1
|
Ohlendieck CM, Matellan C, Manresa MC. Regulation of pathologic fibroblast functions in digestive diseases: a role for hypoxia? Am J Physiol Gastrointest Liver Physiol 2025; 328:G229-G242. [PMID: 39873349 DOI: 10.1152/ajpgi.00277.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/14/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
The recent uncovering of fibroblast heterogeneity has given great insight into the versatility of the stroma. Among other cellular processes, fibroblasts are now thought to contribute to the coordination of immune responses in a range of chronic inflammatory diseases and cancer. Although the pathologic roles of myofibroblasts, inflammatory fibroblasts, and cancer-associated fibroblasts in disease are reasonably well understood, the mechanisms behind their activation remain to be uncovered. In the gastrointestinal (GI) tract, several interleukins and tumor necrosis factor superfamily members have been identified as possible mediators driving the acquisition of inflammatory and fibrotic properties in fibroblasts. In addition to cytokines, other microenvironmental factors such as nutrient and oxygen availability are likely contributors to this process. In this respect, the phenomenon of low cellular oxygen levels known as hypoxia is common in a plethora of GI diseases. Indeed, the cross talk between hypoxia and inflammation is well-documented, with an abundance of studies suggesting that oxygen-sensing enzymes may have regulatory effects on inflammatory signaling pathways such as NF-κB. However, the impact that this has in GI fibroblasts in the context of chronic diseases has not been fully uncovered. Here we discuss the role of fibroblasts in GI diseases, the mediators that have emerged as regulators of their functions and the potential impact of hypoxia in this process, highlighting areas that require further investigation.
Collapse
Affiliation(s)
- Cian M Ohlendieck
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Carlos Matellan
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Mario C Manresa
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
2
|
Skorup I, Valentino G, Aleandri S, Gelli R, Ganguin AA, Felli E, Selicean SE, Marxer RA, Teworte S, Lucić A, Gracia-Sancho J, Berzigotti A, Ridi F, Luciani P. Polyenylphosphatidylcholine as bioactive excipient in tablets for the treatment of liver fibrosis. Int J Pharm 2023; 646:123473. [PMID: 37788730 DOI: 10.1016/j.ijpharm.2023.123473] [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: 04/23/2023] [Revised: 08/24/2023] [Accepted: 09/30/2023] [Indexed: 10/05/2023]
Abstract
Liver fibrosis is a condition characterized by the accumulation of extracellular matrix (ECM) arising from the myofibroblastic transdifferentiation of hepatic stellate cells (HSCs) occurring as the natural response to liver damage. To date, no pharmacological treatments have been specifically approved for liver fibrosis. We recently reported a beneficial effect of polyenylphosphatidylcholines (PPCs)-rich formulations in reverting fibrogenic features of HSCs. However, unsaturated phospholipids' properties pose a constant challenge to the development of tablets as preferred patient-centric dosage form. Profiting from the advantageous physical properties of the PPCs-rich Soluthin® S 80 M, we developed a tablet formulation incorporating 70% w/w of this bioactive lipid. Tablets were characterized via X-ray powder diffraction, thermogravimetry, and Raman confocal imaging, and passed the major compendial requirements. To mimic physiological absorption after oral intake, phospholipids extracted from tablets were reconstituted as protein-free chylomicron (PFC)-like emulsions and tested on the fibrogenic human HSC line LX-2 and on primary cirrhotic rat hepatic stellate cells (PRHSC). Lipids extracted from tablets and reconstituted in buffer or as PFC-like emulsions exerted the same antifibrotic effect on both activated LX-2 and PRHSCs as observed with plain S 80 M liposomes, showing that the manufacturing process did not interfere with the bioactivity of PPCs.
Collapse
Affiliation(s)
- Ivo Skorup
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Gina Valentino
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Rita Gelli
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Aymar Abel Ganguin
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Eric Felli
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, Hepatology, University of Bern, Bern, Switzerland
| | - Sonia Emilia Selicean
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, Hepatology, University of Bern, Bern, Switzerland
| | - Rosanne Angela Marxer
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Sarah Teworte
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Ana Lucić
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Jordi Gracia-Sancho
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, Hepatology, University of Bern, Bern, Switzerland; Liver Vascular Biology Research Group, CIBEREHD, IDIBAPS Research Institute, Barcelona, Spain
| | - Annalisa Berzigotti
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, Hepatology, University of Bern, Bern, Switzerland
| | - Francesca Ridi
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
3
|
Mohammad Omar J, Hai Y, Jin S. Hypoxia-induced factor and its role in liver fibrosis. PeerJ 2022; 10:e14299. [PMID: 36523459 PMCID: PMC9745792 DOI: 10.7717/peerj.14299] [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: 05/16/2022] [Accepted: 10/04/2022] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis develops as a result of severe liver damage and is considered a major clinical concern throughout the world. Many factors are crucial for liver fibrosis progression. While advancements have been made to understand this disease, no effective pharmacological drug and treatment strategies have been established that can effectively prevent liver fibrosis or even could halt the fibrotic process. Most of those advances in curing liver fibrosis have been aimed towards mitigating the causes of fibrosis, including the development of potent antivirals to inhibit the hepatitis virus. It is not practicable for many individuals; however, a liver transplant becomes the only suitable alternative. A liver transplant is an expensive procedure. Thus, there is a significant need to identify potential targets of liver fibrosis and the development of such agents that can effectively treat or reverse liver fibrosis by targeting them. Researchers have identified hypoxia-inducible factors (HIFs) in the last 16 years as important transcription factors driving several facets of liver fibrosis, making them possible therapeutic targets. The latest knowledge on HIFs and their possible role in liver fibrosis, along with the cell-specific activities of such transcription factors that how they play role in liver fibrosis progression, is discussed in this review.
Collapse
Affiliation(s)
- Jan Mohammad Omar
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical, Harbin, Heilongjiang, China
| | - Yang Hai
- College of International Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shizhu Jin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical, Harbin, Heilongjiang, China
| |
Collapse
|
4
|
Abd El-Fattah EE, Zakaria AY. Targeting HSP47 and HSP70: promising therapeutic approaches in liver fibrosis management. J Transl Med 2022; 20:544. [DOI: 10.1186/s12967-022-03759-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/06/2022] [Indexed: 11/28/2022] Open
Abstract
AbstractLiver fibrosis is a liver disease in which there is an excessive buildup of extracellular matrix proteins, including collagen. By regulating cytokine production and the inflammatory response, heat shock proteins (HSPs) contribute significantly to a wider spectrum of fibrotic illnesses, such as lung, liver, and idiopathic pulmonary fibrosis by aiding in the folding and assembly of freshly synthesized proteins, HSPs serve as chaperones. HSP70 is one of the key HSPs in avoiding protein aggregation which induces its action by sending unfolded and/or misfolded proteins to the ubiquitin–proteasome degradation pathway and antagonizing influence on epithelial-mesenchymal transition. HSP47, on the other hand, is crucial for boosting collagen synthesis, and deposition, and fostering the emergence of fibrotic disorders. The current review aims to provide light on how HSP70 and HSP47 affect hepatic fibrogenesis. Additionally, our review looks into new therapeutic approaches that target HSP70 and HSP47 and could potentially be used as drug candidates to treat liver fibrosis, especially in cases of comorbidities.
Collapse
|
5
|
Perramón M, Carvajal S, Reichenbach V, Fernández‐Varo G, Boix L, Macias‐Muñoz L, Melgar‐Lesmes P, Bruix J, Melmed S, Lamas S, Jiménez W. The pituitary tumour-transforming gene 1/delta-like homologue 1 pathway plays a key role in liver fibrogenesis. Liver Int 2022; 42:651-662. [PMID: 35050550 PMCID: PMC9303549 DOI: 10.1111/liv.15165] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND AIMS PTTG1 is almost undetectable in adult livers but is highly expressed in hepatocarcinoma. While little is known about its involvement in liver fibrosis, PTTG1 expression is associated with DLK1. We assessed the role of the PTTG1/DLK1 pathway in fibrosis progression and the potential therapeutic effect of PTTG1 silencing in fibrosis. METHODS Pttg1 and Dlk1 were studied in liver and isolated cell populations of control and fibrotic rats and in human liver biopsies. The fibrotic molecular signature was analysed in Pttg1-/- and Pttg1+/+ fibrotic mice. Finally, Pttg1 silencing was evaluated in rats as a novel antifibrotic therapy. RESULTS Pttg1 and Dlk1 mRNA selectively increased in fibrotic rats paralleling fibrosis progression. Serum DLK1 concentrations correlated with hepatic collagen content and systemic and portal haemodynamics. Human cirrhotic livers showed greater PTTG1 and DLK1 transcript abundance than non-cirrhotic, and reduced collagen was observed in Pttg1 Pttg1-/- mice. The liver fibrotic molecular signature revealed lower expression of genes related to extracellular matrix remodelling including Mmp8 and 9 and Timp4 and greater eotaxin and Mmp13 than fibrotic Pttg1+/+ mice. Finally, interfering Pttg1 resulted in reduced liver fibrotic area, lower α-Sma and decreased portal pressure than fibrotic animals. Furthermore, Pttg1 silencing decreased the transcription of Dlk1, collagens I and III, Pdgfrβ, Tgfrβ, Timp1, Timp2 and Mmp2. CONCLUSIONS Pttg1/Dlk1 are selectively overexpressed in the cirrhotic liver and participate in ECM turnover regulation. Pttg1 disruption decreases Dlk1 transcription and attenuates collagen deposition. PTTG1/DLK1 signalling is a novel pathway for targeting the progression of liver fibrosis.
Collapse
Affiliation(s)
- Meritxell Perramón
- Biochemistry and Molecular Genetics ServiceHospital Clínic UniversitariBarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD)BarcelonaSpain
| | - Silvia Carvajal
- Biochemistry and Molecular Genetics ServiceHospital Clínic UniversitariBarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD)BarcelonaSpain
| | - Vedrana Reichenbach
- Biochemistry and Molecular Genetics ServiceHospital Clínic UniversitariBarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD)BarcelonaSpain
| | - Guillermo Fernández‐Varo
- Biochemistry and Molecular Genetics ServiceHospital Clínic UniversitariBarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD)BarcelonaSpain
| | - Loreto Boix
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD)BarcelonaSpain,Department of MedicineUniversity of BarcelonaBarcelonaSpain,Barcelona‐Clínic Liver Cancer GroupHospital Clínic UniversitariBarcelonaSpain
| | - Laura Macias‐Muñoz
- Biochemistry and Molecular Genetics ServiceHospital Clínic UniversitariBarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD)BarcelonaSpain
| | - Pedro Melgar‐Lesmes
- Biochemistry and Molecular Genetics ServiceHospital Clínic UniversitariBarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD)BarcelonaSpain,Department of BiomedicineUniversity of BarcelonaBarcelonaSpain,Institute for Medical Engineering and ScienceMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Jordi Bruix
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD)BarcelonaSpain,Department of MedicineUniversity of BarcelonaBarcelonaSpain,Barcelona‐Clínic Liver Cancer GroupHospital Clínic UniversitariBarcelonaSpain
| | - Shlomo Melmed
- Department of Medicine, Cedars‐Sinai Research InstituteUniversity of California School of MedicineLos AngelesCAUSA
| | - Santiago Lamas
- Centro de Biología Molecular Severo Ochoa (CSIC‐UAM)MadridSpain
| | - Wladimiro Jiménez
- Biochemistry and Molecular Genetics ServiceHospital Clínic UniversitariBarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEReHD)BarcelonaSpain,Department of BiomedicineUniversity of BarcelonaBarcelonaSpain
| |
Collapse
|
6
|
Sun J, Shi L, Xiao T, Xue J, Li J, Wang P, Wu L, Dai X, Ni X, Liu Q. microRNA-21, via the HIF-1α/VEGF signaling pathway, is involved in arsenite-induced hepatic fibrosis through aberrant cross-talk of hepatocytes and hepatic stellate cells. CHEMOSPHERE 2021; 266:129177. [PMID: 33310519 DOI: 10.1016/j.chemosphere.2020.129177] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/17/2020] [Accepted: 11/29/2020] [Indexed: 06/12/2023]
Abstract
Long-term exposure to arsenic, a widely distributed environmental toxicant, may result in damage to various organs, including the liver. Mice exposed chronically to arsenite developed hepatic damage, inflammation, and fibrosis, as well as increased levels of microRNA-21 (miR-21) and hypoxia-inducible factor (HIF)-1α. The levels of miR-21 and HIF-1α were also enhanced in primary hepatocytes and L-02 cells exposed to arsenite. The culture media from these cells induced the activation of hepatic stellate cells (HSCs), as demonstrated by up-regulation of the protein levels of α-smooth muscle actin (α-SMA) and collagen1A2 (COL1A2) and by increased activity in gel contractility assays. For L-02 cells, knockdown of miR-21 blocked the arsenite-induced up-regulation of HIF-1α and vascular endothelial growth factor (VEGF), which prevented the activation of LX-2 cells induced by medium from arsenite-exposed L-02 cells. However, these effects were reversed by down-regulation of von Hippel Lindau protein (pVHL). In arsenite-treated L-02 cells, miR-21 knockdown elevated the levels of ubiquitination and accelerated the degradation of HIF-1α via pVHL. In the livers of miR-21-/- mice exposed chronically to arsenite, there were less hepatic damage, lower fibrosis, lower levels of HIF-1α and VEGF, and higher levels of pVHL than for wild-type mice. In summary, we propose that miR-21, acting via the HIF-1α/VEGF signaling pathway, is involved in arsenite-induced hepatic fibrosis through mediating aberrant cross-talk of hepatocytes and HSCs. The findings provide evidence relating to the pathogenesis of hepatic fibrosis induced by exposure to arsenic.
