1
|
Macrophage polarization is involved in liver fibrosis induced by β 1-adrenoceptor autoantibody. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1100-1112. [PMID: 35983976 PMCID: PMC9828683 DOI: 10.3724/abbs.2022102] [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] [Indexed: 11/25/2022] Open
Abstract
Accumulating evidence suggests that liver injury can be induced by the over-expression of β 1-adrenergic receptors (β 1-ARs). High titers of autoantibodies specific to β 1-adrenergic receptors (β 1-AA) are detected in the sera of heart failure patients, potentially playing agonist-like roles. However, the role of β 1-AA in liver function has not been characterized. In this study, we collect the sera of primary biliary cholangitis (PBC) patients, a condition which easily develops into liver fibrosis, and analyze the relationship between PBC and β 1-AA. A passive immunization model is established to assess the effect of β 1-AA on the liver. Subsequently, the effect of β 1-AA on macrophages is investigated in vitro. Results show that PBC patients have a high titer and ratio of β 1-AA, compared to controls. Liver injury and fibrosis are induced by β 1-AA. In vitro experiments with ROS probe demonstrate that β 1-AA induces macrophages to produce ROS and secrete TNFα. These effects can be partially reversed by metoprolol, a blocker for β 1-AR. Results from the transwell and phagocytosis assays show that β 1-AA promotes macrophage migration and phagocytosis. FCM tests suggest that β 1-AA induces the alteration of M1 rather than M2 markers in macrophages. Finally, the Annexin V/PI assay indicates that macrophage culture supernatants stimulated by β 1-AA cause hepatocyte apoptosis. Overall, these results suggest that β 1-AA is involved in PBC. The β 1-AA-induced activation, phagocytosis and phenotypic modification of macrophages may play an important role in the development of hepatic fibrosis and injury.
Collapse
|
2
|
Three-dimensional (3D) liver cell models - a tool for bridging the gap between animal studies and clinical trials when screening liver accumulation and toxicity of nanobiomaterials. Drug Deliv Transl Res 2022; 12:2048-2074. [PMID: 35507131 PMCID: PMC9066991 DOI: 10.1007/s13346-022-01147-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 12/13/2022]
Abstract
Despite the exciting properties and wide-reaching applications of nanobiomaterials (NBMs) in human health and medicine, their translation from bench to bedside is slow, with a predominant issue being liver accumulation and toxicity following systemic administration. In vitro 2D cell-based assays and in vivo testing are the most popular and widely used methods for assessing liver toxicity at pre-clinical stages; however, these fall short in predicting toxicity for NBMs. Focusing on in vitro and in vivo assessment, the accurate prediction of human-specific hepatotoxicity is still a significant challenge to researchers. This review describes the relationship between NBMs and the liver, and the methods for assessing toxicity, focusing on the limitations they bring in the assessment of NBM hepatotoxicity as one of the reasons defining the poor translation for NBMs. We will then present some of the most recent advances towards the development of more biologically relevant in vitro liver methods based on tissue-mimetic 3D cell models and how these could facilitate the translation of NBMs going forward. Finally, we also discuss the low public acceptance and limited uptake of tissue-mimetic 3D models in pre-clinical assessment, despite the demonstrated technical and ethical advantages associated with them.
Collapse
|
3
|
Salvati A, Poelstra K. Drug Targeting and Nanomedicine: Lessons Learned from Liver Targeting and Opportunities for Drug Innovation. Pharmaceutics 2022; 14:217. [PMID: 35057111 PMCID: PMC8777931 DOI: 10.3390/pharmaceutics14010217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 02/08/2023] Open
Abstract
Drug targeting and nanomedicine are different strategies for improving the delivery of drugs to their target. Several antibodies, immuno-drug conjugates and nanomedicines are already approved and used in clinics, demonstrating the potential of such approaches, including the recent examples of the DNA- and RNA-based vaccines against COVID-19 infections. Nevertheless, targeting remains a major challenge in drug delivery and different aspects of how these objects are processed at organism and cell level still remain unclear, hampering the further development of efficient targeted drugs. In this review, we compare properties and advantages of smaller targeted drug constructs on the one hand, and larger nanomedicines carrying higher drug payload on the other hand. With examples from ongoing research in our Department and experiences from drug delivery to liver fibrosis, we illustrate opportunities in drug targeting and nanomedicine and current challenges that the field needs to address in order to further improve their success.
Collapse
Affiliation(s)
- Anna Salvati
- Correspondence: (A.S.); (K.P.); Tel.: +31-503639831 (A.S.); +31-503633287 (K.P.)
| | - Klaas Poelstra
- Correspondence: (A.S.); (K.P.); Tel.: +31-503639831 (A.S.); +31-503633287 (K.P.)
| |
Collapse
|
4
|
Kumar V, Xin X, Ma J, Tan C, Osna N, Mahato RI. Therapeutic targets, novel drugs, and delivery systems for diabetes associated NAFLD and liver fibrosis. Adv Drug Deliv Rev 2021; 176:113888. [PMID: 34314787 PMCID: PMC8440458 DOI: 10.1016/j.addr.2021.113888] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/12/2021] [Accepted: 07/18/2021] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes mellitus (T2DM) associated non-alcoholic fatty liver disease (NAFLD) is the fourth-leading cause of death. Hyperglycemia induces various complications, including nephropathy, cirrhosis and eventually hepatocellular carcinoma (HCC). There are several etiological factors leading to liver disease development, which involve insulin resistance and oxidative stress. Free fatty acid (FFA) accumulation in the liver exerts oxidative and endoplasmic reticulum (ER) stresses. Hepatocyte injury induces release of inflammatory cytokines from Kupffer cells (KCs), which are responsible for activating hepatic stellate cells (HSCs). In this review, we will discuss various molecular targets for treating chronic liver diseases, including homeostasis of FFA, lipid metabolism, and decrease in hepatocyte apoptosis, role of growth factors, and regulation of epithelial-to-mesenchymal transition (EMT) and HSC activation. This review will also critically assess different strategies to enhance drug delivery to different cell types. Targeting nanocarriers to specific liver cell types have the potential to increase efficacy and suppress off-target effects.
