1
|
Ning Y, Zhou IY, Schaub JR, Rotile NJ, Boice A, Ay I, Turner S, Caravan P. Multimodal Imaging Demonstrates Antifibrotic Effects of Targeting αvβ6/αvβ1 Integrins in Biliary Fibrosis. Invest Radiol 2025:00004424-990000000-00333. [PMID: 40306257 DOI: 10.1097/rli.0000000000001202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
OBJECTIVES Development of molecular therapies for liver fibrosis is slowed by a lack of noninvasive methods addressing questions of target expression, target engagement, and treatment response. Integrin αvβ6 is a biomarker of liver fibrosis that is upregulated in livers of patients with primary sclerosing cholangitis. It activates latent TGF-β and plays a critical role in regulating extracellular matrix expression, especially collagen. In this study, our aim was to use combined αvβ6 integrin-targeted positron emission tomography (PET) and collagen-specific magnetic resonance imaging (MRI) to measure target expression/engagement and liver fibrosis reduction with a αvβ6 integrin inhibitor. MATERIALS AND METHODS We conducted a treatment study in bile duct-ligated (BDL) rats using a small molecule inhibitor to αvβ6/αvβ1. 68Ga-DOTA-R01-MG, an αvβ6-specific PET probe, was used to noninvasively measure αvβ6 expression and target engagement in the liver. CM-101, a type I collagen MRI probe, was used to quantify fibrosis. RESULTS 68Ga-DOTA-R01-MG PET showed 3-fold higher liver uptake in BDL rats compared with sham rats at 17 days after surgery. Pretreatment with high dose αvβ6/αvβ1 inhibitor 1 hour before imaging significantly decreased liver PET uptake in BDL rats (31%, P = 0.012). Two weeks of daily dosing with an αvβ6/αvβ1 inhibitor attenuated αvβ6 expression in BDL rat liver as assessed by αvβ6 PET (0.27 ± 0.07 percent injected dose [%ID]/mL compared with 0.40 ± 0.09 %ID/mL in vehicle-treated group, P = 0.014) and reduced liver fibrosis as assessed by collagen MRI (liver relaxation rate change ΔR1 = 0.14 ± 0.11 vs 0.36 ± 0.06, P = 0.0037). Imaging findings were confirmed by histology (collagen proportionate area 10.7 ± 2.8% vs 22.5 ± 6.1%, P < 0.001). CONCLUSIONS A single imaging protocol combining molecular MRI and PET can be used to effectively monitor integrin inhibitor treatment by measuring target expression/engagement and treatment outcomes. Multimodality molecular imaging may be valuable in accelerating drug development in molecular therapies for liver fibrosis.
Collapse
Affiliation(s)
- Yingying Ning
- From the Athinoula A. Martinos Center for Biomedical Imaging, Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA (Y.N., I.Y.Z., N.J.R., A.B., I.A., P.C.); and Pliant Therapeutics, Inc., South San Francisco, CA (J.R.S., S.T.)
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Jirouskova M, Harant K, Cejnar P, Ojha S, Korelova K, Sarnova L, Sticova E, Mayr CH, Schiller HB, Gregor M. Dynamics of compartment-specific proteomic landscapes of hepatotoxic and cholestatic models of liver fibrosis. eLife 2025; 13:RP98023. [PMID: 40197391 PMCID: PMC11978302 DOI: 10.7554/elife.98023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025] Open
Abstract
Accumulation of extracellular matrix (ECM) in liver fibrosis is associated with changes in protein abundance and composition depending upon etiology of the underlying liver disease. Current efforts to unravel etiology-specific mechanisms and pharmacological targets rely on several models of experimental fibrosis. Here, we characterize and compare dynamics of hepatic proteome remodeling during fibrosis development and spontaneous healing in experimental mouse models of hepatotoxic (carbon tetrachloride [CCl4] intoxication) and cholestatic (3,5-diethoxycarbonyl-1,4-dihydrocollidine [DDC] feeding) injury. Using detergent-based tissue extraction and mass spectrometry, we identified compartment-specific changes in the liver proteome with detailed attention to ECM composition and changes in protein solubility. Our analysis revealed distinct time-resolved CCl4 and DDC signatures, with identified signaling pathways suggesting limited healing and a potential for carcinogenesis associated with cholestasis. Correlation of protein abundance profiles with fibrous deposits revealed extracellular chaperone clusterin with implicated role in fibrosis resolution. Dynamics of clusterin expression was validated in the context of human liver fibrosis. Atomic force microscopy of fibrotic livers complemented proteomics with profiles of disease-associated changes in local liver tissue mechanics. This study determined compartment-specific proteomic landscapes of liver fibrosis and delineated etiology-specific ECM components, providing thus a foundation for future antifibrotic therapies.