Collapse
Affiliation(s)
- Jing Sun
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Le Shi
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Tian Xiao
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Junchao Xue
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Junjie Li
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Peiwen Wang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Lu Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Xiangyu Dai
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Xinye Ni
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, 213003, Jiangsu, People's Republic of China.
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
| |
Collapse
|
7
|
Abstract
Hepatic fibrosis is a complex mechanism defined by the net deposition of the extracellular matrix (ECM) owing to liver injury caused by multiple etiologies such as viral hepatitis and nonalcoholic fatty liver disease. Many cell types are implicated in liver fibrosis development and progression. In general, liver fibrosis starts with the recruitment of inflammatory immune cells to generate cytokines, growth factors, and other activator molecules. Such chemical mediators drive the hepatic stellate cells (HSCs) to activate the production of the ECM component. The activation of HSC is thus a crucial event in the fibrosis initiation, and a significant contributor to collagen deposition (specifically type I). This review explores the causes and mechanisms of hepatic fibrosis and focuses on the roles of key molecules involved in liver fibro genesis, some of which are potential targets for therapeutics to hamper liver fibro genesis.
Collapse
Affiliation(s)
- Reham M Dawood
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Mai A El-Meguid
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Ghada Maher Salum
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Mostafa K El Awady
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| |
Collapse
|
8
|
Pesce A, Ciurleo R, Bramanti A, Armeli Iapichino EC, Petralia MC, Magro GG, Fagone P, Bramanti P, Nicoletti F, Mangano K. Effects of Combined Admistration of Imatinib and Sorafenib in a Murine Model of Liver Fibrosis. Molecules 2020; 25:4310. [PMID: 32962198 PMCID: PMC7571085 DOI: 10.3390/molecules25184310] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is defined as excessive extracellular matrix deposition in the hepatic parenchyma as a consequence of complex interactions among matrix-producing hepatic stellate cells (HSCs) and liver-resident and infiltrating cells. In addition to the liver, the process of fibrosis may represent end-stage disease of several diseases including kidneys, lungs, spleens, heart, muscles and at certain extent, the central nervous system and the peripheral nerves. To date, antifibrotic treatment of fibrosis represents an unconquered area for drug development. The aim of the present study was to test the efficacy of a new drug combination for the treatment of hepatic fibrosis in order to provide a proof-of-concept for the use of therapeutic agents in clinical practice. For this purpose, we have studied the effects of the PDGF inhibitor imatinib and the angiogenesis inhibitor sorafenib, administered alone or in combination, in reducing the progression of the fibrogenetic process in a pre-clinical model of liver damage induced in mice by repeated administration of Concanavalin A (ConA), resembling long-tern autoimmune hepatitis. Our results suggest that treatments with imatinib and sorafenib can modulate potently and, in a superimposable fashion, the fibrinogenic process when administered alone. However, and in agreement with the computational data presently generated, they only exert partial overlapping antifibrotic effects in modulating the main pathways involved in the process of liver fibrosis, without significant additive or synergist effects, when administered in combination.
Collapse
Affiliation(s)
- Antonio Pesce
- Department of Medical and Surgical Sciences and Advanced Technology G.F. Ingrassia, University of Catania, Via Santa Sofia 86, 95123 Catania, Italy; (A.P.); (G.G.M.)
| | - Rosella Ciurleo
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (A.B.); (P.B.)
| | - Alessia Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (A.B.); (P.B.)
| | | | - Maria Cristina Petralia
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.P.); (P.F.); (K.M.)
| | - Gaetano Giuseppe Magro
- Department of Medical and Surgical Sciences and Advanced Technology G.F. Ingrassia, University of Catania, Via Santa Sofia 86, 95123 Catania, Italy; (A.P.); (G.G.M.)
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.P.); (P.F.); (K.M.)
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy; (R.C.); (A.B.); (P.B.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.P.); (P.F.); (K.M.)
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (M.C.P.); (P.F.); (K.M.)
| |
Collapse
|
9
|
Mai ZH, Huang Y, Huang D, Huang ZS, He ZX, Li PL, Zhang S, Weng JF, Gu WL. Reversine and herbal Xiang-Sha-Liu-Jun-Zi decoction ameliorate thioacetamide-induced hepatic injury by regulating the RelA/NF-κB/caspase signaling pathway. Open Life Sci 2020; 15:696-710. [PMID: 33817258 PMCID: PMC7747499 DOI: 10.1515/biol-2020-0059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/30/2020] [Accepted: 05/30/2020] [Indexed: 02/07/2023] Open
Abstract
This study investigated the anti-fibrotic effects of reversine and Chinese medicine Xiang–Sha–Liu–Jun–Zi decoction (XSLJZD) on thioacetamide (TAA)-induced hepatic injury. Sprague-Dawley rats were intraperitoneally administered with TAA, then injected with reversine intraperitoneally, and/or orally provided with XSLJZD. TAA resulted in liver injury with increases in the liver index and levels of serum aspartate aminotransferase (AST) and alanine aminotransferase. Reversine alleviated the liver index and AST level and improved TAA-induced pathological changes but decreased TAA-induced collagen deposition, and α-smooth muscle actin and transforming growth factor-β1 expression. Reversine also modulated the mRNA levels of inflammatory cytokines, such as RelA, interleukin (IL)-17A, IL-22, IL-1β, IL-6, NLR family pyrin domain containing 3, platelet-derived growth factor, and monocyte chemoattractant protein, and suppressed nuclear factor (NF)-κB (p65) phosphorylation and caspase 1 activation. Meanwhile, XSLJZD protected TAA-injured liver without increasing fibrosis and enhanced the regulating effect of reversine on RelA, IL-17A, IL-1β, and MCP-1 cytokines. In conclusion, reversine ameliorates liver injury and inhibits inflammation reaction by regulating NF-κB, and XSLJZD protects the liver through its synergistic effect with reversine on regulating inflammatory cytokines.
Collapse
Affiliation(s)
- Zhen-Hao Mai
- Department of Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Yuexiu District, Guangzhou, Guangdong 518180, People's Republic of China.,Guangzhou Medical University, Guangzhou, Guangdong 510180, People's Republic of China
| | - Yu Huang
- Department of Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Yuexiu District, Guangzhou, Guangdong 518180, People's Republic of China.,Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, People's Republic of China
| | - Di Huang
- Department of Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Yuexiu District, Guangzhou, Guangdong 518180, People's Republic of China.,Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, People's Republic of China
| | - Zi-Sheng Huang
- Department of Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Yuexiu District, Guangzhou, Guangdong 518180, People's Republic of China.,Guangzhou Medical University, Guangzhou, Guangdong 510180, People's Republic of China
| | - Zhi-Xiang He
- Department of Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Yuexiu District, Guangzhou, Guangdong 518180, People's Republic of China
| | - Pei-Lin Li
- Department of Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Yuexiu District, Guangzhou, Guangdong 518180, People's Republic of China
| | - Shuai Zhang
- Department of Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Yuexiu District, Guangzhou, Guangdong 518180, People's Republic of China.,Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, People's Republic of China
| | - Jie-Feng Weng
- Department of Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Yuexiu District, Guangzhou, Guangdong 518180, People's Republic of China.,Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, People's Republic of China
| | - Wei-Li Gu
- Department of Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Yuexiu District, Guangzhou, Guangdong 518180, People's Republic of China.,Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, Guangzhou, Guangdong 510180, People's Republic of China
| |
Collapse
|
10
|
Predictive Value of Precision-Cut Kidney Slices as an Ex Vivo Screening Platform for Therapeutics in Human Renal Fibrosis. Pharmaceutics 2020; 12:pharmaceutics12050459. [PMID: 32443499 PMCID: PMC7285118 DOI: 10.3390/pharmaceutics12050459] [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: 04/08/2020] [Revised: 05/10/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Animal models are a valuable tool in preclinical research. However, limited predictivity of human biological responses in the conventional models has stimulated the search for reliable preclinical tools that show translational robustness. Here, we used precision-cut kidney slices (PCKS) as a model of renal fibrosis and investigated its predictive capacity for screening the effects of anti-fibrotics. Murine and human PCKS were exposed to TGFβ or PDGF pathway inhibitors with established anti-fibrotic efficacy. For each treatment modality, we evaluated whether it affected: (1) culture-induced collagen type I gene expression and interstitial accumulation; (2) expression of markers of TGFβ and PDGF signaling; and (3) expression of inflammatory markers. We summarized the outcomes of published in vivo animal and human studies testing the three inhibitors in renal fibrosis, and drew a parallel to the PCKS data. We showed that the responses of murine PCKS to anti-fibrotics highly corresponded with the known in vivo responses observed in various animal models of renal fibrosis. Moreover, our results suggested that human PCKS can be used to predict drug efficacy in clinical trials. In conclusion, our study demonstrated that the PCKS model is a powerful predictive tool for ex vivo screening of putative drugs for renal fibrosis.
Collapse
|
11
|
|
12
|
ERK Pathway in Activated, Myofibroblast-Like, Hepatic Stellate Cells: A Critical Signaling Crossroad Sustaining Liver Fibrosis. Int J Mol Sci 2019; 20:ijms20112700. [PMID: 31159366 PMCID: PMC6600376 DOI: 10.3390/ijms20112700] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/21/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022] Open
Abstract
Fibrogenic progression of chronic liver disease, whatever the etiology, is characterized by persistent chronic parenchymal injury, chronic activation of inflammatory response, and sustained activation of liver fibrogenesis, and of pathological wound healing response. A critical role in liver fibrogenesis is played by hepatic myofibroblasts (MFs), a heterogeneous population of α smooth-muscle actin—positive cells that originate from various precursor cells through a process of activation and transdifferentiation. In this review, we focus the attention on the role of extracellular signal-regulated kinase (ERK) signaling pathway as a critical one in modulating selected profibrogenic phenotypic responses operated by liver MFs. We will also analyze major therapeutic antifibrotic strategies developed in the last two decades in preclinical studies, some translated to clinical conditions, designed to interfere directly or indirectly with the Ras/Raf/MEK/ERK signaling pathway in activated hepatic MFs, but that also significantly increased our knowledge on the biology and pathobiology of these fascinating profibrogenic cells.
Collapse
|
13
|
Stefanovic B, Manojlovic Z, Vied C, Badger CD, Stefanovic L. Discovery and evaluation of inhibitor of LARP6 as specific antifibrotic compound. Sci Rep 2019; 9:326. [PMID: 30674965 PMCID: PMC6344531 DOI: 10.1038/s41598-018-36841-y] [Citation(s) in RCA: 35] [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: 08/06/2018] [Accepted: 11/27/2018] [Indexed: 01/17/2023] Open
Abstract
Fibrosis is characterized by excessive production of type I collagen. Biosynthesis of type I collagen in fibrosis is augmented by binding of protein LARP6 to the 5' stem-loop structure (5'SL), which is found exclusively in type I collagen mRNAs. A high throughput screen was performed to discover inhibitors of LARP6 binding to 5'SL, as potential antifibrotic drugs. The screen yielded one compound (C9) which was able to dissociate LARP6 from 5' SL RNA in vitro and to inactivate the binding of endogenous LARP6 in cells. Treatment of hepatic stellate cells (liver cells responsible for fibrosis) with nM concentrations of C9 reduced secretion of type I collagen. In precision cut liver slices, as an ex vivo model of hepatic fibrosis, C9 attenuated the profibrotic response at 1 μM. In prophylactic and therapeutic animal models of hepatic fibrosis C9 prevented development of fibrosis or hindered the progression of ongoing fibrosis when administered at 1 mg/kg. Toxicogenetics analysis revealed that only 42 liver genes changed expression after administration of C9 for 4 weeks, suggesting minimal off target effects. Based on these results, C9 represents the first LARP6 inhibitor with significant antifibrotic activity.