Collapse
Affiliation(s)
- Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Xiaofei Xin
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jingyi Ma
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, University, MS 38677, USA
| | - Natalia Osna
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
5
|
Böttger R, Pauli G, Chao PH, AL Fayez N, Hohenwarter L, Li SD. Lipid-based nanoparticle technologies for liver targeting. Adv Drug Deliv Rev 2020; 154-155:79-101. [PMID: 32574575 DOI: 10.1016/j.addr.2020.06.017] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/26/2020] [Accepted: 06/16/2020] [Indexed: 12/18/2022]
Abstract
Liver diseases such as hepatitis, cirrhosis, and hepatocellular carcinoma are global health problems accounting for approximately 800 million cases and over 2 million deaths per year worldwide. Major drawbacks of standard pharmacological therapies are the inability to deliver a sufficient concentration of a therapeutic agent to the diseased liver, and nonspecific drug delivery leading to undesirable systemic side effects. Additionally, depending on the specific liver disease, drug delivery to a subset of liver cells is required. In recent years, lipid nanoparticles have been developed to passively and actively target drugs to the liver. The success of this approach has been highlighted by the FDA-approval of the first liver-targeting lipid nanoparticle, ONPATTRO, in 2018 and many other promising candidate technologies are expected to follow. This review summarizes recent developments of various lipid-based liver-targeting technologies, namely solid-lipid nanoparticles, liposomes, niosomes and micelles, and discusses the challenges and future perspectives in this field.
Collapse
|
6
|
Kamimura K, Yokoo T, Abe H, Terai S. Gene Therapy for Liver Cancers: Current Status from Basic to Clinics. Cancers (Basel) 2019; 11:cancers11121865. [PMID: 31769427 PMCID: PMC6966544 DOI: 10.3390/cancers11121865] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023] Open
Abstract
The liver is a key organ for metabolism, protein synthesis, detoxification, and endocrine function, and among liver diseases, including hepatitis, cirrhosis, malignant tumors, and congenital disease, liver cancer is one of the leading causes of cancer-related deaths worldwide. Conventional therapeutic options such as embolization and chemotherapy are not effective against advanced-stage liver cancer; therefore, continuous efforts focus on the development of novel therapeutic options, including molecular targeted agents and gene therapy. In this review, we will summarize the progress toward the development of gene therapies for liver cancer, with an emphasis on recent clinical trials and preclinical studies.
Collapse
Affiliation(s)
- Kenya Kamimura
- Correspondence: ; Tel.: +81-25-227-2207; Fax: +81-25-227-0776
| | | | | | | |
Collapse
|
7
|
Zhang Z, Wen H, Weng J, Feng L, Liu H, Hu X, Zeng F. Silencing of EPCAM suppresses hepatic fibrosis and hepatic stellate cell proliferation in mice with alcoholic hepatitis via the PI3K/Akt/mTOR signaling pathway. Cell Cycle 2019; 18:2239-2254. [PMID: 31378124 PMCID: PMC6738525 DOI: 10.1080/15384101.2019.1642067] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Alcoholic hepatitis (AH) is a severe condition developed in patients with underlying alcoholic liver disease. Epithelial cell adhesion molecule (EPCAM) plays a role in hepatitis. Therefore, the current study aimed to explore the effect of EPCAM and its potential mechanism in AH. Bioinformatic analysis was performed to screen differentially expressed genes associated with AH. AH mouse models were established through a Lieber-DeCarli liquid diet containing 4% ethanol, which were co-treated with siRNA against EPCAM or the PI3K/Akt/mTOR signaling pathway inhibitor in order to investigate the effects of EPCAM and the PI3K/Akt/mTOR signaling pathway on hepatic fibrosis, hepatic stellate cell (HSC) proliferation and apoptosis. The relationship between EPCAM and the PI3K/Akt/mTOR signaling pathway was investigated for the purposes of elucidating the potential mechanism of EPCAM in AH. EPCAM was predicted to regulate AH progression through the PI3K/Akt/mTOR signaling pathway. Silencing EPCAM or inhibition of the PI3K/Akt/mTOR signaling pathway inhibited the hepatic fibrosis and HSC proliferation yet induced HSC apoptosis. Moreover, silencing EPCAM was found to repress the PI3K/Akt/mTOR signaling pathway as evidenced by decreased levels of Bcl2 yet increased levels of caspase-3. Collectively, silencing EPCAM could hinder AH progression by inhibiting the PI3K/Akt/mTOR signaling pathway, which might serve as a potential therapeutic target for AH treatment.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, P. R. China,CONTACT Zhi Zhang
| | - Huiqing Wen
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Jun Weng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Lei Feng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Hongya Liu
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Xiaojun Hu
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Fanhong Zeng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
8
|
Li Y, Pu S, Liu Q, Li R, Zhang J, Wu T, Chen L, Li H, Yang X, Zou M, Xiao J, Xie W, He J. An integrin-based nanoparticle that targets activated hepatic stellate cells and alleviates liver fibrosis. J Control Release 2019; 303:77-90. [PMID: 31004666 DOI: 10.1016/j.jconrel.2019.04.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/05/2019] [Accepted: 04/16/2019] [Indexed: 02/06/2023]
Abstract
Activation of hepatic stellate cells (HSCs) contributes to the development of liver fibrosis. Because of a relatively small population of HSCs in the liver and the lack of specific membrane targeting proteins, HSC-targeted therapy remains a major clinical challenge. Here we first showed that a hallmark of activated HSC (aHSC) is their increased expression of integrin αvβ3. Thus we established sterically stable liposomes that contain the cyclic peptides (cRGDyK) with a high affinity to αvβ3 to achieve aHSC-specific delivery. Our results showed that the cRGDyK-guided liposomes were preferentially internalized by activated HSCs in vitro and in vivo, and the internalization was abolished by excess free cRGDyK or knockdown of αvβ3. In contrast, quiescent HSCs, hepatocytes, Kupffer cells, sinusoidal endothelial cells, or biliary cells showed minimal uptake of the cRGDyK-guided liposomes. When loaded with the hedgehog inhibitor vismodegib, the cRGDyK-guided liposomes inhibited hedgehog pathway signaling specifically in activated HSCs. Moreover, treatment of mice with vismodegib-loaded cRGDyK-liposomes markedly inhibited the fibrogenic phenotype in bile duct ligation- or thioacetamide-treated mice. We conclude that the cRGDyK-guided liposomes can specifically target the activated HSCs, but not quiescent HSCs. This nanoparticle system showed great promise to deliver therapeutic agents to aHSC to treat liver fibrosis.