Collapse
Affiliation(s)
- Marketa Jirouskova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Karel Harant
- Laboratory of Mass Spectrometry, BIOCEV, Faculty of Science, Charles UniversityPragueCzech Republic
| | - Pavel Cejnar
- Department of Mathematics, Informatics and Cybernetics, University of Chemistry and TechnologyPragueCzech Republic
| | - Srikant Ojha
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
- Department of Animal Physiology, Faculty of Science, Charles UniversityPragueCzech Republic
| | - Katerina Korelova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Lenka Sarnova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Eva Sticova
- Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental MedicinePragueCzech Republic
- Department of Pathology, The Third Faculty of Medicine, Charles University and University Hospital Kralovske VinohradyPragueCzech Republic
| | - Christoph H Mayr
- Helmholtz Munich, Research Unit Precision Regenerative Medicine; Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Herbert B Schiller
- Helmholtz Munich, Research Unit Precision Regenerative Medicine; Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL)MunichGermany
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians UniversityMunichGermany
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| |
Collapse
|
3
|
Yan X, Han Q, Wu W, Li H, Zhang W, Wang Y, Chen W, Yang A, You H. ITGA8 deficiency in hepatic stellate cells attenuates CCl 4-Induced liver fibrosis via suppression of COL11A1. Biochem Biophys Res Commun 2025; 756:151522. [PMID: 40056500 DOI: 10.1016/j.bbrc.2025.151522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND AND OBJECTIVE Liver fibrosis is a pathological process driven by chronic liver injury, characterized by excessive extracellular matrix (ECM) deposition due to hepatic stellate cell (HSC) activation. Integrins are critical regulators of ECM remodeling and HSC activation, yet the role of integrin α8(ITGA8) in liver fibrosis remains unclear. This study aims to investigate the function and underlying mechanisms of HSC-derived ITGA8 in liver fibrosis and evaluate the therapeutic potential of ITGA8-targeted intervention. METHODS A CCl4-induced mouse liver fibrosis model and public database analysis were used to assess ITGA8 expression and localization in liver fibrosis. AAV2/6-shItga8 was utilized to selectively silence HSC-derived ITGA8, and its effects on HSC activation and ECM accumulation were examined. In addition, in vitro ITGA8 knockdown combined with proteomic analysis was performed to explore the molecular mechanisms linking ITGA8 to ECM remodeling. RESULTS ITGA8 expression was significantly upregulated in fibrotic liver tissues across different etiologies, with a strong colocalization with HSCs. Silencing ITGA8 using AAV2/6-shItga8 effectively reduced liver fibrosis, as indicated by decreased hepatic inflammation, lower serum ALT levels, reduced inflammatory cell infiltration, and downregulated expression of pro-inflammatory cytokines. Fibrosis markers, including Sirius Red staining, type I collagen deposition, and α-SMA expression, were all reduced upon Itga8 silencing. Proteomic analysis revealed that ITGA8 regulates liver fibrosis through the ECM-receptor interaction pathway, with COL11A1 identified as a key downstream target. ITGA8 knockdown significantly suppressed COL11A1 expression, and reduced HSC-mediated collagen contraction, suggesting that ITGA8 contributes to ECM cross-linking and fibrosis progression via COL11A1 regulation. CONCLUSION This study demonstrates that HSC-derived ITGA8 promotes ECM accumulation and liver fibrosis progression by regulating COL11A1. Targeted silencing of ITGA8 via AAV2/6-shItga8 effectively alleviates liver fibrosis, providing new insights into ITGA8 as a potential therapeutic target for antifibrotic treatment.