Collapse
Affiliation(s)
- Branko Stefanovic
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL, 32306, USA.
| | - Zarko Manojlovic
- Keck School of Medicine of University of Southern California, 1450 Biggy Street, NRT 4510, Los Angeles, CA, 90033, USA
| | - Cynthia Vied
- Translational Science Laboratory, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL, 32306, USA
| | - Crystal-Dawn Badger
- Translational Science Laboratory, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL, 32306, USA
- Proteomics and Metabolomics Facility, Colorado State University, 401 West Pitkin Street, Fort Collins, CO, 80521, USA
| | - Lela Stefanovic
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL, 32306, USA
| |
Collapse
|
14
|
Peng F, Tee JK, Setyawati MI, Ding X, Yeo HLA, Tan YL, Leong DT, Ho HK. Inorganic Nanomaterials as Highly Efficient Inhibitors of Cellular Hepatic Fibrosis. ACS APPLIED MATERIALS & INTERFACES 2018; 10:31938-31946. [PMID: 30156820 DOI: 10.1021/acsami.8b10527] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Chronic liver dysfunction usually begins with hepatic fibrosis. To date, no effective anti-fibrotic drugs have been approved for clinical use in humans. In the current work, titanium dioxide (TiO2) nanoparticles (NPs) and silicon dioxide (SiO2) NPs are used as active inhibitors with intrinsic chemico-physico properties to block fibrosis and the associated phenotypes through acting on hepatic stellate cells (HSCs, the liver machinery for depositing scar tissues seen in fibrosis). Using LX-2 cells as the HSC model, internalized nanomaterials are found to suppress classical outcomes of cellular fibrosis, for example, inhibiting the expression of collagen I (Col-I) and alpha smooth muscle actin (α-SMA), initiated by transforming growth factor β (TGF-β)-activated HSCs in both a concentration-dependent and a time-dependent manner. Biochemically, these nanomaterials could also facilitate the proteolytic breakdown of collagen by up-regulation of matrix metalloproteinases (MMPs) and down-regulation of tissue inhibitors of MMPs (TIMPs). Furthermore, through regulating epithelial-mesenchymal transition (EMT) genes [e.g., E-cadherin (E-Cad) and N-cadherin (N-Cad)], the adhesion and migration profiles of TGF-β-activated LX-2 cells treated with nanomaterials were further inhibited, reverting them to a more quiescent state. Thus, the collective results pave the new way that nanomaterials can be used as potential therapeutic inhibitors for the treatment of in vivo fibrosis.
Collapse
Affiliation(s)
- Fei Peng
- Department of Pharmacy, Faculty of Science , National University of Singapore , 18 Science Drive 4 , Singapore 117543 , Singapore
| | - Jie Kai Tee
- Department of Pharmacy, Faculty of Science , National University of Singapore , 18 Science Drive 4 , Singapore 117543 , Singapore
- NUS Graduate School for Integrative Sciences & Engineering , Centre for Life Sciences , 28 Medical Drive , Singapore 117456 , Singapore
| | - Magdiel Inggrid Setyawati
- Department of Chemical and Biomolecular Engineering , National University of Singapore , 4 Engineering Drive 4 , Singapore 117585 , Singapore
| | - Xianguang Ding
- Department of Chemical and Biomolecular Engineering , National University of Singapore , 4 Engineering Drive 4 , Singapore 117585 , Singapore
| | - Hui Ling Angie Yeo
- Department of Pharmacy, Faculty of Science , National University of Singapore , 18 Science Drive 4 , Singapore 117543 , Singapore
| | - Yeong Lan Tan
- Department of Pharmacy, Faculty of Science , National University of Singapore , 18 Science Drive 4 , Singapore 117543 , Singapore
- NUS Graduate School for Integrative Sciences & Engineering , Centre for Life Sciences , 28 Medical Drive , Singapore 117456 , Singapore
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering , National University of Singapore , 4 Engineering Drive 4 , Singapore 117585 , Singapore
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science , National University of Singapore , 18 Science Drive 4 , Singapore 117543 , Singapore
| |
Collapse
|
15
|
Papadopoulos N, Lennartsson J. The PDGF/PDGFR pathway as a drug target. Mol Aspects Med 2018; 62:75-88. [DOI: 10.1016/j.mam.2017.11.007] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023]
|
16
|
Angiogenesis and Hepatic Fibrosis: Western and Chinese Medicine Therapies on the Road. Chin J Integr Med 2018; 24:713-720. [DOI: 10.1007/s11655-018-3007-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2016] [Indexed: 02/07/2023]
|
17
|
Abstract
Fibrosis is part of a tissue repair response to injury, defined as increased deposition of extracellular matrix. In some instances, fibrosis is beneficial; however, in the majority of diseases fibrosis is detrimental. Virtually all chronic progressive diseases are associated with fibrosis, representing a huge number of patients worldwide. Fibrosis occurs in all organs and tissues, becomes irreversible with time and further drives loss of tissue function. Various cells types initiate and perpetuate pathological fibrosis by paracrine activation of the principal cellular executors of fibrosis, i.e. stromal mesenchymal cells like fibroblasts, pericytes and myofibroblasts. Multiple pathways are involved in fibrosis, platelet-derived growth factor (PDGF)-signaling being one of the central mediators. Stromal mesenchymal cells express both PDGF receptors (PDGFR) α and β, activation of which drives proliferation, migration and production of extracellular matrix, i.e. the principal processes of fibrosis. Here, we review the role of PDGF signaling in organ fibrosis, with particular focus on the more recently described ligands PDGF-C and -D. We discuss the potential challenges, opportunities and open questions in using PDGF as a potential target for anti-fibrotic therapies.
Collapse
Affiliation(s)
| | - Jürgen Floege
- Division of Nephrology, RWTH University of Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH University of Aachen, Germany; Division of Nephrology, RWTH University of Aachen, Germany.
| |
Collapse
|
18
|
Cannito S, Novo E, Parola M. Therapeutic pro-fibrogenic signaling pathways in fibroblasts. Adv Drug Deliv Rev 2017; 121:57-84. [PMID: 28578015 DOI: 10.1016/j.addr.2017.05.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/28/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023]
Abstract
Myofibroblasts (MFs) play a critical role in the progression of chronic inflammatory and fibroproliferative diseases in different tissues/organs, whatever the etiology. Fibrosis is preceded and sustained by persistent injury and inflammatory response in a profibrogenic scenario involving mutual interactions, operated by several mediators and pathways, of MFs and related precursor cells with innate immunity cells and virtually any cell type in a defined tissue. These interactions, mediators and related signaling pathways are critical in initiating and perpetuating the differentiation of precursor cells into MFs that in different tissues share peculiar traits and phenotypic responses, including the ability to proliferate, produce ECM components, migrate and contribute to the modulation of inflammatory response and tissue angiogenesis. Literature studies related to liver, lung and kidney fibrosis have outlined a number of MF-related core regulatory fibrogenic signaling pathways conserved across these different organs and potentially targetable in order to develop effective antifibrotic therapeutic strategies.
Collapse
|
19
|
Rókusz A, Veres D, Szücs A, Bugyik E, Mózes M, Paku S, Nagy P, Dezső K. Ductular reaction correlates with fibrogenesis but does not contribute to liver regeneration in experimental fibrosis models. PLoS One 2017; 12:e0176518. [PMID: 28445529 PMCID: PMC5405957 DOI: 10.1371/journal.pone.0176518] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/12/2017] [Indexed: 12/13/2022] Open
Abstract
Background and aims Ductular reaction is a standard component of fibrotic liver tissue but its function is largely unknown. It is supposed to interact with the matrix producing myofibroblasts and compensate the declining regenerative capacity of hepatocytes. The relationship between the extent of fibrosis—ductular reaction, proliferative activity of hepatocytes and ductular reaction were studied sequentially in experimental hepatic fibrosis models. Methods Liver fibrosis/cirrhosis was induced in wild type and TGFβ overproducing transgenic mice by carbon tetrachloride and thioacetamide administration. The effect of thioacetamide was modulated by treatment with imatinib and erlotinib. The extent of ductular reaction and fibrosis was measured by morphometry following cytokeratin 19 immunofluorescent labeling and Picro Sirius staining respectively. The proliferative activity of hepatocytes and ductular reaction was evaluated by BrdU incorporation. The temporal distribution of the parameters was followed and compared within and between different experimental groups. Results There was a strong significant correlation between the extent of fibrosis and ductular reaction in each experimental group. Although imatinib and erlotinib temporarily decreased fibrosis this effect later disappeared. We could not observe negative correlation between the proliferation of hepatocytes and ductular reaction in any of the investigated models. Conclusions The stringent connection between ductular reaction and fibrosis, which cannot be influenced by any of our treatment regimens, suggests that there is a close mutual interaction between them instead of a unidirectional causal relationship. Our results confirm a close connection between DR and fibrogenesis. However, since the two parameters changed together we could not establish a causal relationship and were unable to reveal which was the primary event. The lack of inverse correlation between the proliferation of hepatocytes and ductular reaction questions that ductular reaction can compensate for the failing regenerative activity of hepatocytes. No evidences support the persistent antifibrotic property of imatinib or erlotinib.
Collapse
Affiliation(s)
- András Rókusz
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Dániel Veres
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Armanda Szücs
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Edina Bugyik
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Miklós Mózes
- Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - Sándor Paku
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.,Tumor Progression Research Group, Joint Research Organization of the Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Péter Nagy
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Katalin Dezső
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
20
|
Abstract
Liver fibrosis resulting from chronic liver injury are major causes of morbidity and mortality worldwide. Among causes of hepatic fibrosis, viral infection is most common (hepatitis B and C). In addition, obesity rates worldwide have accelerated the risk of liver injury due to nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). Also liver fibrosis is associated with the consumption of alcohol, or autoimmune hepatitis and chronic cholangiophaties. The response of hepatocytes to inflammation plays a decisive role in the physiopathology of hepatic fibrosis, which involves the recruitment of both pro- and anti-inflammatory cells such as monocytes and macrophages. As well as the production of other cytokines and chemokines, which increase the stimulus of hepatic stellate cells by activating proinflammatory cells. The aim of this review is to identify the therapeutic options available for the treatment of the liver fibrosis, enabling the prevention of progression when is detected in time.
Collapse
Affiliation(s)
| | - Beatriz Barranco-Fragoso
- Department of Gastroenterology, National Medical Center "20 Noviembre", 03229 Mexico, DF, Mexico
| | | |
Collapse
|
21
|
Rókusz A, Bugyik E, Szabó V, Szücs A, Paku S, Nagy P, Dezső K. Imatinib accelerates progenitor cell-mediated liver regeneration in choline-deficient ethionine-supplemented diet-fed mice. Int J Exp Pathol 2016; 97:389-396. [PMID: 27918111 DOI: 10.1111/iep.12209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/10/2016] [Indexed: 12/12/2022] Open
Abstract
Severe chronic hepatic injury can induce complex reparative processes. Ductular reaction and the appearance of small hepatocytes are standard components of this response, which is thought to have both adverse (e.g. fibrosis, carcinogenesis) and beneficial (regeneration) consequences. This complex tissue reaction is regulated by orchestrated cytokine action. We have investigated the influence of the tyrosine kinase inhibitor imatinib on a regenerative process. Ductular reaction was induced in mice by the widely used choline-deficient ethionine-supplemented diet (CDE). Test animals were treated daily with imatinib. After 6 weeks of treatment, imatinib successfully reduced the extent of ductular reaction and fibrosis in the CDE model. Furthermore, the number of small hepatocytes increased, and these cells had high proliferative activity, were positive for hepatocyte nuclear factor 4 and expressed high levels of albumin and peroxisome proliferator-activated receptor alpha. The overall functional zonality of the hepatic parenchyma (cytochrome P450 2E1 and glucose 6 phosphatase activity; endogenous biotin content) was maintained. The expression of platelet-derived growth factor receptor beta, which is the major target of imatinib, was downregulated. The anti-fibrotic activity of imatinib has already been reported in several experimental models. Additionally, in the CDE model imatinib was able to enhance regeneration and preserve the functional arrangement of hepatic lobules. These results suggest that imatinib might promote the recovery of the liver following parenchymal injury through the inhibition of platelet-derived growth factor receptor beta.
