101
|
Zhou L, Chen TW, Zhang XM, Yang Z, Tang HJ, Deng D, Zeng NL, Wang LY, Chen XL, Li H, Li CP, Li L, Xie XY, Hu J. Liver dynamic contrast-enhanced MRI for staging liver fibrosis in a piglet model. J Magn Reson Imaging 2014; 39:872-878. [PMID: 24123400 DOI: 10.1002/jmri.24248] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 05/07/2013] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To determine whether dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) could monitor progression of liver fibrosis in a piglet model, and which DCE-MRI parameter is most accurate for staging this disease. MATERIALS AND METHODS Sixteen piglets were prospectively used to model liver fibrosis and underwent liver DCE-MRI followed by biopsy on the 0, 5th, 9th, 16th, and 21st weekends after modeling of fibrosis. Time of peak (TOP), time to peak (TTP), positive enhancement integral (PEI), maximum slope of increase (MSI), and maximum slope of decrease (MSD) were measured and statistically analyzed for the monitoring and staging. RESULTS As fibrosis progresses, TOP and TTP tended to increase, whereas MSI, MSD, and PEI tended to decrease (all P < 0.05). TOP, TTP, and MSI could discriminate fibrosis stage 0 from 1-4, 0-1 from 2-4, 0-2 from 3-4, and 0-3 from 4; PEI could distinguish the above-mentioned stages except 0-3 from 4; and MSD could distinguish stage 0-3 from 4, and 0 from 1-4 (all P < 0.05). For predicting stage ≥1, ≥2, and ≥3, the area under receiver operating characteristic curve (AUC) of MSI was largest among all parameters; for stage 4 AUC of TTP was largest. CONCLUSION DCE-MRI has the potential to dynamically stage progression of liver fibrosis.
Collapse
Affiliation(s)
- Li Zhou
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Deb A, Ubil E. Cardiac fibroblast in development and wound healing. J Mol Cell Cardiol 2014; 70:47-55. [PMID: 24625635 DOI: 10.1016/j.yjmcc.2014.02.017] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 02/27/2014] [Accepted: 02/28/2014] [Indexed: 01/14/2023]
Abstract
Cardiac fibroblasts are the most abundant cell type in the mammalian heart and comprise approximately two-thirds of the total number of cardiac cell types. During development, epicardial cells undergo epithelial-mesenchymal-transition to generate cardiac fibroblasts that subsequently migrate into the developing myocardium to become resident cardiac fibroblasts. Fibroblasts form a structural scaffold for the attachment of cardiac cell types during development, express growth factors and cytokines and regulate proliferation of embryonic cardiomyocytes. In post natal life, cardiac fibroblasts play a critical role in orchestrating an injury response. Fibroblast activation and proliferation early after cardiac injury are critical for maintaining cardiac integrity and function, while the persistence of fibroblasts long after injury leads to chronic scarring and adverse ventricular remodeling. In this review, we discuss the physiologic function of the fibroblast during cardiac development and wound healing, molecular mediators of activation that could be possible targets for drug development for fibrosis and finally the use of reprogramming technologies for reversing scar. This article is part of a Special Issue entitled "Myocyte-Fibroblast Signalling in Myocardium."
Collapse
Affiliation(s)
- Arjun Deb
- Division of Cardiology, Department of Medicine, Cardiovascular Research Laboratory, David Geffen School of Medicine at University of California, Los Angeles, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at University of California, Los Angeles, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California, Los Angeles, USA; Molecular Biology Institute, David Geffen School of Medicine at University of California, Los Angeles, USA; Program in Molecular Cellular & Integrative Physiology, David Geffen School of Medicine at University of California, Los Angeles, USA.
| | - Eric Ubil
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| |
Collapse
|
103
|
Wang XR, Lu YH. Role of Wnt/β-catenin pathway in activation and proliferation of hepatic stellate cells. Shijie Huaren Xiaohua Zazhi 2014; 22:609-614. [DOI: 10.11569/wcjd.v22.i5.609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis refers to a pathological state in which a variety of pathogenic factors lead to hepatocyte inflammation and necrosis, the imbalance between degradation and deposition of collagen and other extracellular matrix (ECM) molecules, and the abnormal proliferation of liver connective tissue. In the process of the formation of hepatic fibrosis, hepatic stellate cells are the major contributor. Activated hepatic stellate cells secrete extracellular matrix components, and excessive deposition of extracellular matrix is the central event in liver fibrosis. Currently, there have been many articles reporting that the Wnt/β-catenin signaling pathway is involved in organ fibrosis and liver fibrosis, but the underlying mechanism has not been clearly clarified. Some studies have also confirmed that there is a certain relationship between the Wnt/β-catenin pathway and activation of hepatic stellate cells in liver fibrosis.
Collapse
|
104
|
Protection effect of kallistatin on carbon tetrachloride-induced liver fibrosis in rats via antioxidative stress. PLoS One 2014; 9:e88498. [PMID: 24558397 PMCID: PMC3928242 DOI: 10.1371/journal.pone.0088498] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/06/2014] [Indexed: 12/24/2022] Open
Abstract
Prolonged inflammation and oxidative stress are emerging as key causes of pathological wound healing and the development of liver fibrosis. We have investigated the effects of recombinant human kallistatin, produced in Pichia. pastoris, on preventing carbon tetrachloride (CCl4)-induced liver fibrosis in rats. Daily administration of kallistatin prevented development of CCl4-induced liver fibrosis, which was evidenced by histological study. In all kallistatin treated rats, activation of hepatic stellate cells (HSC) as assessed by s-smooth muscle actin staining was attenuated, TGF- β1 expression was inhibited, class I serum biomarkers associated with the process of fibrogenesis, such as hyaluronic acid, laminin, and procollagen III, were lowered, compared with that in the model control group. Furthermore, residual hepatic functional reserve was improved by kallistatin treatment. CCl4 induced elevation of malondialdehyde level and reduced superoxide dismutase activity in the liver, while kallistatin reduced these oxidative parameters. We also investigated the effects of kallistatin on rat primary HSC and LX-2, the human HSC cell line. Kallistatin scavenged H2O2-induced ROS in the LX-2 cells, and suppressed the activation of primary HSC. These results suggest recombinant human kallistatin might be a promising drug candidate for therapeutic intervention of liver fibrosis.
Collapse
|
105
|
Kong XY, Nesset CK, Damme M, Løberg EM, Lübke T, Mæhlen J, Andersson KB, Lorenzo PI, Roos N, Thoresen GH, Rustan AC, Kase ET, Eskild W. Loss of lysosomal membrane protein NCU-G1 in mice results in spontaneous liver fibrosis with accumulation of lipofuscin and iron in Kupffer cells. Dis Model Mech 2014; 7:351-62. [PMID: 24487409 PMCID: PMC3944495 DOI: 10.1242/dmm.014050] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human kidney predominant protein, NCU-G1, is a highly conserved protein with an unknown biological function. Initially described as a nuclear protein, it was later shown to be a bona fide lysosomal integral membrane protein. To gain insight into the physiological function of NCU-G1, mice with no detectable expression of this gene were created using a gene-trap strategy, and Ncu-g1gt/gt mice were successfully characterized. Lysosomal disorders are mainly caused by lack of or malfunctioning of proteins in the endosomal-lysosomal pathway. The clinical symptoms vary, but often include liver dysfunction. Persistent liver damage activates fibrogenesis and, if unremedied, eventually leads to liver fibrosis/cirrhosis and death. We demonstrate that the disruption of Ncu-g1 results in spontaneous liver fibrosis in mice as the predominant phenotype. Evidence for an increased rate of hepatic cell death, oxidative stress and active fibrogenesis were detected in Ncu-g1gt/gt liver. In addition to collagen deposition, microscopic examination of liver sections revealed accumulation of autofluorescent lipofuscin and iron in Ncu-g1gt/gt Kupffer cells. Because only a few transgenic mouse models have been identified with chronic liver injury and spontaneous liver fibrosis development, we propose that the Ncu-g1gt/gt mouse could be a valuable new tool in the development of novel treatments for the attenuation of fibrosis due to chronic liver damage.
Collapse
Affiliation(s)
- Xiang Y Kong
- Department of Bioscience, University of Oslo, 0316 Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Oikawa H, Maesawa C, Tatemichi Y, Nishinari Y, Nishiya M, Mizugai H, Ikeda A, Oikawa K, Takikawa Y, Masuda T. A disintegrin and metalloproteinase 17 (ADAM17) mediates epidermal growth factor receptor transactivation by angiotensin II on hepatic stellate cells. Life Sci 2014; 97:137-44. [PMID: 24412389 DOI: 10.1016/j.lfs.2013.12.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 12/02/2013] [Accepted: 12/18/2013] [Indexed: 02/04/2023]
Abstract
AIMS Epidermal growth factor receptor (EGFR) transactivation induced by angiotensin II (Ang II) participates in the progression of various diseases. A disintegrin and metalloproteinase 17 (ADAM17) is thought to promote renal fibrosis, cardiac hypertrophy with fibrosis and atherosclerosis by activation of the EGFR through secretion of EGFR ligands. The purpose of this study was to investigate whether Ang II-induced EGFR transactivation occurs on hepatic stellate cells (HSCs) and whether the reaction is mediated via ADAM17. MAIN METHODS Ang II-induced EGFR transactivation and cellular proliferation of the human HSC line LI90 were investigated using Western blotting and ATP assay, respectively. Ang II-induced secretion of mature amphiregulin into the cell culture medium was evaluated by enzyme-linked immunosorbent assay (ELISA). KEY FINDINGS An inhibitor of ADAM17, TAPI-1, as well as antagonists of EGFR and angiotensin II type-1 receptor (AT1), attenuated Ang II-induced EGFR transactivation and proliferation of LI90 cells. Furthermore, silencing of ADAM17 inhibited Ang II-induced secretion of mature amphiregulin in addition to EGFR transactivation. SIGNIFICANCE These results indicate that ADAM17 mediates Ang II-induced EGFR transactivation on HSCs, and that this process may participate in the progression of liver fibrosis.
