1
|
Sorrentino G. Microenvironmental control of the ductular reaction: balancing repair and disease progression. Cell Death Dis 2025; 16:246. [PMID: 40180915 PMCID: PMC11968979 DOI: 10.1038/s41419-025-07590-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
The ductular reaction (DR) is a dynamic adaptive cellular response within the liver, triggered by various hepatic insults and characterized by an expansion of dysmorphic biliary epithelial cells and liver progenitors. This complex response presents a dual role, playing a pivotal function in liver regeneration but, paradoxically, contributing to the progression of liver diseases, depending upon specific contextual factors and signaling pathways involved. This comprehensive review aims to offer a holistic perspective on the DR, focusing into its intricate cellular and molecular mechanisms, highlighting its pathological significance, and exploring its potential therapeutic implications. An up-to-date understanding of the DR in the context of different liver injuries is provided, analyzing its contributions to liver regeneration, inflammation, fibrosis, and ultimately carcinogenesis. Moreover, the review highlights the role of multiple microenvironmental factors, including the influence of extracellular matrix, tissue mechanics and the interplay with the intricate hepatic cell ecosystem in shaping the DR's regulation. Finally, in vitro and in vivo experimental models of the DR will be discussed, providing insights into how researchers can study and manipulate this critical cellular response. By comprehensively addressing the multifaceted nature of the DR, this review contributes to a more profound understanding of its pathophysiological role in liver diseases, thus offering potential therapeutic avenues for hepatic disorders and improving patient outcomes.
Collapse
Affiliation(s)
- Giovanni Sorrentino
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.
| |
Collapse
|
2
|
Senavirathna T, Shafaei A, Lareu R, Balmer L. Unlocking the Therapeutic Potential of Ellagic Acid for Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis. Antioxidants (Basel) 2024; 13:485. [PMID: 38671932 PMCID: PMC11047720 DOI: 10.3390/antiox13040485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity is in epidemic proportions in many parts of the world, contributing to increasing rates of non-alcoholic fatty liver disease (NAFLD). NAFLD represents a range of conditions from the initial stage of fatty liver to non-alcoholic steatohepatitis (NASH), which can progress to severe fibrosis, through to hepatocellular carcinoma. There currently exists no treatment for the long-term management of NAFLD/NASH, however, dietary interventions have been investigated for the treatment of NASH, including several polyphenolic compounds. Ellagic acid is one such polyphenolic compound. Nutraceutical food abundant in ellagic acid undergoes initial hydrolysis to free ellagic acid within the stomach and small intestine. The proposed mechanism of action of ellagic acid extends beyond its initial therapeutic potential, as it is further broken down by the gut microbiome into urolithin. Both ellagic acid and urolithin have been found to alleviate oxidative stress, inflammation, and fibrosis, which are associated with NAFLD/NASH. While progress has been made in understanding the pharmacological and biological activity of ellagic acid and its involvement in NAFLD/NASH, it has yet to be fully elucidated. Thus, the aim of this review is to summarise the currently available literature elucidating the therapeutic potential of ellagic acid and its microbial-derived metabolite urolithin in NAFLD/NASH.
Collapse
Affiliation(s)
- Tharani Senavirathna
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia;
| | - Armaghan Shafaei
- Centre for Integrative Metabolomics and Computational Biology, School of Science, Edith Cowan University, Perth, WA 6027, Australia;
| | - Ricky Lareu
- Curtin Medical School and Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA 6845, Australia
| | - Lois Balmer
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia;
| |
Collapse
|
3
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:1-95. [DOI: 10.1016/b978-0-7020-8228-3.00001-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Song Y, Lu Z, Shu W, Xiang Z, Wang Z, Wei X, Xu X. Arouse potential stemness: Intrinsic and acquired stem cell therapeutic strategies for advanced liver diseases. CELL INSIGHT 2023; 2:100115. [PMID: 37719773 PMCID: PMC10502372 DOI: 10.1016/j.cellin.2023.100115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 09/19/2023]
Abstract
Liver diseases are a major health issue, and prolonged liver injury always progresses. Advanced liver disorders impair liver regeneration. Millions of patients die yearly worldwide, even with the available treatments of liver transplantation and artificial liver support system. With its abundant cell resources and significant differentiative potential, stem cell therapy is a viable treatment for various disorders and offers hope to patients waiting for orthotopic liver transplantation. Considering such plight, stem cell therapeutic strategies deliver hope to the patients. Moreover, we conclude intrinsic and acquired perspectives based on stem cell sources. The properties and therapeutic uses of these stem cells' specific types or sources were then reviewed. Owing to the recent investigations of the above cells, a safe and effective therapy will emerge for advanced liver diseases soon.
Collapse
Affiliation(s)
- Yisu Song
- Department of Hepatobiliary and Pancreatic Surgery Affiliated Hangzhou First People’s Hospital Zhejiang University School of Medicine Hangzhou, Zhejiang, 310006, China
- Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Zhengyang Lu
- Department of Hepatobiliary and Pancreatic Surgery Affiliated Hangzhou First People’s Hospital Zhejiang University School of Medicine Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
- Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Wenzhi Shu
- Department of Hepatobiliary and Pancreatic Surgery Affiliated Hangzhou First People’s Hospital Zhejiang University School of Medicine Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Ze Xiang
- Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengxin Wang
- Department of General Surgery, Huashan Hospital, Fudan University Shanghai, 200040, China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic Surgery Affiliated Hangzhou First People’s Hospital Zhejiang University School of Medicine Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
5
|
Gromowski T, Lukacs-Kornek V, Cisowski J. Current view of liver cancer cell-of-origin and proposed mechanisms precluding its proper determination. Cancer Cell Int 2023; 23:3. [PMID: 36609378 PMCID: PMC9824961 DOI: 10.1186/s12935-022-02843-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023] Open
Abstract
Hepatocellular carcinoma and intrahepatic cholangiocarcinoma are devastating primary liver cancers with increasing prevalence in many parts of the world. Despite intense investigation, many aspects of their biology are still largely obscure. For example, numerous studies have tackled the question of the cell-of-origin of primary liver cancers using different experimental approaches; they have not, however, provided a clear and undisputed answer. Here, we will review the evidence from animal models supporting the role of all major types of liver epithelial cells: hepatocytes, cholangiocytes, and their common progenitor as liver cancer cell-of-origin. Moreover, we will also propose mechanisms that promote liver cancer cell plasticity (dedifferentiation, transdifferentiation, and epithelial-to-mesenchymal transition) which may contribute to misinterpretation of the results and which make the issue of liver cancer cell-of-origin particularly complex.
Collapse
Affiliation(s)
- Tomasz Gromowski
- grid.5522.00000 0001 2162 9631Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Veronika Lukacs-Kornek
- grid.10388.320000 0001 2240 3300Institute of Experimental Immunology, University Hospital of the Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Jaroslaw Cisowski
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
6
|
Mowla A, Belford R, Köhn-Gaone J, Main N, Tirnitz-Parker JEE, Yeoh GC, Kennedy BF. Biomechanical assessment of chronic liver injury using quantitative micro-elastography. BIOMEDICAL OPTICS EXPRESS 2022; 13:5050-5066. [PMID: 36187256 PMCID: PMC9484444 DOI: 10.1364/boe.467684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 06/16/2023]
Abstract
Hepatocellular carcinoma is one of the most lethal cancers worldwide, causing almost 700,000 deaths annually. It mainly arises from cirrhosis, which, in turn, results from chronic injury to liver cells and corresponding fibrotic changes. Although it is known that chronic liver injury increases the elasticity of liver tissue, the role of increased elasticity of the microenvironment as a possible hepatocarcinogen is yet to be investigated. One reason for this is the paucity of imaging techniques capable of mapping the micro-scale elasticity variation in liver and correlating that with cancerous mechanisms on the cellular scale. The clinical techniques of ultrasound elastography and magnetic resonance elastography typically do not provide micro-scale resolution, while atomic force microscopy can only assess the elasticity of a limited number of cells. We propose quantitative micro-elastography (QME) for mapping the micro-scale elasticity of liver tissue into images known as micro-elastograms, and therefore, as a technique capable of correlating the micro-environment elasticity of tissue with cellular scale cancerous mechanisms in liver. We performed QME on 13 freshly excised healthy and diseased mouse livers and present micro-elastograms, together with co-registered histology, in four representative cases. Our results indicate a significant increase in the mean (×6.3) and standard deviation (×6.0) of elasticity caused by chronic liver injury and demonstrate that the onset and progression of pathological features such as fibrosis, hepatocyte damage, and immune cell infiltration correlate with localized variations in micro-elastograms.