Collapse
Affiliation(s)
- András Rókusz
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Edina Bugyik
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Vanessza Szabó
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Armanda Szücs
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Sándor Paku
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.,Tumor Progression Research Group, Joint Research Organization of the Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Péter Nagy
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Katalin Dezső
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
22
|
Qu K, Liu T, Lin T, Zhang X, Cui R, Liu S, Meng F, Zhang J, Tai M, Wan Y, Liu C. Tyrosine kinase inhibitors: friends or foe in treatment of hepatic fibrosis? Oncotarget 2016; 7:67650-67660. [PMID: 27588502 PMCID: PMC5341902 DOI: 10.18632/oncotarget.11767] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 08/29/2016] [Indexed: 12/21/2022] Open
Abstract
Aberrant activity of tyrosine kinases has been proved to be associated with multiple diseases including fibrotic diseases. Tyrosine kinases inhibitors (TKIs) might be a novel approach to transform the anti-fibrotic treatment. However, both beneficial and adverse effects are observed by researchers when using these TKIs in either preclinical animal models or patients with hepatic fibrosis. Since hepatotoxicity of TKIs is the leading cause for drug withdrawals thus limits its application in anti-fibrosis, not only efficacy but also safety of TKIs should be paid great concerns. It has been observed in a few studies that TKIs could induce relatively high rate of hepatic biochemical markers elevations and even result in liver failure. Fortunately, several strategies have been adopt to handle with the hepatotoxicity. Accumulating evidences suggest that hepatic stellate cells (HSC) play a pivotal role in hepatic fibrogenesis, so it might be a good option to develop selective TKIs specifically targeting HSCs. The present review will briefly summarize the anti-fibrotic mechanism of TKIs, adverse effects of TKIs as well as the novel developed selective delivery of TKIs.
Collapse
Affiliation(s)
- Kai Qu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tian Liu
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ting Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Surgical Intensive Care Unit (SICU), The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xing Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ruixia Cui
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Sinan Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Surgical Intensive Care Unit (SICU), The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fandi Meng
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingyao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Surgical Intensive Care Unit (SICU), The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Minghui Tai
- Department of Ultrasound, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yong Wan
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Surgical Intensive Care Unit (SICU), The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
23
|
Leiting S, Seidl S, Martinez-Palacian A, Muhl L, Kanse SM. Transforming Growth Factor-β (TGF-β) Inhibits the Expression of Factor VII-activating Protease (FSAP) in Hepatocytes. J Biol Chem 2016; 291:21020-21028. [PMID: 27462075 DOI: 10.1074/jbc.m116.744631] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Indexed: 01/09/2023] Open
Abstract
Deletion of the Habp2 gene encoding Factor VII-activating protease (FSAP) increases liver fibrosis in mice. A single nucleotide polymorphism (G534E) in HABP2 leads to lower enzymatic activity and is associated with enhanced liver fibrosis in humans. Liver fibrosis is associated with a decrease in FSAP expression but, to date, nothing is known about how this might be regulated. Primary mouse hepatocytes or the hepatocyte cell line, AML12, were treated with different factors, and expression of FSAP was determined. Of the various regulatory factors tested, only transforming growth factor-β (TGF-β) demonstrated a concentration- and time-dependent inhibition of FSAP expression at the mRNA and protein level. The TGF-β-Type I receptor (ALK-5) antagonist SB431542 and Smad2 siRNA, but neither SIS3, which inhibits SMAD3, nor siRNA against Smad3 could block this effect. Various regions of the HABP2 promoter region were cloned into reporter constructs, and the promoter activity was determined. Accordingly, the promoter activity, which could phenocopy changes in Habp2 mRNA in response to TGF-β, was found to be located in the 177-bp region upstream of the transcription start site, and this region did not contain any SMAD binding sites. Mutation analysis of the promoter and chromatin immunoprecipitation assays were performed to identify an important role for the ATF3 binding element. Thus, TGF-β is the most likely mediator responsible for the decrease in FSAP expression in liver fibrosis.
Collapse
Affiliation(s)
- Silke Leiting
- From the Institute for Biochemistry, Justus-Liebig-University, 35392 Giessen, Germany
| | | | | | - Lars Muhl
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
| | - Sandip M Kanse
- From the Institute for Biochemistry, Justus-Liebig-University, 35392 Giessen, Germany, Oslo University Hospital and Institute for Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway, and
| |
Collapse
|
24
|
Wang P, Koyama Y, Liu X, Xu J, Ma HY, Liang S, Kim IH, Brenner DA, Kisseleva T. Promising Therapy Candidates for Liver Fibrosis. Front Physiol 2016; 7:47. [PMID: 26909046 PMCID: PMC4754444 DOI: 10.3389/fphys.2016.00047] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/01/2016] [Indexed: 12/11/2022] Open
Abstract
Liver fibrosis is a wound-healing process in response to repeated and chronic injury to hepatocytes and/or cholangiocytes. Ongoing hepatocyte apoptosis or necrosis lead to increase in ROS production and decrease in antioxidant activity, which recruits inflammatory cells from the blood and activate hepatic stellate cells (HSCs) changing to myofibroblasts. Injury to cholangiocytes also recruits inflammatory cells to the liver and activates portal fibroblasts in the portal area, which release molecules to activate and amplify cholangiocytes. No matter what origin of myofibroblasts, either HSCs or portal fibroblasts, they share similar characteristics, including being positive for α-smooth muscle actin and producing extracellular matrix. Based on the extensive pathogenesis knowledge of liver fibrosis, therapeutic strategies have been designed to target each step of this process, including hepatocyte apoptosis, cholangiocyte proliferation, inflammation, and activation of myofibroblasts to deposit extracellular matrix, yet the current therapies are still in early-phase clinical development. There is an urgent need to translate the molecular mechanism of liver fibrosis to effective and potent reagents or therapies in human.
Collapse
Affiliation(s)
- Ping Wang
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA; Liver Research Center, Beijing Friendship Hospital, Capital Medical UniversityBeijing, China
| | - Yukinori Koyama
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Surgery, Graduate School of Medicine, Kyoto UniversityKyoto, Japan
| | - Xiao Liu
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Jun Xu
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Hsiao-Yen Ma
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - Shuang Liang
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - In H Kim
- Department of Surgery, University of CaliforniaSan Diego, La Jolla, CA, USA; Department of Medicine, University of CaliforniaSan Diego, La Jolla, CA, USA
| | - David A Brenner
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
25
|
Qu K, Huang Z, Lin T, Liu S, Chang H, Yan Z, Zhang H, Liu C. New Insight into the Anti-liver Fibrosis Effect of Multitargeted Tyrosine Kinase Inhibitors: From Molecular Target to Clinical Trials. Front Pharmacol 2016; 6:300. [PMID: 26834633 PMCID: PMC4716646 DOI: 10.3389/fphar.2015.00300] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/08/2015] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinases (TKs) is a family of tyrosine protein kinases with important functions in the regulation of a broad variety of physiological cell processes. Overactivity of TK disturbs cellular homeostasis and has been linked to the development of certain diseases, including various fibrotic diseases. In regard to liver fibrosis, several TKs, such as vascular endothelial growth factor receptor, platelet-derived growth factor receptor, fibroblast growth factor receptor, and epidermal growth factor receptor kinases, have been identified as central mediators in collagen production and potential targets for anti-liver fibrosis therapies. Given the essential role of TKs during liver fibrogenesis, multitargeted inhibitors of aberrant TK activity, including sorafenib, erlotinib, imatinib, sunitinib, nilotinib, brivanib and vatalanib, have been shown to have potential for treating liver fibrosis. Beneficial effects are observed by researchers of this field using these multitargeted TK inhibitors in preclinical animal models and in patients with liver fibrosis. The present review will briefly summarize the anti-liver fibrosis effects of multitargeted TK inhibitors and molecular mechanisms.
Collapse
Affiliation(s)
- Kai Qu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an, China
| | - Zichao Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong UniversityXi'an, China; Department of General Surgery, Shaanxi Cancer HospitalXi'an, China
| | - Ting Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an, China
| | - Sinan Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an, China
| | - Hulin Chang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong UniversityXi'an, China; Department of Hepatobiliary Surgery, Shaanxi Provincial People's HospitalXi'an, China
| | - Zhaoyong Yan
- Department of Pain Treatment, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Hongxin Zhang
- Department of Pain Treatment, Tangdu Hospital, Fourth Military Medical University Xi'an, China
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an, China
| |
Collapse
|
26
|
Peng ZW, Ikenaga N, Liu SB, Sverdlov DY, Vaid KA, Dixit R, Weinreb PH, Violette S, Sheppard D, Schuppan D, Popov Y. Integrin αvβ6 critically regulates hepatic progenitor cell function and promotes ductular reaction, fibrosis, and tumorigenesis. Hepatology 2016; 63:217-32. [PMID: 26448099 PMCID: PMC5312042 DOI: 10.1002/hep.28274] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 09/22/2015] [Accepted: 10/05/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED Integrin αvβ6 is rapidly up-regulated on cells of epithelial lineage during tissue injury, where one of its primary functions is activation of latent transforming growth factor beta 1 (TGFβ1). In human liver cirrhosis, αvβ6 is overexpressed by cells comprising the ductular reaction, and its inhibition suppresses experimental biliary fibrosis in rodents. Here, we show that αvβ6 is expressed on the actively proliferating subset of hepatic progenitor cells and is required for their progenitor function in vivo and in vitro through integrin αvβ6-dependent TGFβ1 activation. Freshly isolated αvβ6(+) liver cells demonstrate clonogenic potential and differentiate into cholangiocytes and functional hepatocytes in vitro, whereas colony formation by epithelial cell adhesion molecule-positive progenitor cells is blocked by αvβ6-neutralizing antibody and in integrin beta 6-deficient cells. Inhibition of progenitors by anti-αvβ6 antibody is recapitulated by TGFβ1 neutralization and rescued by addition of bioactive TGFβ1. Genetic disruption or selective targeting of αvβ6 with 3G9 antibody potently inhibits progenitor cell responses in mouse models of chronic biliary injury and protects from liver fibrosis and tumorigenesis, two conditions clinically associated with exacerbated ductular reaction. CONCLUSION These results suggest that αvβ6 is a promising target for chronic fibrotic liver diseases and associated cancers.