Collapse
Affiliation(s)
- Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China; Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Shiyun Pu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China; Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Rui Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China; Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Jinhang Zhang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China; Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Tong Wu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China; Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Lei Chen
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China; Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Hong Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China; Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Xuping Yang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China; Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Min Zou
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Jia Xiao
- Institute of Clinical Science, Guangzhou Overseas Chinese Hospital, Jinan University, Guangzhou, China
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Jinhan He
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China; Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Morsy MA, Nair AB. Prevention of rat liver fibrosis by selective targeting of hepatic stellate cells using hesperidin carriers. Int J Pharm 2018; 552:241-250. [DOI: 10.1016/j.ijpharm.2018.10.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/29/2018] [Accepted: 10/02/2018] [Indexed: 12/15/2022]
|
10
|
Anavi S, Madar Z, Tirosh O. Non-alcoholic fatty liver disease, to struggle with the strangle: Oxygen availability in fatty livers. Redox Biol 2017; 13:386-392. [PMID: 28667907 PMCID: PMC5493836 DOI: 10.1016/j.redox.2017.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 06/18/2017] [Accepted: 06/20/2017] [Indexed: 12/29/2022] Open
Abstract
Nonalcoholic fatty liver diseases (NAFLD) is one of the most common chronic liver disease in Western countries. Oxygen is a central component of the cellular microenvironment, which participate in the regulation of cell survival, differentiation, functions and energy metabolism. Accordingly, sufficient oxygen supply is an important factor for tissue durability, mainly in highly metabolic tissues, such as the liver. Accumulating evidence from the past few decades provides strong support for the existence of interruptions in oxygen availability in fatty livers. This outcome may be the consequence of both, impaired systemic microcirculation and cellular membrane modifications which occur under steatotic conditions. This review summarizes current knowledge regarding the main factors which can affect oxygen supply in fatty liver.
Collapse
Affiliation(s)
- Sarit Anavi
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Israel; Peres Academic Center, Rehovot, Israel
| | - Zecharia Madar
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Israel
| | - Oren Tirosh
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|
11
|
Schon HT, Bartneck M, Borkham-Kamphorst E, Nattermann J, Lammers T, Tacke F, Weiskirchen R. Pharmacological Intervention in Hepatic Stellate Cell Activation and Hepatic Fibrosis. Front Pharmacol 2016; 7:33. [PMID: 26941644 PMCID: PMC4764688 DOI: 10.3389/fphar.2016.00033] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/08/2016] [Indexed: 12/17/2022] Open
Abstract
The activation and transdifferentiation of hepatic stellate cells (HSCs) into contractile, matrix-producing myofibroblasts (MFBs) are central events in hepatic fibrogenesis. These processes are driven by autocrine- and paracrine-acting soluble factors (i.e., cytokines and chemokines). Proof-of-concept studies of the last decades have shown that both the deactivation and removal of hepatic MFBs as well as antagonizing profibrogenic factors are in principle suitable to attenuate ongoing hepatic fibrosis. Although several drugs show potent antifibrotic activities in experimental models of hepatic fibrosis, there is presently no effective pharmaceutical intervention specifically approved for the treatment of liver fibrosis. Pharmaceutical interventions are generally hampered by insufficient supply of drugs to the diseased liver tissue and/or by adverse effects as a result of affecting non-target cells. Therefore, targeted delivery systems that bind specifically to receptors solely expressed on activated HSCs or transdifferentiated MFBs and delivery systems that can improve drug distribution to the liver in general are urgently needed. In this review, we summarize current strategies for targeted delivery of drugs to the liver and in particular to pro-fibrogenic liver cells. The applicability and efficacy of sequestering molecules, selective protein carriers, lipid-based drug vehicles, viral vectors, transcriptional targeting approaches, therapeutic liver- and HSC-specific nanoparticles, and miRNA-based strategies are discussed. Some of these delivery systems that had already been successfully tested in experimental animal models of ongoing hepatic fibrogenesis are expected to translate into clinically useful therapeutics specifically targeting HSCs.
Collapse
Affiliation(s)
- Hans-Theo Schon
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen Aachen, Germany
| | - Matthias Bartneck
- Department of Medicine III, University Hospital RWTH Aachen Aachen, Germany
| | - Erawan Borkham-Kamphorst
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen Aachen, Germany
| | - Jacob Nattermann
- Department of Internal Medicine I, University of Bonn Bonn, Germany
| | - Twan Lammers
- Department for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University Hospital RWTH Aachen Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen Aachen, Germany
| |
Collapse
|
12
|
Zhang YN, Poon W, Tavares AJ, McGilvray ID, Chan WCW. Nanoparticle-liver interactions: Cellular uptake and hepatobiliary elimination. J Control Release 2016; 240:332-348. [PMID: 26774224 DOI: 10.1016/j.jconrel.2016.01.020] [Citation(s) in RCA: 806] [Impact Index Per Article: 100.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/04/2016] [Accepted: 01/11/2016] [Indexed: 12/31/2022]
Abstract
30-99% of administered nanoparticles will accumulate and sequester in the liver after administration into the body. This results in reduced delivery to the targeted diseased tissue and potentially leads to increased toxicity at the hepatic cellular level. This review article focuses on the inter- and intra-cellular interaction between nanoparticles and hepatic cells, the elimination mechanism of nanoparticles through the hepatobiliary system, and current strategies to manipulate liver sequestration. The ability to solve the "nanoparticle-liver" interaction is critical to the clinical translation of nanotechnology for diagnosing and treating cancer, diabetes, cardiovascular disorders, and other diseases.
