in ALI mice displayed the remarkable ALI-targeting ability of MVs-DiD-NPs, with the highest fluorescence intensity observed in liver tissue. This biomimetic approach combining macrophage membranes with nanoparticle delivery, holds great potential for targeted ALI treatment.
Collapse
Affiliation(s)
- Jinlu Shang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, People's Republic of China
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou 646000, People's Republic of China
| | - Tiantian Liang
- Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang 621000, People's Republic of China
| | - Daiqing Wei
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, People's Republic of China
| | - Feiyang Qin
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou 646000, People's Republic of China
| | - Jing Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, People's Republic of China
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou 646000, People's Republic of China
| | - Yun Ye
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, People's Republic of China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, People's Republic of China
| |
Collapse
|
7
|
Liu L, Zhang Y, Li X, Deng J. Microenvironment of pancreatic inflammation: calling for nanotechnology for diagnosis and treatment. J Nanobiotechnology 2023; 21:443. [PMID: 37996911 PMCID: PMC10666376 DOI: 10.1186/s12951-023-02200-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Acute pancreatitis (AP) is a common and life-threatening digestive disorder. However, its diagnosis and treatment are still impeded by our limited understanding of its etiology, pathogenesis, and clinical manifestations, as well as by the available detection methods. Fortunately, the progress of microenvironment-targeted nanoplatforms has shown their remarkable potential to change the status quo. The pancreatic inflammatory microenvironment is typically characterized by low pH, abundant reactive oxygen species (ROS) and enzymes, overproduction of inflammatory cells, and hypoxia, which exacerbate the pathological development of AP but also provide potential targeting sites for nanoagents to achieve early diagnosis and treatment. This review elaborates the various potential targets of the inflammatory microenvironment of AP and summarizes in detail the prospects for the development and application of functional nanomaterials for specific targets. Additionally, it presents the challenges and future trends to develop multifunctional targeted nanomaterials for the early diagnosis and effective treatment of AP, providing a valuable reference for future research.
Collapse
Affiliation(s)
- Lu Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Street, Nanchong, 637001, China
| | - Yiqing Zhang
- Institute of Burn Research Southwest Hospital State Key Lab of Trauma Burn and Combined Injury Chongqing Key Laboratory for Disease Proteomics Army Medical University, Chongqing, 400038, China
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese PLA General Hospita, PLA Medical College, 28 Fu Xing Road, Beijing, 100853, China
| | - Xinghui Li
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Street, Nanchong, 637001, China.
| | - Jun Deng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Street, Nanchong, 637001, China.
- Institute of Burn Research Southwest Hospital State Key Lab of Trauma Burn and Combined Injury Chongqing Key Laboratory for Disease Proteomics Army Medical University, Chongqing, 400038, China.
| |
Collapse
|