Collapse
Affiliation(s)
- Alireza Mowla
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
| | - Rose Belford
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
| | - Julia Köhn-Gaone
- Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Nathan Main
- Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Janina E. E. Tirnitz-Parker
- Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
- Centre for Medical Research, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - George C. Yeoh
- Centre for Medical Research, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA 6009, Australia
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Brendan F. Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| |
Collapse
|
7
|
Makino Y, Hikita H, Fukumoto K, Sung JH, Sakano Y, Murai K, Sakane S, Kodama T, Sakamori R, Kondo J, Kobayashi S, Tatsumi T, Takehara T. Constitutive Activation of the Tumor Suppressor p53 in Hepatocytes Paradoxically Promotes Non-Cell Autonomous Liver Carcinogenesis. Cancer Res 2022; 82:2860-2873. [PMID: 35696550 PMCID: PMC9379366 DOI: 10.1158/0008-5472.can-21-4390] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/27/2022] [Accepted: 06/08/2022] [Indexed: 01/07/2023]
Abstract
In chronic liver diseases (CLD), p53 is constitutively activated in hepatocytes due to various etiologies as viral infection, ethanol exposure, or lipid accumulation. This study was aimed to clarify the significance of p53 activation on the pathophysiology of CLDs. In Kras-mutant liver cancer model, murine double minute 2 (Mdm2), a negative regulator of p53, was specifically deleted in hepatocytes [Alb-Cre KrasLSL-G12D Mdm2fl/fl (LiKM; KrasG12D mutation and Mdm2 loss in the liver)]. Accumulation of p53 and upregulation of its downstream genes were observed in hepatocytes in LiKM mice. LiKM mice showed liver inflammation accompanied by hepatocyte apoptosis, senescence-associated secretory phenotype (SASP), and the emergence of hepatic progenitor cells (HPC). More importantly, Mdm2 deletion promoted non-cell autonomous development of liver tumors. Organoids generated from HPCs harbored tumor-formation ability when subcutaneously inoculated into NOD/Shi-scid/IL2Rγ (null) mice. Treatment with acyclic retinoid suppressed growth of HPCs in vitro and inhibited tumorigenesis in LiKM mice. All of the phenotypes in LiKM mice, including accelerated liver tumorigenesis, were negated by further deletion of p53 in hepatocytes (Alb-Cre KrasLSL-G12D Mdm2fl/fl p53fl/fl). Activation of hepatic p53 was noted in liver biopsy samples obtained from 182 patients with CLD, in comparison with 23 normal liver samples without background liver diseases. In patients with CLD, activity of hepatic p53 was positively correlated with the expression of apoptosis, SASP, HPC-associated genes and tumor incidence in the liver after biopsy. In conclusion, activation of hepatocyte p53 creates a microenvironment prone to tumor formation from HPCs. Optimization of p53 activity in hepatocytes is important to prevent patients with CLD from hepatocarcinogenesis. SIGNIFICANCE This study reveals that activation of p53 in hepatocytes promotes liver carcinogenesis derived from HPCs, which elucidates a paradoxical aspect of a tumor suppressor p53 and novel mechanism of liver carcinogenesis. See related commentary by Barton and Lozano, p. 2824.
Collapse
Affiliation(s)
- Yuki Makino
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenji Fukumoto
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ji Hyun Sung
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihiro Sakano
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuhiro Murai
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sadatsugu Sakane
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takahiro Kodama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryotaro Sakamori
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jumpei Kondo
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohide Tatsumi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan.,Corresponding Author: Tetsuo Takehara, Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan. Phone: 816-6879-3621; Fax: 816-6879-3629; E-mail:
| |
Collapse
|
8
|
Fu Q, Ohnishi S, Suda G, Sakamoto N. Small-molecule inhibitor cocktail promotes the proliferation of pre-existing liver progenitor cells. Stem Cell Reports 2022; 17:1589-1603. [PMID: 35777357 PMCID: PMC9287679 DOI: 10.1016/j.stemcr.2022.05.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 11/09/2022] Open
Abstract
A recent study showed that a cocktail of three small molecules, Y-27632, A83-01, and CHIR99021 (YAC), converts mature hepatocytes (MHs) into proliferative bipotent cells that can be induced into MHs and cholangiocytes in rats. However, when we reproduced these experiments, it was found that bipotent cells may be derived from resident liver progenitor cells (LPCs), whose proliferative activity was promoted by YAC. A simple and efficient sorting scheme was also developed in this study to harvest high-purity and high-yield LPCs. The inducible bipotency of purified LPCs was verified; in addition, they were found to spontaneously differentiate into hepatocytes and cholangiocytes due to changes in proliferative status even without induction. Moreover, during the differentiation process, some hepatocytes spontaneously reconverted to LPCs under certain conditions, such as the release of contact inhibition. These findings may improve our understanding of LPCs and provide a cell source for regenerative medicine. A small-molecule cocktail promotes rat liver progenitor proliferation in vitro Highly purified progenitors can be simply obtained by their physical properties Purified progenitors preferentially proliferate and then spontaneously differentiate Progenitor differentiation is closely related to varied proliferation signals
Collapse
Affiliation(s)
- Qingjie Fu
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Shunsuke Ohnishi
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan; Laboratory of Molecular and Cellular Medicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan.
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| |
Collapse
|
9
|
Shu Y, Xu Q, Xu Y, Tao Q, Shao M, Cao X, Chen Y, Wu Z, Chen M, Zhou Y, Zhou P, Shi Y, Bu H. Loss of Numb promotes hepatic progenitor expansion and intrahepatic cholangiocarcinoma by enhancing Notch signaling. Cell Death Dis 2021; 12:966. [PMID: 34667161 PMCID: PMC8526591 DOI: 10.1038/s41419-021-04263-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 09/26/2021] [Accepted: 10/06/2021] [Indexed: 02/05/2023]
Abstract
Numb, a stem cell fate determinant, acts as a tumor suppressor and is closely related to a wide variety of malignancies. Intrahepatic cholangiocarcinoma (iCCA) originates from hepatic progenitors (HPCs); however, the role of Numb in HPC malignant transformation and iCCA development is still unclear. A retrospective cohort study indicated that Numb was frequently decreased in tumor tissues and suggests poor prognosis in iCCA patients. Consistently, in a chemically induced iCCA mouse model, Numb was downregulated in tumor cells compared to normal cholangiocytes. In diet-induced chronic liver injury mouse models, Numb ablation significantly promoted histological impairment, HPC expansion, and tumorigenesis. Similarly, Numb silencing in cultured iCCA cells enhanced cell spheroid growth, invasion, metastasis, and the expression of stem cell markers. Mechanistically, Numb was found to bind to the Notch intracellular domain (NICD), and Numb ablation promoted Notch signaling; this effect was reversed when Notch signaling was blocked by γ-secretase inhibitor treatment. Our results suggested that loss of Numb plays an important role in promoting HPC expansion, HPC malignant transformation, and, ultimately, iCCA development in chronically injured livers. Therapies targeting suppressed Numb are promising for the treatment of iCCA.
Collapse
Affiliation(s)
- Yuke Shu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing Xu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yahong Xu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing Tao
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingyang Shao
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoyue Cao
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuwei Chen
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenru Wu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Menglin Chen
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yongjie Zhou
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Transplantation, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Zhou
- Department of Pathology, Sichuan Tumor Hospital, Chengdu, 610041, China
| | - Yujun Shi
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Transplantation, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Hong Bu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
10
|
Khoo T, Lam D, Olynyk JK. Impact of modern antiviral therapy of chronic hepatitis B and C on clinical outcomes of liver disease. World J Gastroenterol 2021; 27:4831-4845. [PMID: 34447229 PMCID: PMC8371504 DOI: 10.3748/wjg.v27.i29.4831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/14/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic infections with the hepatitis B and C viruses have significant worldwide health and economic impacts. Previous treatments for hepatitis C such as interferon and ribavirin therapy were ineffective and poorly tolerated by patients. The introduction of directly acting curative antiviral therapy for hepatitis C and the wider use of nucleos(t)ide analogues for suppression of chronic Hepatitis B infection have resulted in many positive developments. Decreasing the prevalence of hepatitis B and C have concurrently reduced transmission rates and hence, the number of new infections. Antiviral treatments have decreased the rates of liver decompensation and as a result, lowered hospitalisation and mortality rates for both chronic hepatitis B and C infection. The quality of life of chronically infected patients has also been improved significantly by modern treatment. Antiviral therapy has stopped the progression of liver disease to cirrhosis in certain patient cohorts and prevented ongoing hepatocellular damage in patients with existing cirrhosis. Longer term benefits of antiviral therapy include a reduced risk of developing hepatocellular carcinoma and decreased number of patients requiring liver transplantation. This review article assesses the literature and summarises the impact of modern antiviral therapy of chronic hepatitis B and C on clinical outcomes from liver disease.
Collapse
Affiliation(s)
- Tiffany Khoo
- Department of Gastroenterology and Hepatology, Fiona Stanley Hospital, Murdoch 6150, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
| | - Danielle Lam
- Department of Gastroenterology and Hepatology, Fiona Stanley Hospital, Murdoch 6150, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
| | - John K Olynyk
- Department of Gastroenterology and Hepatology, Fiona Stanley Hospital, Murdoch 6150, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
| |
Collapse
|
11
|
Kim A, Wu X, Allende DS, Nagy LE. Gene Deconvolution Reveals Aberrant Liver Regeneration and Immune Cell Infiltration in Alcohol-Associated Hepatitis. Hepatology 2021; 74:987-1002. [PMID: 33619773 PMCID: PMC8475730 DOI: 10.1002/hep.31759] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/23/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Acute liver damage causes hepatocyte stress and death, but in chronic liver disease impaired hepatocyte regeneration and immune cell infiltration prevents recovery. While the roles of both impaired liver regeneration and immune infiltration have been studied extensively in chronic liver diseases, the differential contribution of these factors is difficult to assess. APPROACH AND RESULTS We combined single-cell RNA-sequencing (RNA-seq) data from healthy livers and peripheral immune cells to measure cell proportions in chronic liver diseases. Using bulk RNA-seq data from patients with early alcohol-associated hepatitis, severe AH (sAH), HCV, HCV with cirrhosis, and NAFLD, we performed gene deconvolution to predict the contribution of different cell types in each disease. Patients with sAH had the greatest change in cell composition, with increases in both periportal hepatocytes and cholangiocyte populations. Interestingly, while central vein hepatocytes were decreased, central vein endothelial cells were expanded. Endothelial cells are thought to regulate liver regeneration through WNT signaling. WNT2, important in central vein hepatocyte development, was down in sAH, while multiple other WNTs and WNT receptors were up-regulated. Immunohistochemistry revealed up-regulation of FZD6, a noncanonical WNT receptor, in hepatocytes in sAH. Immune cell populations also differed in disease. In sAH, a specific group of inflammatory macrophages was increased and distinct from the macrophage population in patients with HCV. Network and correlation analyses revealed that changes in the cell types in the liver were highly correlated with clinical liver function tests. CONCLUSIONS These results identify distinct changes in the liver cell populations in chronic liver disease and illustrate the power of using single-cell RNA-seq data from a limited number of samples in understanding multiple different diseases.
