1
|
Li F, Yuan R, Zhang J, Su B, Qi X. Advances in nanotechnology for the diagnosis and management of metabolic dysfunction-associated steatotic liver disease. Asian J Pharm Sci 2025; 20:101025. [PMID: 40182137 PMCID: PMC11964547 DOI: 10.1016/j.ajps.2025.101025] [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: 08/18/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 04/05/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has a high global incidence and associated with increased lipid accumulation in hepatocytes, elevated hepatic enzyme levels, liver fibrosis, and hepatic carcinoma. Despite decades of research and significant advancements, the treatment of MASLD still faces formidable challenges. Nanoprobes for diagnostics and nanomedicine for targeted drug delivery to the liver present promising options for MASLD diagnosis and treatment, enhancing both imaging contrast and bioavailability. Here, we review recent advances in nanotechnology applied to MASLD diagnosis and treatment, specifically focusing on drug delivery systems targeting hepatocytes, hepatic stellate cells, Kupffer cells, and liver sinusoidal endothelial cells. This review aims to provide an overview of nanomedicine's potential in early MASLD diagnosis and therapeutic interventions, addressing related complications.
Collapse
Affiliation(s)
- Fenfen Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ruyan Yuan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jiamin Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Bing Su
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaolong Qi
- Liver Disease Center of Integrated Traditional Chinese and Western Medicine, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Nanjing 210009, China
- Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University; State Key Laboratory of Digital Medical Engineering, Nanjing 210009, China
| |
Collapse
|
2
|
Zhang G, Wu K, Jiang X, Gao Y, Ding D, Wang H, Yu C, Wang X, Jia N, Zhu L. The role of ferroptosis-related non-coding RNA in liver fibrosis. Front Cell Dev Biol 2024; 12:1517401. [PMID: 39717848 PMCID: PMC11663870 DOI: 10.3389/fcell.2024.1517401] [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: 10/26/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Liver fibrosis represents a reversible pathophysiological process, caused by chronic inflammation stemming from hepatocyte damage. It delineates the initial stage in the progression of chronic liver disease. This pathological progression is characterized by the excessive accumulation of the extracellular matrix (ECM), which leads to significant structural disruption and ultimately impairs liver function. To date, no specific antifibrotic drugs have been developed, and advanced liver fibrosis remains largely incurable. Liver transplantation remains the sole efficacious intervention for advanced liver fibrosis; nevertheless, it is constrained by exorbitant costs and the risk of postoperative immune rejection, underscoring the imperative for novel therapeutic strategies. Ferroptosis, an emergent form of regulated cell death, has been identified as a pivotal regulatory mechanism in the development of liver fibrosis and is intricately linked with the progression of liver diseases. Recent investigations have elucidated that a diverse array of non-coding RNAs (ncRNAs), including microRNAs, long non-coding RNAs, and circular RNAs, are involved in the ferroptosis pathway, thereby modulating the progression of various diseases, including liver fibrosis. In recent years, the roles of ferroptosis and ferroptosis-related ncRNAs in liver fibrosis have attracted escalating scholarly attention. This paper elucidates the pathophysiology of liver fibrosis, explores the mechanisms underlying ferroptosis, and delineates the involvement of ncRNA-mediated ferroptosis pathways in the pathology of liver fibrosis. It aims to propose novel strategies for the prevention and therapeutic intervention of liver fibrosis.
Collapse
Affiliation(s)
- Guozhu Zhang
- Department of Emergency Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Kejia Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaobo Jiang
- Kunshan Zhenchuan Community Health Service Center, Kunshan, Jiangsu, China
| | - Yuan Gao
- Department of Hepato-Biliary-Pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Dong Ding
- Department of Hepato-Biliary-Pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hao Wang
- Department of Emergency Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Chongyuan Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaozhong Wang
- Department of General Surgery, Wujin Affiliated Hospital of Jiangsu University and the Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Naixin Jia
- Department of Hepatobiliary Surgery, Kunshan First People’s Hospital affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Li Zhu
- Department of Emergency Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
3
|
Yuan Y, Li J, Chen M, Zhao Y, Zhang B, Chen X, Zhao J, Liang H, Chen Q. Nano-encapsulation of drugs to target hepatic stellate cells: Toward precision treatments of liver fibrosis. J Control Release 2024; 376:318-336. [PMID: 39413846 DOI: 10.1016/j.jconrel.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Liver fibrosis is characterized by excessive extracellular matrix (ECM) deposition triggered by hepatic stellate cells (HSCs). As central players in fibrosis progression, HSCs are the most important therapeutic targets for antifibrotic therapy. However, owing to the limitations of systemic drug administration, there is still no suitable and effective clinical treatment. In recent years, nanosystems have demonstrated expansive therapeutic potential and evolved into a clinical modality. In liver fibrosis, nanosystems have undergone a paradigm shift from targeting the whole liver to locally targeted modifying processes. Nanomedicine delivered to HSCs has significant potential in managing liver fibrosis, where optimal management would benefit from targeted delivery, personalized therapy based on the specific site of interest, and minor side effects. In this review, we present a brief overview of the role of HSCs in the pathogenesis of liver fibrosis, summarize the different types of nanocarriers and their specific delivery applications in liver fibrosis, and highlight the biological barriers associated with the use of nanosystems to target HSCs and approaches available to solve this issue. We further discuss in-depth all the molecular target receptors overexpressed during HSC activation in liver fibrosis and their corresponding ligands that have been used for drug or gene delivery targeting HSCs.
Collapse
Affiliation(s)
- Yue Yuan
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Jiaxuan Li
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Min Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Ying Zhao
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Jianping Zhao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| | - Qian Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China.
| |
Collapse
|
4
|
Yıldırım M, Acet Ö, Önal Acet B, Karakoç V, Odabaşı M. Innovative approach against cancer: Thymoquinone-loaded PHEMA-based magnetic nanoparticles and their effects on MCF-7 breast cancer. Biochem Biophys Res Commun 2024; 734:150464. [PMID: 39083970 DOI: 10.1016/j.bbrc.2024.150464] [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: 05/29/2024] [Revised: 07/20/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Breast cancer is most common cancer among women in the World. Thymoquinone (TQ) exhibits a wide range of biological activities such as anticancer, antidiabetic, antimicrobial, analgesic, antioxidant, and anti-inflammatory effects. However, its effectiveness in cancer treatment is hindered by its poor bioavailability, attributed to its limited solubility in water. Hence, novel strategies are required to enhance the bioavailability of TQ, which possesses remarkable anticancer characteristics. The aim of this study is to prepare pHEMA-based magnetic nanoparticles carrying TQ (TQ-MNPs) to improve bioavailability, and therapeutic efficacy against breast cancer. For this purpose, TQ-MNPs were synthesized and characterized with Fourier transform infrared spectrophotometer (FTIR), scanning electron microscopy (SEM), dynamic light scattering (DLS), magnetic field using a vibrating sample magnetometer (VSM). The loading capabilities of synthesized magentic nanostructures were evaluated, and release investigations were conducted under experimental conditions that mimic the cellular environment. The findings of the studies indicated that the TQ carrying capacity of MNPs was deemed satisfactory, and the release efficiency was adequate. MNPs and TQ-MNPs showed biocompatibility against HDFa cells. TQ-MNPs showed stronger anti-proliferative activity against MCF-7 breast cancer cells compared to free TQ (p < 0.05). TQ-MNPs induced apoptosis in MCF-7 breast cancer cells.
Collapse
Affiliation(s)
- Metin Yıldırım
- Faculty of Pharmacy, Department of Biochemistry, Harran University, Şanlıurfa, Turkey.
| | - Ömür Acet
- Department of Pharmacy Services, Vocational School of Health Services, Tarsus University, Mersin, Turkey.
| | - Burcu Önal Acet
- Department of Chemistry, Arts and Science Faculty, Aksaray University, Aksaray, Turkey
| | - Veyis Karakoç
- Eldivan Vocational School of Health Services, Çankırı Karatekin University, Çankırı, Turkey
| | - Mehmet Odabaşı
- Department of Chemistry, Arts and Science Faculty, Aksaray University, Aksaray, Turkey
| |
Collapse
|
5
|
Kurbatova IV, Topchieva LV, Dudanova OP, Shipovskaya AA. Role of MMP-2 and MMP-9 in the Relationship between Inflammation, Fibrosis, and Apoptosis during Progression of Non-Alcoholic Fatty Liver Disease and Diagnostic Significance of Plasma Levels of Their Active Forms. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1998-2022. [PMID: 39647828 DOI: 10.1134/s0006297924110130] [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: 04/02/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 12/10/2024]
Abstract
MMP-2 and MMP-9 play an important role in pathogenesis of chronic liver diseases, participating in the processes of inflammation and fibrosis. Their role in progression of non-alcoholic fatty liver disease (NAFLD) is poorly understood. Analysis of MMP-2, -9 levels in the blood plasma of patients with different forms of NAFLD [liver steatosis (LS) and non-alcoholic steatohepatitis (NASH) of weak (-WA), moderate (MA), high (-HA) activity without pronounced fibrosis] was performed. Correlations between the levels of MMP-2, -9 and mRNA of the genes MMP2, MMP9, ADAM17, NLRP3, caspase 3 activity in peripheral blood leukocytes (PBL), TNFα, IL-6, sIL-6R, cytokeratin-18 fragments in plasma were assessed. In steatosis, the levels of MMP2 gene mRNA in PBL and MMP-2 in plasma are lower than in the control, and expression of the NLRP3 gene in PBL is increased relative to other groups. In the NASH-WA, the level of MMP-9 is higher than in the control, in LS, and in NASH-MA, which could be associated with activation of inflammation during transformation of LS into NASH. The plasma level of MMP-9 over 389.50 pg/ml has been shown to be diagnostically significant for identification of NASH-WA among the patients with steatosis (AUC ROC = 0.818, 95% CI = 0.689-0.948, p < 0.001). In NAFLD, the level of MMP-9 could be associated not only with inflammation, but also with apoptosis. ADAM17 probably plays a certain role in this regard. In the advanced NASH, hepatocyte apoptosis is increased, the level of caspase 3 activity in PBL is increased, the level of MMP-9 in the blood is reduced to the level of the control and LS. In the NASH-HA, the level of mRNA of the ADAM17 gene in PBL is increased compared to the control, NASH-WA, and NASH-MA. Thus, MMP-2 and MMP-9 are involved in pathogenesis of NAFLD already at the early stages and their level in blood could be associated with the presence and severity of inflammation in the liver parenchyma.