Collapse
Affiliation(s)
- Xuzhen Yan
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China; Beijing Clinical Medicine Institute, Beijing, PR China; National Clinical Research Center of Digestive Diseases, Beijing, PR China
| | - Qi Han
- Liver Research Center & Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China; National Clinical Research Center of Digestive Diseases, Beijing, PR China
| | - Wenyue Wu
- Liver Research Center & Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China; National Clinical Research Center of Digestive Diseases, Beijing, PR China
| | - Hong Li
- Liver Research Center & Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China; National Clinical Research Center of Digestive Diseases, Beijing, PR China
| | - Wen Zhang
- Liver Research Center & Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China; National Clinical Research Center of Digestive Diseases, Beijing, PR China
| | - Yiwen Wang
- Liver Research Center & Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China; National Clinical Research Center of Digestive Diseases, Beijing, PR China
| | - Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China; Beijing Clinical Medicine Institute, Beijing, PR China; National Clinical Research Center of Digestive Diseases, Beijing, PR China
| | - Aiting Yang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China; Beijing Clinical Medicine Institute, Beijing, PR China; National Clinical Research Center of Digestive Diseases, Beijing, PR China.
| | - Hong You
- Liver Research Center & Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China; Beijing Clinical Medicine Institute, Beijing, PR China; National Clinical Research Center of Digestive Diseases, Beijing, PR China.
| |
Collapse
|
4
|
Gupta V, Sehrawat TS, Pinzani M, Strazzabosco M. Portal Fibrosis and the Ductular Reaction: Pathophysiological Role in the Progression of Liver Disease and Translational Opportunities. Gastroenterology 2025; 168:675-690. [PMID: 39251168 PMCID: PMC11885590 DOI: 10.1053/j.gastro.2024.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/27/2024] [Accepted: 07/20/2024] [Indexed: 09/11/2024]
Abstract
A consistent feature of chronic liver diseases and the hallmark of pathologic repair is the so-called "ductular reaction." This is a histologic abnormality characterized by an expansion of dysmorphic cholangiocytes inside and around portal spaces infiltrated by inflammatory, mesenchymal, and vascular cells. The ductular reaction is a highly regulated response based on the reactivation of morphogenetic signaling mechanisms and a complex crosstalk among a multitude of cell types. The nature and mechanism of these exchanges determine the difference between healthy regenerative liver repair and pathologic repair. An orchestrated signaling among cell types directs mesenchymal cells to deposit a specific extracellular matrix with distinct physical and biochemical properties defined as portal fibrosis. Progression of fibrosis leads to vast architectural and vascular changes known as "liver cirrhosis." The signals regulating the ecology of this microenvironment are just beginning to be addressed. Contrary to the tumor microenvironment, immune modulation inside this "benign" microenvironment is scarcely known. One of the reasons for this is that both the ductular reaction and portal fibrosis have been primarily considered a manifestation of cholestatic liver disease, whereas this phenomenon is also present, albeit with distinctive features, in all chronic human liver diseases. Novel human-derived cellular models and progress in "omics" technologies are increasing our knowledge at a fast pace. Most importantly, this knowledge is on the edge of generating new diagnostic and therapeutic advances. Here, we will critically review the latest advances, in terms of mechanisms, pathophysiology, and treatment prospects. In addition, we will delineate future avenues of research, including innovative translational opportunities.