Collapse
Affiliation(s)
- Yi-Nan Zhang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada
| | - Wilson Poon
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada
| | - Anthony J Tavares
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada
| | - Ian D McGilvray
- Multi Organ Transport Program, Toronto General Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada; Toronto General Research Institute, University Health Network, 585 University Avenue, Toronto, ON M5G 2N2, Canada
| | - Warren C W Chan
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Department of Chemistry, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Department of Chemical Engineering & Applied Chemistry, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Department of Materials Science and Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada.
| |
Collapse
|
13
|
Abe H, Kamimura K, Kobayashi Y, Ohtsuka M, Miura H, Ohashi R, Yokoo T, Kanefuji T, Suda T, Tsuchida M, Aoyagi Y, Zhang G, Liu D, Terai S. Effective Prevention of Liver Fibrosis by Liver-targeted Hydrodynamic Gene Delivery of Matrix Metalloproteinase-13 in a Rat Liver Fibrosis Model. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e276. [PMID: 26730813 PMCID: PMC5012547 DOI: 10.1038/mtna.2015.49] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/16/2015] [Indexed: 02/07/2023]
Abstract
Liver fibrosis is the final stage of liver diseases that lead to liver failure and cancer. While various diagnostic methods, including the use of serum marker, have been established, no standard therapy has been developed. The objective of this study was to assess the approach of overexpressing matrix metalloproteinase-13 gene (MMP13) in rat liver to prevent liver fibrosis progression. A rat liver fibrosis model was established by ligating the bile duct, followed by liver-targeted hydrodynamic gene delivery of a MMP13 expression vector, containing a CAG promoter-MMP13-IRES-tdTomato-polyA cassette. After 14 days, the serum level of MMP13 peaked at 71.7 pg/ml in MMP13-treated group, whereas the nontreated group only showed a level of ~5 pg/ml (P < 0.001). These levels were sustained for the next 60 days. The statistically lower level of the hyaluronic acids in treated group versus the nontreated group (P < 0.05) reveals the therapeutic effect of MMP13 overexpression. Quantitative analysis of tissue stained with sirius red showed a statistically larger volume of fibrotic tissue in the nontreated group compared to that of MMP13-treated rats (P < 0.05). These results suggest that the liver-targeted hydrodynamic delivery of MMP13 gene could be effective in the prevention of liver fibrosis.
Collapse
Affiliation(s)
- Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1–757 Asahimachi–dori, Chuo–ku, Niigata, Niigata 9518510, Japan. E-mail:
| | - Yuji Kobayashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Masato Ohtsuka
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, School of Medicine, Tokai University, Isehara, Kanagawa, Japan
| | - Hiromi Miura
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, School of Medicine, Tokai University, Isehara, Kanagawa, Japan
| | - Riuko Ohashi
- Department of Pathology, Niigata University Medical and Dental Hospital, Niigata, Niigata, Japan
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Tsutomu Kanefuji
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Takeshi Suda
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Masanori Tsuchida
- Division of Thoracic and Cardiovascular Surgery, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Yutaka Aoyagi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Guisheng Zhang
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, USA
| | - Dexi Liu
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, USA
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| |
Collapse
|
14
|
Abstract
Nanoscale systems are currently under investigation for multiple different diagnostic and therapeutic applications. These systems can be used to identify pathologically changed tissues or to selectively deliver drugs to these sites; both applications have an extremely high potential to ameliorate therapeutic outcomes for patients. Tissues as well as single cells can be targeted because of the small size of these systems, which enables enhanced diagnosis and increased specificity of therapy. Drug loads can be delivered directly to the site of action, which can result in a reduction in incidence and severity of adverse systemic effects. Several nano-based platform technologies are currently under investigation for use in therapeutic approaches, mainly for anti-inflammatory and anti-cancer therapies. Although many nanoscale systems show promising therapeutic outcomes in preclinical studies, only a limited number are ready for clinical use. This Review will discuss the diverse nanomaterials currently available and the first specific uses for select gastroenterological and hepatological pathologies. The discussion of diagnostic and therapeutic applications will consider realities of market introduction of these sometimes very complex systems in light of remaining regulatory challenges and hurdles for industrial production.
Collapse
|
15
|
Wang H, Thorling CA, Liang X, Bridle KR, Grice JE, Zhu Y, Crawford DHG, Xu ZP, Liu X, Roberts MS. Diagnostic imaging and therapeutic application of nanoparticles targeting the liver. J Mater Chem B 2015; 3:939-958. [PMID: 32261972 DOI: 10.1039/c4tb01611d] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Liver diseases, particularly viral hepatitis, cirrhosis and hepatocellular carcinoma, are common in clinical practice with high morbidity and mortality worldwide. Many substances for diagnostic imaging and therapy of liver diseases may have either severe adverse effects or insufficient effectiveness in vivo because of their nonspecific uptake. Therefore, by targeting the delivery of drugs into the liver or specific liver cells, drug efficiency may be largely improved. This review summarizes the up-to-date research progress focusing on nanoparticles targeting the liver for both diagnostic and therapeutic purposes. Targeting strategies, mechanisms of enhanced effects, and clinical applications of nanoparticles are discussed specifically. We believe that new targeting nanotechnology such as nanoprobes for multi-modality imaging and multifunctional nanoparticles would facilitate significant advancements in this active research area in the near future.
Collapse
Affiliation(s)
- Haolu Wang
- Therapeutics Research Centre, School of Medicine, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, QLD 4102, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kumar P, Smith T, Rahman K, Thorn NE, Anania FA. Adiponectin agonist ADP355 attenuates CCl4-induced liver fibrosis in mice. PLoS One 2014; 9:e110405. [PMID: 25310107 PMCID: PMC4195748 DOI: 10.1371/journal.pone.0110405] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/15/2014] [Indexed: 12/29/2022] Open
Abstract
Liver fibrosis is a growing global health problem characterized by excess deposition of fibrillar collagen, and activation of hepatic stellate cells (HSCs). Adiponectin is known to possess anti-fibrotic properties; however a high physiological concentration and multiple forms circulating in blood prohibit clinical use. Recently, an adiponectin-like small synthetic peptide agonist (ADP355: H-DAsn-Ile-Pro-Nva-Leu-Tyr-DSer-Phe-Ala-DSer-NH2) was synthesized for the treatment of murine breast cancer. The present study was designed to evaluate the efficacy of ADP355 as an anti-fibrotic agent in the in vivo carbon tetrachloride (CCl4)-induced liver fibrosis model. Liver fibrosis was induced in eight-week old male C57BL/6J mice by CCl4-gavage every other day for four weeks before injection of a nanoparticle-conjugated with ADP355 (nano-ADP355). Control gold nanoparticles and nano-ADP355 were administered by intraperitoneal injection for two weeks along with CCl4-gavage. All mice were sacrificed after 6 weeks, and serum and liver tissue were collected for biochemical, histopathologic and molecular analyses. Biochemical studies suggested ADP355 treatment attenuates liver fibrosis, determined by reduction of serum aspartate aminotransferase (AST), alanine aminotransferase ALT) and hydroxyproline. Histopathology revealed chronic CCl4-treatment results in significant fibrosis, while ADP355 treatment induced significantly reversed fibrosis. Key markers for fibrogenesis-α-smooth muscle actin (α-SMA), transforming growth factor-beta1 (TGF-β1), connective tissue growth factor (CTGF), and the tissue inhibitor of metalloproteinase I (TIMP1) were also markedly attenuated. Conversely, liver lysates from ADP355 treated mice increased phosphorylation of both endothelial nitric oxide synthase (eNOS) and AMPK while AKT phosphorylation was diminished. These findings suggest ADP355 is a potent anti-fibrotic agent that can be an effective intervention against liver fibrosis.