Collapse
Affiliation(s)
- Hiroki Oikawa
- Department of Pathology, School of Medicine, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-Cho, Shiwa-Gun, Iwate 028-3694, Japan.
| | - Chihaya Maesawa
- Department of Pathology, School of Medicine, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-Cho, Shiwa-Gun, Iwate 028-3694, Japan; Department of Tumor Biology, Institute of Biomedical Science, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-Cho, Shiwa-Gun, Iwate 028-3694, Japan
| | - Yoshinori Tatemichi
- Department of Internal Medicine, School of Medicine, Iwate Medical University, Uchimaru 19-1, Morioka, Iwate 020-8505, Japan
| | - Yutaka Nishinari
- Department of Tumor Biology, Institute of Biomedical Science, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-Cho, Shiwa-Gun, Iwate 028-3694, Japan; Department of Surgery, School of Medicine, Iwate Medical University, Uchimaru 19-1, Morioka, Iwate 020-8505, Japan
| | - Masao Nishiya
- Department of Pathology, School of Medicine, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-Cho, Shiwa-Gun, Iwate 028-3694, Japan
| | - Hisata Mizugai
- Department of Pathology, School of Medicine, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-Cho, Shiwa-Gun, Iwate 028-3694, Japan; Department of Internal Medicine, School of Medicine, Iwate Medical University, Uchimaru 19-1, Morioka, Iwate 020-8505, Japan
| | - Aya Ikeda
- Department of Pathology, School of Medicine, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-Cho, Shiwa-Gun, Iwate 028-3694, Japan
| | - Kanta Oikawa
- Department of Internal Medicine, School of Medicine, Iwate Medical University, Uchimaru 19-1, Morioka, Iwate 020-8505, Japan
| | - Yasuhiro Takikawa
- Department of Internal Medicine, School of Medicine, Iwate Medical University, Uchimaru 19-1, Morioka, Iwate 020-8505, Japan
| | - Tomoyuki Masuda
- Department of Pathology, School of Medicine, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-Cho, Shiwa-Gun, Iwate 028-3694, Japan
| |
Collapse
|
107
|
Yovchev MI, Xue Y, Shafritz DA, Locker J, Oertel M. Repopulation of the fibrotic/cirrhotic rat liver by transplanted hepatic stem/progenitor cells and mature hepatocytes. Hepatology 2014; 59:284-95. [PMID: 23840008 PMCID: PMC4091900 DOI: 10.1002/hep.26615] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 06/26/2013] [Indexed: 12/14/2022]
Abstract
UNLABELLED Considerable progress has been made in developing antifibrotic agents and other strategies to treat liver fibrosis; however, significant long-term restoration of functional liver mass has not yet been achieved. Therefore, we investigated whether transplanted hepatic stem/progenitor cells can effectively repopulate the liver with advanced fibrosis/cirrhosis. Stem/progenitor cells derived from fetal livers or mature hepatocytes from DPPIV(+) F344 rats were transplanted into DPPIV(-) rats with thioacetamide (TAA)-induced fibrosis/cirrhosis; rats were sacrificed 1, 2, or 4 months later. Liver tissues were analyzed by histochemistry, hydroxyproline determination, reverse-transcription polymerase chain reaction (RT-PCR), and immunohistochemistry. After chronic TAA administration, DPPIV(-) F344 rats exhibited progressive fibrosis, cirrhosis, and severe hepatocyte damage. Besides stellate cell activation, increased numbers of stem/progenitor cells (Dlk-1(+), AFP(+), CD133(+), Sox-9(+), FoxJ1(+)) were observed. In conjunction with partial hepatectomy (PH), transplanted stem/progenitor cells engrafted, proliferated competitively compared to host hepatocytes, differentiated into hepatocytic and biliary epithelial cells, and generated new liver mass with extensive long-term liver repopulation (40.8 ± 10.3%). Remarkably, more than 20% liver repopulation was achieved in the absence of PH, associated with reduced fibrogenic activity (e.g., expression of alpha smooth muscle actin, platelet-derived growth factor receptor β, desmin, vimentin, tissue inhibitor of metalloproteinase-1) and fibrosis (reduced collagen). Furthermore, hepatocytes can also replace liver mass with advanced fibrosis/cirrhosis, but to a lesser extent than fetal liver stem/progenitor cells. CONCLUSION This study is a proof of principle demonstration that transplanted epithelial stem/progenitor cells can restore injured parenchyma in a liver environment with advanced fibrosis/cirrhosis and exhibit antifibrotic effects.
Collapse
Affiliation(s)
- Mladen I. Yovchev
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine of Yeshiva University,Dept. of Medicine (Division of Gastroenterology & Liver Diseases), Albert Einstein College of Medicine of Yeshiva University
| | - Yuhua Xue
- Dept. of Pathology (Division of Experimental Pathology), University of Pittsburgh
| | - David A. Shafritz
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine of Yeshiva University,Dept. of Medicine (Division of Gastroenterology & Liver Diseases), Albert Einstein College of Medicine of Yeshiva University
| | - Joseph Locker
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine of Yeshiva University,Department of Pathology, Albert Einstein College of Medicine of Yeshiva University,Dept. of Pathology (Division of Experimental Pathology), University of Pittsburgh
| | - Michael Oertel
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine of Yeshiva University,Dept. of Medicine (Division of Gastroenterology & Liver Diseases), Albert Einstein College of Medicine of Yeshiva University,Dept. of Pathology (Division of Experimental Pathology), University of Pittsburgh,McGowan Institute for Regenerative Medicine, University of Pittsburgh,Corresponding author: Michael Oertel, Ph.D.; University of Pittsburgh; School of Medicine; Dept. of Pathology; 200 Lothrop Street – BST S-404; Pittsburgh, PA 15261; Telephone: 412-648-9727; Fax: 412-648-1916;
| |
Collapse
|
108
|
Iwaisako K, Taura K, Koyama Y, Takemoto K, Asagiri M. Strategies to Detect Hepatic Myofibroblasts in Liver Cirrhosis of Different Etiologies. CURRENT PATHOBIOLOGY REPORTS 2014; 2:209-215. [PMID: 25401051 PMCID: PMC4223535 DOI: 10.1007/s40139-014-0057-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Liver cirrhosis, a late stage of hepatic fibrosis, is an increasing cause of morbidity and mortality worldwide. Hepatic fibrosis is mainly caused by alcoholic or non-alcoholic steatohepatitis, chronic viral hepatitis, or autoimmune and biliary diseases. Myofibroblasts, which are absent from the normal liver, are differentiated from heterogeneous cell populations in response to a liver injury of any etiology and produce the extracellular matrix. Hepatic stellate cells are considered the main source of myofibroblasts. However, the origin of hepatic myofibroblasts remains unresolved, and despite considerable research, only a limited success has been achieved by existing anti-fibrotic therapies. The question remains whether these limitations are caused by lack of attention to the critical targets, the myofibroblasts derived from cells of other mesenchymal origins. Therefore, identifying the origin of myofibroblasts may provide insight into the mechanisms underlying liver fibrosis, and may lead to the development of more effective therapies. This review will examine our current strategies for detecting hepatic myofibroblasts of different origins.
Collapse
Affiliation(s)
- Keiko Iwaisako
- Department of Target Therapy Oncology, Kyoto University Graduate School of Medicine, 54 Kawaharacho, Shogoin, Sakyo-Ku, Kyoto, 606-8507 Japan
| | - Kojiro Taura
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507 Japan
| | - Yukinori Koyama
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507 Japan
| | - Kenji Takemoto
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507 Japan
- Innovation Center for Immunoregulation and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507 Japan
| | - Masataka Asagiri
- Innovation Center for Immunoregulation and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507 Japan
| |
Collapse
|
109
|
de Franchis R, Dell’Era A. Pre-primary and Primary Prophylaxis of Variceal Hemorrhage. VARICEAL HEMORRHAGE 2014. [PMCID: PMC7121476 DOI: 10.1007/978-1-4939-0002-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Variceal hemorrhage is a life-threatening complication of portal hypertension. Thus, prevention of variceal formation (pre-primary prophylaxis) or at least prevention of variceal bleeding are important goals to improve life quality and—if possible—survival of patients with liver cirrhosis. Interruption of the underlying cause of liver disease is the most successful approach, which, however, often fails. For this situation interruption or modulation of different pathophysiological mechanisms leading to fibrosis, hyperdynamic circulation and portal hypertension have been shown effective in animal models. But few could be translated to humans. By contrast, different steps to prevent first bleeding from varices have proven successful in many clinical trials. These applied mainly drugs to lower portal pressure, such as nonselective β-blockers, or endoscopic obliteration of varices, while prophylactic shunt procedures are not advised.