Collapse
Affiliation(s)
- Vikas Gupta
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Tejasav S Sehrawat
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Massimo Pinzani
- UCL Institute for Liver & Digestive Health, Royal Free Hospital, London, United Kingdom; University of Pittsburgh Medical Center-Mediterranean Institute for Transplantation and Highly Specialized Therapies, Palermo, Italy
| | - Mario Strazzabosco
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut.
| |
Collapse
|
5
|
Cao L, Zhou Y, Lin S, Yang C, Guan Z, Li X, Yang S, Gao T, Zhao J, Fan N, Song Y, Li D, Li X, Li Z, Guan F, Tan Z. The trajectory of vesicular proteomic signatures from HBV-HCC by chitosan-magnetic bead-based separation and DIA-proteomic analysis. J Extracell Vesicles 2024; 13:e12499. [PMID: 39207047 PMCID: PMC11359709 DOI: 10.1002/jev2.12499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/04/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent primary liver cancer often associated with chronic hepatitis B virus infection (CHB) and liver cirrhosis (LC), underscoring the critical need for biomarker discovery to improve patient outcomes. Emerging as a promising avenue for biomarker development, proteomic technology leveraging liquid biopsy from small extracellular vesicles (sEV) offers new insights. Here, we evaluated various methods for sEV isolation and identified polysaccharide chitosan (CS) as an optimal approach. Subsequently, we employed optimized CS-based magnetic beads (Mag-CS) for sEV separation from serum samples of healthy controls, CHB, LC, and HBV-HCC patients. Leveraging data-independent acquisition mass spectrometry coupled with machine learning, we uncovered potential vesicular protein biomarker signatures (KNG1, F11, KLKB1, CAPNS1, CDH1, CPN2, NME2) capable of distinguishing HBV-HCC from CHB, LC, and non-HCC conditions. Collectively, our findings highlight the utility of Mag-CS-based sEV isolation for identifying early detection biomarkers in HBV-HCC.
Collapse
Affiliation(s)
- Lin Cao
- Institute of HematologyProvincial Key Laboratory of Biotechnology, School of MedicineNorthwest UniversityXi'anShaanxiChina
| | - Yue Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life SciencesNorthwest UniversityXi'anShaanxiChina
| | - Shuai Lin
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Chunyan Yang
- Institute of Basic and Translational MedicineXi'an Medical UniversityXi'anShaanxiChina
| | - Zixuan Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life SciencesNorthwest UniversityXi'anShaanxiChina
| | - Xiaofan Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life SciencesNorthwest UniversityXi'anShaanxiChina
| | - Shujie Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life SciencesNorthwest UniversityXi'anShaanxiChina
| | - Tong Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life SciencesNorthwest UniversityXi'anShaanxiChina
| | - Jiazhen Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life SciencesNorthwest UniversityXi'anShaanxiChina
| | - Ning Fan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life SciencesNorthwest UniversityXi'anShaanxiChina
| | - Yanan Song
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life SciencesNorthwest UniversityXi'anShaanxiChina
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anShaanxiP.R. China
| | - Xiang Li
- Institute of HematologyProvincial Key Laboratory of Biotechnology, School of MedicineNorthwest UniversityXi'anShaanxiChina
| | - Zhuo Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life SciencesNorthwest UniversityXi'anShaanxiChina
- Department of Laboratory MedicineThe First Affiliated Hospital of Xi'an Medical UniversityXi'anShaanxiP.R. China
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life SciencesNorthwest UniversityXi'anShaanxiChina
| | - Zengqi Tan
- Institute of HematologyProvincial Key Laboratory of Biotechnology, School of MedicineNorthwest UniversityXi'anShaanxiChina
| |
Collapse
|
6
|
Banerjee A, Farci P. Fibrosis and Hepatocarcinogenesis: Role of Gene-Environment Interactions in Liver Disease Progression. Int J Mol Sci 2024; 25:8641. [PMID: 39201329 PMCID: PMC11354981 DOI: 10.3390/ijms25168641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
The liver is a complex organ that performs vital functions in the body. Despite its extraordinary regenerative capacity compared to other organs, exposure to chemical, infectious, metabolic and immunologic insults and toxins renders the liver vulnerable to inflammation, degeneration and fibrosis. Abnormal wound healing response mediated by aberrant signaling pathways causes chronic activation of hepatic stellate cells (HSCs) and excessive accumulation of extracellular matrix (ECM), leading to hepatic fibrosis and cirrhosis. Fibrosis plays a key role in liver carcinogenesis. Once thought to be irreversible, recent clinical studies show that hepatic fibrosis can be reversed, even in the advanced stage. Experimental evidence shows that removal of the insult or injury can inactivate HSCs and reduce the inflammatory response, eventually leading to activation of fibrolysis and degradation of ECM. Thus, it is critical to understand the role of gene-environment interactions in the context of liver fibrosis progression and regression in order to identify specific therapeutic targets for optimized treatment to induce fibrosis regression, prevent HCC development and, ultimately, improve the clinical outcome.