Collapse
Affiliation(s)
- Pradeep Kumar
- Emory University School of Medicine, Department of Medicine, Division of Digestive Diseases, Atlanta, GA, United States of America
| | - Tekla Smith
- Emory University School of Medicine, Department of Medicine, Division of Digestive Diseases, Atlanta, GA, United States of America
| | - Khalidur Rahman
- Emory University School of Medicine, Department of Medicine, Division of Digestive Diseases, Atlanta, GA, United States of America
| | - Natalie E. Thorn
- Emory University School of Medicine, Department of Medicine, Division of Digestive Diseases, Atlanta, GA, United States of America
| | - Frank A. Anania
- Emory University School of Medicine, Department of Medicine, Division of Digestive Diseases, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
17
|
Glycosylation-mediated targeting of carriers. J Control Release 2014; 190:542-55. [DOI: 10.1016/j.jconrel.2014.06.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 05/29/2014] [Accepted: 06/02/2014] [Indexed: 12/24/2022]
|
18
|
Abstract
Nonviral vectors which offer a safer and versatile alternative to viral vectors have been developed to overcome problems caused by viral carriers. However, their transfection efficacy or level of expression is substantially lower than viral vectors. Among various nonviral gene vectors, lipid nanoparticles are an ideal platform for the incorporation of safety and efficacy into a single delivery system. In this chapter, we highlight current lipidic vectors that have been developed for gene therapy of tumors and other diseases. The pharmacokinetic, toxic behaviors and clinic trials of some successful lipids particles are also presented.
Collapse
|
19
|
Narmada BC, Kang Y, Venkatraman L, Peng Q, Sakban RB, Nugraha B, Jiang X, Bunte RM, So PTC, Tucker-Kellogg L, Mao HQ, Yu H. Hepatic stellate cell-targeted delivery of hepatocyte growth factor transgene via bile duct infusion enhances its expression at fibrotic foci to regress dimethylnitrosamine-induced liver fibrosis. Hum Gene Ther 2013; 24:508-19. [PMID: 23527815 DOI: 10.1089/hum.2012.158] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Liver fibrosis generates fibrotic foci with abundant activated hepatic stellate cells and excessive collagen deposition juxtaposed with healthy regions. Targeted delivery of antifibrotic therapeutics to hepatic stellate cells (HSCs) might improve treatment outcomes and reduce adverse effects on healthy tissue. We delivered the hepatocyte growth factor (HGF) gene specifically to activated hepatic stellate cells in fibrotic liver using vitamin A-coupled liposomes by retrograde intrabiliary infusion to bypass capillarized hepatic sinusoids. The antifibrotic effects of DsRed2-HGF vector encapsulated within vitamin A-coupled liposomes were validated by decreases in fibrotic markers in vitro. Fibrotic cultures transfected with the targeted transgene showed a significant decrease in fibrotic markers such as transforming growth factor-β1. In rats, dimethylnitrosamine-induced liver fibrosis is manifested by an increase in collagen deposition and severe defenestration of sinusoidal endothelial cells. The HSC-targeted transgene, administered via retrograde intrabiliary infusion in fibrotic rats, successfully reduced liver fibrosis markers alpha-smooth muscle actin and collagen, accompanied by an increase in the expression of DsRed2-HGF near the fibrotic foci. Thus, targeted delivery of HGF gene to hepatic stellate cells increased the transgene expression at the fibrotic foci and strongly enhanced its antifibrotic effects.
Collapse
|
20
|
Chen HJ, Liang TM, Lee IJ, Huang YT, Lin YL. Scutellariae radix suppresses LPS-induced liver endothelial cell activation and inhibits hepatic stellate cell migration. JOURNAL OF ETHNOPHARMACOLOGY 2013; 150:835-842. [PMID: 24036165 DOI: 10.1016/j.jep.2013.08.049] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 08/08/2013] [Accepted: 08/26/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liver fibrosis is the result of long-term liver damage and the wound-healing process, in which the hepatic stellate cell (HSC) plays a crucial role during fibrogenesis. The liver sinusoidal endothelial cell (LSEC) is a liver-resident scavenger, contributing to sinusoidal remodeling, HSC activation and liver fibrosis. Lipopolysaccharide (LPS) causes an inflammatory reaction associated with portal circulation and LSECs signaling. Scutellariae radix, the root of Scutellaria baicalensis Georgi, is a Chinese herb widely used for liver diseases. However, its effect on LSEC activation and HSC migration in liver fibrosis has not been investigated yet. AIM OF THIS STUDY LPS-induced rat LSEC (rLSEC) activation was used as a model to screen and explore the active components of Scutellariae radix. The anti-fibrotic effect of Scutellariae radix on rLSEC activation and rHSC migration was further investigated. MATERIALS AND METHODS LPS-induced rLSEC mRNA expression, including VEGF, VEGFR, MCP-1, and TGF-β1, were examined by real-time PCR analyses. MCP-1 protein levels were measured by an ELISA kit. rLSEC conditioned medium on rHSC migration was measured by wound-healing assay and transwell chemoattraction assay. RESULTS Results showed LPS-induced rLSEC activation with upregulated MCP-1 mRNA and protein expressions, and that rLSEC-condition medium enhanced rHSC migration. Both baicalein and wogonin from the active subfraction significantly reduced MCP-1 expression, but only baicalein markedly inhibited rHSC migration in rLSEC conditioned medium. CONCLUSION This study demonstrated that Scutellariae radix attenuates LPS-induced rLSEC activation and HSC migration with downregulation of MCP-1 expression. The results provide supporting evidence that Scutellariae radix may be beneficial for the amelioration of liver fibrosis.