Collapse
Affiliation(s)
| | - Alessandra Dell’Era
- Ospedale Universitario Luigi Sacco, Universitá degli Studi di Milano, UOC Gastroenterologia, Milano, Italy
| |
Collapse
|
110
|
Kramann R, DiRocco DP, Humphreys BD. Understanding the origin, activation and regulation of matrix-producing myofibroblasts for treatment of fibrotic disease. J Pathol 2013; 231:273-89. [PMID: 24006178 DOI: 10.1002/path.4253] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 08/26/2013] [Indexed: 12/19/2022]
Abstract
Fibrosis and scar formation results from chronic progressive injury in virtually every tissue and affects a growing number of people around the world. Myofibroblasts drive fibrosis, and recent work has demonstrated that mesenchymal cells, including pericytes and perivascular fibroblasts, are their main progenitors. Understanding the cellular mechanisms of pericyte/fibroblast-to-myofibroblast transition, myofibroblast proliferation and the key signalling pathways that regulate these processes is essential to develop novel targeted therapeutics for the growing patient population suffering from solid organ fibrosis. In this review, we summarize the current knowledge about different progenitor cells of myofibroblasts, discuss major pathways that regulate their transdifferentiation and discuss the current status of novel targeted anti-fibrotic therapeutics in development.
Collapse
Affiliation(s)
- Rafael Kramann
- Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; RWTH Aachen University, Division of Nephrology, Aachen, Germany
| | | | | |
Collapse
|
111
|
Zhou L, Chen TW, Zhang XM, Li CJ, Yang ZF, Zeng NL, Wang LY, Li T, Wang D, Li J, Li CP, Li L, Xie XY. Spleen dynamic contrast-enhanced magnetic resonance imaging as a new method for staging liver fibrosis in a piglet model. PLoS One 2013; 8:e83697. [PMID: 24376732 PMCID: PMC3869810 DOI: 10.1371/journal.pone.0083697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 11/11/2013] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To explore spleen hemodynamic alteration in liver fibrosis with dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), and to determine how to stage liver fibrosis with spleen DCE-MRI parameters. MATERIALS AND METHODS Sixteen piglets were prospectively used to model liver fibrosis staged by liver biopsy, and underwent spleen DCE-MRI on 0, 5th, 9th, 16th and 21st weekend after modeling this disease. DCE-MRI parameters including time to peak (TTP), positive enhancement integral (PEI), maximum slope of increase (MSI) and maximum slope of decrease (MSD) of spleen were measured, and statistically analyzed to stage this disease. RESULTS Spearman's rank correlation tests showed that TTP tended to increase with increasing stages of liver fibrosis (r = 0.647, P<0.001), and that PEI tended to decrease from stage 0 to 4 (r = -0.709, P<0.001). MSD increased slightly from stage 0 to 2 (P>0.05), and decreased from stage 2 to 4 (P<0.05). MSI increased from stage 0 to 1, and decreased from stage 1 to 4 (all P>0.05). Mann-Whitney tests demonstrated that TTP and PEI could classify fibrosis between stage 0 and 1-4, between 0-1 and 2-4, between 0-2 and 3-4, or between 0-3 and 4 (all P<0.01). MSD could discriminate between 0-2 and 3-4 (P = 0.006), or between 0-3 and 4 (P = 0.012). MSI could not differentiate between any two stages. Receiver operating characteristic analysis illustrated that area under receiver operating characteristic curve (AUC) of TTP was larger than of PEI for classifying stage ≥1 and ≥2 (AUC = 0.851 and 0.783, respectively). PEI could best classify stage ≥3 and 4 (AUC = 0.903 and 0.96, respectively). CONCLUSION Spleen DCE-MRI has potential to monitor spleen hemodynamic alteration and classify liver fibrosis stages.
Collapse
Affiliation(s)
- Li Zhou
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Tian-wu Chen
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiao-ming Zhang
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Cheng-jun Li
- Department of Anatomy, and Morphometric Research Laboratory, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zhen-feng Yang
- Department of Radiology, Third Affiliated Hospital of Xinxiang Medical College, Xinxiang, Henan, China
| | - Nan-lin Zeng
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Li-ying Wang
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Ting Li
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Dan Wang
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Li
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Chun-ping Li
- Sichuan Key Laboratory of Medical Imaging, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Li Li
- Department of Pathology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xian-yong Xie
- Department of Pathology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
112
|
Deng G, Huang XJ, Luo HW, Huang FZ, Liu XY, Wang YH. Amelioration of carbon tetrachloride-induced cirrhosis and portal hypertension in rat using adenoviral gene transfer of Akt. World J Gastroenterol 2013; 19:7778-7787. [PMID: 24431897 PMCID: PMC3837279 DOI: 10.3748/wjg.v19.i43.7778] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/28/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether a virus constitutively expressing active Akt is useful to prevent cirrhosis induced by carbon tetrachloride (CCl4).
METHODS: Using cre-loxp technique, we created an Ad-myr-HA-Akt virus, in which Akt is labeled by a HA tag and its expression is driven by myr promoter. Further, through measuring enzyme levels and histological structure, we determined the efficacy of this Ad-myr-HA-Akt virus in inhibiting the development of cirrhosis induced by CCl4 in rats. Lastly, using western blotting, we examined the expression levels and/or phosphorylation status of Akt, apoptotic mediators, endothelial nitric oxide synthase (eNOS), and markers for hepatic stellate cells activation to understand the underlying mechanisms of protective role of this virus.
RESULTS: The Ad-myr-HA-Akt virus was confirmed using polymerase chain reaction amplification of inserted Akt gene and sequencing for full length of inserted fragment, which was consistent with the sequence reported in the GenBank. The concentrations of Ad-myr-HA-Akt and adenoviral enhanced green fluorescent protein (Ad-EGFP) virus used in the current study were 5.5 × 1011 vp/mL. The portal vein diameter, peak velocity of blood flow, portal blood flow and congestion index were significantly increased in untreated, saline and Ad-EGFP cirrhosis groups when compared to normal control after the virus was introduced to animal through tail veil injection. In contrast, these parameters in the Akt cirrhosis group were comparable to normal control group. Compared to the normal control, the liver function (Alanine aminotransferase, Aspartate aminotransferase and Albumin) was significantly impaired in the untreated, saline and Ad-EGFP cirrhosis groups. The Akt cirrhosis group showed significant improvement of liver function when compared to the untreated, saline and Ad-EGFP cirrhosis groups. The Hyp level and portal vein pressure in Akt cirrhosis groups were also significantly lower than other cirrhosis groups. The results of HE and Van Gieson staining indicated that Akt group has better preservation of histological structure and less fibrosis than other cirrhosis groups. The percentage of apoptotic cell was greatly less in Akt cirrhosis group than in other cirrhosis groups. Akt group showed positive HA tag and an increased level of phosphorylated Akt as well as decreased levels of Fas. In contrast, Caspase-3 and Caspase-9 levels in Akt group were significantly lower than other cirrhosis groups. Noticeable decrease of DR5 and α-SMA and increase of phosphorylated eNOS were observed in the Akt group when compared to other cirrhosis groups. The NO level in liver was significantly higher in Akt group than other cirrhosis groups, which was consistent with the level of phosphorylated eNOS in these groups.
CONCLUSION: This study suggest that Ad-myr-HA-Akt virus is a useful tool to prevent CCl4-induced cirrhosis in rat model and Akt pathway may be a therapeutic target for human cirrhosis.
Collapse
|
113
|
Brenner DA. Reversibility of liver fibrosis. Gastroenterol Hepatol (N Y) 2013; 9:737-739. [PMID: 24764791 PMCID: PMC3995196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Affiliation(s)
- David A Brenner
- Vice Chancellor for Health Sciences Dean of the School of Medicine University of California, San Diego San Diego, California
| |
Collapse
|
114
|
Angiotensin II facilitates fibrogenic effect of TGF-β1 through enhancing the down-regulation of BAMBI caused by LPS: a new pro-fibrotic mechanism of angiotensin II. PLoS One 2013; 8:e76289. [PMID: 24155898 PMCID: PMC3796560 DOI: 10.1371/journal.pone.0076289] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 08/22/2013] [Indexed: 02/06/2023] Open
Abstract
Angiotensin II has progressively been considered to play an important role in the development of liver fibrosis, although the mechanism isn't fully understood. The aim of this study was to investigate a possible pro-fibrotic mechanism, by which angiotensin II would enhance the pro-fibrotic effect of transforming growth factor beta 1 (TGF-β1) through up-regulation of toll-like receptor 4 (TLR4) and enhancing down-regulation of TGF-β1 inhibitory pseudo-receptor-BAMBI caused by LPS in hepatic stellate cells (HSCs). Firstly, the synergistic effects of angiotensin II, TGF-β1 and LPS on collagen 1α production were confirmed in vitro by ELISA, in which angiotensin II, LPS and TGF-β1 were treated sequentially, and in vivo by immunofluorescence, in the experiments single or multiple intra-peritoneally implanted osmotic mini-pumps administrating angiotensin II or LPS combined with intra-peritoneal injections of TGF-β1 were used. We also found that only LPS and TGF-β1 weren't enough to induce obvious fibrogenesis without angiotensin II. Secondly, to identify the reason of why angiotensin II is so important, the minute level of TLR4 in activated HSCs - T6 and primary quiescent HSCs of rat, up-regulation of TLR4 by angiotensin II and blockage by different angiotensin II receptor type 1 (AT1) blockers in HSCs were assayed by western blotting in vitro and immunofluorescence in vivo. Finally, BAMBI expression level, which is regulated by LPS-TLR4 pathway, was detected by qRT-PCR and results showed angiotensin II enhanced the down-regulation of BAMBI mRNA caused by LPS in vitro and in vivo, and TLR4 neutralization antibody blocked this interactive effect. These data demonstrated that angiotensin II enhances LPS-TLR4 pathway signaling and further down-regulates expression of BAMBI through up-regulation of TLR4, which results in facilitation of pro-fibrotic activity of TGF-β1. Angiotensin II, LPS and TGF-β1 act synergistically during hepatic fibrogenesis, showing crosstalks between angiotensin II-AT1, LPS-TLR4 and TGF-β1-BAMBI signal pathways in rat HSCs.