Collapse
Affiliation(s)
- Adam Kim
- Northern Ohio Alcohol Center, Center for Liver Disease Research, Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Xiaoqin Wu
- Northern Ohio Alcohol Center, Center for Liver Disease Research, Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Laura E. Nagy
- Northern Ohio Alcohol Center, Center for Liver Disease Research, Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, United States
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
12
|
Gratte FD, Pasic S, Abu Bakar NDB, Gogoi-Tiwari J, Liu X, Carlessi R, Kisseleva T, Brenner DA, Ramm GA, Olynyk JK, Tirnitz-Parker JEE. Previous liver regeneration induces fibro-protective mechanisms during thioacetamide-induced chronic liver injury. Int J Biochem Cell Biol 2021; 134:105933. [PMID: 33540107 DOI: 10.1016/j.biocel.2021.105933] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 12/29/2022]
Abstract
Chronic liver injury is characterised by continuous or repeated epithelial cell loss and inflammation. Hepatic wound healing involves matrix deposition through activated hepatic stellate cells (HSCs) and the expansion of closely associated Ductular Reactions and liver progenitor cells (LPCs), which are thought to give rise to new epithelial cells. In this study, we used the murine thioacetamide (TAA) model to reliably mimic these injury and regeneration dynamics and assess the impact of a recovery phase on subsequent liver injury and fibrosis. Age-matched naïve or 6-week TAA-treated/4-week recovered mice (C57BL/6 J, n = 5-9) were administered TAA for six weeks (C57BL/6 J, n = 5-9). Sera and liver tissues were harvested at key time points to assess liver injury biochemically, by real-time PCR for fibrotic mediators, Sirius Red staining and hydroxyproline assessment for collagen deposition as well as immunofluorescence for inflammatory, HSC and LPC markers. In addition, primary HSCs and the HSC cell line LX-2 were co-cultured with the well-characterised LPC line BMOL and analysed for potential changes in expression of fibrogenic mediators. Our data demonstrate that recovery from a previous TAA insult, with LPCs still present on day 0 of the second treatment, led to a reduced TAA-induced disease progression with less severe fibrosis than in naïve TAA-treated animals. Importantly, primary activated HSCs significantly reduced pro-fibrogenic gene expression when co-cultured with LPCs. Taken together, previous TAA injury established a fibro-protective molecular and cellular microenvironment. Our proof-of principle HSC/LPC co-culture data demonstrate that LPCs communicate with HSCs to regulate fibrogenesis, highlighting a key role for LPCs as regulatory cells during chronic liver disease.
Collapse
Affiliation(s)
- Francis D Gratte
- School of Veterinary and Life Sciences, Murdoch University, Perth, WA, Australia; Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia.
| | - Sara Pasic
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia.
| | - N Dianah B Abu Bakar
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia.
| | - Jully Gogoi-Tiwari
- School of Veterinary and Life Sciences, Murdoch University, Perth, WA, Australia; Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia.
| | - Xiao Liu
- Department of Surgery, University of California, San Diego, La Jolla, CA, USA.
| | - Rodrigo Carlessi
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia.
| | - Tatiana Kisseleva
- Department of Surgery, University of California, San Diego, La Jolla, CA, USA.
| | - David A Brenner
- School of Medicine, University of California, San Diego, La Jolla, CA, USA.
| | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; The University of Queensland, Brisbane, QLD, Australia.
| | - John K Olynyk
- Fiona Stanley and Fremantle Hospital Group, Perth, WA, Australia; School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia.
| | - Janina E E Tirnitz-Parker
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia.
| |
Collapse
|
13
|
Frohlich J, Vinciguerra M. Candidate rejuvenating factor GDF11 and tissue fibrosis: friend or foe? GeroScience 2020; 42:1475-1498. [PMID: 33025411 PMCID: PMC7732895 DOI: 10.1007/s11357-020-00279-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Growth differentiation factor 11 (GDF11 or bone morphogenetic protein 11, BMP11) belongs to the transforming growth factor-β superfamily and is closely related to other family member-myostatin (also known as GDF8). GDF11 was firstly identified in 2004 due to its ability to rejuvenate the function of multiple organs in old mice. However, in the past few years, the heralded rejuvenating effects of GDF11 have been seriously questioned by many studies that do not support the idea that restoring levels of GDF11 in aging improves overall organ structure and function. Moreover, with increasing controversies, several other studies described the involvement of GDF11 in fibrotic processes in various organ setups. This review paper focuses on the GDF11 and its pro- or anti-fibrotic actions in major organs and tissues, with the goal to summarize our knowledge on its emerging role in regulating the progression of fibrosis in different pathological conditions, and to guide upcoming research efforts.
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.
- Institute for Liver and Digestive Health, Division of Medicine, University College London (UCL), London, UK.
| |
Collapse
|
14
|
Lim HK, Jeffrey GP, Ramm GA, Soekmadji C. Pathogenesis of Viral Hepatitis-Induced Chronic Liver Disease: Role of Extracellular Vesicles. Front Cell Infect Microbiol 2020; 10:587628. [PMID: 33240824 PMCID: PMC7683521 DOI: 10.3389/fcimb.2020.587628] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles are encapsulated lipid nanoparticles secreted by a variety of cell types in living organisms. They are known to carry proteins, metabolites, nucleic acids, and lipids as their cargoes and are important mediators of intercellular communication. The role of extracellular vesicles in chronic liver disease has been reported. Chronic liver disease such as viral hepatitis accounts for a significant mortality and morbidity burden worldwide. Hepatic fibrosis has been commonly associated with the chronic form of viral hepatitis, which results in end-stage liver disease, including cirrhosis, liver failure, and carcinoma in some patients. In this review, we discuss the potential role of extracellular vesicles in mediating communication between infectious agents (hepatitis B and C viruses) and host cells, and how these complex cell-cell interactions may facilitate the development of chronic liver disease. We will further discuss how understanding their biological mechanism of action might be beneficial for developing therapeutic strategies to treat chronic liver disease.
Collapse
Affiliation(s)
- Hong Kiat Lim
- Hepatic Fibrosis Group, Department of Cellular and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Gary P Jeffrey
- Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia.,Sir Charles Gairdner Hospital, Nedlands, Hepatology Department and Liver Transplant Service, Perth, WA, Australia
| | - Grant A Ramm
- Hepatic Fibrosis Group, Department of Cellular and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Carolina Soekmadji
- Hepatic Fibrosis Group, Department of Cellular and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
15
|
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide and the third leading cause of cancer related mortality with a 10 year survival rate of merely 22-35%. Tumorigenesis frequently occurs in patients with chronic liver disease where continued liver cell damage, compensatory proliferation and inflammation provide the basis for tumor initiation, promotion and progression. Animal models of HCC are particularly useful to better understand molecular events underlying liver tumorigenesis. To this end, chemical carcinogenesis protocols based on the injection of genotoxic compounds such as diethylnitrosamine (DEN) are widely used to model liver tumorigenesis in rodents. DEN injection into 2 week old mice is sufficient to cause liver tumorigenesis after 8-10 months. When injected into older mice, DEN has to be combined with administration of tumor promoting agents such as phenobarbital or feeding high fat diet. Such protocols allow to dissect the different steps of tumor formation (i.e., tumor initiation and promotion) experimentally and to model liver pathologies in mice which frequently lead to HCC in human patients such as non-alcoholic fatty liver disease. Here, we review several established chemical carcinogenesis protocols based on DEN injection into mice and discuss their advantages as well as potential limitations.
Collapse
Affiliation(s)
- Isabel Schulien
- Department of Medicine II, Medical Center-University of Freiburg and Faculty of Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Peter Hasselblatt
- Department of Medicine II, Medical Center-University of Freiburg and Faculty of Medicine, University Hospital Freiburg, Freiburg, Germany.
| |
Collapse
|
16
|
Novo E, Bocca C, Foglia B, Protopapa F, Maggiora M, Parola M, Cannito S. Liver fibrogenesis: un update on established and emerging basic concepts. Arch Biochem Biophys 2020; 689:108445. [PMID: 32524998 DOI: 10.1016/j.abb.2020.108445] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/20/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023]
Abstract
Liver fibrogenesis is defined as a dynamic and highly integrated process occurring during chronic injury to liver parenchyma that can result in excess deposition of extracellular matrix (ECM) components (i.e., liver fibrosis). Liver fibrogenesis, together with chronic inflammatory response, is then primarily involved in the progression of chronic liver diseases (CLD) irrespective of the specific etiology. In the present review we will first offer a synthetic and updated overview of major basic concepts in relation to the role of myofibroblasts (MFs), macrophages and other hepatic cell populations involved in CLD to then offer an overview of established and emerging issues and mechanisms that have been proposed to favor and/or promote CLD progression. A special focus will be dedicated to selected issues that include emerging features in the field of cholangiopathies, the emerging role of genetic and epigenetic factors as well as of hypoxia, hypoxia-inducible factors (HIFs) and related mediators.