Collapse
Affiliation(s)
- Zhen-Wei Peng
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Naoki Ikenaga
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Susan B. Liu
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Deanna Y. Sverdlov
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Kahini A. Vaid
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Richa Dixit
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | | | - Dean Sheppard
- Lung Biology Center, University of California, San Francisco School of Medicine, San Francisco, CA
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Mainz, Germany
| | - Yury Popov
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
27
|
PDGF-Mediated Regulation of Liver Fibrosis. CURRENT PATHOBIOLOGY REPORTS 2015. [DOI: 10.1007/s40139-015-0096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
28
|
Lin TT, Gao DY, Liu YC, Sung YC, Wan D, Liu JY, Chiang T, Wang L, Chen Y. Development and characterization of sorafenib-loaded PLGA nanoparticles for the systemic treatment of liver fibrosis. J Control Release 2015; 221:62-70. [PMID: 26551344 DOI: 10.1016/j.jconrel.2015.11.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 07/19/2015] [Accepted: 11/05/2015] [Indexed: 11/18/2022]
Abstract
Sorafenib is a tyrosine kinase inhibitor that has recently been shown to be a potential antifibrotic agent. However, a narrow therapeutic window limits the clinical use and therapeutic efficacy of sorafenib. Herein, we have developed and optimized nanoparticle (NP) formulations prepared from a mixture of poly(ethylene glycol)-b-poly(lactic-co-glycolic acid) (PEG-PLGA) copolymers with poly(lactic-co-glycolic acid) (PLGA) for the systemic delivery of sorafenib into the fibrotic livers of CCl4-induced fibrosis mouse models. We characterized and compared the pharmaceutical and biological properties of two different PLGA nanoparticles (NPs)--PEG-PLGA NPs (PEG-PLGA/PLGA=10/0) and PEG-PLGA/PLGA NPs (PEG-PLGA/PLGA=5/5). Increasing the PLGA content in the PEG-PLGA/PLGA mixture led to increases in the particle size and drug encapsulation efficacy and a decrease in the drug release rate. Both PEG-PLGA and PEG-PLGA/PLGA NPs significantly prolonged the blood circulation of the cargo and increased the uptake by the fibrotic livers. The systemic administration of PEG-PLGA or PEG-PLGA/PLGA NPs containing sorafenib twice per week for a period of 4 weeks efficiently ameliorated liver fibrosis, as indicated by decreased α-smooth muscle actin (α-SMA) content and collagen production in the livers of CCl4-treated mice. Furthermore, sorafenib-loaded PLGA NPs significantly shrank the abnormal blood vessels and decreased microvascular density (MVD), leading to vessel normalization in the fibrotic livers. In conclusion, our results reflect the clinical potential of sorafenib-loaded PLGA NPs for the prevention and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ts-Ting Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Dong-Yu Gao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Chi Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yun-Chieh Sung
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Dehui Wan
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Jia-Yu Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Tsaiyu Chiang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Liying Wang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
29
|
Borkham-Kamphorst E, Weiskirchen R. The PDGF system and its antagonists in liver fibrosis. Cytokine Growth Factor Rev 2015; 28:53-61. [PMID: 26547628 DOI: 10.1016/j.cytogfr.2015.10.002] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/19/2015] [Indexed: 01/18/2023]
Abstract
Platelet derived growth factor (PDGF) signaling plays an important role in activated hepatic stellate cells and portal fibroblast proliferation, chemotaxis, migration and cell survival. PDGF receptors and ligands are upregulated in experimental liver fibrotic models as well as in human liver fibrotic diseases. Blocking of PDGF signaling ameliorates experimental liver fibrogenesis. The plurality of molecular and cellular activities of PDGF and its involvement in initiation, progression and resolution of hepatic fibrogenesis offers an infinite number of therapeutic possibilities. These include the application of therapeutic antibodies (e.g. AbyD3263, MOR8457) which specifically sequester individual PDGF isoforms or the inhibition of PDGF isoforms by synthetic aptamers. In particular, the isolation of innovative slow off-rate modified aptamers (e.g., SOMAmer SL1 and SL5) that carry functional groups absent in natural nucleic acids by the Systematic Evolution of Ligands by EXponential (SELEX) enrichment technique offers the possibility to design high affinity aptamers that target PDGF isoforms for clinical purposes. Dominant-negative soluble PDGF receptors are also effective in attenuation of hepatic stellate cell proliferation and hepatic fibrogenesis. Moreover, some multikinase inhibitors targeting PDGF signaling have been intensively tested during the last decade and are on the way into advanced preclinical studies and clinical trials. This narrative review aims to gauge the recent progression of research into PDGF systems and liver fibrosis.
Collapse
Affiliation(s)
- Erawan Borkham-Kamphorst
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany.
| |
Collapse
|
30
|
Choi JS, Kim JK, Yang YJ, Kim Y, Kim P, Park SG, Cho EY, Lee DH, Choi JW. Identification of cromolyn sodium as an anti-fibrotic agent targeting both hepatocytes and hepatic stellate cells. Pharmacol Res 2015; 102:176-83. [PMID: 26453959 DOI: 10.1016/j.phrs.2015.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 10/02/2015] [Accepted: 10/05/2015] [Indexed: 11/30/2022]
Abstract
Liver fibrosis and cirrhosis, the late stage of fibrosis, are threatening diseases that lead to liver failure and patient death. Although aberrantly activated hepatic stellate cells (HSCs) are the main cause of disease initiation, the symptoms are primarily related to damaged hepatocytes. Thus, damaged hepatocytes, as well as HSCs, need to be simultaneously considered as therapeutic targets to develop more efficient treatments. Here, we suggest cromolyn sodium as an anti-fibrotic agent to commonly modulate hepatocytes and hepatic stellate cells. The differentially expressed genes from 6 normal and 40 cirrhotic liver tissues which were collected from GEO data were assessed by pharmacokinetic analysis using a connectivity map to identify agents that commonly revert abnormal hepatocytes and HSCs to normal conditions. Based on a series of analyses, a few candidates were selected. Candidates were tested in vitro to determine their anti-fibrotic efficacy on HSCs and hepatocytes. Cromolyn, which was originally developed as a mast cell stabilizer, showed the potential to ameliorate activated HSCs in vitro. The activation and collagen accumulation for HSC cell lines LX2 and HSC-T6 were reduced by 50% after cromolyn treatment at a low concentration without apoptosis. Furthermore, cromolyn treatment compromised the TGF-β-induced epithelial mesenchyme transition and replicative senescence rate of hepatocytes, which are generally associated with fibrogenesis. Taken together, cromolyn may be the basis for an effective cure for fibrosis and cirrhosis because it targets both HSCs and hepatocytes.
Collapse
Affiliation(s)
- Joon-Seok Choi
- College of Pharmacy, Catholic University of Daegu, Gyeongbuk 712-702, Republic of Korea
| | - Jun Ki Kim
- Biomedical Engineering Center, Asan Institute for Life Sciences, Asan Medical Center and University of Ulsan, College of Medicine, Seoul, 138-736, Republic of Korea
| | - Yoon Jung Yang
- Wonkwang Institute of Integrative Biomedical Science and Dental Research Institute, School of Dentistry, Wonkwang University, Iksan, Chonbuk 570-711, Republic of Korea
| | - Yeseul Kim
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 305-338, Republic of Korea
| | - Pilhan Kim
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology, Daejeon, 305-338, Republic of Korea
| | - Sang Gyu Park
- Department of Pharmacy, Ajou University, Suwon, Gyenggi-do 443-270, Republic of Korea
| | - Eun-Young Cho
- Department of Internal Medicine, Wonkwang University School of Medicine & Hospital, Iksan, Jeonbuk 570-711, Republic of Korea
| | - Dae Ho Lee
- Department of Internal Medicine, Wonkwang University School of Medicine & Hospital, Iksan, Jeonbuk 570-711, Republic of Korea
| | - Jin Woo Choi
- Wonkwang Institute of Integrative Biomedical Science and Dental Research Institute, School of Dentistry, Wonkwang University, Iksan, Chonbuk 570-711, Republic of Korea; Advanced Institute of Convergence Technology, Seoul National University, Suwon, Gyeonggi-do 443-270, Republic of Korea.
| |
Collapse
|
31
|
van Dijk F, Olinga P, Poelstra K, Beljaars L. Targeted Therapies in Liver Fibrosis: Combining the Best Parts of Platelet-Derived Growth Factor BB and Interferon Gamma. Front Med (Lausanne) 2015; 2:72. [PMID: 26501061 PMCID: PMC4594310 DOI: 10.3389/fmed.2015.00072] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/18/2015] [Indexed: 12/11/2022] Open
Abstract
Cytokines, growth factors, and other locally produced mediators play key roles in the regulation of disease progression. During liver fibrosis, these mediators orchestrate the balance between pro- and antifibrotic activities as exerted by the hepatic cells. Two important players in this respect are the profibrotic mediator platelet-derived growth factor BB (PDGF-BB) and the antifibrotic cytokine interferon gamma (IFNγ). PDGF-BB, produced by many resident and infiltrating cells, causes extensive proliferation, migration, and contraction of hepatic stellate cells (HSCs) and myofibroblasts. These cells are the extracellular matrix-producing hepatic cells and they highly express the PDGFβ receptor. On the other hand, IFNγ is produced by natural killer cells in fibrotic livers and is endowed with proinflammatory, antiviral, and antifibrotic activities. This cytokine attracted much attention as a possible therapeutic compound in fibrosis. However, clinical trials yielded disappointing results because of low efficacy and adverse effects, most likely related to the dual role of IFNγ in fibrosis. In our studies, we targeted the antifibrotic IFNγ to the liver myofibroblasts. For that, we altered the cell binding properties of IFNγ, by delivery of the IFNγ-nuclear localization sequence to the highly expressed PDGFβ receptor using a PDGFβ receptor recognizing peptide, thereby creating a construct referred to as “Fibroferon” (i.e., fibroblast-targeted interferon γ). In recent years, we demonstrated that HSC-specific delivery of IFNγ increased its antifibrotic potency and improved its general safety profile in vivo, making Fibroferon highly suitable for the treatment of (fibrotic) diseases associated with elevated PDGFβ receptor expression. The present review summarizes the knowledge on these two key mediators, PDGF-BB and IFNγ, and outlines how we used this knowledge to create the cell-specific antifibrotic compound Fibroferon containing parts of both of these mediators.
Collapse
Affiliation(s)
- Fransien van Dijk
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy , Groningen , Netherlands ; Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute for Pharmacy , Groningen , Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute for Pharmacy , Groningen , Netherlands
| | - Klaas Poelstra
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy , Groningen , Netherlands
| | - Leonie Beljaars
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy , Groningen , Netherlands
| |
Collapse
|
32
|
Roth KJ, Copple BL. Role of Hypoxia-Inducible Factors in the Development of Liver Fibrosis. Cell Mol Gastroenterol Hepatol 2015; 1:589-597. [PMID: 28210703 PMCID: PMC5301877 DOI: 10.1016/j.jcmgh.2015.09.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/16/2015] [Indexed: 02/08/2023]
Abstract
Liver fibrosis remains a significant clinical problem in the United States and throughout the world. Although important advances in the understanding of this disease have been made, no effective pharmacologic agents have been developed that directly prevent or reverse the fibrotic process. Many of the successes in liver fibrosis treatment have been targeted toward treating the cause of fibrosis, such as the development of new antivirals that eradicate hepatitis virus. For many patients, however, this is not feasible, so a liver transplant remains the only viable option. Thus, there is a critical need to identify new therapeutic targets that will slow or reverse the progression of fibrosis in such patients. Research over the last 16 years has identified hypoxia-inducible factors (HIFs) as key transcription factors that drive many aspects of liver fibrosis, making them potential targets of therapy. In this review, we discuss the latest work on HIFs and liver fibrosis, including the cell-specific functions of these transcription factors in the development of liver fibrosis.
Collapse
Key Words
- BDL, bile duct ligation
- CCl4, carbon tetrachloride
- Ccr, C-C chemokine receptor
- FGF, fibroblast growth factor
- HGF, hepatocyte growth factor
- HIFs, hypoxia-inducible factors
- HSC, hepatic stellate cell
- Hepatic Stellate Cells
- Hypoxia-Inducible Factors
- Jmjd, Jumonji domain-containing
- Kupffer Cells
- Liver Fibrosis
- PAI-1, plasminogen activator inhibitor-1
- PDGF, platelet-derived growth factor
- Rgs, regulator of G-protein signaling
- TGF-β, transforming growth factor β
- VEGF, vascular endothelial growth factor
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
| | - Bryan L. Copple
- Correspondence Address correspondence to: Bryan L. Copple, PhD, Department of Pharmacology and Toxicology, Michigan State University, 1355 Bogue Street, B403 Life Sciences Building, East Lansing, Michigan 48824.Department of Pharmacology and ToxicologyMichigan State University1355 Bogue Street, B403 Life Sciences BuildingEast LansingMichigan 48824
| |
Collapse
|
33
|
Gacic J, Vorkapic E, Olsen RS, Söderberg D, Gustafsson T, Geffers R, Skoglund K, Matussek A, Wågsäter D. Imatinib reduces cholesterol uptake and matrix metalloproteinase activity in human THP-1 macrophages. Pharmacol Rep 2015; 68:1-6. [PMID: 26721343 DOI: 10.1016/j.pharep.2015.05.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 04/30/2015] [Accepted: 05/27/2015] [Indexed: 11/20/2022]
Abstract
BACKGROUND Imatinib mesylate (Glivec®, formerly STI-571) is a selective tyrosine kinase inhibitor used for the treatment of chronic myeloid leukemia and gastrointestinal stromal tumors. However, there are reports suggesting that imatinib could be atheroprotective by lowering plasma low-density lipoprotein (LDL). AIM To investigate the potential inhibitory effect of imatinib on cholesterol uptake in human macrophages as well as its effect on matrix metalloproteinase (MMP) activity. METHODS AND RESULTS Uptake of fluorescence-labeled LDL was analyzed using flow cytometry. Macrophages treated with imatinib showed a 23.5%, 27%, and 15% decrease in uptake of native LDL (p<0.05), acetylated LDL (p<0.01), and copper-modified oxidized LDL (p<0.01), respectively. Gel-based zymography showed that secretion and activity of MMP-2 and MMP-9 were inhibited by imatinib. Using GeneChip Whole Transcript Expression array analysis, no obvious gene candidates involved in the mechanisms of cholesterol metabolism or MMP regulation were found to be affected by imatinib. Instead, we found that imatinib up-regulated microRNA 155 (miR155) by 43.8% and down-regulated ADAM metallopeptidase domain 28 (ADAM28) by 41.4%. Both genes could potentially play an atheroprotective role and would be interesting targets in future studies. CONCLUSION Our results indicate that imatinib causes post-translational inhibition with respect to cholesterol uptake and regulation of MMP-2 and MMP-9. More research is needed to further evaluate the role of imatinib in the regulation of other genes and processes.