Collapse
|
115
|
Liedtke C, Luedde T, Sauerbruch T, Scholten D, Streetz K, Tacke F, Tolba R, Trautwein C, Trebicka J, Weiskirchen R. Experimental liver fibrosis research: update on animal models, legal issues and translational aspects. FIBROGENESIS & TISSUE REPAIR 2013; 6:19. [PMID: 24274743 PMCID: PMC3850878 DOI: 10.1186/1755-1536-6-19] [Citation(s) in RCA: 251] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 09/11/2013] [Indexed: 12/13/2022]
Abstract
Liver fibrosis is defined as excessive extracellular matrix deposition and is based on complex interactions between matrix-producing hepatic stellate cells and an abundance of liver-resident and infiltrating cells. Investigation of these processes requires in vitro and in vivo experimental work in animals. However, the use of animals in translational research will be increasingly challenged, at least in countries of the European Union, because of the adoption of new animal welfare rules in 2013. These rules will create an urgent need for optimized standard operating procedures regarding animal experimentation and improved international communication in the liver fibrosis community. This review gives an update on current animal models, techniques and underlying pathomechanisms with the aim of fostering a critical discussion of the limitations and potential of up-to-date animal experimentation. We discuss potential complications in experimental liver fibrosis and provide examples of how the findings of studies in which these models are used can be translated to human disease and therapy. In this review, we want to motivate the international community to design more standardized animal models which might help to address the legally requested replacement, refinement and reduction of animals in fibrosis research.
Collapse
Affiliation(s)
- Christian Liedtke
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Tom Luedde
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Tilman Sauerbruch
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - David Scholten
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Konrad Streetz
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - René Tolba
- Institute of Laboratory Animal Science, RWTH University Hospital Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Ralf Weiskirchen
- Institute of Clinical Chemistry and Pathobiochemistry, RWTH University Hospital Aachen, Aachen D-52074, Germany
| |
Collapse
|
116
|
Gobejishvili L, Barve S, Breitkopf-Heinlein K, Li Y, Zhang J, Avila DV, Dooley S, McClain CJ. Rolipram attenuates bile duct ligation-induced liver injury in rats: a potential pathogenic role of PDE4. J Pharmacol Exp Ther 2013; 347:80-90. [PMID: 23887098 PMCID: PMC3781411 DOI: 10.1124/jpet.113.204933] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 07/18/2013] [Indexed: 12/19/2022] Open
Abstract
Anti-inflammatory and antifibrotic effects of the broad spectrum phosphodiesterase (PDE) inhibitor pentoxifylline have suggested an important role for cyclic nucleotides in the pathogenesis of hepatic fibrosis; however, studies examining the role of specific PDEs are lacking. Endotoxemia and Toll-like receptor 4 (TLR4)-mediated inflammatory and profibrotic signaling play a major role in the development of hepatic fibrosis. Because cAMP-specific PDE4 critically regulates lipopolysaccharide (LPS)-TLR4-induced inflammatory cytokine expression, its pathogenic role in bile duct ligation-induced hepatic injury and fibrogenesis in Sprague-Dawley rats was examined. Initiation of cholestatic liver injury and fibrosis was accompanied by a significant induction of PDE4A, B, and D expression and activity. Treatment with the PDE4-specific inhibitor rolipram significantly decreased liver PDE4 activity, hepatic inflammatory and profibrotic cytokine expression, injury, and fibrosis. At the cellular level, in relevance to endotoxemia and inflammatory cytokine production, PDE4B was observed to play a major regulatory role in the LPS-inducible tumor necrosis factor (TNF) production by isolated Kupffer cells. Moreover, PDE4 expression was also involved in the in vitro activation and transdifferentiation of isolated hepatic stellate cells (HSCs). Particularly, PDE4A, B, and D upregulation preceded induction of the HSC activation marker α-smooth muscle actin (α-SMA). In vitro treatment of HSCs with rolipram effectively attenuated α-SMA, collagen expression, and accompanying morphologic changes. Overall, these data strongly suggest that upregulation of PDE4 expression during cholestatic liver injury plays a potential pathogenic role in the development of inflammation, injury, and fibrosis.
Collapse
Affiliation(s)
- Leila Gobejishvili
- Department of Internal Medicine (L.G., S.B., J.Z., D.V.A., C.J.M.) and Department of Pharmacology and Toxicology (S.B., D.V.A., C.J.M.), University of Louisville, Louisville, Kentucky; Department of Surgery, University of Louisville Medical Center, Louisville, Kentucky (Y.L.); VA Medical Center, Louisville, Kentucky (C.J.M.); University of Louisville Alcohol Research Center Louisville, Kentucky (L.G., S.B., J.Z., D.V.A., C.J.M.); and Department of Medicine II, Section Molecular Hepatology-Alcohol Associated Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (K.B.-H., S.D.)
| | | | | | | | | | | | | | | |
Collapse
|
117
|
Fausther M, Lavoie EG, Dranoff JA. Contribution of Myofibroblasts of Different Origins to Liver Fibrosis. CURRENT PATHOBIOLOGY REPORTS 2013; 1:225-230. [PMID: 23997993 DOI: 10.1007/s40139-013-0020-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The most common cause of liver failure is cirrhosis, due to progressive liver fibrosis and other architectural changes in the liver. Fibrosis occurs after liver injury or stress and results directly from an imbalance between the processes of extracellular matrix synthesis (fibrogenesis) and degradation (fibrolysis). Although research studies have identified several promising targets at the molecular level, current therapies to prevent and treat hepatic fibrosis in patients have only shown limited success. It is well established that liver myofibroblasts are the primary effector cells responsible for the extensive extracellular matrix accumulation and scar formation observed during hepatic fibrosis, in both clinical and experimental settings. Thus, as the major fibrogenic cells implicated in wound healing and tissue repair response, liver myofibroblasts could represent excellent targets for antifibrotic therapies. Still, the exact natures and identities of liver myofibroblasts precursors have yet to be resolved, and their relative contribution to hepatic fibrosis to be determined. The goal of this review is to examine the relative importance of liver myofibroblast precursors in the pathogenesis of liver fibrosis.
Collapse
Affiliation(s)
- Michel Fausther
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock AR 72205, USA
| | | | | |
Collapse
|
118
|
Liu X, Xu J, Brenner DA, Kisseleva T. Reversibility of Liver Fibrosis and Inactivation of Fibrogenic Myofibroblasts. CURRENT PATHOBIOLOGY REPORTS 2013; 1:209-214. [PMID: 24000319 DOI: 10.1007/s40139-013-0018-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many studies have demonstrated that hepatic fibrosis is reversible. Regression of liver fibrosis is associated with resorption of fibrous scar and disappearance of collagen producing myofibroblasts. The fate of these myofibroblasts has been recently revealed: Some myofibroblasts undergo senescence and apoptose during reversal of fibrosis, while other myofibroblasts revert to a quiescent-like phenotype. Inactivation of myofibroblasts is a newly described phenomenon1 which now requires mechanistic investigation. Understanding of the mechanism of HSC inactivation upon cessation of fibrogenic stimuli may identify new approaches to revert already existing aHSCs/myofibroblasts into a quiescent-like state. This review summarizes the research on the inactivation of hepatic myofibroblasts.
Collapse
Affiliation(s)
- Xiao Liu
- Dept. of Medicine, University of California, San Diego, CA, USA
| | | | | | | |
Collapse
|
119
|
Wedelolactone exhibits anti-fibrotic effects on human hepatic stellate cell line LX-2. Eur J Pharmacol 2013; 714:105-11. [PMID: 23791612 DOI: 10.1016/j.ejphar.2013.06.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 05/31/2013] [Accepted: 06/08/2013] [Indexed: 01/07/2023]
Abstract
Wedelolactone is a major coumarin of Eclipta prostrata, which is used for preventing liver damage. However the effects of wedelolactone on hepatic fibrosis remained unexplored. The purpose of this study was to demonstrate the anti-fibrotic effects of wedelolactone on activated human hepatic stellate cell (HSC) line LX-2 and the possible underlying mechanisms by means of MTT assay, Hoechst staining, as well as real-time quantitative PCR and western blot. The results showed that wedelolactone reduced the cellular viability of LX-2 in a time and dose-dependent manner. After treatment of wedelolactone, the expressions of collagen I and α-smooth muscle actin, two biomarkers of LX-2 activation, were remarkably decreased. The apoptosis of LX-2 cells was induced by wedelolactone accompanied with the decreasing expression of anti-apoptotic Bcl-2 and increasing expression of pro-apoptotic Bax. In addition, phosphorylated status of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) was up-regulated, but not in p38. Moreover, wedelolactone significantly repressed the level of phosphorylated inhibitor of nuclear factor κB (IκB) and p65 in nucleus in spite of tumor necrosis factor-α stimulation. In conclusion, wedelolactone could significantly inhibit the activation of LX-2 cells, the underlying mechanisms of which included inducing Bcl-2 family involved apoptosis, up-regulating phosphorylated status of ERK and JNK expressions, and inhibiting nuclear factor-κB (NF-κB) mediated activity. Wedelolactone might present as a useful tool for the prevention and treatment of hepatic fibrosis.