Collapse
Affiliation(s)
- Irina V Kurbatova
- Institute of Biology, Karelian Research Centre, Russian Academy of Sciences, Petrozavodsk, Karelia, 185910, Russia.
| | - Lyudmila V Topchieva
- Institute of Biology, Karelian Research Centre, Russian Academy of Sciences, Petrozavodsk, Karelia, 185910, Russia
| | - Olga P Dudanova
- Zilber Medical Institute, Petrozavodsk State University, Petrozavodsk, Karelia, 185910, Russia
| | - Anastasia A Shipovskaya
- Zilber Medical Institute, Petrozavodsk State University, Petrozavodsk, Karelia, 185910, Russia
| |
Collapse
|
6
|
Meng X, Zhu G, Yang YG, Sun T. Targeted delivery strategies: The interactions and applications of nanoparticles in liver diseases. Biomed Pharmacother 2024; 175:116702. [PMID: 38729052 DOI: 10.1016/j.biopha.2024.116702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
In recent years, nanoparticles have been broadly utilized in various drugs delivery formulations. Nanodelivery systems have shown promise in solving problems associated with the distribution of hydrophobic drugs and have promoted the accumulation of nanomedicines in the circulation or in organs. However, the injection dose of nanoparticles (NPs) is much greater than that needed by diseased tissues or organs. In other words, most of the NPs are localized off-target and do not reach the desired tissue or organs. With the rapid development of biodegradable and biosafety nanomaterials, the nanovectors represent assurance of safety. However, the off-target effects also induce concerns about the application of NPs, especially in the delivery of gene editing tools. Therefore, a complete understanding of the biological responses to NPs in the body will clearly guide the design of targeted delivery of NPs. The different properties of various nanodelivery systems may induce diverse interactions between carriers and organs. In this review, we describe the relationship between the liver, the most influenced organ of systemic administration of NPs, and targeted delivery nanoplatforms. Various transport vehicles have adopted multiple delivery strategies for the targeted delivery to the cells in the homeostasis liver and in diseased liver. Additionally, nanodelivery systems provide a novel strategy for treating incurable diseases. The appearance of a targeted delivery has profoundly improved the application of NPs to liver diseases.
Collapse
Affiliation(s)
- Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Ge Zhu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
7
|
Fateh ST, Aghaii AH, Aminzade Z, Shahriari E, Roohpour N, Koosha F, Dezfuli AS. Inorganic nanoparticle-cored dendrimers for biomedical applications: A review. Heliyon 2024; 10:e29726. [PMID: 38694058 PMCID: PMC11061704 DOI: 10.1016/j.heliyon.2024.e29726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 05/03/2024] Open
Abstract
Hybrid nanostructures exhibit a synergistic combination of features derived from their individual components, showcasing novel characteristics resulting from their distinctive structure and chemical/physical properties. Surface modifiers play a pivotal role in shaping INPs' primary attributes, influencing their physicochemical properties, stability, and functional applications. Among these modifiers, dendrimers have gained attention as highly effective multifunctional agents for INPs, owing to their unique structural qualities, dendritic effects, and physicochemical properties. Dendrimers can be seamlessly integrated with diverse inorganic nanostructures, including metal NPs, carbon nanostructures, silica NPs, and QDs. Two viable approaches to achieving this integration involve either growing or grafting dendrimers, resulting in inorganic nanostructure-cored dendrimers. The initial step involves functionalizing the nanostructures' surface, followed by the generation of dendrimers through stepwise growth or attachment of pre-synthesized dendrimer branches. This hybridization imparts superior qualities to the resulting structure, including biocompatibility, solubility, high cargo loading capacity, and substantial functionalization potential. Combining the unique properties of dendrimers with those of the inorganic nanostructure cores creates a multifunctional system suitable for diverse applications such as theranostics, bio-sensing, component isolation, chemotherapy, and cargo-carrying applications. This review summarizes the recent developments, with a specific focus on the last five years, within the realm of dendrimers. It delves into their role as modifiers of INPs and explores the potential applications of INP-cored dendrimers in the biomedical applications.
Collapse
Affiliation(s)
- Sepand Tehrani Fateh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Ronash Technology Pars Company(AMINBIC), Tehran, Iran
| | - Amir Hossein Aghaii
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
- Ronash Technology Pars Company(AMINBIC), Tehran, Iran
| | - Zahra Aminzade
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Shahriari
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Fereshteh Koosha
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
8
|
Wang L, Zhou J, Wang J, Wang X, Dong H, Zhao L, Wu J, Peng J. Hepatic Stellate Cell-Targeting Micelle Nanomedicine for Early Diagnosis and Treatment of Liver Fibrosis. Adv Healthc Mater 2024; 13:e2303710. [PMID: 38293743 DOI: 10.1002/adhm.202303710] [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: 11/22/2023] [Revised: 01/23/2024] [Indexed: 02/01/2024]
Abstract
Diagnosing and treating liver fibrosis is a challenging yet crucial endeavor due to its complex pathogenesis and risk of deteriorating into cirrhosis, liver failure, and even hepatic cancer. Herein, a silica cross-linked micelles (SCLMs) based nano-system is developed for both diagnosing and treating liver fibrosis. The SCLMs are first modified with peptide CTCE9908 (CT-SCLMs) and can actively target CXCR4, which is overexpressed in activated hepatic stellate cells (HSCs). To enable diagnosis, an ONOO--responded near-infrared fluorescent probe NOF2 is loaded into the CT-SCLMs. This nano-system can target the aHSCs and diagnose the liver fibrosis particularly in CCl4-induced liver damage, by monitoring the reactive nitrogen species. Furthermore, a step is taken toward treatment by co-encapsulating two anti-fibrosis drugs, silibinin and sorafenib, within the CT-SCLMs. This combined approach results in a significant alleviation of liver injury. Symptoms associated with liver fibrosis, such as deposition of collagen, expression of hydroxyproline, and raised serological indicators show notable improvement. In summary, the CXCR4-targeted nano-system can serve as a promising theragnostic system of early warning and diagnosis for liver fibrosis, offering hope against progression of this serious liver condition.
Collapse
Affiliation(s)
- Lei Wang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jieying Zhou
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, 33199, USA
| | - Jian Wang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Xiaotang Wang
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, 33199, USA
| | - Haijuan Dong
- The Public Laboratory Platform, China Pharmaceutical University, Nanjing, 211198, China
| | - Lingzhi Zhao
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Junchen Wu
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Juanjuan Peng
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| |
Collapse
|
9
|
Zaid A, Ariel A. Harnessing anti-inflammatory pathways and macrophage nano delivery to treat inflammatory and fibrotic disorders. Adv Drug Deliv Rev 2024; 207:115204. [PMID: 38342241 DOI: 10.1016/j.addr.2024.115204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/08/2023] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
Targeting specific organs and cell types using nanotechnology and sophisticated delivery methods has been at the forefront of applicative biomedical sciences lately. Macrophages are an appealing target for immunomodulation by nanodelivery as they are heavily involved in various aspects of many diseases and are highly plastic in their nature. Their continuum of functional "polarization" states has been a research focus for many years yielding a profound understanding of various aspects of these cells. The ability of monocyte-derived macrophages to metamorphose from pro-inflammatory to reparative and consequently to pro-resolving effectors has raised significant interest in its therapeutic potential. Here, we briefly survey macrophages' ontogeny and various polarization phenotypes, highlighting their function in the inflammation-resolution shift. We review their inducing mediators, signaling pathways, and biological programs with emphasis on the nucleic acid sensing-IFN-I axis. We also portray the polarization spectrum of macrophages and the characteristics of their transition between different subtypes. Finally, we highlighted different current drug delivery methods for targeting macrophages with emphasis on nanotargeting that might lead to breakthroughs in the treatment of wound healing, bone regeneration, autoimmune, and fibrotic diseases.