Collapse
Affiliation(s)
- Jelena Gacic
- Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Emina Vorkapic
- Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Renate Slind Olsen
- Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden; Department of Laboratory Services, County Hospital Ryhov, Jönköping, Sweden
| | - Daniel Söderberg
- Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Therese Gustafsson
- Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Robert Geffers
- Genome Analytics Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Karin Skoglund
- Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Andreas Matussek
- Department of Laboratory Services, County Hospital Ryhov, Jönköping, Sweden
| | - Dick Wågsäter
- Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
34
|
Aono Y, Kishi M, Yokota Y, Azuma M, Kinoshita K, Takezaki A, Sato S, Kawano H, Kishi J, Goto H, Uehara H, Izumi K, Nishioka Y. Role of platelet-derived growth factor/platelet-derived growth factor receptor axis in the trafficking of circulating fibrocytes in pulmonary fibrosis. Am J Respir Cell Mol Biol 2015; 51:793-801. [PMID: 24885373 DOI: 10.1165/rcmb.2013-0455oc] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Circulating fibrocytes have been reported to migrate into the injured lungs, and contribute to fibrogenesis via CXCL12-CXCR4 axis. In contrast, we report that imatinib mesylate prevented bleomycin (BLM)-induced pulmonary fibrosis in mice by inhibiting platelet-derived growth factor receptor (PDGFR), even when it was administered only in the early phase. The goal of this study was to test the hypothesis that platelet-derived growth factor (PDGF) might directly contribute to the migration of fibrocytes to the injured lungs. PDGFR expression in fibrocytes was examined by flow cytometry and RT-PCR. The migration of fibrocytes was evaluated by using a chemotaxis assay for human fibrocytes isolated from peripheral blood. The numbers of fibrocytes triple-stained for CD45, collagen-1, and CXCR4 were also examined in lung digests of BLM-treated mice. PDGFR mRNA levels in fibrocytes isolated from patients with idiopathic pulmonary fibrosis were investigated by real-time PCR. Fibrocytes expressed both PDGFR-α and -β, and migrated in response to PDGFs. PDGFR inhibitors (imatinib, PDGFR-blocking antibodies) suppressed fibrocyte migration in vitro, and reduced the number of fibrocytes in the lungs of BLM-treated mice. PDGF-BB was a stronger chemoattractant than the other PDGFs in vitro, and anti-PDGFR-β-blocking antibody decreased the numbers of fibrocytes in the lungs compared with anti-PDGFR-α antibody in vivo. Marked expression of PDGFR-β was observed in fibrocytes from patients with idiopathic pulmonary fibrosis compared with healthy subjects. These results suggest that PDGF directly functions as a strong chemoattractant for fibrocytes. In particular, the PDGF-BB-PDGFR-β biological axis might play a critical role in fibrocyte migration into the fibrotic lungs.
Collapse
Affiliation(s)
- Yoshinori Aono
- Departments of 1 Respiratory Medicine and Rheumatology, and
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yoshida S, Ikenaga N, Liu SB, Peng ZW, Chung J, Sverdlov DY, Miyamoto M, Kim YO, Ogawa S, Arch RH, Schuppan D, Popov Y. Extrahepatic platelet-derived growth factor-β, delivered by platelets, promotes activation of hepatic stellate cells and biliary fibrosis in mice. Gastroenterology 2014; 147:1378-92. [PMID: 25173753 DOI: 10.1053/j.gastro.2014.08.038] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 08/20/2014] [Accepted: 08/23/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Platelet-derived growth factor-β (PDGFB) is a mitogen for hepatic stellate cells (HSCs). We studied the cellular sources of PDGFB and the effects of a high-affinity monoclonal antibody against PDGFB (MOR8457) in mouse models of biliary fibrosis. METHODS Cellular sources of PDGFB were identified using quantitative reverse-transcription polymerase chain reaction, biochemical, and immunohistologic methods. Mice with advanced biliary fibrosis, MDR2(Abcb4)-null mice, and C57Bl/6 (control) mice were placed on 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-supplemented diets and were given weekly intraperitoneal injections of MOR8457. Platelets were depleted from MDR2-null mice by injection of an antibody against CD41, or inhibited with diets containing low-dose aspirin. Liver tissues were collected and analyzed by quantitative reverse-transcription PCR and histologic and biochemical analyses. RESULTS Levels of PDGFB protein, but not messenger RNA, were increased in fibrotic livers of MDR2-null mice, compared with control mice. Platelet clusters were detected in the hepatic endothelium, in close proximity to HSCs, and were identified as a source of PDGFB protein in MDR2-null mice. Levels of the PDGFB were increased in serum samples from patients with early stages of liver fibrosis of various etiologies (F1-2, n = 16; P < .05), compared with nonfibrotic liver tissue (F0, n = 12). Depletion of platelets from MDR2-null mice normalized hepatic levels of PDGFB within 48 hours, reducing levels of a marker of HSC activation (α-smooth muscle actin) and expression of genes that promote fibrosis. Diets supplemented with low-dose aspirin reduced circulating serum and hepatic levels of PDGFB and significantly reduced progression of fibrosis in MDR2-null mice over 1 year. MOR8457 produced a dose-dependent decrease in liver fibrosis in MDR2-null mice, reducing collagen deposition by 45% and expression of fibrosis-associated genes by 50%, compared with mice given a control antibody. In vitro, platelets activated freshly isolated HSCs (induction of α-smooth muscle actin and fibrosis-associated genes) via a PDGFB-dependent mechanism. MOR8457 also reduced liver fibrosis in mice placed on DDC-supplemented diets. CONCLUSIONS Platelets produce PDGFB to activate HSC and promote fibrosis in MDR2-null mice and mice on DDC-supplemented diets. Antiplatelet therapy or selective inhibition of PDGFB might reduce biliary fibrosis in patients with liver disease.
Collapse
Affiliation(s)
- Shuhei Yoshida
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Naoki Ikenaga
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Susan B Liu
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Zhen-Wei Peng
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jeanhee Chung
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Deanna Y Sverdlov
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Makoto Miyamoto
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Yong Ook Kim
- Institute of Translational Immunology, University Medical Center, Mainz, Germany
| | | | | | - Detlef Schuppan
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; Institute of Translational Immunology, University Medical Center, Mainz, Germany
| | - Yury Popov
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
36
|
Nishioka Y, Azuma M, Kishi M, Aono Y. Targeting platelet-derived growth factor as a therapeutic approach in pulmonary fibrosis. THE JOURNAL OF MEDICAL INVESTIGATION 2014; 60:175-83. [PMID: 24190033 DOI: 10.2152/jmi.60.175] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal lung disease characterized by the proliferation of fibroblasts and deposition of extracellular matrix. Since the prognosis of IPF is still poor, novel therapeutic modalities are strongly required. For this reason, to find molecular target for therapy of IPF is of much importance. The recent understanding of pathogenesis in IPF indicates the critical role of alveolar epithelial type II cells (AECII) and fibroblasts. Although the detailed mechanisms involved in IPF is still unclear, various profibrotic mediators which are produced by the injured AECII are thought to play a role in the progression of pulmonary fibrosis via stimulating fibroblasts. Among them, platelet-derived growth factor (PDGF) is one of critical growth factors by stimulating the proliferation and migration of fibroblasts. In this review, we discuss the role of PDGF in pulmonary fibrosis and the possibility as a therapeutic target for IPF.
Collapse
Affiliation(s)
- Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Institute of Health Biosciences, the University of Tokushima Graduate School
| | | | | | | |
Collapse
|
37
|
Westra IM, Oosterhuis D, Groothuis GMM, Olinga P. The effect of antifibrotic drugs in rat precision-cut fibrotic liver slices. PLoS One 2014; 9:e95462. [PMID: 24755660 PMCID: PMC3995767 DOI: 10.1371/journal.pone.0095462] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/26/2014] [Indexed: 12/16/2022] Open
Abstract
Two important signaling pathways in liver fibrosis are the PDGF- and TGFβ pathway and compounds inhibiting these pathways are currently developed as antifibrotic drugs. Testing antifibrotic drugs requires large numbers of animal experiments with high discomfort. Therefore, a method to study these drugs ex vivo was developed using precision-cut liver slices from fibrotic rat livers (fPCLS), representing an ex vivo model with a multicellular fibrotic environment. We characterized the fibrotic process in fPCLS from rat livers after 3 weeks of bile duct ligation (BDL) during incubation and tested compounds predominantly inhibiting the TGFβ pathway (perindopril, valproic acid, rosmarinic acid, tetrandrine and pirfenidone) and PDGF pathway (imatinib, sorafenib and sunitinib). Gene expression of heat shock protein 47 (Hsp47), α smooth muscle actin (αSma) and pro-collagen 1A1 (Pcol1A1) and protein expression of collagens were determined. During 48 hours of incubation, the fibrosis process continued in control fPCLS as judged by the increased gene expression of the three fibrosis markers, and the protein expression of collagen 1, mature fibrillar collagen and total collagen. Most PDGF-inhibitors and TGFβ-inhibitors significantly inhibited the increase in gene expression of Hsp47, αSma and Pcol1A1. Protein expression of collagen 1 was significantly reduced by all PDGF-inhibitors and TGFβ-inhibitors, while total collagen was decreased by rosmarinic acid and tetrandrine only. However, fibrillar collagen expression was not changed by any of the drugs. In conclusion, rat fPCLS can be used as a functional ex vivo model of established liver fibrosis to test antifibrotic compounds inhibiting the PDGF- and TGFβ signalling pathway.
Collapse
Affiliation(s)
- Inge M. Westra
- Pharmacokinetics, Toxicology and Targeting, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
- Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Dorenda Oosterhuis
- Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Geny M. M. Groothuis
- Pharmacokinetics, Toxicology and Targeting, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Peter Olinga
- Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
38
|
Westra IM, Oosterhuis D, Groothuis GMM, Olinga P. Precision-cut liver slices as a model for the early onset of liver fibrosis to test antifibrotic drugs. Toxicol Appl Pharmacol 2014; 274:328-38. [PMID: 24321339 DOI: 10.1016/j.taap.2013.11.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 11/19/2013] [Accepted: 11/25/2013] [Indexed: 01/26/2023]
Abstract
Induction of fibrosis during prolonged culture of precision-cut liver slices (PCLS) was reported. In this study, the use of rat PCLS was investigated to further characterize the mechanism of early onset of fibrosis in this model and the effects of antifibrotic compounds. Rat PCLS were incubated for 48h, viability was assessed by ATP and gene expression of PDGF-B and TGF-β1 and the fibrosis markers Hsp47, αSma and Pcol1A1 and collagen1 protein expressions were determined. The effects of the antifibrotic drugs imatinib, sorafenib and sunitinib, PDGF-pathway inhibitors, and perindopril, valproic acid, rosmarinic acid, tetrandrine and pirfenidone, TGFβ-pathway inhibitors, were determined. After 48h of incubation, viability of the PCLS was maintained and gene expression of PDGF-B was increased while TGF-β1 was not changed. Hsp47, αSma and Pcol1A1 gene expressions were significantly elevated in PCLS after 48h, which was further increased by PDGF-BB and TGF-β1. The increased gene expression of fibrosis markers was inhibited by all three PDGF-inhibitors, while TGFβ-inhibitors showed marginal effects. The protein expression of collagen 1 was inhibited by imatinib, perindopril, tetrandrine and pirfenidone. In conclusion, the increased gene expression of PDGF-B and the down-regulation of fibrosis markers by PDGF-pathway inhibitors, together with the absence of elevated TGF-β1 gene expression and the limited effect of the TGFβ-pathway inhibitors, indicated the predominance of the PDGF pathway in the early onset of fibrosis in PCLS. PCLS appear a useful model for research of the early onset of fibrosis and for testing of antifibrotic drugs acting on the PDGF pathway.