Collapse
Affiliation(s)
- Erica Novo
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| | - Claudia Bocca
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| | - Beatrice Foglia
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| | - Francesca Protopapa
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| | - Marina Maggiora
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| | - Maurizio Parola
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy.
| | - Stefania Cannito
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| |
Collapse
|
17
|
Addante A, Roncero C, Lazcanoiturburu N, Méndez R, Almalé L, García-Álvaro M, ten Dijke P, Fabregat I, Herrera B, Sánchez A. A Signaling Crosstalk between BMP9 and HGF/c-Met Regulates Mouse Adult Liver Progenitor Cell Survival. Cells 2020; 9:cells9030752. [PMID: 32204446 PMCID: PMC7140668 DOI: 10.3390/cells9030752] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/17/2022] Open
Abstract
During chronic liver disease, hepatic progenitor cells (HPC, oval cells in rodents) become activated, proliferate, and differentiate into cholangiocytes and/or hepatocytes contributing to the final outcome of the regenerative process in a context-dependent fashion. Here, we analyze the crosstalk between the hepatocyte growth factor (HGF)/c-Met signaling axis, key for liver regeneration, and bone morphogenetic protein (BMP)9, a BMP family ligand that has emerged as a critical regulator of liver pathology. Our results show that HGF/c-Met signaling blocks BMP9-mediated apoptotic cell death, while it potentiates small mothers against decapentaplegic (SMAD)1 signaling triggered by BMP9 in oval cells. Interestingly, HGF-induced overactivation of SMAD1, -5, -8 requires the upregulation of TGF-β type receptor activin receptor-like kinase (ALK)1, and both ALK1 and SMAD1 are required for the counteracting effect of HGF on BMP9 apoptotic activity. On the other hand, we also prove that BMP9 triggers the activation of p38MAPK in oval cells, which drives BMP9-apoptotic cell death. Therefore, our data support a model in which BMP9 and HGF/c-Met signaling axes establish a signaling crosstalk via ALK1 that modulates the balance between the two pathways with opposing activities, SMAD1 (pro-survival) and p38 mitogen-activated protein kinases (p38MAPK; pro-apoptotic), which determines oval cell fate. These data help delineate the complex signaling network established during chronic liver injury and its impact on the oval cell regenerative response.
Collapse
Affiliation(s)
- Annalisa Addante
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - Cesáreo Roncero
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - Nerea Lazcanoiturburu
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - Rebeca Méndez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - Laura Almalé
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - María García-Álvaro
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08907 L’Hospitalet de Llobregat, Barcelona, Spain;
- School of Medicine and Health Sciences, University of Barcelona, 08007 Barcelona, Spain
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Blanca Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
- Correspondence: (B.H.); (A.S.); Tel.: +34 913941855 (A.S.)
| | - Aránzazu Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
- Correspondence: (B.H.); (A.S.); Tel.: +34 913941855 (A.S.)
| |
Collapse
|
18
|
Regulation of Fibrotic Processes in the Liver by ADAM Proteases. Cells 2019; 8:cells8101226. [PMID: 31601007 PMCID: PMC6830092 DOI: 10.3390/cells8101226] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/18/2022] Open
Abstract
Fibrosis in the liver is mainly associated with the activation of hepatic stellate cells (HSCs). Both activation and clearance of HSCs can be mediated by ligand–receptor interactions. Members of the a disintegrin and metalloprotease (ADAM) family are involved in the proteolytic release of membrane-bound ligands and receptor ectodomains and the remodelling of the extracellular matrix. ADAM proteases are therefore major regulators of intercellular signalling pathways. In the present review we discuss how ADAM proteases modulate pro- and anti-fibrotic processes and how ADAM proteases might be harnessed therapeutically in the future.
Collapse
|
19
|
A critical role of autophagy in regulating the mesenchymal transition of ductular cells in liver cirrhosis. Sci Rep 2019; 9:10673. [PMID: 31337842 PMCID: PMC6650611 DOI: 10.1038/s41598-019-46764-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 07/05/2019] [Indexed: 02/07/2023] Open
Abstract
Our previous studies have shown that autophagy mediates the link between ductular reaction (DR) and liver cirrhosis. Whether the subsequent fibrogenic response is regulated by increased autophagy in DR remains unclear. Here, using both human liver specimens and a rat model of liver cirrhosis induced by 2-acetylaminofluorene (AAF) and carbon tetrachloride (CCL4), we explored the involvement of autophagy in regulating mesenchymal transition of ductular cells. Ductular cells from AAF/CCL4 livers exhibited increased autophagy compared to those of normal livers. These cells showed morphological and functional characteristics of mesenchymal cells. Blocking autophagy using bafilomycin A1 or siRNA targeting ATG7 reduced the expression of mesenchymal markers in these ductular cells from AAF/CCL4 livers, indicating a role for autophagy in regulating the mesenchymal phenotype of ductular cells. Furthermore, we show that the mesenchymal transition in DR requires the activation of transforming growth factor-β (TGF-β) signaling in an autophagy-dependent manner. Importantly, in cirrhotic human livers, ductular cells that are positive for LC3B also showed increased expression of TGF-β and fibroblast-specific protein-1. Our data suggest activation of autophagy in ductular cells, and also demonstrate that it is required for the mesenchymal transition during the DR, processes that are critically involved in the pathogenesis of cirrhosis.
Collapse
|
20
|
Takemura A, Gong S, Sekine S, Ito K. Inhibition of biliary network reconstruction by benzbromarone delays recovery from pre-existing liver injury. Toxicology 2019; 423:32-41. [DOI: 10.1016/j.tox.2019.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/09/2019] [Accepted: 05/13/2019] [Indexed: 01/23/2023]
|
21
|
An Immunohistochemical Study of Gastric Mucosa and Critical Review Indicate that the Subepithelial Telocytes are Prelymphatic Endothelial Cells. ACTA ACUST UNITED AC 2019; 55:medicina55070316. [PMID: 31252668 PMCID: PMC6680827 DOI: 10.3390/medicina55070316] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/26/2019] [Accepted: 06/20/2019] [Indexed: 12/18/2022]
Abstract
Background and Objectives: There are only a few studies regarding gut subepithelial telocytes (TCs). The telopodes, namely peculiar TCs’ prolongations described on two-dimensional cuts, are not enough to differentiate this specific cell type. Subepithelial TCs were associated with the intestinal stem niche but a proper differential diagnosis with lymphatic endothelial cells (LECs) was not performed. In this study, we will also critically review studies suggesting that distinctive TCs could be positioned within the lamina propria. Materials and Methods: We performed an immunohistochemical study of human gastric mucosa to test the expression of D2-40, the lymphatic marker, as well as that of CD31, CD34, CD44, CD117/c-kit, α-smooth muscle actin (α-SMA) and vimentin in the gastric subepithelial niche. Results: The results support the poorly investigated anatomy of intramural gastric lymphatics, with circumferential collectors located on both sides of the muscularis mucosae (mucosal and then submucosal) and myenteric collectors in the muscularis propria. We also found superficial epithelial prelymphatic channels bordered by D2-40+ but CD31–TC-like cells. Deep epithelial lymphatic collectors drain in collectors within the lamina propria. Blood endothelial cells expressed CD31, CD34, CD44, and vimentin. Conclusions: Therefore, the positive diagnosis of TC for subepithelial CD34+ cells should be regarded with caution, as they could also be artefacts, resulting from the two-dimensional examination of three dimensional structures, or as LECs. Lymphatic markers should be routinely used to discriminate TCs from LECs.
Collapse
|
22
|
Almalé L, García-Álvaro M, Martínez-Palacián A, García-Bravo M, Lazcanoiturburu N, Addante A, Roncero C, Sanz J, de la O López M, Bragado P, Mikulits W, Factor VM, Thorgeirsson SS, Casal JI, Segovia JC, Rial E, Fabregat I, Herrera B, Sánchez A. c-Met Signaling Is Essential for Mouse Adult Liver Progenitor Cells Expansion After Transforming Growth Factor-β-Induced Epithelial-Mesenchymal Transition and Regulates Cell Phenotypic Switch. Stem Cells 2019; 37:1108-1118. [PMID: 31108004 DOI: 10.1002/stem.3038] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/08/2019] [Accepted: 04/29/2019] [Indexed: 01/10/2023]
Abstract
Adult hepatic progenitor cells (HPCs)/oval cells are bipotential progenitors that participate in liver repair responses upon chronic injury. Recent findings highlight HPCs plasticity and importance of the HPCs niche signals to determine their fate during the regenerative process, favoring either fibrogenesis or damage resolution. Transforming growth factor-β (TGF-β) and hepatocyte growth factor (HGF) are among the key signals involved in liver regeneration and as component of HPCs niche regulates HPCs biology. Here, we characterize the TGF-β-triggered epithelial-mesenchymal transition (EMT) response in oval cells, its effects on cell fate in vivo, and the regulatory effect of the HGF/c-Met signaling. Our data show that chronic treatment with TGF-β triggers a partial EMT in oval cells based on coexpression of epithelial and mesenchymal markers. The phenotypic and functional profiling indicates that TGF-β-induced EMT is not associated with stemness but rather represents a step forward along hepatic lineage. This phenotypic transition confers advantageous traits to HPCs including survival, migratory/invasive and metabolic benefit, overall enhancing the regenerative potential of oval cells upon transplantation into a carbon tetrachloride-damaged liver. We further uncover a key contribution of the HGF/c-Met pathway to modulate the TGF-β-mediated EMT response. It allows oval cells expansion after EMT by controlling oxidative stress and apoptosis, likely via Twist regulation, and it counterbalances EMT by maintaining epithelial properties. Our work provides evidence that a coordinated and balanced action of TGF-β and HGF are critical for achievement of the optimal regenerative potential of HPCs, opening new therapeutic perspectives. Stem Cells 2019;37:1108-1118.