Collapse
Affiliation(s)
- Ahmad Zaid
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel
| | - Amiram Ariel
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel.
| |
Collapse
|
10
|
Martinez-Campanario MC, Cortés M, Moreno-Lanceta A, Han L, Ninfali C, Domínguez V, Andrés-Manzano MJ, Farràs M, Esteve-Codina A, Enrich C, Díaz-Crespo FJ, Pintado B, Escolà-Gil JC, García de Frutos P, Andrés V, Melgar-Lesmes P, Postigo A. Atherosclerotic plaque development in mice is enhanced by myeloid ZEB1 downregulation. Nat Commun 2023; 14:8316. [PMID: 38097578 PMCID: PMC10721632 DOI: 10.1038/s41467-023-43896-7] [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: 01/25/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
Accumulation of lipid-laden macrophages within the arterial neointima is a critical step in atherosclerotic plaque formation. Here, we show that reduced levels of the cellular plasticity factor ZEB1 in macrophages increase atherosclerotic plaque formation and the chance of cardiovascular events. Compared to control counterparts (Zeb1WT/ApoeKO), male mice with Zeb1 ablation in their myeloid cells (Zeb1∆M/ApoeKO) have larger atherosclerotic plaques and higher lipid accumulation in their macrophages due to delayed lipid traffic and deficient cholesterol efflux. Zeb1∆M/ApoeKO mice display more pronounced systemic metabolic alterations than Zeb1WT/ApoeKO mice, with higher serum levels of low-density lipoproteins and inflammatory cytokines and larger ectopic fat deposits. Higher lipid accumulation in Zeb1∆M macrophages is reverted by the exogenous expression of Zeb1 through macrophage-targeted nanoparticles. In vivo administration of these nanoparticles reduces atherosclerotic plaque formation in Zeb1∆M/ApoeKO mice. Finally, low ZEB1 expression in human endarterectomies is associated with plaque rupture and cardiovascular events. These results set ZEB1 in macrophages as a potential target in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- M C Martinez-Campanario
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036, Barcelona, Spain
| | - Marlies Cortés
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036, Barcelona, Spain
| | - Alazne Moreno-Lanceta
- Department of Biomedicine, University of Barcelona School of Medicine, 08036, Barcelona, Spain
| | - Lu Han
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036, Barcelona, Spain
| | - Chiara Ninfali
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036, Barcelona, Spain
| | - Verónica Domínguez
- Transgenesis Facility, National Center of Biotechnology (CNB) and Center for Molecular Biology Severo Ochoa (UAM-CBMSO), Spanish National Research Council (CSIC) and Autonomous University of Madrid (UAM), Cantoblanco, 28049, Madrid, Spain
| | - María J Andrés-Manzano
- Group of Molecular and Genetic Cardiovascular Pathophysiology, Spanish National Center for Cardiovascular Research (CNIC), 28029, Madrid, Spain
- Center for Biomedical, Research Network in Cardiovascular Diseases (CIBERCV), Carlos III Health Institute, 28029, Madrid, Spain
| | - Marta Farràs
- Department of Biochemistry and Molecular Biology, Institute of Biomedical Research Sant Pau, University Autonomous of Barcelona, 08041, Barcelona, Spain
- Center for Biomedical Research Network in Diabetes and Associated Metabolic Diseases (CIBERDEM), Carlos III Health Institute, 28029, Madrid, Spain
| | | | - Carlos Enrich
- Department of Biomedicine, University of Barcelona School of Medicine, 08036, Barcelona, Spain
- Group of signal transduction, intracellular compartments and cancer, IDIBAPS, 08036, Barcelona, Spain
| | - Francisco J Díaz-Crespo
- Department of Pathology, Hospital General Universitario Gregorio Marañón, 28007, Madrid, Spain
| | - Belén Pintado
- Transgenesis Facility, National Center of Biotechnology (CNB) and Center for Molecular Biology Severo Ochoa (UAM-CBMSO), Spanish National Research Council (CSIC) and Autonomous University of Madrid (UAM), Cantoblanco, 28049, Madrid, Spain
| | - Joan C Escolà-Gil
- Department of Biochemistry and Molecular Biology, Institute of Biomedical Research Sant Pau, University Autonomous of Barcelona, 08041, Barcelona, Spain
- Center for Biomedical Research Network in Diabetes and Associated Metabolic Diseases (CIBERDEM), Carlos III Health Institute, 28029, Madrid, Spain
| | - Pablo García de Frutos
- Center for Biomedical, Research Network in Cardiovascular Diseases (CIBERCV), Carlos III Health Institute, 28029, Madrid, Spain
- Department Of Cell Death and Proliferation, Institute for Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036, Barcelona, Spain
- Group of Hemotherapy and Hemostasis, IDIBAPS, 08036, Barcelona, Spain
| | - Vicente Andrés
- Group of Molecular and Genetic Cardiovascular Pathophysiology, Spanish National Center for Cardiovascular Research (CNIC), 28029, Madrid, Spain
- Center for Biomedical, Research Network in Cardiovascular Diseases (CIBERCV), Carlos III Health Institute, 28029, Madrid, Spain
| | - Pedro Melgar-Lesmes
- Department of Biomedicine, University of Barcelona School of Medicine, 08036, Barcelona, Spain
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, 08036, Barcelona, Spain
- Center for Biomedical Research Network in Gastrointestinal and Liver Diseases (CIBEREHD), Carlos III Health Institute, 28029, Madrid, Spain
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, 02139, USA
| | - Antonio Postigo
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036, Barcelona, Spain.
- Center for Biomedical Research Network in Gastrointestinal and Liver Diseases (CIBEREHD), Carlos III Health Institute, 28029, Madrid, Spain.
- Molecular Targets Program, Division of Oncology, Department of Medicine, J.G. Brown Cancer Center, Louisville, KY, 40202, USA.
- ICREA, 08010, Barcelona, Spain.
| |
Collapse
|
11
|
Stangel C, Kagkoura A, Pippa N, Stellas D, Zhang M, Okazaki T, Demetzos C, Tagmatarchis N. Preclinical evaluation of modified carbon nanohorns and their complexation with insulin. NANOSCALE ADVANCES 2023; 5:6847-6857. [PMID: 38059018 PMCID: PMC10696926 DOI: 10.1039/d3na00471f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/23/2023] [Indexed: 12/08/2023]
Abstract
The current study emphasizes the minimal toxicity observed in vitro and in vivo for carbon nanohorns (CNHs) modified with third generation polyamidoamine (PAMAM) dendrimers. Initially, we investigated the interactions between CNH-PAMAM and lipid bilayers, which were utilized as representative models of cellular membranes for the evaluation of their toxicity in vitro. We found that the majority of those interactions occur between the modified CNHs and the polar groups of phospholipids, meaning that CNH-PAMAM does not incorporate into the lipid chains, and thus, disruption of the lipid bilayer structure is avoided. This outcome is a very important observation for further evaluation of CNH-PAPAM in cell lines and in animal models. Next, we demonstrated the potential of CNH-PAMAM for complexation with insulin, as a proof of concept for its employment as a delivery platform. Importantly, our study provides comprehensive evidence of low toxicity for CNH-PAMAM both in vitro and in vivo. The assessment of cellular toxicity revealed that the modified CNHs exhibited minimal toxicity, with concentrations of 151 μg mL-1 and 349 μg mL-1, showing negligible harm to EO771 cells and mouse embryonic fibroblasts (MEFs), respectively. Moreover, the histological analysis of the mouse livers demonstrated no evidence of tissue necrosis and inflammation, or any visible signs of severe toxicity. These findings collectively indicate the safe profile of CNH-PAMAM and further contribute to the growing body of knowledge on the safe and efficient utilization of CNH-based nanomaterials in drug and protein delivery applications.
Collapse
Affiliation(s)
- Christina Stangel
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation 48 Vassileos Constantinou Avenue Athens 11635 Greece
| | - Antonia Kagkoura
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation 48 Vassileos Constantinou Avenue Athens 11635 Greece
| | - Natassa Pippa
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens Athens 15771 Greece
| | - Dimitris Stellas
- Institute of Chemical Biology, National Hellenic Research Foundation 48 Vassileos Constantinou Avenue Athens 11635 Greece
| | - Minfang Zhang
- Nano Carbon Device Research Center, National Institute of Advanced Industrial Science and Technology (AIST) Tsukuba 305-8565 Japan
| | - Toshiya Okazaki
- Nano Carbon Device Research Center, National Institute of Advanced Industrial Science and Technology (AIST) Tsukuba 305-8565 Japan
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens Athens 15771 Greece
| | - Nikos Tagmatarchis
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation 48 Vassileos Constantinou Avenue Athens 11635 Greece
| |
Collapse
|
12
|
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
|
13
|
Ou L, Tan X, Qiao S, Wu J, Su Y, Xie W, Jin N, He J, Luo R, Lai X, Liu W, Zhang Y, Zhao F, Liu J, Kang Y, Shao L. Graphene-Based Material-Mediated Immunomodulation in Tissue Engineering and Regeneration: Mechanism and Significance. ACS NANO 2023; 17:18669-18687. [PMID: 37768738 DOI: 10.1021/acsnano.3c03857] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Tissue engineering and regenerative medicine hold promise for improving or even restoring the function of damaged organs. Graphene-based materials (GBMs) have become a key player in biomaterials applied to tissue engineering and regenerative medicine. A series of cellular and molecular events, which affect the outcome of tissue regeneration, occur after GBMs are implanted into the body. The immunomodulatory function of GBMs is considered to be a key factor influencing tissue regeneration. This review introduces the applications of GBMs in bone, neural, skin, and cardiovascular tissue engineering, emphasizing that the immunomodulatory functions of GBMs significantly improve tissue regeneration. This review focuses on summarizing and discussing the mechanisms by which GBMs mediate the sequential regulation of the innate immune cell inflammatory response. During the process of tissue healing, multiple immune responses, such as the inflammatory response, foreign body reaction, tissue fibrosis, and biodegradation of GBMs, are interrelated and influential. We discuss the regulation of these immune responses by GBMs, as well as the immune cells and related immunomodulatory mechanisms involved. Finally, we summarize the limitations in the immunomodulatory strategies of GBMs and ideas for optimizing GBM applications in tissue engineering. This review demonstrates the significance and related mechanism of the immunomodulatory function of GBM application in tissue engineering; more importantly, it contributes insights into the design of GBMs to enhance wound healing and tissue regeneration in tissue engineering.