Collapse
Affiliation(s)
- Inge M Westra
- Division of Pharmacokinetics, Toxicology and Targeting, Department of Pharmacy, University of Groningen, The Netherlands
| | - Dorenda Oosterhuis
- Division of Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, The Netherlands
| | - Geny M M Groothuis
- Division of Pharmacokinetics, Toxicology and Targeting, Department of Pharmacy, University of Groningen, The Netherlands
| | - Peter Olinga
- Division of Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, The Netherlands.
| |
Collapse
|
39
|
Tibaldi E, Zonta F, Bordin L, Magrin E, Gringeri E, Cillo U, Idotta G, Pagano MA, Brunati AM. The tyrosine phosphatase SHP-1 inhibits proliferation of activated hepatic stellate cells by impairing PDGF receptor signaling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:288-98. [PMID: 24140598 DOI: 10.1016/j.bbamcr.2013.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 09/20/2013] [Accepted: 10/11/2013] [Indexed: 02/08/2023]
Abstract
The dimerization and auto-transphosphorylation of platelet-derived growth factor receptor (PDGFR) upon engagement by platelet-derived growth factor (PDGF) activates signals promoting the mitogenic response of hepatic stellate cells (HSCs) due to liver injury, thus contributing to the development of hepatic fibrosis. We demonstrate that the tyrosine phosphatases Src homology 2 domain-containing phosphatase 1 and 2 (SHP-1 and SHP-2) act as crucial regulators of a complex signaling network orchestrated by PDGFR activation in a spatio-temporal manner with diverse and opposing functions in HSCs. In fact, silencing of either phosphatase shows that SHP-2 is committed to PDGFR-mediated cell proliferation, whereas SHP-1 dephosphorylates PDGFR hence abrogating the downstream signaling pathways that result in HSC activation. In this regard, SHP-1 as an off-switch of PDGFR signaling appears to emerge as a valuable molecular target to trigger as to prevent HSC proliferation and the fibrogenic effects of HSC activation. We show that boswellic acid, a multitarget compound with potent anti-inflammatory action, exerts an anti-proliferative effect on HSCs, as in other cell models, by upregulating SHP-1 with subsequent dephosphorylation of PDGFR-β and downregulation of PDGF-dependent signaling after PDGF stimulation. Moreover, the synergism resulting from the combined use of boswellic acid and imatinib, which directly inhibits PDGFR-β activity, on activated HSCs offers new perspectives for the development of therapeutic strategies that could implement molecules affecting diverse players of this molecular circuit, thus paving the way to multi-drug low-dose regimens for liver fibrosis.
Collapse
Affiliation(s)
- Elena Tibaldi
- Department of Molecular Medicine, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy
| | - Francesca Zonta
- Department of Molecular Medicine, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy
| | - Luciana Bordin
- Department of Molecular Medicine, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy
| | - Elisa Magrin
- Department of Molecular Medicine, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy
| | - Enrico Gringeri
- Department of Surgical and Gastroenterological Sciences, University of Padua, Via Giustiniani 2, 35128 Padua, Italy
| | - Umberto Cillo
- Department of Surgical and Gastroenterological Sciences, University of Padua, Via Giustiniani 2, 35128 Padua, Italy
| | - Giuseppe Idotta
- Department of Medical Specialties, Hospital of Cittadella, Via Casa di Ricovero 40, 35013 Padua, Italy
| | - Mario Angelo Pagano
- Department of Molecular Medicine, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy.
| | - Anna Maria Brunati
- Department of Molecular Medicine, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy.
| |
Collapse
|
40
|
Abstract
Fibrosis is an intrinsic response to chronic injury, maintaining organ integrity when extensive necrosis or apoptosis occurs. With protracted damage, fibrosis can progress toward excessive scarring and organ failure, as in liver cirrhosis. To date, antifibrotic treatment of fibrosis represents an unconquered area for drug development, with enormous potential but also high risks. Preclinical research has yielded numerous targets for antifibrotic agents, some of which have entered early-phase clinical studies, but progress has been hampered due to the relative lack of sensitive and specific biomarkers to measure fibrosis progression or reversal. Here we focus on antifibrotic approaches for liver that address specific cell types and functional units that orchestrate fibrotic wound healing responses and have a sound preclinical database or antifibrotic activity in early clinical trials. We also touch upon relevant clinical study endpoints, optimal study design, and developments in fibrosis imaging and biomarkers.
Collapse
Affiliation(s)
- Detlef Schuppan
- Institute of Molecular and Translational Medicine and Department of Medicine I, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany.
| | | |
Collapse
|
41
|
Kim Y, Fiel MI, Albanis E, Chou HI, Zhang W, Khitrov G, Friedman SL. Anti-fibrotic activity and enhanced interleukin-6 production by hepatic stellate cells in response to imatinib mesylate. Liver Int 2012; 32:1008-17. [PMID: 22507133 PMCID: PMC3370152 DOI: 10.1111/j.1478-3231.2012.02806.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 03/14/2012] [Indexed: 02/13/2023]
Abstract
OBJECTIVE To examine imatinib mesylate's effects on stellate cell responses in vivo and in vitro. The hepatic stellate cell (HSC) is a key target of anti-fibrotic therapies. Imatinib mesylate is a small molecule receptor tyrosine kinase inhibitor indicated for treatment of chronic myelogenous leukaemia and GI stromal tumours. DESIGN Because imatinib inhibits β-PDGFR signalling, which stimulates HSC proliferation, we assessed its activity in culture and in vivo, and examined downstream targets in a human stellate cell line (LX-2) using cDNA microarray. METHODS AND RESULTS Imatinib inhibited proliferation of LX-2 cells (0.5-10 mM) but not primary human stellate cells, with no effect on viability, associated with attenuated β-PDGFR phosphorylation. Mitochondrial activity and superoxide anion production were decreased in response to imatinib. cDNA microarray uncovered up-regulation of 29 genes in response to imatinib, including interleukin-6 (IL-6) mRNA, which was correlated with progressive IL-6 secretion. Imatinib also decreased gene expression of collagen α(1) (I), alpha smooth muscle actin, β-PDGFR, transforming growth factor β receptor type 1, matrix metalloproteinase 2 and tissue inhibitor of metalloproteinase 2. In vivo, imatinib administered to rats beginning 4 weeks after starting thioacetamide (TAA) led to reduced collagen content, with significant reductions in portal pressure and down-regulation of fibrogenic genes in whole liver. Importantly, hepatic IL-6 mRNA levels were significantly increased in TAA-treated animals receiving imatinib. CONCLUSIONS These findings reinforce the anti-fibrotic activity of imatinib and uncover an unexpected link between inhibition of HSC activation by imatinib and enhanced secretion of IL-6, a regenerative cytokine.
Collapse
Affiliation(s)
- Youngchul Kim
- Division of Liver Diseases, Department of Medicine, The Mount Sinai School of Medicine, New York, NY,College of Oriental Medicine, Kyung Hee University, Seoul, Korea
| | - Maria Isabel Fiel
- Department of Pathology, The Mount Sinai School of Medicine, New York, NY
| | - Efsevia Albanis
- Division of Liver Diseases, Department of Medicine, The Mount Sinai School of Medicine, New York, NY
| | - Hsin I Chou
- Division of Liver Diseases, Department of Medicine, The Mount Sinai School of Medicine, New York, NY
| | - Weijia Zhang
- Bioinformatics Laboratory, Department of Medicine, The Mount Sinai School of Medicine, New York, NY
| | - Gregory Khitrov
- Life Sciences Technology Laboratory Department of Medicine, The Mount Sinai School of Medicine, New York, NY
| | - Scott L. Friedman
- Division of Liver Diseases, Department of Medicine, The Mount Sinai School of Medicine, New York, NY
| |
Collapse
|
42
|
Kuo WL, Yu MC, Lee JF, Tsai CN, Chen TC, Chen MF. Imatinib mesylate improves liver regeneration and attenuates liver fibrogenesis in CCL4-treated mice. J Gastrointest Surg 2012; 16:361-369. [PMID: 22068968 DOI: 10.1007/s11605-011-1764-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 10/16/2011] [Indexed: 01/31/2023]
Abstract
BACKGROUNDS Imatinib mesylate (STI-571), a tyrosine kinase inhibitor, has previously been demonstrated to attenuate liver fibrogenesis through inhibition of the activation of hepatic stellate cells (HSCs) in CCL(4)-treated rat models. AIMS This study aimed to further evaluate the role of STI-571 in liver regeneration. MATERIALS AND METHODS All animals were divided into four groups, and mice were treated with or without CCL(4) and STI-571 (n = 6 for each group). RESULTS Activated cultured HSCs in vitro with STI-571 administration showed increased apoptosis and reduced proliferation, as determined by flow cytometric analysis, 3-(4, 5-cimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide assay, and confocal microscopy. STI-571 treatment attenuated liver fibrosis in vivo, as was evident in the results of histology, mRNA level, and expression analysis of smooth muscle actin and type I collagen. Mice treated with STI-571 had increased liver weight ratio and the improvement in liver regeneration was compatible with the change of serum interleukin 6 levels (p < 0.05). Further, increased apoptosis and a reduced proliferation were observed in the CCL(4)-treated mice after STI-571 treatment based on the immunohistochemical staining of Annexin V, phosphorylated STAT3, and PCNA. CONCLUSION STI-571 treatment effectively attenuated liver fibrogenesis and improved in liver regeneration in vivo and induced apoptosis in HSCs both in vitro and in vivo.
Collapse
Affiliation(s)
- Wen-Ling Kuo
- Department of Surgery, Chang Gung Memorial Hospital and Medical School of Chang Gung University, Kuei-Shan, Taoyuan, Taiwan
| | | | | | | | | | | |
Collapse
|
43
|
van Beuge MM, Poelstra K, Prakash J. Specific delivery of kinase inhibitors in nonmalignant and malignant diseases. Expert Opin Drug Deliv 2011; 9:59-70. [PMID: 22111941 DOI: 10.1517/17425247.2012.638625] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Kinase inhibitors have been hailed as a breakthrough in the treatment of cancer. Extensive research is now being devoted to the development of kinase inhibitors as a treatment for many nonmalignant diseases. However, the use of kinase inhibitors in both malignant and nonmalignant diseases is also associated with side effects and the development of resistance. It may be worthwhile to explore whether cell-specific delivery of kinase inhibitors improves therapeutic efficacy and reduces side effects. AREAS COVERED This review aims to provide an overview of the preclinical studies performed to examine the specific targeting of kinase inhibitors in vitro and in vivo. It gives an introduction to kinase signaling pathways induced during disease, along with the possible problems associated with their inhibition. It also discusses the studies on specific delivery and shows that altering the specificity of kinase inhibitors by targeting methods improves their effectivity and safety. EXPERT OPINION Compared with the delivery of cytotoxic compounds, the specific delivery of kinase inhibitors has not yet been studied extensively. The studies discussed in this review provide an insight into methods used to target kinase inhibitors to different organs. The targeting of different kinase inhibitors has improved their therapeutic possibilities, but many questions still remain to be studied.