Collapse
|
120
|
Wuestefeld T, Pesic M, Rudalska R, Dauch D, Longerich T, Kang TW, Yevsa T, Heinzmann F, Hoenicke L, Hohmeyer A, Potapova A, Rittelmeier I, Jarek M, Geffers R, Scharfe M, Klawonn F, Schirmacher P, Malek NP, Ott M, Nordheim A, Vogel A, Manns MP, Zender L. A Direct in vivo RNAi screen identifies MKK4 as a key regulator of liver regeneration. Cell 2013; 153:389-401. [PMID: 23582328 DOI: 10.1016/j.cell.2013.03.026] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 12/27/2012] [Accepted: 03/19/2013] [Indexed: 10/27/2022]
Abstract
The liver harbors a distinct capacity for endogenous regeneration; however, liver regeneration is often impaired in disease and therefore insufficient to compensate for the loss of hepatocytes and organ function. Here we describe a functional genetic approach for the identification of gene targets that can be exploited to increase the regenerative capacity of hepatocytes. Pools of small hairpin RNAs (shRNAs) were directly and stably delivered into mouse livers to screen for genes modulating liver regeneration. Our studies identify the dual-specific kinase MKK4 as a master regulator of liver regeneration. MKK4 silencing robustly increased the regenerative capacity of hepatocytes in mouse models of liver regeneration and acute and chronic liver failure. Mechanistically, induction of MKK7 and a JNK1-dependent activation of the AP1 transcription factor ATF2 and the Ets factor ELK1 are crucial for increased regeneration of hepatocytes with MKK4 silencing.
Collapse
Affiliation(s)
- Torsten Wuestefeld
- Department of Gastroenterology, Hepatology & Endocrinology, Medical School Hannover, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Abstract
PURPOSE OF REVIEW Pathological deposition of fibrous matrix in organs is a major problem and contributes to as many as 45% of all natural deaths. Chronic kidney disease affects 8% of the US population, and is characterized by fibrotic processes. It frequently progresses to organ failure and is a major cause of cardiovascular death; yet it lacks therapies. Understanding the pathological mechanisms of fibrosis in the kidney and other organs is central to the development of new therapeutics. RECENT FINDINGS Pericytes are mesenchymal cells that partially cover capillary walls. Pericytes play critical roles in micro-vessel formation, maturation and stability. New genetic fate-mapping studies have identified pericytes and the closely related resident fibroblasts as the major progenitors of scar-forming myofibroblasts in multiple organs including the kidney, appearing in response to tissue injury. When pericytes become myofibroblasts they lose pericyte functions. Capillaries become unstable with deleterious consequences for the kidney. The cellular and molecular mechanisms underpinning these processes are starting to unravel, leading to new therapeutics for chronic fibrosing diseases of the kidney and potentially other organs. SUMMARY This review focuses on pericytes in the kidney and other organs, their role in fibrogenesis and their role in regulation of the microvasculature.
Collapse
|
122
|
Nowatzky J, Knorr A, Hirth-Dietrich C, Siegling A, Volk HD, Limmer A, Knolle P, Weber O. Inactivated Orf virus (Parapoxvirus ovis) elicits antifibrotic activity in models of liver fibrosis. Hepatol Res 2013; 43:535-46. [PMID: 22971208 DOI: 10.1111/j.1872-034x.2012.01086.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 07/17/2012] [Accepted: 07/30/2012] [Indexed: 02/08/2023]
Abstract
AIM Inactivated Orf virus (ORFV, Parapoxvirus ovis) demonstrates strong antiviral activity in animal models including a human hepatitis B virus (HBV)-transgenic mouse. In addition, expression of interferon (IFN)-γ and interleukin-10 (IL-10) was induced after administration of inactivated ORFV in these mice. IFN-γ and IL-10 are known to elicit antifibrotic activity. We therefore aimed to study antifibrotic activity of inactivated ORFV in models of liver fibrosis. METHODS We characterized ORFV-induced hepatic cytokine expression in rats. We then studied ORFV in two models of liver fibrosis in rats, pig serum-induced liver fibrosis and carbon tetrachloride (CCL4 )-induced liver fibrosis. RESULTS ORFV induced hepatic expression of IFN-γ and IL-10 in rats. ORFV mediated antifibrotic activity when administrated concomitantly with the fibrosis-inducing agents in both models of liver fibrosis. Importantly, when CCL4 -induced liver fibrosis was already established, ORFV application still showed significant antifibrotic activity. In addition, we were able to demonstrate a direct antifibrotic effect of ORFV on stellate cells. CONCLUSION These results establish a potential novel antifibrotic therapeutic approach that not only prevents but also resolves established liver fibrosis. Further studies are required to unravel the details of the mechanisms involved.
Collapse
Affiliation(s)
- Janina Nowatzky
- Bayer HealthCare, Wuppertal; Medical Faculty University of Witten-Herdecke, Witten
| | | | | | | | | | | | | | | |
Collapse
|
123
|
Nunes de Carvalho S, da Cunha Lira D, Costa Cortez EA, de Andrade DC, Thole AA, Stumbo AC, de Carvalho L. Bone marrow cell transplantation is associated with fibrogenic cells apoptosis during hepatic regeneration in cholestatic rats. Biochem Cell Biol 2013; 91:88-94. [DOI: 10.1139/bcb-2012-0045] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Liver fibrosis is accompanied by hepatocyte death and proliferation of α-SMA+ fibrogenic cells (activated hepatic stellate cells and myofibroblasts), which synthesize extracellular matrix components that contribute to disorganization of the hepatic parenchyma and loss of liver function. Therefore, apoptosis of these fibrogenic cells is important to hepatic regeneration. This study aimed to analyze the effect of cell therapy using bone marrow mononuclear cell (BMMNC) transplantation on α-SMA expression and on apoptosis of hepatic cells during liver fibrosis induced by bile duct ligation (BDL). Livers were collected from normal rats, fibrotic rats after 14 and 21 days of BDL, and rats that received BMMNC at 14 days of BDL and were analyzed after 7 days. Apoptosis in fibrogenic cells was analyzed by immunoperoxidase, confocal microscopy, and Western blotting, and liver regeneration was assessed by proliferating cell nuclear antigen staining. Results showed that caspase-3 and proliferating cell nuclear antigen expression were significantly increased in the BMMNC-treated group. Additionally, confocal microscopy analysis showed cells coexpressing α-SMA and caspase-3 in these animals, suggesting fibrogenic cell death. These results suggest a novel role for BMMNC in liver regeneration during fibrotic disease by stimulating fibrogenic cells apoptosis and hepatocyte proliferation, probably through secretion of specific cytokines that modulate the hepatic microenvironment toward an antifibrogenic balance.
Collapse
Affiliation(s)
- Simone Nunes de Carvalho
- Laboratório Cultura de Células, Departamento de Histologia e Embriologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, UERJ, Av. Prof. Manoel de Abreu 444, 3° andar, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Dalvaci da Cunha Lira
- Laboratório Cultura de Células, Departamento de Histologia e Embriologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, UERJ, Av. Prof. Manoel de Abreu 444, 3° andar, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Erika Afonso Costa Cortez
- Laboratório Cultura de Células, Departamento de Histologia e Embriologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, UERJ, Av. Prof. Manoel de Abreu 444, 3° andar, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Daniela Caldas de Andrade
- Laboratório Cultura de Células, Departamento de Histologia e Embriologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, UERJ, Av. Prof. Manoel de Abreu 444, 3° andar, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Alessandra Alves Thole
- Laboratório Cultura de Células, Departamento de Histologia e Embriologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, UERJ, Av. Prof. Manoel de Abreu 444, 3° andar, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Ana Carolina Stumbo
- Laboratório Cultura de Células, Departamento de Histologia e Embriologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, UERJ, Av. Prof. Manoel de Abreu 444, 3° andar, 20550-170 Rio de Janeiro, RJ, Brazil
| | - Lais de Carvalho
- Laboratório Cultura de Células, Departamento de Histologia e Embriologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, UERJ, Av. Prof. Manoel de Abreu 444, 3° andar, 20550-170 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
124
|
Nilotinib induces apoptosis and autophagic cell death of activated hepatic stellate cells via inhibition of histone deacetylases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1992-2003. [PMID: 23499874 DOI: 10.1016/j.bbamcr.2013.02.033] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 02/07/2013] [Accepted: 02/27/2013] [Indexed: 12/21/2022]
Abstract
Increasing hepatic stellate cell (HSC) death is a very attractive approach for limiting liver fibrosis. Tyrosine kinase inhibitors have been shown to have anti-fibrotic properties, but the mechanisms are poorly understood. Here, we identified the mechanism of action of the second-generation tyrosine kinase inhibitor nilotinib in inducing HSC death. Human HSC line (LX-2) and rat HSCs were treated with nilotinib and its predecessor, imatinib, in the absence or presence of various blockers, known to interfere with death signaling pathways. Nilotinib, but not imatinib, induced progressive cell death of activated, but not quiescent, HSCs in a dose-dependent manner. Activated HSCs died through apoptosis, as denoted by increased DNA fragmentation and caspase activation, and through autophagy, as indicated by the accumulation of autophagic markers, light chain (LC)3A-II and LC3B-II. Although inhibition of caspases with Z-VAD-FMK suppressed nilotinib-induced HSCs' apoptosis, there was no increase in HSCs' survival, because autophagy was exacerbated. However, blocking the mitochondrial permeability transition pore (mPTP) opening with cyclosporin A completely abolished both apoptosis and autophagy due to nilotinib. Moreover, nilotinib treatment decreased the protein expression of histone deacetylases 1, 2 and 4. Interestingly, pretreament with C646, a selective p300/CBP histone acetyl transferase inhibitor, resulted in diverting nilotinib-induced apoptosis and autophagy towards necrosis. In conclusion, the identification of mPTP as a target of nilotinib in activated HSCs suggests coordination with histone deacetylases inhibition to induce apoptosis and autophagy. Thus, our study provides novel insights into the anti-fibrotic effects of nilotinib.