Collapse
Affiliation(s)
- Lingling Ou
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiner Tan
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shijia Qiao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Junrong Wu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuan Su
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528399, China
| | - Wenqiang Xie
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Nianqiang Jin
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jiankang He
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ruhui Luo
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xuan Lai
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Wenjing Liu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Fujian Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yiyuan Kang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
14
|
Luangmonkong T, Parichatikanond W, Olinga P. Targeting collagen homeostasis for the treatment of liver fibrosis: Opportunities and challenges. Biochem Pharmacol 2023; 215:115740. [PMID: 37567319 DOI: 10.1016/j.bcp.2023.115740] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Liver fibrosis is an excessive production, aberrant deposition, and deficit degradation of extracellular matrix (ECM). Patients with unresolved fibrosis ultimately undergo end-stage liver diseases. To date, the effective and safe strategy to cease fibrosis progression remains an unmet clinical need. Since collagens are the most abundant ECM protein which play an essential role in fibrogenesis, the suitable regulation of collagen homeostasis could be an effective strategy for the treatment of liver fibrosis. Therefore, this review provides a brief overview on the dysregulation of ECM homeostasis, focusing on collagens, in the pathogenesis of liver fibrosis. Most importantly, promising therapeutic mechanisms related to biosynthesis, deposition and extracellular interactions, and degradation of collagens, together with preclinical and clinical antifibrotic evidence of drugs affecting each target are orderly criticized. In addition, challenges for targeting collagen homeostasis in the treatment of liver fibrosis are discussed.
Collapse
Affiliation(s)
- Theerut Luangmonkong
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Thailand; Centre of Biopharmaceutical Science for Healthy Ageing (BSHA), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand.
| | - Warisara Parichatikanond
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Thailand; Centre of Biopharmaceutical Science for Healthy Ageing (BSHA), Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, The Netherlands
| |
Collapse
|
15
|
Moreno-Lanceta A, Medrano-Bosch M, Fundora Y, Perramón M, Aspas J, Parra-Robert M, Baena S, Fondevila C, Edelman ER, Jiménez W, Melgar-Lesmes P. RNF41 orchestrates macrophage-driven fibrosis resolution and hepatic regeneration. Sci Transl Med 2023; 15:eabq6225. [PMID: 37437019 DOI: 10.1126/scitranslmed.abq6225] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/16/2023] [Indexed: 07/14/2023]
Abstract
Hepatic inflammation is a common trigger of chronic liver disease. Macrophage activation is a predictive parameter for survival in patients with cirrhosis. Ring finger protein 41 (RNF41) negatively regulates proinflammatory cytokines and receptors; however, the precise involvement of macrophage RNF41 in liver cirrhosis remains unknown. Here, we sought to understand how RNF41 dictates macrophage fate in hepatic fibrosis and repair within the inflammatory milieu. We found that RNF41 expression is down-regulated in CD11b+ macrophages recruited to mouse fibrotic liver and to patient cirrhotic liver regardless of cirrhosis etiology. Prolonged inflammation with TNF-α progressively reduced macrophage RNF41 expression. We designed a macrophage-selective gene therapy with dendrimer-graphite nanoparticles (DGNPs) to explore the influence of macrophage RNF41 restoration and depletion in liver fibrosis and regeneration. RNF41 expression induced in CD11b+ macrophages by DGNP-conjugated plasmids ameliorated liver fibrosis, reduced liver injury, and stimulated hepatic regeneration in fibrotic mice with or without hepatectomy. This therapeutic effect was mainly mediated by the induction of insulin-like growth factor 1. Conversely, depletion of macrophage RNF41 worsened inflammation, fibrosis, hepatic damage, and survival. Our data reveal implications of macrophage RNF41 in the control of hepatic inflammation, fibrosis, and regeneration and provide a rationale for therapeutic strategies in chronic liver disease and potentially other diseases characterized by inflammation and fibrosis.
Collapse
Affiliation(s)
- Alazne Moreno-Lanceta
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona 08036, Spain
- Institut d'Investigacions Biomèdiques August Pi-Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona 08036, Spain
| | - Mireia Medrano-Bosch
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona 08036, Spain
| | - Yilliam Fundora
- Institut d'Investigacions Biomèdiques August Pi-Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona 08036, Spain
- Liver Transplant Unit, Institut Clínic de Malalties Digestives I Metabòliques, Hospital Clínic, University of Barcelona, Barcelona 08036, Spain
| | - Meritxell Perramón
- Institut d'Investigacions Biomèdiques August Pi-Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona 08036, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Barcelona 08036, Spain
| | - Jessica Aspas
- Liver Transplant Unit, Institut Clínic de Malalties Digestives I Metabòliques, Hospital Clínic, University of Barcelona, Barcelona 08036, Spain
| | - Marina Parra-Robert
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Barcelona 08036, Spain
| | - Sheila Baena
- Liver Transplant Unit, Institut Clínic de Malalties Digestives I Metabòliques, Hospital Clínic, University of Barcelona, Barcelona 08036, Spain
| | - Constantino Fondevila
- Institut d'Investigacions Biomèdiques August Pi-Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona 08036, Spain
- Liver Transplant Unit, Institut Clínic de Malalties Digestives I Metabòliques, Hospital Clínic, University of Barcelona, Barcelona 08036, Spain
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wladimiro Jiménez
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona 08036, Spain
- Institut d'Investigacions Biomèdiques August Pi-Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona 08036, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Barcelona 08036, Spain
| | - Pedro Melgar-Lesmes
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona 08036, Spain
- Institut d'Investigacions Biomèdiques August Pi-Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona 08036, Spain
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
16
|
Medrano-Bosch M, Simón-Codina B, Jiménez W, Edelman ER, Melgar-Lesmes P. Monocyte-endothelial cell interactions in vascular and tissue remodeling. Front Immunol 2023; 14:1196033. [PMID: 37483594 PMCID: PMC10360188 DOI: 10.3389/fimmu.2023.1196033] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
Monocytes are circulating leukocytes of innate immunity derived from the bone marrow that interact with endothelial cells under physiological or pathophysiological conditions to orchestrate inflammation, angiogenesis, or tissue remodeling. Monocytes are attracted by chemokines and specific receptors to precise areas in vessels or tissues and transdifferentiate into macrophages with tissue damage or infection. Adherent monocytes and infiltrated monocyte-derived macrophages locally release a myriad of cytokines, vasoactive agents, matrix metalloproteinases, and growth factors to induce vascular and tissue remodeling or for propagation of inflammatory responses. Infiltrated macrophages cooperate with tissue-resident macrophages during all the phases of tissue injury, repair, and regeneration. Substances released by infiltrated and resident macrophages serve not only to coordinate vessel and tissue growth but cellular interactions as well by attracting more circulating monocytes (e.g. MCP-1) and stimulating nearby endothelial cells (e.g. TNF-α) to expose monocyte adhesion molecules. Prolonged tissue accumulation and activation of infiltrated monocytes may result in alterations in extracellular matrix turnover, tissue functions, and vascular leakage. In this review, we highlight the link between interactions of infiltrating monocytes and endothelial cells to regulate vascular and tissue remodeling with a special focus on how these interactions contribute to pathophysiological conditions such as cardiovascular and chronic liver diseases.
Collapse
Affiliation(s)
- Mireia Medrano-Bosch
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Blanca Simón-Codina
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Wladimiro Jiménez
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Elazer R. Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Pedro Melgar-Lesmes
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
17
|
Moreno-Lanceta A, Medrano-Bosch M, Simón-Codina B, Barber-González M, Jiménez W, Melgar-Lesmes P. PPAR-γ Agonist GW1929 Targeted to Macrophages with Dendrimer-Graphene Nanostars Reduces Liver Fibrosis and Inflammation. Pharmaceutics 2023; 15:pharmaceutics15051452. [PMID: 37242695 DOI: 10.3390/pharmaceutics15051452] [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: 03/29/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Macrophages play essential roles during the progression of chronic liver disease. They actively participate in the response to liver damage and in the balance between fibrogenesis and regression. The activation of the PPARγ nuclear receptor in macrophages has traditionally been associated with an anti-inflammatory phenotype. However, there are no PPARγ agonists with high selectivity for macrophages, and the use of full agonists is generally discouraged due to severe side effects. We designed dendrimer-graphene nanostars linked to a low dose of the GW1929 PPARγ agonist (DGNS-GW) for the selective activation of PPARγ in macrophages in fibrotic livers. DGNS-GW preferentially accumulated in inflammatory macrophages in vitro and attenuated macrophage pro-inflammatory phenotype. The treatment with DGNS-GW in fibrotic mice efficiently activated liver PPARγ signaling and promoted a macrophage switch from pro-inflammatory M1 to anti-inflammatory M2 phenotype. The reduction of hepatic inflammation was associated with a significant reduction in hepatic fibrosis but did not alter liver function or hepatic stellate cell activation. The therapeutic antifibrotic utility of DGNS-GW was attributed to an increased expression of hepatic metalloproteinases that allowed extracellular matrix remodeling. In conclusion, the selective activation of PPARγ in hepatic macrophages with DGNS-GW significantly reduced hepatic inflammation and stimulated extracellular matrix remodeling in experimental liver fibrosis.