Collapse
Affiliation(s)
- Anindita Banerjee
- Department of Transfusion Transmitted Diseases, ICMR-National Institute of Immunohaematology, Mumbai 400012, Maharashtra, India;
| | - Patrizia Farci
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Peng Z, Wei G, Huang P, Matta H, Gao W, An P, Zhao S, Lin Y, Tan L, Vaid K, Skelton-Badlani D, Nasser I, Budas G, Lopez D, Li L, Breckenridge D, Myers R, McHutchison J, Kuang M, Popov YV. ASK1/ p38 axis inhibition blocks the release of mitochondrial "danger signals" from hepatocytes and suppresses progression to cirrhosis and liver cancer. Hepatology 2024; 80:346-362. [PMID: 38377458 PMCID: PMC11477174 DOI: 10.1097/hep.0000000000000801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 12/14/2023] [Indexed: 02/22/2024]
Abstract
BACKGROUND AND AIMS Apoptosis Signal-regulating Kinase 1 (ASK1) is activated by various pathological stimuli and induces cell apoptosis through downstream p38 activation. We studied the effect of pharmacological ASK1 inhibition on cirrhosis and its sequelae using comprehensive preclinical in vivo and in vitro systems. APPROACH AND RESULTS Short-term (4-6 wk) and long-term (24-44 wk) ASK1 inhibition using small molecule GS-444217 was tested in thioacetamide-induced and BALB/c. Mdr2-/- murine models of cirrhosis and HCC, and in vitro using primary hepatocyte cell death assays. Short-term GS-444217 therapy in both models strongly reduced phosphorylated p38, hepatocyte death, and fibrosis by up to 50%. Profibrogenic release of mitochondrial DAMP mitochondrial deoxyribonucleic acid from dying hepatocytes was blocked by ASK1 or p38 inhibition. Long-term (24 wk) therapy in BALBc.Mdr2 - / - model resulted in a moderate 25% reduction in bridging fibrosis, but not in net collagen deposition. Despite this, the development of cirrhosis was effectively prevented, with strongly reduced p21 + hepatocyte staining (by 72%), serum ammonia levels (by 46%), and portal pressure (average 6.07 vs. 8.53 mm Hg in controls). Extended ASK1 inhibition for 44 wk in aged BALB/c. Mdr2-/- mice resulted in markedly reduced tumor number and size by ~50% compared to the control group. CONCLUSIONS ASK1 inhibition suppresses the profibrogenic release of mitochondrial deoxyribonucleic acid from dying hepatocytes in a p38-dependent manner and protects from liver fibrosis. Long-term ASK1 targeting resulted in diminished net antifibrotic effect, but the progression to liver cirrhosis and cancer in BALBc/ Mdr2- / - mice was effectively inhibited. These data support the clinical evaluation of ASK1 inhibitors in fibrotic liver diseases.