Collapse
|
125
|
Cong M, Liu T, Wang P, Fan X, Yang A, Bai Y, Peng Z, Wu P, Tong X, Chen J, Li H, Cong R, Tang S, Wang B, Jia J, You H. Antifibrotic effects of a recombinant adeno-associated virus carrying small interfering RNA targeting TIMP-1 in rat liver fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1607-16. [PMID: 23474083 DOI: 10.1016/j.ajpath.2013.01.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 01/08/2013] [Accepted: 01/10/2013] [Indexed: 12/13/2022]
Abstract
Elevated tissue inhibitor of metalloproteinase 1 (TIMP-1) expression contributes to excess production of extracellular matrix in liver fibrosis. Herein, we constructed a recombinant adeno-associated virus (rAAV) carrying siRNA of the TIMP-1 gene (rAAV/siRNA-TIMP-1) and investigated its effects on liver fibrosis in rats. Two models of rat liver fibrosis, the carbon tetrachloride and bile duct ligation models, were treated with rAAV/siRNA-TIMP-1. In the carbon tetrachloride model, rAAV/siRNA-TIMP-1 administration attenuated fibrosis severity, as determined by histologic analysis of hepatic collagen accumulation, hydroxyproline content, and concentrations of types I and III collagen in livers and sera. Levels of mRNA and active matrix metalloproteinase (MMP) 13 were elevated, whereas levels of mRNA and active MMP-2 were decreased. Moreover, a marked decrease was noted in the expression of α-smooth muscle actin, a biomarker of activated hepatic stellate cells (HSCs), and transforming growth factor-β1, critical for the development of liver fibrosis. Similarly, rAAV/siRNA-TIMP-1 treatment significantly alleviated bile duct ligation-induced liver fibrosis. Furthermore, this treatment dramatically suppressed TIMP-1 expression in HSCs from both model rats. These data indicate that the administration of rAAV/siRNA-TIMP-1 attenuated liver fibrosis by directly elevating the function of MMP-13 and diminishing activated HSCs. It also resulted in indirect decreased expression of type I collagen, MMP-2, and transforming growth factor-β1. In conclusion, rAAV/siRNA-TIMP-1 may be an effective antifibrotic gene therapy agent.
Collapse
Affiliation(s)
- Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Friedman SL, Sheppard D, Duffield JS, Violette S. Therapy for Fibrotic Diseases: Nearing the Starting Line. Sci Transl Med 2013; 5:167sr1. [DOI: 10.1126/scitranslmed.3004700] [Citation(s) in RCA: 480] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
127
|
Van Beneden K, Mannaerts I, Pauwels M, Van den Branden C, van Grunsven LA. HDAC inhibitors in experimental liver and kidney fibrosis. FIBROGENESIS & TISSUE REPAIR 2013; 6:1. [PMID: 23281659 PMCID: PMC3564760 DOI: 10.1186/1755-1536-6-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 11/29/2012] [Indexed: 02/08/2023]
Abstract
Histone deacetylase (HDAC) inhibitors have been extensively studied in experimental models of cancer, where their inhibition of deacetylation has been proven to regulate cell survival, proliferation, differentiation and apoptosis. This in turn has led to the use of a variety of HDAC inhibitors in clinical trials. In recent years the applicability of HDAC inhibitors in other areas of disease has been explored, including the treatment of fibrotic disorders. Impaired wound healing involves the continuous deposition and cross-linking of extracellular matrix governed by myofibroblasts leading to diseases such as liver and kidney fibrosis; both diseases have high unmet medical needs which are a burden on health budgets worldwide. We provide an overview of the potential use of HDAC inhibitors against liver and kidney fibrosis using the current understanding of these inhibitors in experimental animal models and in vitro models of fibrosis.
Collapse
Affiliation(s)
- Katrien Van Beneden
- Department of Human Anatomy, Liver Cell Biology Lab, Vrije Universiteit Brussel, Brussels, Belgium
| | - Inge Mannaerts
- Department of Cell Biology, Liver Cell Biology Lab, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marina Pauwels
- Department of Human Anatomy, Liver Cell Biology Lab, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Leo A van Grunsven
- Department of Cell Biology, Liver Cell Biology Lab, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
128
|
Strategies for anti-fibrotic therapies. Biochim Biophys Acta Mol Basis Dis 2012; 1832:1088-103. [PMID: 23266403 DOI: 10.1016/j.bbadis.2012.12.007] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/07/2012] [Accepted: 12/08/2012] [Indexed: 02/07/2023]
Abstract
The fibrotic diseases encompass a wide spectrum of entities including such multisystemic diseases as systemic sclerosis, nephrogenic systemic fibrosis and sclerodermatous graft versus host disease, as well as organ-specific disorders such as pulmonary, liver, and kidney fibrosis. Collectively, given the wide variety of affected organs, the chronic nature of the fibrotic processes, and the large number of individuals suffering their devastating effects, these diseases pose one of the most serious health problems in current medicine and a serious economic burden to society. Despite these considerations there is currently no accepted effective treatment. However, remarkable progress has been achieved in the elucidation of their pathogenesis including the identification of the critical role of myofibroblasts and the determination of molecular mechanisms that result in the transcriptional activation of the genes responsible for the fibrotic process. Here we review the origin of the myofibroblast and discuss the crucial regulatory pathways involving multiple growth factors and cytokines that participate in the pathogenesis of the fibrotic process. Potentially effective therapeutic strategies based upon this new information are considered in detail and the major challenges that remain and their possible solutions are presented. It is expected that translational efforts devoted to convert this new knowledge into novel and effective anti-fibrotic drugs will be forthcoming in the near future. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
|
129
|
Wahl K, Rosenberg W, Vaske B, Manns MP, Schulze-Osthoff K, Bahr MJ, Bantel H. Biopsy-controlled liver fibrosis staging using the enhanced liver fibrosis (ELF) score compared to transient elastography. PLoS One 2012; 7:e51906. [PMID: 23284811 PMCID: PMC3526479 DOI: 10.1371/journal.pone.0051906] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 11/07/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND AIMS Chronic liver diseases are characterized by inflammatory and fibrotic liver injuries that often result in liver cirrhosis with its associated complications such as portal hypertension and hepatocellular carcinoma. Liver biopsy still represents the reference standard for fibrosis staging, although transient elastography is increasingly used for non-invasive monitoring of fibrosis progression. However, this method is not generally available and is associated with technical limitations emphasizing the need for serological biomarkers staging of liver fibrosis. The enhanced liver fibrosis (ELF) score was shown to accurately predict significant liver fibrosis in different liver diseases, although extracellular matrix components detected by this score may not only mirror the extent of liver fibrosis but also inflammatory processes. METHODS In this prospective biopsy-controlled study we evaluated the utility of the ELF score in comparison to transient elastography to predict different stages of fibrosis in 102 patients with chronic liver diseases. RESULTS Both techniques revealed similar area under receiver operating characteristic curve values for prediction of advanced fibrosis stages. Compared to transient elastography, the ELF score showed a broader overlap between low and moderate fibrosis stages and a stronger correlation with inflammatory liver injury. CONCLUSIONS Both the ELF score as well as transient elastography allowed for high quality fibrosis staging. However, the ELF score was less discriminative in low and moderate fibrosis stages and appeared more strongly influenced by inflammatory liver injury. This should be considered when making clinical interpretations on the basis of ELF score values.
Collapse
Affiliation(s)
- Kristin Wahl
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - William Rosenberg
- Centre for Hepatology, University College London Institute of Liver and Digestive Health, Division of Medicine, University College London, London, United Kingdom
| | - Bernhard Vaske
- Institute of Biometry, Hannover Medical School, Hannover, Germany
| | - Michael P. Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | | | - Heike Bantel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
130
|
Zhang DW, Zhao YX, Wei D, Li YL, Zhang Y, Wu J, Xu J, Chen C, Tang H, Zhang W, Gong L, Han Y, Chen ZN, Bian H. HAb18G/CD147 promotes activation of hepatic stellate cells and is a target for antibody therapy of liver fibrosis. J Hepatol 2012; 57:1283-91. [PMID: 22878468 DOI: 10.1016/j.jhep.2012.07.042] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 07/23/2012] [Accepted: 07/27/2012] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Activated hepatic stellate cells (HSCs) located in the Disse's space play a crucial role in liver fibrosis. HAb18G/CD147, a tumor-related glycoprotein, is highly expressed in hepatocellular carcinoma cells and fibroblasts. Whether HAb18G/CD147 plays an important role in the hepatic fibrogenesis is unknown. METHODS Immunohistochemistry for HAb18G/CD147 and α-smooth muscle actin expression in diseased liver tissues was used for correlation analysis. The function of HAb18G/CD147 in fibrogenesis was evaluated with the human HSCs LX-2 cell line and carbon tetrachloride-induced mouse liver fibrosis model. The specific antibody HAb18 targeting HAb18G/CD147 was injected intravenously into the mouse to investigate whether HAb18G/CD147 could be a potential target for liver fibrosis treatment. RESULTS HAb18G/CD147 is highly expressed on activated HSCs in the sinusoid. The positive rates of HAb18G/CD147 expression in human HBV-related liver cirrhosis, liver biopsy with HBV and liver adjacent to hemangioma were 95.6% (65/68), 14.8% (8/54) and 6.4% (8/125), respectively. HAb18G/CD147 expression was significantly correlated with the Child-Pugh grade (r=0.2848, p=0.0186) and with the expression of α-smooth muscle actin in HSCs (r=0.4434, p=0.0002) in liver cirrhosis. Transforming growth factor-β1 upregulated HAb18G/CD147 expression in LX-2 cells. Transfection of HAb18G/CD147 promoted the profibrogenic genes expression. In mouse liver fibrosis model, HAb18G/CD147 expression increased with the development of fibrogenesis and decreased during the liver fibrosis spontaneous recovery. The HAb18 targeting HAb18G/CD147 could attenuate liver fibrosis. CONCLUSIONS These data suggest that HAb18G/CD147 plays a role in HSC activation and is a potential therapeutic target in fibrosis/cirrhosis.