Collapse
Affiliation(s)
- Alazne Moreno-Lanceta
- Department of Biomedicine, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
| | - Mireia Medrano-Bosch
- Department of Biomedicine, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Blanca Simón-Codina
- Department of Biomedicine, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | | | - Wladimiro Jiménez
- Department of Biomedicine, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
| | - Pedro Melgar-Lesmes
- Department of Biomedicine, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
18
|
Shan L, Wang F, Zhai D, Meng X, Liu J, Lv X. Matrix metalloproteinases induce extracellular matrix degradation through various pathways to alleviate hepatic fibrosis. Biomed Pharmacother 2023; 161:114472. [PMID: 37002573 DOI: 10.1016/j.biopha.2023.114472] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/20/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Liver fibrosis is the common consequence of various chronic liver injuries and is mainly characterized by the imbalance between the production and degradation of extracellular matrix, which leads to the accumulation of interstitial collagen and other matrix components. Matrix metalloproteinases (MMPs) and their specific inhibitors, that is, tissue inhibitors of metalloproteinases (TIMPs), play a crucial role in collagen synthesis and lysis. Previous in vivo and in vitro studies of our laboratory found repressing extracellular matrix (ECM) accumulation by restoring the balance between MMPs and TIMPs can alleviate liver fibrosis. We conducted a review of articles published in PubMed and Science Direct in the last decade until February 1, 2023, which were searched for using these words "MMPs/TIMPs" and "Hepatic Fibrosis." Through a literature review, this article reviews the experimental studies of liver fibrosis based on MMPs/TIMPs, summarizes the components that may exert an anti-liver fibrosis effect by affecting the expression or activity of MMPs/TIMPs, and attempts to clarify the mechanism of MMPs/TIMPs in regulating collagen homeostasis, so as to provide support for the development of anti-liver fibrosis drugs. We found the MMP-TIMP-ECM interaction can result in better understanding of the pathogenesis and progression of hepatic fibrosis from a different angle, and targeting this interaction may be a promising therapeutic strategy for hepatic fibrosis. Additionally, we summarized and analyzed the drugs that have been found to reduce liver fibrosis by changing the ratio of MMPs/TIMPs, including medicine natural products.
Collapse
Affiliation(s)
- Liang Shan
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China; The Key Laboratory of Major Autoimmune Diseases, Hefei 230032, Anhui, China
| | - Fengling Wang
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Dandan Zhai
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Xiangyun Meng
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Jianjun Liu
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China.
| | - Xiongwen Lv
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China; The Key Laboratory of Major Autoimmune Diseases, Hefei 230032, Anhui, China.
| |
Collapse
|
19
|
Anwar I, Ashfaq UA. Impact of Nanotechnology on Differentiation and Augmentation of Stem Cells for Liver Therapy. Crit Rev Ther Drug Carrier Syst 2023; 40:89-116. [PMID: 37585310 DOI: 10.1615/critrevtherdrugcarriersyst.2023042400] [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: 08/18/2023]
Abstract
The liver is one of the crucial organs of the body that performs hundreds of chemical reactions needed by the body to survive. It is also the largest gland of the body. The liver has multiple functions, including the synthesis of chemicals, metabolism of nutrients, and removal of toxins. It also acts as a storage unit. The liver has a unique ability to regenerate itself, but it can lead to permanent damage if the injury is beyond recovery. The only possible treatment of severe liver damage is liver transplant which is a costly procedure and has several other drawbacks. Therefore, attention has been shifted towards the use of stem cells that have shown the ability to differentiate into hepatocytes. Among the numerous kinds of stem cells (SCs), the mesenchymal stem cells (MSCs) are the most famous. Various studies suggest that an MSC transplant can repair liver function, improve the signs and symptoms, and increase the chances of survival. This review discusses the impact of combining stem cell therapy with nanotechnology. By integrating stem cell science and nanotechnology, the information about stem cell differentiation and regulation will increase, resulting in a better comprehension of stem cell-based treatment strategies. The augmentation of SCs with nanoparticles has been shown to boost the effect of stem cell-based therapy. Also, the function of green nanoparticles in liver therapies is discussed.
Collapse
Affiliation(s)
- Ifrah Anwar
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
20
|
Jari Litany RI, Praseetha PK. Tiny tots for a big-league in wound repair: Tools for tissue regeneration by nanotechniques of today. J Control Release 2022; 349:443-459. [PMID: 35835401 DOI: 10.1016/j.jconrel.2022.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/13/2022]
Abstract
Overall, chronic injuries place considerable burden on patients and health systems. The skin injuries are exposed to inflammatory bacteria and hinder the healing process. The skin being the biggest tissue of the whole body ensures protection against microbial invasion, dehydration, and against chemical, thermal, bright radiations and mechanical agents. When injured, the skin loses its defensive purpose and the attack of bacterial types arises with the loss of protein, water, and electrolytes. Improved wound closure therapy helps to restore normal skin function by managing wounds with the help of a suitable skin replacement. According to the type of wound and its healing ability, an appropriate skin replacement system must be identified. Nanofibrous layers because of their permeable structure, their large superficial reach and their similarity with the local extracellular network serve as cutaneous substitution for dealing with deep and superficial injuries. By a diminished microbial load without infestation, scab formation and infiltration of defense cells in the initial phase, acute injuries are usually characterized. Here recovery is related with epithelialization, angiogenesis and relocation of fibroblasts. The wound becomes obstinate when microbial biofilms are developed while the immune system does not manage to eliminate the infection. Increased inflammatory process, lower deep tissue oxygenation, fibrin cuffs, fibroblastic senescence, altered angiogenesis, stalled re-epithelialization and chronic infection have been visualized. Conventional wound mending treatments for the most part falling flat to supply a great clinical result, either basically like wound epithelialization and regulation of fluid loss or practically like histological highlights that decide versatility, strength, affectability, etc. Conventional wound therapies commonly fail to offer a better medical output, like wound epithelialization and regulation of fluid reduction or physiologically like cellular features that determine durability, sensitivity, elasticity, etc. Nanotechnology may be a dependable investigation space for wound-healing treatments through their versatile physicochemical properties. Advancing nano platforms with novel solutions for curing chronicdiabetic wounds are discussed in detail that can guide further research in this sector.
Collapse
Affiliation(s)
- R I Jari Litany
- Department of Nanotechnology, Noorul Islam Centre for Higher Education, Kumaracoil, Tamil Nadu 629180, India
| | - P K Praseetha
- Department of Nanotechnology, Noorul Islam Centre for Higher Education, Kumaracoil, Tamil Nadu 629180, India.
| |
Collapse
|
21
|
Yang J, Luo G, Li C, Zhao Z, Ju S, Li Q, Chen Z, Ding C, Tong X, Zhao J. Cystatin SN promotes epithelial-mesenchymal transition and serves as a prognostic biomarker in lung adenocarcinoma. BMC Cancer 2022; 22:589. [PMID: 35637432 PMCID: PMC9150371 DOI: 10.1186/s12885-022-09685-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/17/2022] [Indexed: 11/14/2022] Open
Abstract
Background Cystatins are a class of proteins that can inhibit cysteine protease and are widely distributed in human bodily fluids and secretions. Cystatin SN (CST1), a member of the CST superfamily, is abnormally expressed in a variety of tumors. However, its effect on the occurrence and development of lung adenocarcinoma (LUAD) remains unclear. Methods We obtained transcriptome analysis data of CST1 from The Cancer Genome Atlas (TCGA) and GSE31210 databases. The association of CST1 expression with prognosis, gene mutations and tumor immune microenvironment was analyzed using public databases. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were performed to investigate the potential mechanisms of CST1. Results In this study, we found that CST1 was highly expressed in lung adenocarcinoma and was associated with prognosis and tumor immune microenvironment. Genetic mutations of CST1 were shown to be related to disease-free survival (DFS) by using the c-BioPortal tool. Potential proteins binding to CST1 were identified by constructing a protein-protein interaction (PPI) network. Gene set enrichment analysis (GSEA) of CST1 revealed that CST1 was notably enriched in epithelial-mesenchymal transition (EMT). Cell experiments confirmed that overexpression of CST1 promoted lung adenocarcinoma cells migration and invasion, while knockdown of CST1 significantly inhibited lung adenocarcinoma cells migration and invasion. Conclusions Our comprehensive bioinformatics analyses revealed that CST1 may be a novel prognostic biomarker in LUAD. Experiments confirmed that CST1 promotes epithelial-mesenchymal transition in LUAD cells. These findings will help to better understand the distinct role of CST1 in LUAD. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09685-z.