Collapse
Affiliation(s)
- Laura Almalé
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - María García-Álvaro
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Adoración Martínez-Palacián
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - María García-Bravo
- Cell Differentiation and Cytometry Unit, Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Advanced Therapies Mixed Unit, CIEMAT/IIS Fundación Jiménez Díaz, Madrid, Spain
| | - Nerea Lazcanoiturburu
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Annalisa Addante
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Cesáreo Roncero
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Julián Sanz
- Department of Pathology, Hospital Clínico San Carlos, Madrid, Spain
| | - María de la O López
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Paloma Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Wolfgang Mikulits
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Valentina M Factor
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Snorri S Thorgeirsson
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.,Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - J Ignacio Casal
- Department of Functional Proteomics, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - José-Carlos Segovia
- Cell Differentiation and Cytometry Unit, Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.,Advanced Therapies Mixed Unit, CIEMAT/IIS Fundación Jiménez Díaz, Madrid, Spain
| | - Eduardo Rial
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Blanca Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Aránzazu Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
23
|
Farley AM, Braxton DR, Li J, Trounson K, Sakar-Dey S, Nayer B, Ikeda T, Lau KX, Hardikar W, Hasegawa K, Pera MF. Antibodies to a CA 19-9 Related Antigen Complex Identify SOX9 Expressing Progenitor Cells In Human Foetal Pancreas and Pancreatic Adenocarcinoma. Sci Rep 2019; 9:2876. [PMID: 30814526 PMCID: PMC6393509 DOI: 10.1038/s41598-019-38988-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 01/11/2019] [Indexed: 12/21/2022] Open
Abstract
The Sialyl Lewis A antigen, or CA 19-9, is the prototype serum biomarker for adenocarcinoma of the pancreas. Despite extensive clinical study of CA 19-9 in gastrointestinal malignancies, surprisingly little is known concerning the specific cell types that express this marker during development, tissue regeneration and neoplasia. SOX9 is a transcription factor that plays a key role in these processes in foregut tissues. We report the biochemistry and tissue expression of the GCTM-5 antigen, a pancreatic cancer marker related to, but distinct from, CA19-9. This antigen, defined by two monoclonal antibodies recognising separate epitopes on a large glycoconjugate protein complex, is co-expressed with SOX9 by foregut ductal progenitors in the developing human liver and pancreas, and in pancreatic adenocarcinoma. These progenitors are distinct from cell populations identified by DCLK1, LGR5, or canonical markers of liver and pancreatic progenitor cells. Co-expression of this antigen complex and SOX9 also characterises the ductal metaplasia of submucosal glands that occurs during the development of Barrett’s oesophagus. The GCTM-5 antigen complex can be detected in the sera of patients with pancreatic adenocarcinoma. The GCTM-5 epitope shows a much more restricted pattern of expression in the normal adult pancreas relative to CA19-9. Our findings will aid in the identification, characterisation, and monitoring of ductal progenitor cells during development and progression of pancreatic adenocarcinoma in man.
Collapse
Affiliation(s)
- Alison M Farley
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia.,The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - David R Braxton
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan Li
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Karl Trounson
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Bhavana Nayer
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| | - Tatsuhiko Ikeda
- Institute for Integrated Cell-Materials Science, Kyoto University, Kyoto, Japan
| | - Kevin X Lau
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Winita Hardikar
- Royal Childrens Hospital, Parkville, Victoria, Australia.,Childrens Medical Research Institute, Parkville, Victoria, Australia
| | - Kouichi Hasegawa
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,Institute for Integrated Cell-Materials Science, Kyoto University, Kyoto, Japan
| | - Martin F Pera
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia. .,Florey Neuroscience and Mental Health Institute, Parkville, Victoria, Australia. .,The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
| |
Collapse
|
24
|
Activation of Polyamine Catabolism by N¹,N 11-Diethylnorspermine in Hepatic HepaRG Cells Induces Dedifferentiation and Mesenchymal-Like Phenotype. Cells 2018; 7:cells7120275. [PMID: 30567412 PMCID: PMC6316793 DOI: 10.3390/cells7120275] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/10/2018] [Accepted: 12/15/2018] [Indexed: 12/24/2022] Open
Abstract
Tumorigenesis is accompanied by the metabolic adaptation of cells to support enhanced proliferation rates and to optimize tumor persistence and amplification within the local microenvironment. In particular, cancer cells exhibit elevated levels of biogenic polyamines. Inhibitors of polyamine biosynthesis and inducers of their catabolism have been evaluated as antitumor drugs, however, their efficacy and safety remain controversial. Our goal was to investigate if drug-induced modulation of polyamine metabolism plays a role in dedifferentiation using differentiated human hepatocyte-like HepaRG cell cultures. N1,N11-diethylnorspermine (DENSpm), a potent inducer of polyamine catabolism, triggered an epithelial-mesenchymal transition (EMT)-like dedifferentiation in HepaRG cultures, as shown by down-regulation of mature hepatocytes markers and upregulation of classical EMT markers. Albeit the fact that polyamine catabolism produces H2O2, DENSpm-induced de-differentiation was not affected by antioxidants. Use of a metabolically stable spermidine analogue showed furthermore, that spermidine is a key regulator of hepatocyte differentiation. Comparative transcriptome analyses revealed, that the DENSpm-triggered dedifferentiation of HepaRG cells was accompanied by dramatic metabolic adaptations, exemplified by down-regulation of the genes of various metabolic pathways and up-regulation of the genes involved in signal transduction pathways. These results demonstrate that polyamine metabolism is tightly linked to EMT and differentiation of liver epithelial cells.
Collapse
|
25
|
Addante A, Roncero C, Almalé L, Lazcanoiturburu N, García-Álvaro M, Fernández M, Sanz J, Hammad S, Nwosu ZC, Lee SJ, Fabregat I, Dooley S, ten Dijke P, Herrera B, Sánchez A. Bone morphogenetic protein 9 as a key regulator of liver progenitor cells in DDC-induced cholestatic liver injury. Liver Int 2018; 38:1664-1675. [PMID: 29751359 PMCID: PMC6693351 DOI: 10.1111/liv.13879] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/26/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Bone morphogenetic protein 9 (BMP9) interferes with liver regeneration upon acute injury, while promoting fibrosis upon carbon tetrachloride-induced chronic injury. We have now addressed the role of BMP9 in 3,5 diethoxicarbonyl-1,4 dihydrocollidine (DDC)-induced cholestatic liver injury, a model of liver regeneration mediated by hepatic progenitor cell (known as oval cell), exemplified as ductular reaction and oval cell expansion. METHODS WT and BMP9KO mice were submitted to DDC diet. Livers were examined for liver injury, fibrosis, inflammation and oval cell expansion by serum biochemistry, histology, RT-qPCR and western blot. BMP9 signalling and effects in oval cells were studied in vitro using western blot and transcriptional assays, plus functional assays of DNA synthesis, cell viability and apoptosis. Crosslinking assays and short hairpin RNA approaches were used to identify the receptors mediating BMP9 effects. RESULTS Deletion of BMP9 reduces liver damage and fibrosis, but enhances inflammation upon DDC feeding. Molecularly, absence of BMP9 results in overactivation of PI3K/AKT, ERK-MAPKs and c-Met signalling pathways, which together with an enhanced ductular reaction and oval cell expansion evidence an improved regenerative response and decreased damage in response to DDC feeding. Importantly, BMP9 directly targets oval cells, it activates SMAD1,5,8, decreases cell growth and promotes apoptosis, effects that are mediated by Activin Receptor-Like Kinase 2 (ALK2) type I receptor. CONCLUSIONS We identify BMP9 as a negative regulator of oval cell expansion in cholestatic injury, its deletion enhancing liver regeneration. Likewise, our work further supports BMP9 as an attractive therapeutic target for chronic liver diseases.