Collapse
Affiliation(s)
- Zhenwei Peng
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Guangyan Wei
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Pinzhu Huang
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Heansika Matta
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Wen Gao
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ping An
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Shuangshuang Zhao
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Yi Lin
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Li Tan
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Center of Hepatopbiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kahini Vaid
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Disha Skelton-Badlani
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Imad Nasser
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Grant Budas
- Gilead Sciences, Inc., Foster City, California, USA
| | - David Lopez
- Gilead Sciences, Inc., Foster City, California, USA
| | - Li Li
- Gilead Sciences, Inc., Foster City, California, USA
| | | | - Rob Myers
- Gilead Sciences, Inc., Foster City, California, USA
| | | | - Ming Kuang
- Center of Hepatopbiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Division of Interventional Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yury V Popov
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Schaub JR, Chen JY, Turner SM. Integrins in biliary injury and fibrosis. Curr Opin Gastroenterol 2024; 40:85-91. [PMID: 38190346 DOI: 10.1097/mog.0000000000000995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
PURPOSE OF REVIEW Current treatment options for cholangiopathies are severely limited and there is thus a critical need to identify and develop therapies. This review discusses the role of integrins in biliary injury and fibrosis and their potential as therapeutic targets. RECENT FINDINGS There are a diverse set of roles that integrins play in biliary injury and fibrosis. Some integrins activate TGF-β signaling or are involved in sensing of the extracellular matrix, making them attractive targets for biliary fibrosis. In recent work, autoantibodies to α v β 6 were identified in patients with PSC, supporting the relevance of this integrin in the disease. In addition, a role for α 2 β 1 in cyst formation was identified in a mouse model of polycystic liver disease. Leukocyte integrins (e.g. α E β 7 and α 4 β 7 ) contribute to lymphocyte trafficking, making them potential targets for biliary inflammation; however, this has not yet translated to the clinic. SUMMARY While all members of the same family of proteins, integrins have diverse roles in the pathogenesis of biliary disease. Targeting one or multiple of these integrins may slow or halt the progression of biliary injury and fibrosis by simultaneously impacting different pathologic cells and processes.
Collapse
Affiliation(s)
| | - Jennifer Y Chen
- Department of Medicine
- The Liver Center, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | | |
Collapse
|
9
|
Shakour N, Karami S, Iranshahi M, Butler AE, Sahebkar A. Antifibrotic effects of sodium-glucose cotransporter-2 inhibitors: A comprehensive review. Diabetes Metab Syndr 2024; 18:102934. [PMID: 38154403 DOI: 10.1016/j.dsx.2023.102934] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/25/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND AND AIMS Scar tissue accumulation in organs is the underlying cause of many fibrotic diseases. Due to the extensive array of organs affected, the long-term nature of fibrotic processes and the large number of people who suffer from the negative impact of these diseases, they constitute a serious health problem for modern medicine and a huge economic burden on society. Sodium-glucose cotransporter-2 inhibitors (SGLT2is) are a relatively new class of anti-diabetic pharmaceuticals that offer additional benefits over and above their glucose-lowering properties; these medications modulate a variety of diseases, including fibrosis. Herein, we have collated and analyzed all available research on SGLT2is and their effects on organ fibrosis, together with providing a proposed explanation as to the underlying mechanisms. METHODS PubMed, ScienceDirect, Google Scholar and Scopus were searched spanning the period from 2012 until April 2023 to find relevant articles describing the antifibrotic effects of SGLT2is. RESULTS The majority of reports have shown that SGLT2is are protective against lung, liver, heart and kidney fibrosis as well as arterial stiffness. According to the results of clinical trials and animal studies, many SGLT2 inhibitors are promising candidates for the treatment of fibrosis. Recent studies have demonstrated that SGLT2is affect an array of cellular processes, including hypoxia, inflammation, oxidative stress, the renin-angiotensin system and metabolic activities, all of which have been linked to fibrosis. CONCLUSION Extensive evidence indicates that SGLT2is are promising treatments for fibrosis, demonstrating protective effects in various organs and influencing key cellular processes linked to fibrosis.