Collapse
Affiliation(s)
- Hong-Jhang Chen
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
21
|
Yang J, Hou Y, Ji G, Song Z, Liu Y, Dai G, Zhang Y, Chen J. Targeted delivery of the RGD-labeled biodegradable polymersomes loaded with the hydrophilic drug oxymatrine on cultured hepatic stellate cells and liver fibrosis in rats. Eur J Pharm Sci 2013; 52:180-90. [PMID: 24296297 DOI: 10.1016/j.ejps.2013.11.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 11/21/2013] [Accepted: 11/24/2013] [Indexed: 01/06/2023]
Abstract
Oxymatrine (OM) is an alkaloid extracted from a Chinese herb that has been found to possess an anti-hepatic fibrosis effect, although its anti-fibrotic potential is limited due to a lack of targeting specificity, a short half-life and adverse effects. Polymersomes (PM) assembled from amphiphilic block copolymers represent promising vesicles for applications that include drug delivery and surface functionalization. The aim of this study was to develop a novel drug carrier based on PM modified with the peptide RGD and evaluate its therapeutic effect on liver fibrosis. A series of PM based on poly(ethylene glycol)-b-poly(ε-caprolactone) (PEG-b-PCL) were prepared and characterized. OM was loaded into PM by a pH-gradient method then the OM-loaded PM was modified with RGD peptide to obtain RGD-PM-OM. The average drug loading of RGD-PM-OM, with a size of 95 nm, was 6.8%. The targeting effects of the system were determined in cultured hepatic stellate cells (HSCs) and bile duct-ligated rats (BLD). RGD-PM-OM displayed better suppression of HSCs proliferation and significantly reduced the expression of the genes for α-SMA and collagen lα1 in cultured HSCs. Furthermore, RGD-PM-OM exhibited markedly superior anti-fibrosis activity by reducing the levels of PC-III and IV-C in serum and connective tissue deposition in BLD compared with PM-OM and OM. These results indicate that targeted RGD-PM-OM markedly attenuates the effects of hepatic fibrosis.
Collapse
Affiliation(s)
- Jianhong Yang
- Department of Pharmaceutical Science, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Ave., Guangzhou 510515, PR China; Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Yanhui Hou
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Gangjian Ji
- Department of Pharmaceutical Science, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Ave., Guangzhou 510515, PR China
| | - Zhihua Song
- Department of Pharmaceutical Science, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Ave., Guangzhou 510515, PR China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Guidong Dai
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Yajun Zhang
- College of Life Sciences of Northwest University, No. 229 Taibai North Road, Xian 710069, PR China
| | - Jianhai Chen
- Department of Pharmaceutical Science, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Ave., Guangzhou 510515, PR China.
| |
Collapse
|
22
|
Poelstra K, Prakash J, Beljaars L. Drug targeting to the diseased liver. J Control Release 2012; 161:188-97. [DOI: 10.1016/j.jconrel.2012.02.011] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 02/08/2012] [Accepted: 02/11/2012] [Indexed: 02/07/2023]
|
23
|
Li F, Li QH, Wang JY, Zhan CY, Xie C, Lu WY. Effects of interferon-gamma liposomes targeted to platelet-derived growth factor receptor-beta on hepatic fibrosis in rats. J Control Release 2011; 159:261-70. [PMID: 22226772 DOI: 10.1016/j.jconrel.2011.12.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 10/11/2011] [Accepted: 12/19/2011] [Indexed: 12/13/2022]
Abstract
No drugs have been approved clinically for the therapy of hepatic fibrosis. Though interferon-γ (IFN-γ) is a highly effective anti-fibrotic agent in vitro and in some animal models in vivo, its anti-fibrotic potential in clinical trials has been disappointing, due to unwanted off-target effects and a short half-life period which results in poor efficacy. The aims of this study are to develop a new targeted drug delivery system to selectively deliver IFN-γ to hepatic stellate cells (HSCs) and to investigate whether it will improve the anti-fibrotic effect of IFN-γ and reduce its side effects in fibrotic livers. Sterically stable liposomes (SSLs) were modified by cyclic peptides (pPB) with a specific affinity for platelet-derived growth factor receptor-β (PDGFR-β), and then IFN-γ was encapsulated in the targeted liposomes (pPB-SSL-IFN-γ). In vitro, pPB-SSL was found to be taken up and internalized by cultured activated HSCs. The binding of FITC-labeled pPB-SSL to activated HSCs was in a time-dependent and concentration-dependent manner, which could be inhibited by excess unlabelled pPB-SSL, PDGF-BB, suramin or monensin. The inhibitory effect of pPB-SSL-IFN-γ on the proliferation of activated HSCs was respectively 7.24-fold and 2.95-fold higher than that of free IFN-γ and IFN-γ encapsulated in untargeted SSLs. In healthy rats, the tissue distribution, living-body tracing image analyses and pharmacokinetics study showed that pPB-SSL-IFN-γ accumulated mainly in the livers and had a longer half-life than free IFN-γ (3.98±0.52h vs. 0.21±0.03h). Furthermore, in rats with hepatic fibrosis induced by thioacetamide injection, FITC-labeled pPB-SSL was found to predominantly localize in activated HSCs by immunofluorescent double staining for FITC and albumin or α-smooth muscle actin (α-SMA). The enhanced anti-fibrotic effect of pPB-SSL-IFN-γ treatnment was indicated by significant decreases in the histologic Ishak stage, collagen I-staining positive areas, and α-SMA expression levels in fibrotic livers. In addition, pPB-SSL-IFN-γ treatment improved the leukopenia caused by low- and high-dosage free IFN-γ treatments. In conclusion, IFN-γ encapsulated in pPB-SSL had an extended circulation half-life and was selectively delivered to activated HSCs, which enhanced the anti-fibrotic effect of IFN-γ and reduced its side-effects in rats with hepatic fibrosis. Thus, pPB-SSL-IFN-γ may be an effective agent for the therapy of hepatic fibrosis.