Collapse
Affiliation(s)
- Annalisa Addante
- Faculty of Pharmacy, Department Biochemistry and Molecular Biology, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Cesáreo Roncero
- Faculty of Pharmacy, Department Biochemistry and Molecular Biology, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Laura Almalé
- Faculty of Pharmacy, Department Biochemistry and Molecular Biology, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Nerea Lazcanoiturburu
- Faculty of Pharmacy, Department Biochemistry and Molecular Biology, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - María García-Álvaro
- Faculty of Pharmacy, Department Biochemistry and Molecular Biology, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Margarita Fernández
- Faculty of Pharmacy, Department Biochemistry and Molecular Biology, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Julián Sanz
- Department Pathology, Hospital Clínico San Carlos, Madrid, Spain
| | - Seddik Hammad
- Medical Faculty Mannheim, Department Medicine II, Heidelberg University, Manhheim, Germany
| | - Zeribe C. Nwosu
- Medical Faculty Mannheim, Department Medicine II, Heidelberg University, Manhheim, Germany
| | - Se-Jin Lee
- Department Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Isabel Fabregat
- Bellvitge Biomedical Research Institute, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Steven Dooley
- Medical Faculty Mannheim, Department Medicine II, Heidelberg University, Manhheim, Germany
| | - Peter ten Dijke
- Department Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, RC Leiden, The Netherlands
| | - Blanca Herrera
- Faculty of Pharmacy, Department Biochemistry and Molecular Biology, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Aránzazu Sánchez
- Faculty of Pharmacy, Department Biochemistry and Molecular Biology, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
26
|
Chen D, Yu D, Wang X, Liu Y, He Y, Deng R, Jiang Y, Zhang F, Liu Y, Xu M, Li J, Luo J, Wang S. Epithelial to mesenchymal transition is involved in ethanol promoted hepatocellular carcinoma cells metastasis and stemness. Mol Carcinog 2018; 57:1358-1370. [PMID: 30295962 DOI: 10.1002/mc.22850] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/04/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022]
Abstract
Hepatocellular Carcinoma (HCC) is a malignant tumor with high rate of relapse and metastasis. Ethanol is a well-known risk factor for HCC; it promotes the progression and aggressiveness of HCC. However, the underlying mechanism remains unclear. In clinic studies, we showed that alcohol consumption is positively correlated with TNM stage and vessel invasion; HCC patients with chronic drinking history had faster progression rate and poorer prognosis compared to non-drinkers. In experimental models, ethanol exposure enhanced the metastasis, and invasion of HCC cells. Ethanol exposure increased cancer stem cells (CSC) population and enhanced stemness of HCC cells in vitro and in vivo. Mechanically, we found that ethanol exposure induced epithelial to mesenchymal transition (EMT) through activating Wnt/β-catenin signaling pathway in HCC cells. We further demonstrated that β-catenin siRNA or salinomycin (an inhibitor of Wnt/β-catenin pathway) partially rescued ethanol-induced EMT. In conclusion, this study suggested that ethanol exposure promotes the metastasis and stemness of HCC cells by inducing EMT.
Collapse
MESH Headings
- Alcohol Drinking
- Animals
- Anti-Infective Agents, Local/pharmacology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Epithelial-Mesenchymal Transition/drug effects
- Ethanol/pharmacology
- Female
- Hep G2 Cells
- Humans
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Male
- Mice, Inbred C57BL
- Mice, Nude
- Middle Aged
- Neoplasm Metastasis
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Danlei Chen
- Schoolof Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Dandan Yu
- Schoolof Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Xinyi Wang
- Schoolof Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Yan Liu
- Schoolof Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, P. R. China
- The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P. R. China
| | - Yongjing He
- The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P. R. China
| | - Ruiqing Deng
- Schoolof Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, P. R. China
- The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P. R. China
| | - Yu Jiang
- Schoolof Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Fengyun Zhang
- Schoolof Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Yakun Liu
- Schoolof Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, P. R. China
| | - Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, College of Medicine, Lexington, Kentucky
| | - Jiabin Li
- The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P. R. China
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, College of Medicine, Lexington, Kentucky
| | - Siying Wang
- Schoolof Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, P. R. China
| |
Collapse
|
27
|
Loo CK, Danieletto S, Friedlander M, Pearen MA, Ramm GA. Peritoneal hepatoid carcinoma with chemotherapy response and possible stem cell involvement. HUMAN PATHOLOGY: CASE REPORTS 2018. [DOI: 10.1016/j.ehpc.2018.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
28
|
Gratte FD, Pasic S, Olynyk JK, Yeoh GCT, Tosh D, Coombe DR, Tirnitz-Parker JEE. Transdifferentiation of pancreatic progenitor cells to hepatocyte-like cells is not serum-dependent when facilitated by extracellular matrix proteins. Sci Rep 2018. [PMID: 29531353 PMCID: PMC5847606 DOI: 10.1038/s41598-018-22596-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The rising prevalence of chronic liver disease, coupled with a permanent shortage of organs for liver transplantation, has sparked enormous interest in alternative treatment strategies. Previous protocols to generate hepatocyte-like cells (HLCs) via pancreas-to-liver transdifferentiation have utilised fetal bovine serum, introducing unknown variables and severely limiting study reproducibility. Therefore, the main goal of this study was to develop a protocol for transdifferentiation of pancreatic progenitor cells to HLCs in a chemically defined, serum-free culture medium. The clonal pancreatic progenitor cell line AR42J-B13 was cultured in basal growth medium on uncoated plastic culture dishes in the absence or presence of Dexamethasone on uncoated, laminin- or fibronectin-coated culture substrata, with or without serum supplementation. The hepatocytic differentiation potential was evaluated: (i) morphologically through bright-field and scanning electron microscopy, (ii) by assessing pancreatic and hepatic marker expression and (iii) by determining the function of HLCs through their ability to synthesise glycogen or take up and release indocyanine green. Here we demonstrate for the first time that transdifferentiation of pancreatic cells to HLCs is not dependent on serum. These results will assist in converting current differentiation protocols into procedures that are compliant with clinical use in future cell-based therapies to treat liver-related metabolic disorders.
Collapse
Affiliation(s)
- Francis D Gratte
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia.,School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA, Australia
| | - Sara Pasic
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - John K Olynyk
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia.,School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA, Australia.,Department of Gastroenterology and Hepatology, Fiona Stanley and Fremantle Hospitals, Perth, WA, Australia
| | - George C T Yeoh
- Cancer and Cell Biology Division, The Harry Perkins Institute of Medical Research, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - David Tosh
- Centre for Regenerative Medicine, University of Bath, Bath, United Kingdom
| | - Deirdre R Coombe
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia. .,Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia.
| | - Janina E E Tirnitz-Parker
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia. .,Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia. .,School of Medicine and Pharmacology, University of Western Australia, Fremantle, WA, Australia.
| |
Collapse
|
29
|
Whole-exome sequencing reveals the origin and evolution of hepato-cholangiocarcinoma. Nat Commun 2018; 9:894. [PMID: 29497050 PMCID: PMC5832792 DOI: 10.1038/s41467-018-03276-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 02/02/2018] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular-cholangiocarcinoma (H-ChC) is a rare subtype of liver cancer with clinicopathological features of both hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA). To date, molecular mechanisms underlying the co-existence of HCC and iCCA components in a single tumor remain elusive. Here, we show that H-ChC samples contain substantial private mutations from WES analyses, ranging from 33.1 to 86.4%, indicative of substantive intratumor heterogeneity (ITH). However, on the other hand, numerous ubiquitous mutations shared by HCC and iCCA suggest the monoclonal origin of H-ChC. Mutated genes identified herein, e.g., VCAN, ACVR2A, and FCGBP, are speculated to contribute to distinct differentiation of HCC and iCCA within H-ChC. Moreover, immunohistochemistry demonstrates that EpCAM is highly expressed in 80% of H-ChC, implying the stemness of such liver cancer. In summary, our data highlight the monoclonal origin and stemness of H-ChC, as well as substantial intratumoral heterogeneity. Hepatocellular-cholangiocarcinoma (H-ChC) is a rare subtype of liver cancer with features of hepatocellular carcinoma and intrahepatic cholangiocarcinoma. Here, the authors utilize whole exome sequencing to highlight the monoclonal origin and stemness of H-ChC, as well as substantial intratumoral heterogeneity.
Collapse
|
30
|
Kramer AS, Latham B, Diepeveen LA, Mou L, Laurent GJ, Elsegood C, Ochoa-Callejero L, Yeoh GC. InForm software: a semi-automated research tool to identify presumptive human hepatic progenitor cells, and other histological features of pathological significance. Sci Rep 2018; 8:3418. [PMID: 29467378 PMCID: PMC5821869 DOI: 10.1038/s41598-018-21757-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/30/2018] [Indexed: 12/19/2022] Open
Abstract
Hepatic progenitor cells (HPCs) play an important regenerative role in acute and chronic liver pathologies. Liver disease research often necessitates the grading of disease severity, and pathologists’ reports are the current gold-standard for assessment. However, it is often impractical to recruit pathologists in large cohort studies. In this study we utilise PerkinElmer’s “InForm” software package to semi-automate the scoring of patient liver biopsies, and compare outputs to a pathologist’s assessment. We examined a cohort of eleven acute hepatitis samples and three non-alcoholic fatty liver disease (NAFLD) samples, stained with HPC markers (GCTM-5 and Pan Cytokeratin), an inflammatory marker (CD45), Sirius Red to detect collagen and haematoxylin/eosin for general histology. InForm was configured to identify presumptive HPCs, CD45+ve inflammatory cells, areas of necrosis, fat and collagen deposition (p < 0.0001). Hepatitis samples were then evaluated both by a pathologist using the Ishak-Knodell scoring system, and by InForm through customised algorithms. Necroinflammation as evaluated by a pathologist, correlated with InForm outputs (r2 = 0.8192, p < 0.05). This study demonstrates that the InForm software package provides a useful tool for liver disease research, allowing rapid, and objective quantification of the presumptive HPCs and identifies histological features that assist with assessing liver disease severity, and potentially can facilitate diagnosis.