Collapse
Affiliation(s)
- Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Karami
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Li S, Sampson C, Liu C, Piao HL, Liu HX. Integrin signaling in cancer: bidirectional mechanisms and therapeutic opportunities. Cell Commun Signal 2023; 21:266. [PMID: 37770930 PMCID: PMC10537162 DOI: 10.1186/s12964-023-01264-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/09/2023] [Indexed: 09/30/2023] Open
Abstract
Integrins are transmembrane receptors that possess distinct ligand-binding specificities in the extracellular domain and signaling properties in the cytoplasmic domain. While most integrins have a short cytoplasmic tail, integrin β4 has a long cytoplasmic tail that can indirectly interact with the actin cytoskeleton. Additionally, 'inside-out' signals can induce integrins to adopt a high-affinity extended conformation for their appropriate ligands. These properties enable integrins to transmit bidirectional cellular signals, making it a critical regulator of various biological processes.Integrin expression and function are tightly linked to various aspects of tumor progression, including initiation, angiogenesis, cell motility, invasion, and metastasis. Certain integrins have been shown to drive tumorigenesis or amplify oncogenic signals by interacting with corresponding receptors, while others have marginal or even suppressive effects. Additionally, different α/β subtypes of integrins can exhibit opposite effects. Integrin-mediated signaling pathways including Ras- and Rho-GTPase, TGFβ, Hippo, Wnt, Notch, and sonic hedgehog (Shh) are involved in various stages of tumorigenesis. Therefore, understanding the complex regulatory mechanisms and molecular specificities of integrins are crucial to delaying cancer progression and suppressing tumorigenesis. Furthermore, the development of integrin-based therapeutics for cancer are of great importance.This review provides an overview of integrin-dependent bidirectional signaling mechanisms in cancer that can either support or oppose tumorigenesis by interacting with various signaling pathways. Finally, we focus on the future opportunities for emergent therapeutics based on integrin agonists. Video Abstract.
Collapse
Affiliation(s)
- Siyi Li
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Chibuzo Sampson
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Changhao Liu
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Hai-Long Piao
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| | - Hong-Xu Liu
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| |
Collapse
|
11
|
Roy A, Shi L, Chang A, Dong X, Fernandez A, Kraft JC, Li J, Le VQ, Winegar RV, Cherf GM, Slocum D, Poulson PD, Casper GE, Vallecillo-Zúniga ML, Valdoz JC, Miranda MC, Bai H, Kipnis Y, Olshefsky A, Priya T, Carter L, Ravichandran R, Chow CM, Johnson MR, Cheng S, Smith M, Overed-Sayer C, Finch DK, Lowe D, Bera AK, Matute-Bello G, Birkland TP, DiMaio F, Raghu G, Cochran JR, Stewart LJ, Campbell MG, Van Ry PM, Springer T, Baker D. De novo design of highly selective miniprotein inhibitors of integrins αvβ6 and αvβ8. Nat Commun 2023; 14:5660. [PMID: 37704610 PMCID: PMC10500007 DOI: 10.1038/s41467-023-41272-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023] Open
Abstract
The RGD (Arg-Gly-Asp)-binding integrins αvβ6 and αvβ8 are clinically validated cancer and fibrosis targets of considerable therapeutic importance. Compounds that can discriminate between homologous αvβ6 and αvβ8 and other RGD integrins, stabilize specific conformational states, and have high thermal stability could have considerable therapeutic utility. Existing small molecule and antibody inhibitors do not have all these properties, and hence new approaches are needed. Here we describe a generalized method for computationally designing RGD-containing miniproteins selective for a single RGD integrin heterodimer and conformational state. We design hyperstable, selective αvβ6 and αvβ8 inhibitors that bind with picomolar affinity. CryoEM structures of the designed inhibitor-integrin complexes are very close to the computational design models, and show that the inhibitors stabilize specific conformational states of the αvβ6 and the αvβ8 integrins. In a lung fibrosis mouse model, the αvβ6 inhibitor potently reduced fibrotic burden and improved overall lung mechanics, demonstrating the therapeutic potential of de novo designed integrin binding proteins with high selectivity.