Collapse
Affiliation(s)
- Feng Li
- Department of Gastroenterology, Zhongshan Hospital affiliated to Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
24
|
Zhang H, Ma Y, Sun XL. Recent developments in carbohydrate-decorated targeted drug/gene delivery. Med Res Rev 2010; 30:270-89. [PMID: 19626595 DOI: 10.1002/med.20171] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Targeted delivery of a drug or gene to its site of action has clear therapeutic advantages by maximizing its therapeutic efficiency and minimizing its systemic toxicity. Generally, targeted drug or gene delivery is performed by loading a macromolecular carrier with an appropriate drug or gene, and by targeting the drug/gene carrier to specific cell or tissue with the help of specific targeting ligand. The emergence of glycobiology, glycotechnology, and glycomics and their continual adaptation by pharmaceutical scientists have opened exciting avenue of medicinal applications of carbohydrates. Among them, the biocompatibility and specific receptor recognition ability confer the ability of carbohydrates as potential targeting ligands for targeted drug and gene delivery applications. This review summarizes recent progress of carbohydrate-decorated targeted drug/gene delivery applications.
Collapse
Affiliation(s)
- Hailong Zhang
- Department of Chemistry, Cleveland State University, Cleveland, Ohio 44115, USA
| | | | | |
Collapse
|
25
|
Poelstra K, Greupink R, Beljaars L. Reprint of: Targeting fibrosis with selective drug carriers. Arab J Gastroenterol 2010. [DOI: 10.1016/j.ajg.2010.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
26
|
Targeting fibrosis with selective drug carriers. Arab J Gastroenterol 2010. [DOI: 10.1016/j.ajg.2009.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
27
|
Dong MX, Jia Y, Zhang YB, Li CC, Geng YT, Zhou L, Li XY, Liu JC, Niu YC. Emodin protects rat liver from CCl 4-induced fibrogenesis via inhibition of hepatic stellate cells activation. World J Gastroenterol 2009; 15:4753-62. [PMID: 19824107 PMCID: PMC2761551 DOI: 10.3748/wjg.15.4753] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of emodin in protecting the liver against fibrogenesis caused by carbon tetrachloride (CCl4) in rats and to further explore the underlying mechanisms.
METHODS: Rat models of experimental hepatic fibrosis were established by injection with CCl4; the treated rats received emodin via oral administration at a dosage of 20 mg/kg twice a week at the same time. Rats injected with olive oil served as a normal group. Histopathological changes were observed by hematoxylin and eosin staining. The activities of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in serum and hepatic hydroxyproline content were assayed by biochemical analyses. The mRNA and protein relevant to hepatic stellate cell (HSC) activation in the liver were assessed using real-time reverse transcription-polymerase chain reaction (RT-PCR), immunohistochemistry, western blotting and enzyme-linked immunosorbent assay.
RESULTS: The degree of hepatic fibrosis increased markedly in the CCl4 group compared to the normal group (P < 0.01), and decreased markedly in the emodin group compared to the CCl4 group according to METAVIR scale (P < 0.01) compared with those in the normal control group (51.02 ± 10.64 IU/L and 132.28 ± 18.14 IU/L). The activities of serum ALT and AST were significantly higher in rats injected with CCl4 (289.25 ± 68.84 IU/L and 423.89 ± 35.67 IU/L, both P < 0.05). The activities of serum ALT and AST were significantly reduced by administration of emodin (176.34 ± 47.29 IU/L and 226.1 ± 44.52 IU/L, both P < 0.05). Compared with the normal controls (54.53 ± 13.46 mg/g), hepatic hydroxyproline content was significantly higher in rats injected with CCl4 (120.27 ± 28.47 mg/g, P < 0.05). Hepatic hydroxyproline content was significantly reduced in the rats treated with emodin at 20 mg/kg (71.25 ± 17.02 mg/g, P < 0.05). Emodin significantly protected the liver from injury by reducing serum AST and ALT activities and reducing hepatic hydroxyproline content. The mRNA levels of transforming growth factor-β1 (TGF-β1), Smad4 and α-SMA in liver tissues were significantly down-regulated in SD rats that received emodin treatment. Furthermore, significant down-regulation of serum TGF-β1 protein levels and protein expression of Smad4 and α-SMA in liver tissues was also observed in the rats. Emodin inhibited HSC activation by reducing the abundance of TGF-β1 and Smad4.
CONCLUSION: Emodin protects the rat liver from CCl4-induced fibrogenesis by inhibiting HSC activation. Emodin might be a therapeutic antifibrotic agent for the treatment of hepatic fibrosis.
Collapse
|
28
|
Abstract
Liver fibrosis and its end stage disease cirrhosis are a major cause of mortality and morbidity around the world. There is no effective pharmaceutical intervention for liver fibrosis at present. Many drugs that show potent antifibrotic activities in vitro often show only minor effects in vivo because of insufficient concentrations of drugs accumulating around the target cell and their adverse effects as a result of affecting other non-target cells. Hepatic stellate cells (HSC) play a critical role in the fibrogenesis of liver, so they are the target cells of antifibrotic therapy. Several kinds of targeted delivery system that could target the receptors expressed on HSC have been designed, and have shown an attractive targeted potential in vivo. After being carried by these delivery systems, many agents showed a powerful antifibrotic effect in animal models of liver fibrosis. These targeted delivery systems provide a new pathway for the therapy of liver fibrosis. The characteristics of theses targeted carriers are reviewed in this paper.
Collapse
Affiliation(s)
- Feng Li
- Zhongshan Hospital Affiliated to Fudan University, Department of Gastroenterology, 180 Fenglin Road, Shanghai, China.
| | | |
Collapse
|
29
|
Meng Y, Gong YC, Dou Y, Li W. Changes of serum cytokines and expression of inducible nitric oxide synthase mRNA by Kupffer cells after relief from obstructive jaundice in rats. J Gastroenterol Hepatol 2009; 24:1064-9. [PMID: 19220682 DOI: 10.1111/j.1440-1746.2008.05746.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
AIMS To investigate the changes of serum endotoxin, tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6) and inducible nitric oxide synthase (iNOS) expression by Kupffer cells after relief of obstructive jaundice (OJ) by internal biliary drainage (ID) and external biliary drainage (ED) in rats. MATERIALS AND METHODS Forty eight adult Sprague Dawley rats were randomly assigned to four groups: OJ, ID, ED and sham operation (SH). Inducible nitric oxide synthase mRNA by the Kupffer cells was detected by reverse transcription polymerase chain reaction. The serum TNF-alpha and IL-6 were measured by enzyme linked immunosorbent assay and endotoxin by kinetic turbidimetric limulus tests. RESULTS Serum endotoxin, IL-6 and TNF-alpha levels were significantly elevated in OJ rats compared to that of SH rats (P < 0.01). After relief from OJ, the elevated endotoxin, IL-6 and TNF-alpha levels could be significantly depressed by ID (P < 0.01). However, the serum IL-6 level was increased in ED rats (P < 0.05) and the TNF-alpha level was not depressed by ED. Expression of iNOS mRNA by Kupffer cells was markedly stronger in the OJ group than in the SH group (P = 0.005). After relief from OJ, the iNOS mRNA expression was suppressed by ID (P = 0.139, ID vs OJ). However, the iNOS mRNA expression was promoted by ED (P = 0.321 ED vs OJ; P = 0.016 ED vs SH). CONCLUSIONS The levels of serum endotoxin, TNF-alpha and IL-6 and the expression of iNOS mRNA by Kupffer cells were increased in rats with obstructive jaundice. Internal biliary drainage could entirely reverse the changes, but external drainage only partially did.