Collapse
Affiliation(s)
- Anne S Kramer
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Bruce Latham
- PathWest Laboratory Medicine WA, Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Luke A Diepeveen
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia
| | - Lingjun Mou
- WA Liver & Kidney Surgical Transplant Service, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - Geoffrey J Laurent
- Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Caryn Elsegood
- School of Pharmacy and Biomedical Science, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Laura Ochoa-Callejero
- Angiogenesis group, Oncology Area, Centre for Biomedical Research of La Rioja, Logroño, Spain
| | - George C Yeoh
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia. .,School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia. .,Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia.
| |
Collapse
|
31
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function, and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2018:1-87. [DOI: 10.1016/b978-0-7020-6697-9.00001-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
32
|
Herrera B, Addante A, Sánchez A. BMP Signalling at the Crossroad of Liver Fibrosis and Regeneration. Int J Mol Sci 2017; 19:ijms19010039. [PMID: 29295498 PMCID: PMC5795989 DOI: 10.3390/ijms19010039] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022] Open
Abstract
Bone Morphogenetic Proteins (BMPs) belong to the Transforming Growth Factor-β (TGF-β) family. Initially identified due to their ability to induce bone formation, they are now known to have multiple functions in a variety of tissues, being critical not only during development for tissue morphogenesis and organogenesis but also during adult tissue homeostasis. This review focus on the liver as a target tissue for BMPs actions, devoting most efforts to summarize our knowledge on their recently recognized and/or emerging roles on regulation of the liver regenerative response to various insults, either acute or chronic and their effects on development and progression of liver fibrosis in different pathological conditions. In an attempt to provide the basis for guiding research efforts in this field both the more solid and more controversial areas of research were highlighted.
Collapse
Affiliation(s)
- Blanca Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Annalisa Addante
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Aránzazu Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| |
Collapse
|
33
|
Elsegood CL, Tirnitz-Parker JE, Olynyk JK, Yeoh GC. Immune checkpoint inhibition: prospects for prevention and therapy of hepatocellular carcinoma. Clin Transl Immunology 2017; 6:e161. [PMID: 29326816 PMCID: PMC5704099 DOI: 10.1038/cti.2017.47] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 08/10/2017] [Accepted: 09/10/2017] [Indexed: 02/06/2023] Open
Abstract
The global prevalence of liver cancer is rapidly rising, mostly as a result of the amplified incidence rates of viral hepatitis, alcohol abuse and obesity in recent decades. Treatment options for liver cancer are remarkably limited with sorafenib being the gold standard for advanced, unresectable hepatocellular carcinoma but offering extremely limited improvement of survival time. The immune system is now recognised as a key regulator of cancer development through its ability to protect against infection and chronic inflammation, which promote cancer development, and eliminate tumour cells when present. However, the tolerogenic nature of the liver means that the immune response to infection, chronic inflammation and tumour cells within the hepatic environment is usually ineffective. Here we review the roles that immune cells and cytokines have in the development of the most common primary liver cancer, hepatocellular carcinoma (HCC). We then examine how the immune system may be subverted throughout the stages of HCC development, particularly with respect to immune inhibitory molecules, also known as immune checkpoints, such as programmed cell death protein-1, programmed cell death 1 ligand 1 and cytotoxic T lymphocyte antigen 4, which have become therapeutic targets. Finally, we assess preclinical and clinical studies where immune checkpoint inhibitors have been used to modify disease during the carcinogenic process. In conclusion, inhibitory molecule-based immunotherapy for HCC is in its infancy and further detailed research in relevant in vivo models is required before its full potential can be realised.
Collapse
Affiliation(s)
- Caryn L Elsegood
- School of Biomedical Science, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Janina Ee Tirnitz-Parker
- School of Biomedical Science, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - John K Olynyk
- School of Biomedical Science, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,Department of Gastroenterology and Hepatology, Fiona Stanley and Fremantle Hospitals, South Metropolitan Health Service, Murdoch, Western Australia, Australia.,School of Health and Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - George Ct Yeoh
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
34
|
Bian M, Chen X, Zhang C, Jin H, Wang F, Shao J, Chen A, Zhang F, Zheng S. Magnesium isoglycyrrhizinate promotes the activated hepatic stellate cells apoptosis via endoplasmic reticulum stress and ameliorates fibrogenesis in vitro and in vivo. Biofactors 2017; 43:836-846. [PMID: 29048780 DOI: 10.1002/biof.1390] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/21/2017] [Accepted: 08/29/2017] [Indexed: 12/11/2022]
Abstract
Varied pathogenetic elements have been touched upon the liver fibrosis, including inflammatory, stress, apoptosis and unfolded proteins aggregation. Magnesium Isoglycyrrhizinate (MgIG) has been accepted to be a neuroprotective effect, hepatoprotective and anti-inflammatory molecule. In our vitro researches, MgIG was considered to activate hepatic stellate cells (HSCs) apoptosis by promoting endoplasmic reticulum stress (ERS) detrimental response to a certain extent. Consequently, MgIG showed its potential therapeutic capacity in fibrogenesis and counteracted the pathogenetic aspects, which were involved in integrating current treatments correcting liver fibrosis. In addition, we further verificated the behavior and pathogenic mechanisms in the CCl4 -induced liver fibrosis in male mice. What surprised us was that with the treatment of MgIG caused the activation of ERS and resisted the activated HSCs in the protective effects on liver damage. We found MgIG significantly promoted the apoptosis of activated HSCs and protected the CCl4 -induced liver fibrosis. Main molecules came down to the unfolded protein response signaling pathway. Furthermore, MgIG inhibited the levels of the downstream inflammatory cytokines, which were triggered by CCl4 -induced liver fibrosis. Here, we reported that MgIG improved behavioral impairments induced by intraperitoneal injection of CCl4 and decreased the expression of proinflammatory factor, which indicated the preserving effects on liver fibrosis. © 2017 BioFactors, 43(6):836-846, 2017.
Collapse
Affiliation(s)
- Mianli Bian
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xingran Chen
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chenxi Zhang
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Huanhuan Jin
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Feixia Wang
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiangjuan Shao
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, St Louis, MO
| | - Feng Zhang
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shizhong Zheng
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
35
|
Gogoi-Tiwari J, Köhn-Gaone J, Giles C, Schmidt-Arras D, Gratte FD, Elsegood CL, McCaughan GW, Ramm GA, Olynyk JK, Tirnitz-Parker JEE. The Murine Choline-Deficient, Ethionine-Supplemented (CDE) Diet Model of Chronic Liver Injury. J Vis Exp 2017. [PMID: 29155718 DOI: 10.3791/56138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Chronic liver diseases, such as viral hepatitis, alcoholic liver disease, or non-alcoholic fatty liver disease, are characterized by continual inflammation, progressive destruction and regeneration of the hepatic parenchyma, liver progenitor cell proliferation, and fibrosis. The end-stage of every chronic liver disease is cirrhosis, a major risk factor for the development of hepatocellular carcinoma. To study processes regulating disease initiation, establishment, and progression, several animal models are used in laboratories. Here we describe a six-week time course of the choline-deficient and ethionine-supplemented (CDE) mouse model, which involves feeding six-week old male C57BL/6J mice with choline-deficient chow and 0.15% DL-ethionine-supplemented drinking water. Monitoring of animal health and a typical body weight loss curve are explained. The protocol demonstrates the gross examination of a CDE-treated liver and blood collection by cardiac puncture for subsequent serum analyses. Next, the liver perfusion technique and collection of different hepatic lobes for standard evaluations are shown, including liver histology assessments by hematoxylin and eosin or Sirius Red stainings, immunofluorescent detection of hepatic cell populations as well as transcriptome profiling of the liver microenvironment. This mouse model is suitable for studying inflammatory, fibrogenic, and liver progenitor cell dynamics induced through chronic liver disease and can be used to test potential therapeutic agents that may modulate these processes.
Collapse
Affiliation(s)
- Jully Gogoi-Tiwari
- School of Biomedical Sciences & Curtin Health Innovation Research Institute, Curtin University
| | - Julia Köhn-Gaone
- School of Biomedical Sciences & Curtin Health Innovation Research Institute, Curtin University
| | - Corey Giles
- School of Public Health & Curtin Health Innovation Research Institute, Curtin University
| | | | - Francis D Gratte
- School of Biomedical Sciences & Curtin Health Innovation Research Institute, Curtin University; School of Veterinary and Life Sciences, Murdoch University
| | - Caryn L Elsegood
- School of Biomedical Sciences & Curtin Health Innovation Research Institute, Curtin University
| | - Geoffrey W McCaughan
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney; Royal Prince Alfred Hospital; A.W. Morrow Gastroenterology and Liver Centre
| | - Grant A Ramm
- QIMR Berghofer Medical Research Institute; Faculty of Medicine and Biomedical Sciences, The University of Queensland
| | - John K Olynyk
- Fiona Stanley and Fremantle Hospitals; School of Medical and Health Sciences, Edith Cowan University
| | - Janina E E Tirnitz-Parker
- School of Biomedical Sciences & Curtin Health Innovation Research Institute, Curtin University; School of Medicine and Pharmacology, University of Western Australia;
| |
Collapse
|
36
|
Taurocholate Induces Biliary Differentiation of Liver Progenitor Cells Causing Hepatic Stellate Cell Chemotaxis in the Ductular Reaction: Role in Pediatric Cystic Fibrosis Liver Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2744-2757. [PMID: 28935574 DOI: 10.1016/j.ajpath.2017.08.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/14/2017] [Accepted: 08/11/2017] [Indexed: 02/07/2023]
Abstract
Cystic fibrosis liver disease (CFLD) in children causes progressive fibrosis leading to biliary cirrhosis; however, its cause(s) and early pathogenesis are unclear. We hypothesized that a bile acid-induced ductular reaction (DR) drives fibrogenesis. The DR was evaluated by cytokeratin-7 immunohistochemistry in liver biopsies, staged for fibrosis, from 60 children with CFLD, and it demonstrated that the DR was significantly correlated with hepatic fibrosis stage and biliary taurocholate levels. To examine the mechanisms involved in DR induction, liver progenitor cells (LPCs) were treated with taurocholate, and key events in DR evolution were assessed: LPC proliferation, LPC biliary differentiation, and hepatic stellate cell (HSC) chemotaxis. Taurocholate induced a time-dependent increase in LPC proliferation and expression of genes associated with cholangiocyte differentiation (cytokeratin 19, connexin 43, integrin β4, and γ-glutamyltranspeptidase), whereas the hepatocyte specification marker HNF4α was suppressed. Functional cholangiocyte differentiation was demonstrated via increased acetylated α-tubulin and SOX9 proteins, the number of primary cilia+ LPCs, and increased active γ-glutamyltranspeptidase enzyme secretion. Taurocholate induced LPCs to release MCP-1, MIP1α, and RANTES into conditioned medium causing HSC chemotaxis, which was inhibited by anti-MIP1α. Immunofluorescence confirmed chemokine expression localized to CK7+ DR and LPCs in CFLD liver biopsies. This study suggests that taurocholate is involved in initiating functional LPC biliary differentiation and the development of the DR, with subsequent induction of chemokines that drive HSC recruitment in CFLD.