Collapse
Affiliation(s)
- Jian Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gaomeng Luo
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chang Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhunlin Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Sheng Ju
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qifan Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhike Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.,Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Tong
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China. .,Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China. .,Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
22
|
Zhong Y, Yang L, Zhou Y, Peng J. Biomarker-responsive Fluorescent Probes for In Vivo Imaging of Liver Injury. Chem Asian J 2022; 17:e202200038. [PMID: 35182452 DOI: 10.1002/asia.202200038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/16/2022] [Indexed: 11/08/2022]
Abstract
Liver injury-related diseases have aroused widespread concern due to its extreme unpredictability, acute onset, and severe consequences. Nowadays, the clinical prediction and assessment of liver injury mainly focus on histopathological and serological approaches, which undergoes a tedious process and sometimes requires invasive biopsy. Over the past decades, fluorescence imaging technique have emerged as a rising star for the diagnosis of diseases owing to its noninvasiveness, high fidelity and ease of operation. On regard to liver injury, the fluorescent probes have been delicately designed to response a variety of endogenous biomolecules to precisely offer comprehensive information about the lesion site. Herein, we make a brief summary and discussion about the design strategies and applications of the recently reported fluorescent biosensors responsive to a series of biomarkers involved in the liver injury. The potential prospects and remaining challenges are also discussed to promote the progression in this field.
Collapse
Affiliation(s)
- Yang Zhong
- China Pharmaceutical University, State Key Laboratory of Natural Medicines, CHINA
| | - Lulu Yang
- China Pharmaceutical University, School of Basic Medicine and Clinical Pharmacy, CHINA
| | - Yunyun Zhou
- China Pharmaceutical University, State Key Laboratory of Natural Medicines, CHINA
| | - Juanjuan Peng
- China Pharmaceutical University, #24 Tong Jia Xiang, Nanjing, CHINA
| |
Collapse
|
23
|
Keshvari S, Genz B, Teakle N, Caruso M, Cestari MF, Patkar OL, Tse BWC, Sokolowski KA, Ebersbach H, Jascur J, MacDonald KPA, Miller G, Ramm GA, Pettit AR, Clouston AD, Powell EE, Hume DA, Irvine KM. Therapeutic potential of macrophage colony-stimulating factor (CSF1) in chronic liver disease. Dis Model Mech 2022; 15:274391. [PMID: 35169835 PMCID: PMC9044210 DOI: 10.1242/dmm.049387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/08/2022] [Indexed: 11/20/2022] Open
Abstract
Resident and recruited macrophages control the development and proliferation of the liver. We showed previously in multiple species that treatment with a macrophage colony stimulating factor (CSF1)-Fc fusion protein initiated hepatocyte proliferation and promoted repair in models of acute hepatic injury in mice. Here we investigated the impact of CSF1-Fc on resolution of advanced fibrosis and liver regeneration, utilizing a non-resolving toxin-induced model of chronic liver injury and fibrosis in C57BL/6J mice. Co-administration of CSF1-Fc with exposure to thioacetamide (TAA) exacerbated inflammation consistent with monocyte contributions to initiation of pathology. After removal of TAA, either acute or chronic CSF1-Fc treatment promoted liver growth, prevented progression and promoted resolution of fibrosis. Acute CSF1-Fc treatment was also anti-fibrotic and pro-regenerative in a model of partial hepatectomy in mice with established fibrosis. The beneficial impacts of CSF1-Fc treatment were associated with monocyte-macrophage recruitment and increased expression of remodeling enzymes and growth factors. These studies indicate that CSF1-dependent macrophages contribute to both initiation and resolution of fibrotic injury and that CSF1-Fc has therapeutic potential in human liver disease.
Collapse
Affiliation(s)
- Sahar Keshvari
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Berit Genz
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Ngari Teakle
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Melanie Caruso
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Michelle F Cestari
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Omkar L Patkar
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Brian W C Tse
- Preclinical Imaging Facility, Translational Research Institute, Brisbane, Queensland, Australia
| | - Kamil A Sokolowski
- Preclinical Imaging Facility, Translational Research Institute, Brisbane, Queensland, Australia
| | - Hilmar Ebersbach
- Novartis Institutes for Biomedical Research (NIBR), Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Julia Jascur
- Novartis Institutes for Biomedical Research (NIBR), Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | | | | | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Allison R Pettit
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Andrew D Clouston
- Envoi Specialist Pathologists, Brisbane, Qld, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Elizabeth E Powell
- Faculty of Medicine, The University of Queensland, Brisbane, Australia.,Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - David A Hume
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Katharine M Irvine
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
24
|
Liu M, Huang Q, Zhu Y, Chen L, Li Y, Gong Z, Ai K. Harnessing reactive oxygen/nitrogen species and inflammation: Nanodrugs for liver injury. Mater Today Bio 2022; 13:100215. [PMID: 35198963 PMCID: PMC8850330 DOI: 10.1016/j.mtbio.2022.100215] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 12/11/2022] Open
Abstract
Overall, 12% of the global population (800 million) suffers from liver disease, which causes 2 million deaths every year. Liver injury involving characteristic reactive oxygen/nitrogen species (RONS) and inflammation plays a key role in progression of liver disease. As a key metabolic organ of the human body, the liver is susceptible to injury from various sources, including COVID-19 infection. Owing to unique structural features and functions of the liver, most current antioxidants and anti-inflammatory drugs are limited against liver injury. However, the characteristics of the liver could be utilized in the development of nanodrugs to achieve specific enrichment in the liver and consequently targeted treatment. Nanodrugs have shown significant potential in eliminating RONS and regulating inflammation, presenting an attractive therapeutic tool for liver disease through controlling liver injury. Therefore, the main aim of the current review is to provide a comprehensive summary of the latest developments contributing to our understanding of the mechanisms underlying nanodrugs in the treatment of liver injury via harnessing RONS and inflammation. Meanwhile, the prospects of nanodrugs for liver injury therapy are systematically discussed, which provides a sound platform for novel therapeutic insights and inspiration for design of nanodrugs to treat liver disease.
Collapse
Affiliation(s)
- Min Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yan Zhu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Li Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yumei Li
- Department of Assisted Reproduction, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| |
Collapse
|
25
|
Cheng D, Chai J, Wang H, Fu L, Peng S, Ni X. Hepatic macrophages: Key players in the development and progression of liver fibrosis. Liver Int 2021; 41:2279-2294. [PMID: 33966318 DOI: 10.1111/liv.14940] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/15/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022]
Abstract
Hepatic fibrosis is a common pathological process involving persistent liver injury with various etiologies and subsequent inflammatory responses that occur in chronic liver diseases. If left untreated, liver fibrosis can progress to liver cirrhosis, hepatocellular carcinoma and eventually, liver failure. Unfortunately, to date, there is no effective treatment for liver fibrosis, with the exception of liver transplantation. Although the pathophysiology of liver fibrosis is multifactorial and includes the activation of hepatic stellate cells, which are known to drive liver fibrogenesis, hepatic macrophages have emerged as central players in the development of liver fibrosis and regression. Hepatic macrophages, which consist of resident macrophages (Kupffer cells) and monocyte-derived macrophages, have been shown to play an intricate role in the initiation of inflammatory responses to liver injury, progression of fibrosis, and promotion of fibrosis resolution. These features have made hepatic macrophages uniquely attractive therapeutic targets in the fight against hepatic fibrosis. In this review, we synthesised the literature to highlight the functions and regulation of heterogeneity in hepatic macrophages. Furthermore, using the existing findings, we attempt to offer insights into the molecular mechanisms underlying the phenotypic switch from fibrogenic macrophages to restorative macrophages, the regulation of heterogeneity, and modes of action for hepatic macrophages. A better understanding of these mechanisms may guide the development of novel anti-fibrotic therapies (eg macrophage subset-targeted treatments) to combat liver fibrosis in the future.
Collapse
Affiliation(s)
- Da Cheng
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, China
| | - Jin Chai
- Cholestatic Liver Diseases Center, Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Huiwen Wang
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, China
| | - Lei Fu
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| | - Shifang Peng
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| | - Xin Ni
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
- International Collaborative Research Center for Medical Metabolomics, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
26
|
Nanoparticles to Target and Treat Macrophages: The Ockham's Concept? Pharmaceutics 2021; 13:pharmaceutics13091340. [PMID: 34575416 PMCID: PMC8469871 DOI: 10.3390/pharmaceutics13091340] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022] Open
Abstract
Nanoparticles are nanomaterials with three external nanoscale dimensions and an average size ranging from 1 to 1000 nm. Nanoparticles have gained notoriety in technological advances due to their tunable physical, chemical, and biological characteristics. However, the administration of functionalized nanoparticles to living beings is still challenging due to the rapid detection and blood and tissue clearance by the mononuclear phagocytic system. The major exponent of this system is the macrophage. Regardless the nanomaterial composition, macrophages can detect and incorporate foreign bodies by phagocytosis. Therefore, the simplest explanation is that any injected nanoparticle will be probably taken up by macrophages. This explains, in part, the natural accumulation of most nanoparticles in the spleen, lymph nodes, and liver (the main organs of the mononuclear phagocytic system). For this reason, recent investigations are devoted to design nanoparticles for specific macrophage targeting in diseased tissues. The aim of this review is to describe current strategies for the design of nanoparticles to target macrophages and to modulate their immunological function involved in different diseases with special emphasis on chronic inflammation, tissue regeneration, and cancer.