Collapse
Affiliation(s)
- Ying Meng
- Department of Gastroenterology and Hepatology, the General Hospital of the Chinese People's Liberation Army, Beijing, China
| | | | | | | |
Collapse
|
30
|
Pradère JP, Gonzalez J, Klein J, Valet P, Grès S, Salant D, Bascands JL, Saulnier-Blache JS, Schanstra JP. Lysophosphatidic acid and renal fibrosis. Biochim Biophys Acta Mol Cell Biol Lipids 2008; 1781:582-7. [PMID: 18455518 DOI: 10.1016/j.bbalip.2008.04.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Revised: 04/01/2008] [Accepted: 04/01/2008] [Indexed: 02/07/2023]
Abstract
The development of fibrosis involves a multitude of events and molecules. Until now the majority of these molecules were found to be proteins or peptides. But recent data show significant involvement of the phospholipid lysophosphatidic acid (LPA) in the development of pulmonary, liver and renal fibrosis. The latest data on the role of LPA and the G-protein-coupled LPA1 receptor in the development of renal fibrosis will be discussed. LPA1-receptor activation was found to be associated with increased vascular leakage and increased fibroblast recruitment in pulmonary fibrosis. Furthermore, in renal fibrosis LPA1-receptor activation stimulates macrophage recruitment and connective tissue growth factor expression. The observations make this receptor an interesting alternative and new therapeutic target in fibrotic diseases.
Collapse
Affiliation(s)
- Jean-Philippe Pradère
- Inserm, U858/I2MR, Department of Metabolism and Obesity, Team #3, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Adrian JE, Poelstra K, Kamps JAAM. Addressing liver fibrosis with liposomes targeted to hepatic stellate cells. J Liposome Res 2008; 17:205-18. [PMID: 18027241 DOI: 10.1080/08982100701528047] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Liver fibrosis is a chronic disease that results from hepatitis B and C infections, alcohol abuse or metabolic and genetic disorders. Ultimately, progression of fibrosis leads to cirrhosis, a stage of the disease characterized by failure of the normal liver functions. Currently, the treatment of liver fibrosis is mainly based on the removal of the underlying cause of the disease and liver transplantation, which is the only treatment for patients with advanced fibrosis. Hepatic stellate cells (HSC) are considered to be key players in the development of liver fibrosis. Chronically activated HSC produces large amounts of extracellular matrix and enhance fibrosis by secreting a broad spectrum of cytokines that exert pro-fibrotic actions in other cells, and in an autocrine manner perpetuate their own activation. Therefore, therapeutic interventions that inhibit activation of HSC and its pro-fibrotic activities are currently under investigation worldwide. In the present study we applied targeted liposomes as drug carriers to HSC in the fibrotic liver and explored the potential of these liposomes in antifibrotic therapies. Moreover, we investigated effects of bioactive compounds delivered by these liposomes on the progression of liver fibrosis. To our knowledge, this is the first study demonstrating that lipid-based drug carriers can be selectively delivered to HSC in the fibrotic liver. By incorporating the bioactive lipid DLPC, these liposomes can modulate different processes such as inflammation and fibrogenesis in the fibrotic liver. This dual functionality of liposomes as a drug carrier system with intrinsic biological effects may be exploited in new approaches to treat liver fibrosis.
Collapse
Affiliation(s)
- Joanna E Adrian
- Department of Pathology and Laboratory Medicine, Medical Biology section, University Medical Center Groningen, University of Groningen, The Netherlands
| | | | | |
Collapse
|
32
|
Du SL, Pan H, Lu WY, Wang J, Wu J, Wang JY. Cyclic Arg-Gly-Asp peptide-labeled liposomes for targeting drug therapy of hepatic fibrosis in rats. J Pharmacol Exp Ther 2007; 322:560-8. [PMID: 17510318 DOI: 10.1124/jpet.107.122481] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Targeting hepatic stellate cells (HSCs) has been challenging due to the lack of specific receptors or motifs on the cells. The aim of the present study was to develop a HSC-specific system for improving drug delivery to HSCs. The affinity of a cyclic peptide containing Arg-Gly-Asp (cRGD) to collagen type VI receptor on HSCs was examined in both in vitro and in vivo experiments. Sterically stable liposomes (SSLs) were modified with this peptide to yield a new carrier, cRGD-SSL. The targeting efficiency of this carrier in delivering interferon (IFN)-alpha1b was investigated in a rat model of liver fibrosis induced by bile duct ligation (BDL). When incubating HSCs or hepatocytes with cyclic RGD peptide, the peptide was bound preferentially to activated HSCs. Biodistribution study showed that the accumulation of cRGD peptide-labeled liposomes in HSCs isolated from BDL rats was 10-fold more than unlabeled SSLs. BDL rats receiving injections of IFN-alpha1b entrapped in cRGD-SSL exhibited significantly reduced extent of liver fibrosis compared with BDL control rats or BDL rats treated with IFN-alpha1b entrapped in SSLs. Thus, cRGD-SSL is an efficient drug carrier, which selectively targets activated HSCs and improves drug therapy for liver fibrosis to a significant extent. This liposomal formulation represents a new means of targeting drug carrier for the treatment of liver fibrosis, and it may have potential clinical applications.
Collapse
Affiliation(s)
- Shi-Lin Du
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 180 Fenglin Rd., Shanghai 200032, China
| | | | | | | | | | | |
Collapse
|