Collapse
|
37
|
Enane FO, Shuen WH, Gu X, Quteba E, Przychodzen B, Makishima H, Bodo J, Ng J, Chee CL, Ba R, Seng Koh L, Lim J, Cheong R, Teo M, Hu Z, Ng KP, Maciejewski J, Radivoyevitch T, Chung A, Ooi LL, Tan YM, Cheow PC, Chow P, Chan CY, Lim KH, Yerian L, Hsi E, Toh HC, Saunthararajah Y. GATA4 loss of function in liver cancer impedes precursor to hepatocyte transition. J Clin Invest 2017; 127:3527-3542. [PMID: 28758902 DOI: 10.1172/jci93488] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/08/2017] [Indexed: 12/18/2022] Open
Abstract
The most frequent chromosomal structural loss in hepatocellular carcinoma (HCC) is of the short arm of chromosome 8 (8p). Genes on the remaining homologous chromosome, however, are not recurrently mutated, and the identity of key 8p tumor-suppressor genes (TSG) is unknown. In this work, analysis of minimal commonly deleted 8p segments to identify candidate TSG implicated GATA4, a master transcription factor driver of hepatocyte epithelial lineage fate. In a murine model, liver-conditional deletion of 1 Gata4 allele to model the haploinsufficiency seen in HCC produced enlarged livers with a gene expression profile of persistent precursor proliferation and failed hepatocyte epithelial differentiation. HCC mimicked this gene expression profile, even in cases that were morphologically classified as well differentiated. HCC with intact chromosome 8p also featured GATA4 loss of function via GATA4 germline mutations that abrogated GATA4 interactions with a coactivator, MED12, or by inactivating mutations directly in GATA4 coactivators, including ARID1A. GATA4 reintroduction into GATA4-haploinsufficient HCC cells or ARID1A reintroduction into ARID1A-mutant/GATA4-intact HCC cells activated hundreds of hepatocyte genes and quenched the proliferative precursor program. Thus, disruption of GATA4-mediated transactivation in HCC suppresses hepatocyte epithelial differentiation to sustain replicative precursor phenotype.
Collapse
Affiliation(s)
- Francis O Enane
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Wai Ho Shuen
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Xiaorong Gu
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ebrahem Quteba
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Bartlomiej Przychodzen
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hideki Makishima
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Juraj Bodo
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Joanna Ng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Chit Lai Chee
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Rebecca Ba
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Lip Seng Koh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Janice Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Rachael Cheong
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Marissa Teo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Zhenbo Hu
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kwok Peng Ng
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jaroslaw Maciejewski
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tomas Radivoyevitch
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alexander Chung
- Department of Hepato-pancreato-biliary and Transplant Surgery and
| | | | - Yu Meng Tan
- Department of Hepato-pancreato-biliary and Transplant Surgery and
| | - Peng-Chung Cheow
- Department of Hepato-pancreato-biliary and Transplant Surgery and
| | - Pierce Chow
- Department of Hepato-pancreato-biliary and Transplant Surgery and
| | - Chung Yip Chan
- Department of Hepato-pancreato-biliary and Transplant Surgery and
| | - Kiat Hon Lim
- Department of Pathology, Singapore General Hospital, Singapore
| | - Lisa Yerian
- Clinical Pathology, Pathology Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Eric Hsi
- Clinical Pathology, Pathology Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Yogen Saunthararajah
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
38
|
Grzelak CA, Sigglekow ND, Tirnitz-Parker JEE, Hamson EJ, Warren A, Maneck B, Chen J, Patkunanathan B, Boland J, Cheng R, Shackel NA, Seth D, Bowen DG, Martelotto LG, Watkins DN, McCaughan GW. Widespread GLI expression but limited canonical hedgehog signaling restricted to the ductular reaction in human chronic liver disease. PLoS One 2017; 12:e0171480. [PMID: 28187190 PMCID: PMC5302813 DOI: 10.1371/journal.pone.0171480] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/20/2017] [Indexed: 12/13/2022] Open
Abstract
Canonical Hedgehog (Hh) signaling in vertebrate cells occurs following Smoothened activation/translocation into the primary cilia (Pc), followed by a GLI transcriptional response. Nonetheless, GLI activation can occur independently of the canonical Hh pathway. Using a murine model of liver injury, we previously identified the importance of canonical Hh signaling within the Pc+ liver progenitor cell (LPC) population and noted that SMO-independent, GLI-mediated signals were important in multiple Pc-ve GLI2+ intrahepatic populations. This study extends these observations to human liver tissue, and analyses the effect of GLI inhibition on LPC viability/gene expression. Human donor and cirrhotic liver tissue specimens were evaluated for SHH, GLI2 and Pc expression using immunofluorescence and qRT-PCR. Changes to viability and gene expression in LPCs in vitro were assessed following GLI inhibition. Identification of Pc (as a marker of canonical Hh signaling) in human cirrhosis was predominantly confined to the ductular reaction and LPCs. In contrast, GLI2 was expressed in multiple cell populations including Pc-ve endothelium, hepatocytes, and leukocytes. HSCs/myofibroblasts (>99%) expressed GLI2, with only 1.92% displaying Pc. In vitro GLI signals maintained proliferation/viability within LPCs and GLI inhibition affected the expression of genes related to stemness, hepatocyte/biliary differentiation and Hh/Wnt signaling. At least two mechanisms of GLI signaling (Pc/SMO-dependent and Pc/SMO-independent) mediate chronic liver disease pathogenesis. This may have significant ramifications for the choice of Hh inhibitor (anti-SMO or anti-GLI) suitable for clinical trials. We also postulate GLI delivers a pro-survival signal to LPCs whilst maintaining stemness.
Collapse
Affiliation(s)
| | | | | | - Elizabeth Jane Hamson
- Liver Injury and Cancer, Centenary Institute, Camperdown, New South Wales, Australia
| | - Alessandra Warren
- Ageing and Alzheimers Institute, Centre for Education and Research on Ageing, University of Sydney, Concord Hospital, Sydney, New South Wales, Australia
- ANZAC Research Institute, University of Sydney, Concord Hospital, Sydney, New South Wales, Australia
| | - Bharvi Maneck
- Liver Injury and Cancer, Centenary Institute, Camperdown, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, R.P.A.H., Camperdown, New South Wales, Australia
| | - Jinbiao Chen
- Liver Injury and Cancer, Centenary Institute, Camperdown, New South Wales, Australia
| | | | - Jade Boland
- Liver Injury and Cancer, Centenary Institute, Camperdown, New South Wales, Australia
| | - Robert Cheng
- Liver Injury and Cancer, Centenary Institute, Camperdown, New South Wales, Australia
| | - Nicholas Adam Shackel
- Liver Injury and Cancer, Centenary Institute, Camperdown, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, R.P.A.H., Camperdown, New South Wales, Australia
- Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Devanshi Seth
- Liver Injury and Cancer, Centenary Institute, Camperdown, New South Wales, Australia
| | - David Geoffrey Bowen
- Liver Injury and Cancer, Centenary Institute, Camperdown, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, R.P.A.H., Camperdown, New South Wales, Australia
| | - Luciano Gastón Martelotto
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - D. Neil Watkins
- The Kinghorn Cancer Centre, Garvan Institute, Darlinghurst, New South Wales, Australia
| | - Geoffrey William McCaughan
- Liver Injury and Cancer, Centenary Institute, Camperdown, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, R.P.A.H., Camperdown, New South Wales, Australia
- Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
39
|
Köhn-Gaone J, Dwyer BJ, Grzelak CA, Miller G, Shackel NA, Ramm GA, McCaughan GW, Elsegood CL, Olynyk JK, Tirnitz-Parker JE. Divergent Inflammatory, Fibrogenic, and Liver Progenitor Cell Dynamics in Two Common Mouse Models of Chronic Liver Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1762-1774. [DOI: 10.1016/j.ajpath.2016.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/12/2016] [Accepted: 03/10/2016] [Indexed: 12/16/2022]
|