Collapse
|
27
|
Zhao C, Li Z, Chen J, Su L, Wang J, Chen DS, Ye J, Liao N, Yang H, Song J, Shi J. Site-Specific Biomimicry of Antioxidative Melanin Formation and Its Application for Acute Liver Injury Therapy and Imaging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102391. [PMID: 34278624 DOI: 10.1002/adma.202102391] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/29/2021] [Indexed: 06/13/2023]
Abstract
Biocompatible nano-antioxidants composed of natural molecules/materials, such as dopamine and melanin, are of great interest for diverse biomedical applications. However, the lack of understanding of the precise structure of these biomaterials and thus the actual dose of effective components impedes their advancement to translation. Herein, a strategy to mimic in situ melanin formation and explore its antioxidative applications is reported, by developing a PEGylated, phenylboronic-acid-protected L-DOPA precursor (PAD) that can self-assemble into well-defined nanoparticles (PADN). Exposure to oxidative species leads to deprotection of phenylboronic acids, transforming PADN to PEG-L-DOPA, which, similar to the biosynthetic pathway of melanin, can be oxidized and polymerized into an antioxidative melanin-like structure. With ultrahigh stability and superior antioxidative activity, the PADN shows remarkable efficacy in prevention and treatment of acute liver injury/failure. Moreover, the in situ structure transformation enables PADN to visualize damaged tissue noninvasively by photoacoustic imaging. Overall, a bioinspired antioxidant with precise structure and site-specific biological activity for theranostics of oxidative stress-related diseases is described.
Collapse
Affiliation(s)
- Caiyan Zhao
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou University, Fuzhou, 350108, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Zhi Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou University, Fuzhou, 350108, China
| | - Jingxiao Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, China
| | - Lichao Su
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou University, Fuzhou, 350108, China
| | - Junqing Wang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dean Shuailin Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jiamin Ye
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou University, Fuzhou, 350108, China
| | - Naishun Liao
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou University, Fuzhou, 350108, China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou University, Fuzhou, 350108, China
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou University, Fuzhou, 350108, China
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
28
|
Dai Q, Jiang W, Liu H, Qing X, Wang G, Huang F, Yang Z, Wang C, Gu E, Zhao H, Zhang J, Liu X. Kupffer cell-targeting strategy for the protection of hepatic ischemia/reperfusion injury. NANOTECHNOLOGY 2021; 32:265101. [PMID: 33472187 DOI: 10.1088/1361-6528/abde02] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/20/2021] [Indexed: 06/12/2023]
Abstract
The aim of this study is to evaluate the effect of rare earth upconversion nanoparticles (UCNs) on hepatic ischemia reperfusion injury (IRI) and explore its possible mechanism. Hepatic IRI seriously affects the prognosis of patients undergoing liver surgery. Liver-resident Kupffer cells have been reported to promote IRI. Nanomedicines are known to be effective in the treatment of liver diseases, however, Kupffer cell-targeting nanomedicines for the treatment of IRI are yet to be developed. As potential bioimaging nanomaterials, UCNs have been found to specifically deplete Kupffer cells, but the underlying mechanism is unknown. In this study, we found that UCNs specifically depleted Kupffer cells by pyroptosis, while the co-administration of the caspase-1 inhibitor VX-765 rescued the UCN-induced Kupffer cell pyroptosis in mice. Furthermore, the pre-depletion of Kupffer cells by the UCNs significantly suppressed the release of inflammatory cytokines and effectively improved hepatic IRI. The rescue of the pyroptosis of the Kupffer cells by VX-765 abrogated the protective effect of UCNs on the liver. These results suggest that UCNs are highly promising for the development of Kupffer cell-targeting nanomedicines for intraoperative liver protection.
Collapse
Affiliation(s)
- Qingqing Dai
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, People's Republic of China
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, People's Republic of China
| | - Wei Jiang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, People's Republic of China
| | - Hu Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, People's Republic of China
| | - Xin Qing
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, People's Republic of China
| | - Guobin Wang
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, People's Republic of China
| | - Fan Huang
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, People's Republic of China
| | - Zhilai Yang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, People's Republic of China
| | - Chunhui Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, People's Republic of China
| | - Erwei Gu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, People's Republic of China
| | - Hongchuan Zhao
- Department of Hepatopancreatobiliary Surgery and Organ Transplantation Center, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, People's Republic of China
| | - Jiqian Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, People's Republic of China
| | - Xuesheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, People's Republic of China
| |
Collapse
|
29
|
Alijani H, Noori A, Faridi N, Bathaie S, Mousavi MF. Aptamer-functionalized Fe3O4@MOF nanocarrier for targeted drug delivery and fluorescence imaging of the triple-negative MDA-MB-231 breast cancer cells. J SOLID STATE CHEM 2020. [DOI: 10.1016/j.jssc.2020.121680] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
30
|
Ghanbarinia Firozjah R, Sadeghi A, Khoee S. Ultrasonic De-cross-linking of the pH- and Magneto-Responsive PHEMA/PMMA Microgel to Janus Nanoparticles: A New Synthesis Based on "Grafting from"/"Grafting to" Polymerization. ACS OMEGA 2020; 5:27119-27132. [PMID: 33134672 PMCID: PMC7594003 DOI: 10.1021/acsomega.0c02710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/02/2020] [Indexed: 05/05/2023]
Abstract
Stimuli-responsive Janus nanoparticles (NPs) with a two-facial structure have been used widely in biomedical applications. Among several methods to prepare these NPs, surface-initiated atom transfer radical polymerization (SI-ATRP) has received much attention due to the precise deposition of polymers on the surface of the substrate. In this study, Janus nanoparticles with asymmetric surface chemistry were prepared through a masking method in three steps involving the covalent deposition of super paramagnetic iron oxide nanoparticles (SPIONs) on the cross-linked substrate based on methotrexate (MTX)-grafted poly(2-hydroxyethyl methacrylate) (CPM), surface functionalization of unreacted sites of immobilized SPIONs with 2-bromoisobutyryl bromide (BIBB) in order to prepare the macro-initiator (Br-Fe3O4-CPM), growing poly(methyl methacrylate) (PMMA) on the surface of the macro-initiator through the SI-ATRP method. Optical microscopy was utilized to monitor the successful modification of SPIONs. Poly(methyl methacrylate)-iron oxide-poly(2-hydroxyethyl methacrylate) (PMMA-Fe3O4-PHEMA) microgel was exposed to optimum ultrasound (US) waves to prepare the PMMA-Fe3O4-PHEMA nanoparticle. Transmission electron microscopy (TEM) was used to confirm the precise deposition of polymers and the Janus structure. The MTX release of US-synthesized Janus NPs was studied in PBS at pH values of 7.4 and 5.8. The release data were analyzed using the Excel add-in DDSolver program to evaluate the kinetics of the drug release process from the nanocarrier under different pH values.
Collapse
Affiliation(s)
- Rahim Ghanbarinia Firozjah
- Polymer Laboratory, School
of Chemistry, College of Science, University
of Tehran, P.O. Box 14155
6455, Tehran 1417466191, Iran
| | - Amirhossein Sadeghi
- Polymer Laboratory, School
of Chemistry, College of Science, University
of Tehran, P.O. Box 14155
6455, Tehran 1417466191, Iran
| | - Sepideh Khoee
- Polymer Laboratory, School
of Chemistry, College of Science, University
of Tehran, P.O. Box 14155
6455, Tehran 1417466191, Iran
| |
Collapse
|
31
|
Bai X, Su G, Zhai S. Recent Advances in Nanomedicine for the Diagnosis and Therapy of Liver Fibrosis. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1945. [PMID: 33003520 PMCID: PMC7599596 DOI: 10.3390/nano10101945] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/26/2020] [Accepted: 09/27/2020] [Indexed: 12/11/2022]
Abstract
Liver fibrosis, a reversible pathological process of inflammation and fiber deposition caused by chronic liver injury and can cause severe health complications, including liver failure, liver cirrhosis, and liver cancer. Traditional diagnostic methods and drug-based therapy have several limitations, such as lack of precision and inadequate therapeutic efficiency. As a medical application of nanotechnology, nanomedicine exhibits great potential for liver fibrosis diagnosis and therapy. Nanomedicine enhances imaging contrast and improves tissue penetration and cellular internalization; it simultaneously achieves targeted drug delivery, combined therapy, as well as diagnosis and therapy (i.e., theranostics). In this review, recent designs and development efforts of nanomedicine systems for the diagnosis, therapy, and theranostics of liver fibrosis are introduced. Relative to traditional methods, these nanomedicine systems generally demonstrate significant improvement in liver fibrosis treatment. Perspectives and challenges related to these nanomedicine systems translated from laboratory to clinical use are also discussed.
Collapse
Affiliation(s)
- Xue Bai
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China;
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong 226001, China
| | - Shumei Zhai
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China;
| |
Collapse
|
32
|
Single-Walled Carbon Nanohorns as Promising Nanotube-Derived Delivery Systems to Treat Cancer. Pharmaceutics 2020; 12:pharmaceutics12090850. [PMID: 32906852 PMCID: PMC7558911 DOI: 10.3390/pharmaceutics12090850] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/25/2020] [Accepted: 09/04/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer has become one of the most prevalent diseases worldwide, with increasing incidence in recent years. Current pharmacological strategies are not tissue-specific therapies, which hampers their efficacy and results in toxicity in healthy organs. Carbon-based nanomaterials have emerged as promising nanoplatforms for the development of targeted delivery systems to treat diseased cells. Single-walled carbon nanohorns (SWCNH) are graphene-based horn-shaped nanostructure aggregates with a multitude of versatile features to be considered as suitable nanosystems for targeted drug delivery. They can be easily synthetized and functionalized to acquire the desired physicochemical characteristics, and no toxicological effects have been reported in vivo followed by their administration. This review focuses on the use of SWCNH as drug delivery systems for cancer therapy. Their main applications include their capacity to act as anticancer agents, their use as drug delivery systems for chemotherapeutics, photothermal and photodynamic therapy, gene therapy, and immunosensing. The structure, synthesis, and covalent and non-covalent functionalization of these nanoparticles is also discussed. Although SWCNH are in early preclinical research yet, these nanotube-derived nanostructures demonstrate an interesting versatility pointing them out as promising forthcoming drug delivery systems to target and treat cancer cells.