Collapse
Affiliation(s)
- Da-Wei Zhang
- Cell Engineering Research Center and Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Weber O, Mercer AA, Friebe A, Knolle P, Volk HD. Therapeutic immunomodulation using a virus--the potential of inactivated orf virus. Eur J Clin Microbiol Infect Dis 2012; 32:451-60. [PMID: 23179251 DOI: 10.1007/s10096-012-1780-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 11/07/2012] [Indexed: 12/15/2022]
Abstract
Viruses can manipulate the immune response against them by various strategies to influence immune cells, i.e. by over-activation leading to functional inactivation, bypassing antigen presentation or even suppression of effector functions. Little is known, however, about how these features of immune regulation and modulation could be used for therapeutic purposes. Reasons for this include the complexity of immune regulatory mechanisms under certain disease conditions and the risks that infections with viruses pose to human beings. The orf virus (ORFV), a member of the Parapoxvirus genus of the poxvirus family, is known as a common pathogen in sheep and goats worldwide. The inactivated ORFV, however, has been used as a preventative as well as therapeutic immunomodulator in veterinary medicine in different species. Here, we review the key results obtained in pre-clinical studies or clinical studies in veterinary medicine to characterise the therapeutic potential of inactivated ORFV. Inactivated ORFV has strong effects on cytokine secretion in mice and human immune cells, leading to an auto-regulated loop of initial up-regulation of inflammatory and Th1-related cytokines, followed by Th2-related cytokines that attenuate immunopathology. The therapeutic potential of inactivated ORFV has been recognised in several difficult-to-treat disease areas, such as chronic viral diseases, liver fibrosis or various forms of cancer. Further research will be required in order to evaluate the full beneficial potential of inactivated ORFV for therapeutic immunomodulation.
Collapse
Affiliation(s)
- O Weber
- Bayer Pharmaceuticals Global Drug Discovery, Bayer HealthCare AG, Leverkusen, Germany.
| | | | | | | | | |
Collapse
|
132
|
Li R, Xu L, Liang T, Li Y, Zhang S, Duan X. Puerarin mediates hepatoprotection against CCl4-induced hepatic fibrosis rats through attenuation of inflammation response and amelioration of metabolic function. Food Chem Toxicol 2012; 52:69-75. [PMID: 23146695 DOI: 10.1016/j.fct.2012.10.059] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 10/27/2012] [Accepted: 10/29/2012] [Indexed: 12/12/2022]
Abstract
This study was designed to evaluate the potential effects of puerarin (PR), an effective isoflavonoid compound purified from Pueraria lobata, in treating hepatic fibrosis (HF) rats induced by carbon tetrachloride (CCl(4), 2 mL kg(-1) d(-1)). Compared to model control, PR treatment effectively lowered the serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin (Alb), total protein (TP) in HF rats. Masson stained analysis showed that the condition of HF rats was mitigated. Meanwhile, the tumor necrosis factor alpha (TNF-α), nuclear factor-kappa B (NF-κB) expressions were significantly down-regulated at protein level by PR intervention. Additionally, the activity of superoxide dismutase (SOD) was elevated, while the content of malondialdehyde (MDA) was lessened in liver tissue. As revealed by immunohistochemistry assay, PR therapy resulted in reduced production of transforming growth factor-βl (TGF-βl). Moreover, it also was attributed to decreased mRNA level of inducible nitric oxide synthase (iNOS) using RT-PCR analysis. These findings demonstrate that puerarin successfully reverses hepatotoxicity in CCl(4)-induced HF rats via the underlying mechanisms of regulating serum enzymes and attenuating TNF-α/NF-κB pathway for anti-inflammation response, as well as improving metabolic function in liver tissue.
Collapse
Affiliation(s)
- Rong Li
- Department of Physiology, Guilin Medical University, Guilin, Guangxi 541004, PR China
| | | | | | | | | | | |
Collapse
|
133
|
Sohrabpour AA, Mohamadnejad M, Malekzadeh R. Review article: the reversibility of cirrhosis. Aliment Pharmacol Ther 2012; 36:824-832. [PMID: 22966946 DOI: 10.1111/apt.12044] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 04/18/2012] [Accepted: 08/22/2012] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cirrhosis is the end result of many types of chronic liver diseases. Recent developments in the understanding of the process of hepatic fibrogenesis have revealed that the process is a dynamic one and a capacity for recovery from any degree of fibrosis including those associated with cirrhosis is plausible. AIM To review current evidence of histopathological reversibility following drug therapy of more common aetiologies of cirrhosis. METHODS A PubMed search was performed and the evidence for histopathological regression of advanced fibrosis/cirrhosis following drug therapy was reviewed as of the end of February 2012. RESULTS There is abundant clinical evidence in support of the idea of the reversibility of cirrhosis in patients with different aetiologies of advanced hepatic disease including viral, autoimmune and metabolic/infiltrative liver disease. CONCLUSIONS The concept of cirrhosis has changed from being a form of static and irreversible entity to a dynamic and reversible diseases stage. Novel therapeutic strategies are under investigation to target specific steps in the process of fibrogenesis with the aim of reversing advanced fibrosis/cirrhosis.
Collapse
Affiliation(s)
- A A Sohrabpour
- Digestive Disease Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
134
|
Woolbright BL, Jaeschke H. Novel insight into mechanisms of cholestatic liver injury. World J Gastroenterol 2012; 18:4985-93. [PMID: 23049206 PMCID: PMC3460324 DOI: 10.3748/wjg.v18.i36.4985] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 04/12/2012] [Accepted: 04/20/2012] [Indexed: 02/06/2023] Open
Abstract
Cholestasis results in a buildup of bile acids in serum and in hepatocytes. Early studies into the mechanisms of cholestatic liver injury strongly implicated bile acid-induced apoptosis as the major cause of hepatocellular injury. Recent work has focused both on the role of bile acids in cell signaling as well as the role of sterile inflammation in the pathophysiology. Advances in modern analytical methodology have allowed for more accurate measuring of bile acid concentrations in serum, liver, and bile to very low levels of detection. Interestingly, toxic bile acid levels are seemingly far lower than previously hypothesized. The initial hypothesis has been based largely upon the exposure of μmol/L concentrations of toxic bile acids and bile salts to primary hepatocytes in cell culture, the possibility that in vivo bile acid concentrations may be far lower than the observed in vitro toxicity has far reaching implications in the mechanism of injury. This review will focus on both how different bile acids and different bile acid concentrations can affect hepatocytes during cholestasis, and additionally provide insight into how these data support recent hypotheses that cholestatic liver injury may not occur through direct bile acid-induced apoptosis, but may involve largely inflammatory cell-mediated liver cell necrosis.
Collapse
|
135
|
Pratap A, Singh S, Mundra V, Yang N, Panakanti R, Eason JD, Mahato RI. Attenuation of early liver fibrosis by pharmacological inhibition of smoothened receptor signaling. J Drug Target 2012; 20:770-82. [PMID: 22994359 DOI: 10.3109/1061186x.2012.719900] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hedgehog (Hh) signaling is involved in the pathogenesis of liver fibrosis. It has been previously shown that Hh-inhibitor cyclopamine (CYA) can reduce liver fibrosis in rats. However, CYA is not stable in vivo, which limits its clinical application. This study compares the antifibrotic potential of two known Hh antagonists, vismodegib (GDC-0449, abbreviated to GDC) and CYA. GDC is a synthetic molecule presently in clinical cancer trials and has been reported to be safe and efficacious. These drugs attenuated early liver fibrosis in common bile duct ligated rats, improved liver function, and prevented hepatic stellate cell (HSC) activation, thereby suppressing epithelial to mesenchymal transition (EMT). While both CYA and GDC increased the number of proliferating cell nuclear antigen positive liver cells in vivo, only CYA increased Caspase-3 expression in HSCs in rat livers, suggesting that while GDC and CYA effectively attenuate early liver fibrosis, their hepatoprotective effects may be mediated through different modes of action. Thus, GDC has the potential to serve as a new therapeutic agent for treating early liver fibrosis.