Collapse
Affiliation(s)
- Marike Marjolijn van Beuge
- University of Groningen, University Centre for Pharmacy, Department of Pharmacokinetics, Toxicology & Targeting, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| | | | | |
Collapse
|
44
|
El-Agamy DS, Shebl AM, Said SA. Prevention and treatment of Schistosoma mansoni-induced liver fibrosis in mice. Inflammopharmacology 2011; 19:307-16. [DOI: 10.1007/s10787-011-0092-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 08/24/2011] [Indexed: 12/21/2022]
|
45
|
Shaker ME, Shiha GE, Ibrahim TM. Comparison of early treatment with low doses of nilotinib, imatinib and a clinically relevant dose of silymarin in thioacetamide-induced liver fibrosis. Eur J Pharmacol 2011; 670:593-600. [PMID: 21925495 DOI: 10.1016/j.ejphar.2011.08.041] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2011] [Revised: 07/28/2011] [Accepted: 08/27/2011] [Indexed: 11/29/2022]
Abstract
Our previous study has already confirmed a promising anti-fibrotic activity especially for nilotinib; when given at a daily dose of 10 mg/kg during the last 4 weeks of thioacetamide (TAA)-induced liver fibrosis for 12 weeks in rats. Therefore, this study was carried out to compare the prophylactic potential of low dose of nilotinib to that of its predecessor, imatinib, and a clinically relevant dose of the standard hepatoprotective treatment, silymarin, in TAA-intoxication. Male Wistar rats received intraperitoneal injections of TAA (150 mg/kg, twice weekly) for 8 weeks, as well as oral treatments with imatinib (5 mg/kg/day), nilotinib (5 mg/kg/day) and silymarin (50 mg/kg/day) from the first day of TAA-intoxication. At the end of the study, chronic hepatic injury was evaluated by analysis of liver function tests in serum. Hepatic oxidative stress was assessed by measuring malondialdehyde, 4-hydroxynonenal, total nitrate/nitrite and reduced glutathione contents, as well as myeloperoxidase and superoxide dismutase activities. Hepatic fibrosis was evaluated by histopathology and collagen content. Our results suggest that the prophylactic potential of nilotinib (5 mg/kg/day), imatinib (5mg/kg/day) and silymarin (50 mg/kg/day) in TAA-intoxication for 8 weeks is lower than the late treatments of nilotinib (10 mg/kg/day), imatinib (10mg/kg/day) and silymarin (100 mg/kg/day) during the last 4 weeks of TAA-intoxication for 12 weeks in rats. Taken together, this study suggests that nilotinib may have higher anti-fibrotic activity when administered at a significant stage of fibrosis as a result of impairment of its metabolism in the fibrotic livers.
Collapse
Affiliation(s)
- Mohamed E Shaker
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | | | | |
Collapse
|
46
|
Dolman MEM, van Dorenmalen KMA, Pieters EHE, Lacombe M, Pato J, Storm G, Hennink WE, Kok RJ. Imatinib-ULS-lysozyme: a proximal tubular cell-targeted conjugate of imatinib for the treatment of renal diseases. J Control Release 2011; 157:461-8. [PMID: 21911014 DOI: 10.1016/j.jconrel.2011.08.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/26/2011] [Accepted: 08/27/2011] [Indexed: 01/07/2023]
Abstract
The anticancer drug imatinib is an inhibitor of the platelet-derived growth factor receptor (PDGFR) kinases, which are involved in the pathogenesis of fibrotic diseases. In the current study we investigated the delivery of imatinib to the proximal tubular cells of the kidneys and evaluated the potential antifibrotic effects of imatinib in tubulointerstitial fibrosis. Coupling of imatinib to the low molecular weight protein lysozyme via the platinum (II)-based linker ULS yielded a 0.8:1 drug-carrier conjugate that rapidly accumulated in the proximal tubular cells upon intravenous and intraperitoneal administration. The bioavailability of intraperitoneally administered imatinib-ULS-lysozyme was 100%. Renal imatinib levels persisted for up to 3 days after a single injection of imatinib-ULS-lysozyme. Compared with an equal dose imatinib mesylate, imatinib-ULS-lysozyme resulted in a 30- and 15-fold higher renal exposure of imatinib, for intravenous and intraperitoneal administration respectively. Imatinib-ULS-lysozyme could not be detected in the heart, which is the organ at risk for side-effects of prolonged treatment with imatinib. The efficacy of imatinib-ULS-lysozyme in the treatment of tubulointerstitial fibrosis was evaluated in the unilateral ureteral obstruction (UUO) model in mice. Three days UUO resulted in all signs of early fibrosis, i.e. an increased deposition of matrix and production of profibrotic factors. Although a moderately increased activity of PDGFR-β was observed, the profibrotic phenotype could not be inhibited with imatinib mesylate or with imatinib-ULS-lysozyme. Further evaluation of imatinib mesylate and imatinib-ULS-lysozyme is therefore warranted in an animal model of renal disease in which the activation of PDGFR-β is more pronounced.
Collapse
Affiliation(s)
- M E M Dolman
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Shaker ME, Salem HA, Shiha GE, Ibrahim TM. Nilotinib counteracts thioacetamide-induced hepatic oxidative stress and attenuates liver fibrosis progression. Fundam Clin Pharmacol 2011; 25:248-57. [PMID: 20408881 DOI: 10.1111/j.1472-8206.2010.00824.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The aim of this study was to evaluate and compare the effects of imatinib and nilotinib to that of silymarin on established liver fibrosis and oxidative stress in a thioacetamide (TAA) rat model. Male Wistar rats received intraperitoneal (i.p.) injections of TAA (150mg/kg, twice weekly) for 12weeks. Daily treatments with imatinib (10mg/kg), nilotinib (10mg/kg), and silymarin (100mg/kg) were administered orally during the last 4weeks of TAA-administration. At the end of the study, hepatic damage was evaluated by analysis of liver function tests in serum. Hepatic histopathology and collagen content were employed to quantify liver fibrosis. Hepatic oxidative stress was assessed by measuring malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), total nitrate/nitrite (NOx), and reduced glutathione (GSH) contents, as well as myeloperoxidase (MPO) and superoxide dismutase (SOD) activities. Nilotinib, silymarin and, to a lesser extent, imatinib treatments ameliorated TAA-induced hepatic oxidative stress and damage as indicated by hepatic MDA, 4-HNE, NOx, GSH, MPO and SOD levels, as well as liver function tests. Hepatic histopathology results revealed that nilotinib, imatinib, and silymarin treatments decreased the mean score of fibrosis in TAA-treated rats by 24, 14, and 3%, respectively. However, nilotinib and silymarin, but not imatinib, treatments decreased hepatic collagen content in TAA-treated rats by 17 and 36%, respectively. In conclusion, we demonstrated for the first time that nilotinib not only protected against hepatic oxidative stress, but also slowed down liver fibrosis progression. Thus, we provide the first evidence that nilotinib might be a promising anti-fibrotic drug.
Collapse
Affiliation(s)
- Mohamed E Shaker
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt.
| | | | | | | |
Collapse
|
48
|
Shaker ME, Zalata KR, Mehal WZ, Shiha GE, Ibrahim TM. Comparison of imatinib, nilotinib and silymarin in the treatment of carbon tetrachloride-induced hepatic oxidative stress, injury and fibrosis. Toxicol Appl Pharmacol 2011; 252:165-75. [PMID: 21316382 PMCID: PMC3895503 DOI: 10.1016/j.taap.2011.02.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 02/02/2011] [Accepted: 02/03/2011] [Indexed: 11/27/2022]
Abstract
Effective and well-tolerated anti-fibrotic drugs are currently lacking. Therefore, this study was carried out to investigate the potential anti-fibrotic effects of imatinib, nilotinib and silymarin on established hepatic fibrosis in the carbon tetrachloride (CCl(4)) rat model. Male Wistar rats received intraperitoneal injections of CCl(4) twice weekly for 8weeks, as well as daily intraperitoneal treatments of imatinib (10 and 20mg/kg), nilotinib (10 and 20mg/kg) and silymarin (100mg/kg) during the last 4weeks of CCl(4)-intoxication. At the end of the study, hepatic damage was evaluated by analysis of liver function tests and hepatic oxidative stress parameters. Hepatic fibrosis was evaluated by histopathology and morphometry, as well as collagen and 4-hydroxyproline contents. Nilotinib (20mg/kg) was the most effective treatment to counteract CCl(4)-induced hepatic injury as indicated by liver function tests and histopathology. Nilotinib (10mg/kg), nilotinib (20mg/kg) and silymarin (100mg/kg) treatments reduced the mean score of hepatic fibrosis by 31%, 68% and 47%, respectively, and hepatic collagen content by 47%, 49% and 18%, respectively in CCl(4)-treated rats. Hepatic morphometric evaluation and 4-hydroxyproline content revealed that CCl(4)-induced fibrosis was ameliorated significantly by nilotinib (20mg/kg) and imatinib (20mg/kg). Unlike nilotinib, imatinib (20mg/kg) showed some sort of hepatic injury evidenced by elevation of serum aminotransferases and total bilirubin levels, and hepatic total nitrate/nitrite content, as well as characteristic anisonucleosis visualized with the hematoxylin-eosin staining. In conclusion, this study provides the evidence that nilotinib exerts anti-fibrotic activity and suggests that it may be valuable in the treatment of hepatic fibrosis in humans.
Collapse
Affiliation(s)
- Mohamed E Shaker
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | | | | | | | | |
Collapse
|
49
|
Ogawa S, Ochi T, Shimada H, Inagaki K, Fujita I, Nii A, Moffat MA, Katragadda M, Violand BN, Arch RH, Masferrer JL. Anti-PDGF-B monoclonal antibody reduces liver fibrosis development. Hepatol Res 2010; 40:1128-41. [PMID: 20880061 DOI: 10.1111/j.1872-034x.2010.00718.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AIM To evaluate the usefulness of a platelet-derived growth factor (PDGF)-B specific monoclonal antibody (mAb) as a therapeutic agent to treat chronic liver fibrosis. METHODS Liver fibrosis was induced in ICR mice by bile duct ligation (BDL) or BALB/c mice by weekly injection of concanavalin A (ConA) for 4 or 8 weeks. A mAb specific for mouse and human PDGF-B chain, AbyD3263, was generated, tested in vitro and administered twice a week throughout the experimental period. RESULTS AbyD3263 showed neutralizing activity against mouse and human PDGF-B chain in cell-based assays, as measured in vitro by inhibition of phosphorylation of PDGF receptor β and proliferation of hepatic stellate cells induced by PDGF-BB. The half life of AbyD3263 in mice exceeded 7 days and dosing of animals twice a week resulted in constant plasma levels of the mAb. Induction of liver fibrosis by BDL and ConA resulted in elevated levels of alanine aminotransferase (ALT) in plasma and hydroxyproline in the liver. Treatment with AbyD3263 did not modify ALT levels, but significantly reduced hydroxyproline content in the liver with a maximum reduction of 39% and 54% in the BDL and ConA models, respectively, compared to controls. Conclusios: Consistent with the notion that PDGF-BB plays an important role in the progression of liver fibrosis, AbyD3263 exhibits efficacy in pre-clinical disease models suggesting that pharmacological inhibition of PDGF-B chain may be a therapeutic approach to treat liver fibrosis.
Collapse
Affiliation(s)
- Shinji Ogawa
- Inflammation Research Unit Pharmaceutical Sciences Biotherapeutics CoE, St. Louis Laboratories, Pfizer Global Research and Development Asia Research, Pfizer Worldwide Research and Development, Chesterfield Parkway West, Chesterfield, Missouri, USA Discovery Biology Drug Safety Research & Development, Nagoya Laboratories, Pfizer Global Research and Development, Taketoyo, Aichi, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Thabut D, Shah V. Intrahepatic angiogenesis and sinusoidal remodeling in chronic liver disease: new targets for the treatment of portal hypertension? J Hepatol 2010; 53:976-80. [PMID: 20800926 DOI: 10.1016/j.jhep.2010.07.004] [Citation(s) in RCA: 217] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 07/07/2010] [Accepted: 07/12/2010] [Indexed: 02/06/2023]
Abstract
Portal hypertension accounts for the majority of morbidity and mortality that is encountered in patients with cirrhosis. Portal hypertension is initiated in large part through increases in intrahepatic vascular resistance. Fibrosis, regenerative nodule formation, and intrahepatic vasoconstriction are classical mechanisms that account for increased intrahepatic vascular resistance in cirrhosis. Recent data suggest that intrahepatic angiogenesis and sinusoidal remodeling could also be involved in sinusoidal resistance, fibrosis, and portal hypertension. While angiogenesis is defined as the formation of new vessels deriving from existing ones, sinusoidal remodeling in its pathological form associated with cirrhosis is characterized by increased mural coverage of vessels by contractile HSC. Most attention on the mechanisms of these processes has focused on the liver sinusoidal endothelial cell (SEC), the hepatic stellate cell (HSC), and the paracrine signaling pathways between these two cell types. Interventions that target these vascular structural changes have beneficial effects on portal hypertension and fibrosis in some animal studies which has stimulated interest for pursuing parallel studies in humans with portal hypertension.
Collapse
Affiliation(s)
- Dominique Thabut
- Gastroenterology Research Unit, Advanced Liver Disease Study Group, Fiterman Center for Digestive Diseases, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|