Collapse
|
33
|
Mahdinloo S, Kiaie SH, Amiri A, Hemmati S, Valizadeh H, Zakeri-Milani P. Efficient drug and gene delivery to liver fibrosis: rationale, recent advances, and perspectives. Acta Pharm Sin B 2020; 10:1279-1293. [PMID: 32874828 PMCID: PMC7451940 DOI: 10.1016/j.apsb.2020.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/22/2020] [Accepted: 02/28/2020] [Indexed: 12/17/2022] Open
Abstract
Liver fibrosis results from chronic damages together with an accumulation of extracellular matrix, and no specific medical therapy is approved for that until now. Due to liver metabolic capacity for drugs, the fragility of drugs, and the presence of insurmountable physiological obstacles in the way of targeting, the development of efficient drug delivery systems for anti-fibrotics seems vital. We have explored articles with a different perspective on liver fibrosis over the two decades, then collected and summarized the information by providing corresponding in vitro and in vivo cases. We have discussed the mechanism of hepatic fibrogenesis with different ways of fibrosis induction in animals. Furthermore, the critical chemical and herbal anti-fibrotics, biological molecules such as micro-RNAs, siRNAs, and growth factors, which can affect cell division and differentiation, are mentioned. Likewise, drug and gene delivery and therapeutic systems on in vitro and in vivo models are summarized in the data tables. This review article enlightens recent advances in emerging drugs and nanocarriers and represents perspectives on targeting strategies employed in liver fibrosis treatment.
Collapse
Affiliation(s)
- Somayeh Mahdinloo
- Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz 5166616471, Iran
| | - Seyed Hossein Kiaie
- Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz 5166616471, Iran
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Ala Amiri
- Faculty of Basic Sciences, Islamic Azad University, Science and Research Branch, Tehran 1477893855, Iran
| | - Salar Hemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| |
Collapse
|
34
|
Soltani R, Guo S, Bianco A, Ménard‐Moyon C. Carbon Nanomaterials Applied for the Treatment of Inflammatory Diseases: Preclinical Evidence. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Rym Soltani
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572 University of Strasbourg, ISIS Strasbourg 67000 France
| | - Shi Guo
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572 University of Strasbourg, ISIS Strasbourg 67000 France
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572 University of Strasbourg, ISIS Strasbourg 67000 France
| | - Cécilia Ménard‐Moyon
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572 University of Strasbourg, ISIS Strasbourg 67000 France
| |
Collapse
|
35
|
Han XM, Zheng KW, Wang RL, Yue SF, Chen J, Zhao ZW, Song F, Su Y, Ma Q. Functionalization and optimization-strategy of graphene oxide-based nanomaterials for gene and drug delivery. Am J Transl Res 2020; 12:1515-1534. [PMID: 32509159 PMCID: PMC7270027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 04/14/2020] [Indexed: 06/11/2023]
Abstract
Graphene-family nanomaterials (GFNs) have been widely used in cancer therapy, tissue engineering, antibacterial and biological imaging due to their optical, thermal, and drug absorption properties. When used as drug and gene nanocarrier, the major limitations are aggregation, biocompatibility, and inappropriate release of drugs or genes. To overcome these problems, researchers have developed a variety of functionalization processes. In this review, we grouped the functionalization according to the decoration molecules, putting particular emphasis on the gene delivery. Organic and inorganic materials resulted as the major sets to introduce functional sections onto graphene oxide (GO). We also classified the target molecules used in the GO delivery system, as well as introduced other strategies to increase the delivery efficacy such as controlled release and magnetic targeting.
Collapse
Affiliation(s)
- Xiao-Min Han
- School of Basic and Forensic Medicine, Baotou Medical CollegeBaotou 014040, Inner Mongolia Autonomous Region, P. R. China
| | - Ke-Wen Zheng
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, The First Clinical College of Wenzhou Medical UniversityWenzhou 325000, Zhejiang Province, P. R. China
| | - Rui-Long Wang
- No. 5 Railway Middle SchoolBaotou, Inner Mongolia Autonomous Region, P. R. China
| | - Shu-Fen Yue
- School of Basic and Forensic Medicine, Baotou Medical CollegeBaotou 014040, Inner Mongolia Autonomous Region, P. R. China
| | - Jing Chen
- School of Basic and Forensic Medicine, Baotou Medical CollegeBaotou 014040, Inner Mongolia Autonomous Region, P. R. China
| | - Zi-Wei Zhao
- School of Basic and Forensic Medicine, Baotou Medical CollegeBaotou 014040, Inner Mongolia Autonomous Region, P. R. China
| | - Fang Song
- School of Basic and Forensic Medicine, Baotou Medical CollegeBaotou 014040, Inner Mongolia Autonomous Region, P. R. China
| | - Yan Su
- School of Basic and Forensic Medicine, Baotou Medical CollegeBaotou 014040, Inner Mongolia Autonomous Region, P. R. China
- Blood Conservation Institute, Baotou Medical CollegeBaotou 014040, Inner Mongolia Autonomous Region, P. R. China
| | - Qiang Ma
- School of Basic and Forensic Medicine, Baotou Medical CollegeBaotou 014040, Inner Mongolia Autonomous Region, P. R. China
- Blood Conservation Institute, Baotou Medical CollegeBaotou 014040, Inner Mongolia Autonomous Region, P. R. China
| |
Collapse
|
36
|
Gheybi H, Sattari S, Soleimani K, Adeli M. Graphene-dendritic polymer hybrids: synthesis, properties, and applications. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2019. [DOI: 10.1007/s13738-019-01817-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
37
|
Roles of the Hepatic Endocannabinoid and Apelin Systems in the Pathogenesis of Liver Fibrosis. Cells 2019; 8:cells8111311. [PMID: 31653030 PMCID: PMC6912778 DOI: 10.3390/cells8111311] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/17/2019] [Accepted: 10/23/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatic fibrosis is the consequence of an unresolved wound healing process in response to chronic liver injury and involves multiple cell types and molecular mechanisms. The hepatic endocannabinoid and apelin systems are two signalling pathways with a substantial role in the liver fibrosis pathophysiology-both are upregulated in patients with advanced liver disease. Endogenous cannabinoids are lipid-signalling molecules derived from arachidonic acid involved in the pathogenesis of cardiovascular dysfunction, portal hypertension, liver fibrosis, and other processes associated with hepatic disease through their interactions with the CB1 and CB2 receptors. Apelin is a peptide that participates in cardiovascular and renal functions, inflammation, angiogenesis, and hepatic fibrosis through its interaction with the APJ receptor. The endocannabinoid and apelin systems are two of the multiple cell-signalling pathways involved in the transformation of quiescent hepatic stellate cells into myofibroblast like cells, the main matrix-producing cells in liver fibrosis. The mechanisms underlying the control of hepatic stellate cell activity are coincident despite the marked dissimilarities between the endocannabinoid and apelin signalling pathways. This review discusses the current understanding of the molecular and cellular mechanisms by which the hepatic endocannabinoid and apelin systems play a significant role in the pathophysiology of liver fibrosis.
Collapse
|
38
|
Khoee S, Sadeghi A. An NIR-triggered drug release and highly efficient photodynamic therapy from PCL/PNIPAm/porphyrin modified graphene oxide nanoparticles with the Janus morphology. RSC Adv 2019; 9:39780-39792. [PMID: 35541408 PMCID: PMC9076064 DOI: 10.1039/c9ra06058h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/10/2019] [Indexed: 11/21/2022] Open
Abstract
This project aimed to investigate the synthesis and characteristics of stimuli-responsive nanoparticles with different morphologies. In the first step, graphene oxide was synthesized based on the improved Hummers' method. Then, thermo-responsive poly(N-isopropylacrylamide-co-N-(hydroxymethyl)acrylamide), an amphiphilic copolymer, and poly(caprolactone) (PCL), a hydrophobic polymer, were used to prepare Janus and mixed graphene oxide-based nanoparticles. Fluorescence microscopy was utilized to confirm the Janus structure by labeling the mixed and Janus NPs with fluorescent hydrophobic and hydrophilic dyes via a solvent-evaporation method. Then, terminally modified carboxyl porphyrin (TPPC3-COOH), used as the second generation photosensitizer, was grafted to the copolymer surrounding the mixed and Janus NPs. Next, quercetin, a hydrophobic anti-cancer drug, was loaded onto both NPs to accomplish NIR-triggered photodynamic- and chemo-therapy. Finally, the drug loading, encapsulation efficiency, and in vitro release of thermo-responsive NPs were investigated at temperatures of 37 °C and 40 °C as well as under laser irradiation (808 nm). This project aimed to investigate the synthesis and characteristics of stimuli-responsive nanoparticles with different morphologies.![]()
Collapse
Affiliation(s)
- Sepideh Khoee
- Polymer Laboratory
- School of Chemistry
- College of Science
- University of Tehran
- Tehran
| | - Amirhossein Sadeghi
- Polymer Laboratory
- School of Chemistry
- College of Science
- University of Tehran
- Tehran
| |
Collapse
|