Collapse
Affiliation(s)
- Akshay Pratap
- Division of Solid Organ Transplantation, Methodist University Hospital, Memphis, TN, USA
| | | | | | | | | | | | | |
Collapse
|
136
|
Yao QY, Xu BL, Wang JY, Liu HC, Zhang SC, Tu CT. Inhibition by curcumin of multiple sites of the transforming growth factor-beta1 signalling pathway ameliorates the progression of liver fibrosis induced by carbon tetrachloride in rats. Altern Ther Health Med 2012; 12:156. [PMID: 22978413 PMCID: PMC3495222 DOI: 10.1186/1472-6882-12-156] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 09/08/2012] [Indexed: 12/20/2022]
Abstract
Background At present there is no effective and accepted therapy for hepatic fibrosis. Transforming growth factor (TGF)-β1 signaling pathway contributes greatly to hepatic fibrosis. Reducing TGF-β synthesis or inhibiting components of its complex signaling pathway represent important therapeutic targets. The aim of the study was to investigate the effect of curcumin on liver fibrosis and whether curcumin attenuates the TGF-β1 signaling pathway. Methods Sprague–Dawley rat was induced liver fibrosis by carbon tetrachloride (CCl4) for six weeks together with or without curcumin, and hepatic histopathology and collagen content were employed to quantify liver necro-inflammation and fibrosis. Moreover, the mRNA and protein expression levels of TGF-β1, Smad2, phosphorylated Smad2, Smad3, Smad7 and connective tissue growth factor (CTGF) were determined by quantitative real time-PCR, Western blot, or immunohistochemistry. Results Rats treated with curcumin improved liver necro-inflammation, and reduced liver fibrosis in association with decreased α-smooth muscle actin expression, and decreased collagen deposition. Furthermore, curcumin significantly attenuated expressions of TGFβ1, Smad2, phosphorylated Smad2, Smad3, and CTGF and induced expression of the Smad7. Conclusions Curcumin significantly attenuated the severity of CCl4-induced liver inflammation and fibrosis through inhibition of TGF-β1/Smad signalling pathway and CTGF expression. These data suggest that curcumin might be an effective antifibrotic drug in the prevention of liver disease progression.
Collapse
|
137
|
Insel PA, Murray F, Yokoyama U, Romano S, Yun H, Brown L, Snead A, Lu D, Aroonsakool N. cAMP and Epac in the regulation of tissue fibrosis. Br J Pharmacol 2012; 166:447-56. [PMID: 22233238 DOI: 10.1111/j.1476-5381.2012.01847.x] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fibrosis, the result of excess deposition of extracellular matrix (ECM), in particular collagen, leads to scarring and loss of function in tissues that include the heart, lung, kidney and liver. The second messenger cAMP can inhibit the formation and extent of ECM during this late phase of inflammation, but the mechanisms for these actions of cAMP and of agents that elevate tissue cAMP levels are not well understood. In this article, we review the fibrotic process and focus on two recently recognized aspects of actions of cAMP and its effector Epac (Exchange protein activated by cAMP): (a) blunting of epithelial-mesenchymal transformation (EMT) and (b) down-regulation of Epac expression by profibrotic agents (e.g. TGF-β, angiotensin II), which may promote tissue fibrosis by decreasing Epac-mediated antifibrotic actions. Pharmacological approaches that raise cAMP or blunt the decrease in Epac expression by profibrotic agents may thus be strategies to block or perhaps reverse tissue fibrosis. LINKED ARTICLES This article is part of a themed section on Novel cAMP Signalling Paradigms. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.166.issue-2.
Collapse
Affiliation(s)
- Paul A Insel
- Departments of Pharmacology Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Odena G, Bataller R. Actin-binding proteins as molecular targets to modulate hepatic stellate cell proliferation. Focus on "MARCKS actin-binding capacity mediates actin filament assembly during mitosis in human hepatic stellate cells". Am J Physiol Cell Physiol 2012; 303:C355-6. [PMID: 22744005 DOI: 10.1152/ajpcell.00192.2012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
139
|
|
140
|
Ohyama T, Sato K, Kishimoto K, Yamazaki Y, Horiguchi N, Ichikawa T, Kakizaki S, Takagi H, Izumi T, Mori M. Azelnidipine is a calcium blocker that attenuates liver fibrosis and may increase antioxidant defence. Br J Pharmacol 2012; 165:1173-1187. [PMID: 21790536 PMCID: PMC3346246 DOI: 10.1111/j.1476-5381.2011.01599.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2010] [Revised: 06/20/2011] [Accepted: 07/11/2011] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Oxidative stress plays a critical role in liver fibrogenesis. Reactive oxygen species (ROS) stimulate hepatic stellate cells (HSCs), and ROS-mediated increases in calcium influx further increase ROS production. Azelnidipine is a calcium blocker that has been shown to have antioxidant effects in endothelial cells and cardiomyocytes. Therefore, we evaluated the anti-fibrotic and antioxidative effects of azelnidipine on liver fibrosis. EXPERIMENTAL APPROACH We used TGF-β1-activated LX-2 cells (a human HSC line) and mouse models of fibrosis induced by treatment with either carbon tetrachloride (CCl(4) ) or thioacetamide (TAA). KEY RESULTS Azelnidipine inhibited TGF-β1 and angiotensin II (Ang II)-activated α1(I) collagen mRNA expression in HSCs. Furthermore, TGF-β1- and Ang II-induced oxidative stress and TGF-β1-induced p38 and JNK phosphorylation were reduced in HSCs treated with azelnidipine. Azelnidipine significantly decreased inflammatory cell infiltration, pro-fibrotic gene expressions, HSC activation, lipid peroxidation, oxidative DNA damage and fibrosis in the livers of CCl(4) - or TAA-treated mice. Finally, azelnidipine prevented a decrease in the expression of some antioxidant enzymes and accelerated regression of liver fibrosis in CCl(4) -treated mice. CONCLUSIONS AND IMPLICATIONS Azelnidipine inhibited TGF-β1- and Ang II-induced HSC activation in vitro and attenuated CCl(4) - and TAA-induced liver fibrosis, and it accelerated regression of CCl(4) -induced liver fibrosis in mice. The anti-fibrotic mechanism of azelnidipine against CCl(4) -induced liver fibrosis in mice may have been due an increased level of antioxidant defence. As azelnidipine is widely used in clinical practice without serious adverse effects, it may provide an effective new strategy for anti-fibrotic therapy.
Collapse
Affiliation(s)
- T Ohyama
- Departments of Medicine and Molecular Science Biochemistry, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Liu YC, Gu XH. Roles and relation between C/EBPα and PARs in the activation of hepatic stellate cells. Shijie Huaren Xiaohua Zazhi 2011; 19:3656-3660. [DOI: 10.11569/wcjd.v19.i36.3656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Protease activated receptors (PARs) are main components of the fibrotic cascade mediated by the trypsin and thrombin that amplifies liver inflammation and fibrosis. Gene transcription initiation induced by PARs plays an important role in the activation of hepatic stellate cells (HSCs). HSC activation can be inhibited by the expression of transcription factor CCAAT enhancer binding proteins α (C/EBPα). Further research of the relation between C/EBPα and PARs will contribute to the understanding of the pathogenesis of liver fibrosis and provide a theoretical basis for further exploration of anti-fibrotic strategies.
Collapse
|
142
|
Abstract
Chronic liver injuries of different etiologies eventually lead to fibrosis, a scarring process associated with increased and altered deposition of extracellular matrix in the liver. Progression of fibrosis has a major worldwide clinical impact due to the high number of patients affected by chronic liver disease which can lead to severe complications, expensive treatment, a possible need for liver transplantation, and death. Liver fibrogenesis is characterized by activation of hepatic stellate cells and other extracellular matrix producing cells. Liver fibrosis may regress following specific therapeutic interventions. Other than removing agents causing chronic liver damage, no antifibrotic drug is currently available in clinical practice. The extent of liver fibrosis is variable between individuals, even after controlling for exogenous factors. Thus, host genetic factors are considered to play an important role in the process of liver scarring. Until recently it was believed that this process was irreversible. However, emerging experimental and clinical evidence is starting to show that even cirrhosis in its early stages is potentially reversible.
Collapse
Affiliation(s)
- Mona H Ismail
- Department of Internal Medicine, Division of Gastroenterology, King Fahad University Hospital, Al-Khobar, Saudi Arabia
| | - Massimo Pinzani
- Dipartimento di Medicina Interna Center for Research, High Education and Transfer, Università degli Studi di Firenze, Florence, Italy
| |
Collapse
|
143
|
Ismail MH, Pinzani M. Reversal of hepatic fibrosis: pathophysiological basis of antifibrotic therapies. HEPATIC MEDICINE : EVIDENCE AND RESEARCH 2011. [PMID: 24367223 DOI: 10.2147/hmer.s905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Chronic liver injuries of different etiologies eventually lead to fibrosis, a scarring process associated with increased and altered deposition of extracellular matrix in the liver. Progression of fibrosis has a major worldwide clinical impact due to the high number of patients affected by chronic liver disease which can lead to severe complications, expensive treatment, a possible need for liver transplantation, and death. Liver fibrogenesis is characterized by activation of hepatic stellate cells and other extracellular matrix producing cells. Liver fibrosis may regress following specific therapeutic interventions. Other than removing agents causing chronic liver damage, no antifibrotic drug is currently available in clinical practice. The extent of liver fibrosis is variable between individuals, even after controlling for exogenous factors. Thus, host genetic factors are considered to play an important role in the process of liver scarring. Until recently it was believed that this process was irreversible. However, emerging experimental and clinical evidence is starting to show that even cirrhosis in its early stages is potentially reversible.
Collapse
Affiliation(s)
- Mona H Ismail
- Department of Internal Medicine, Division of Gastroenterology, King Fahad University Hospital, Al-Khobar, Saudi Arabia
| | - Massimo Pinzani
- Dipartimento di Medicina Interna Center for Research, High Education and Transfer, Università degli Studi di Firenze, Florence, Italy
| |
Collapse
|