Collapse
Affiliation(s)
- Anindya Roy
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Lei Shi
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Encodia Inc, 5785 Oberlin Drive, San Diego, CA, 92121, USA
| | - Ashley Chang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Xianchi Dong
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Engineering Research Center of Protein and Peptide Medicine, Ministry of Education, Nanjing, China
| | - Andres Fernandez
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - John C Kraft
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
| | - Viet Q Le
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
| | - Rebecca Viazzo Winegar
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Gerald Maxwell Cherf
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Denali Therapeutics, South San Francisco, CA, USA
| | - Dean Slocum
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA
| | - P Daniel Poulson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Garrett E Casper
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | | | - Jonard Corpuz Valdoz
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA
| | - Marcos C Miranda
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Hua Bai
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Yakov Kipnis
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Audrey Olshefsky
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Tanu Priya
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Lauren Carter
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Rashmi Ravichandran
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Cameron M Chow
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Max R Johnson
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Suna Cheng
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - McKaela Smith
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Catherine Overed-Sayer
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Donna K Finch
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Alchemab Therapeutics Ltd, Cambridge, UK
| | - David Lowe
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Evox Therapeutics Limited, Oxford Science Park, Medawar Centre, East Building, Robert Robinson Avenue, Oxford, OX4 4HG, England
| | - Asim K Bera
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Gustavo Matute-Bello
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, USA
| | - Timothy P Birkland
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Frank DiMaio
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Ganesh Raghu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Dept of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Lance J Stewart
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Melody G Campbell
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
| | - Pam M Van Ry
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, 84602, USA.
| | - Timothy Springer
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, MA, USA.
| | - David Baker
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
12
|
Roy A, Shi L, Chang A, Dong X, Fernandez A, Kraft JC, Li J, Le VQ, Winegar RV, Cherf GM, Slocum D, Daniel Poulson P, Casper GE, Vallecillo-Zúniga ML, Valdoz JC, Miranda MC, Bai H, Kipnis Y, Olshefsky A, Priya T, Carter L, Ravichandran R, Chow CM, Johnson MR, Cheng S, Smith M, Overed-Sayer C, Finch DK, Lowe D, Bera AK, Matute-Bello G, Birkland TP, DiMaio F, Raghu G, Cochran JR, Stewart LJ, Campbell MG, Van Ry PM, Springer T, Baker D. De novo design of highly selective miniprotein inhibitors of integrins αvβ6 and αvβ8. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544624. [PMID: 37398153 PMCID: PMC10312613 DOI: 10.1101/2023.06.12.544624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The RGD (Arg-Gly-Asp)-binding integrins αvβ6 and αvβ8 are clinically validated cancer and fibrosis targets of considerable therapeutic importance. Compounds that can discriminate between the two closely related integrin proteins and other RGD integrins, stabilize specific conformational states, and have sufficient stability enabling tissue restricted administration could have considerable therapeutic utility. Existing small molecules and antibody inhibitors do not have all of these properties, and hence there is a need for new approaches. Here we describe a method for computationally designing hyperstable RGD-containing miniproteins that are highly selective for a single RGD integrin heterodimer and conformational state, and use this strategy to design inhibitors of αvβ6 and αvβ8 with high selectivity. The αvβ6 and αvβ8 inhibitors have picomolar affinities for their targets, and >1000-fold selectivity over other RGD integrins. CryoEM structures are within 0.6-0.7Å root-mean-square deviation (RMSD) to the computational design models; the designed αvβ6 inhibitor and native ligand stabilize the open conformation in contrast to the therapeutic anti-αvβ6 antibody BG00011 that stabilizes the bent-closed conformation and caused on-target toxicity in patients with lung fibrosis, and the αvβ8 inhibitor maintains the constitutively fixed extended-closed αvβ8 conformation. In a mouse model of bleomycin-induced lung fibrosis, the αvβ6 inhibitor potently reduced fibrotic burden and improved overall lung mechanics when delivered via oropharyngeal administration mimicking inhalation, demonstrating the therapeutic potential of de novo designed integrin binding proteins with high selectivity.
Collapse
Affiliation(s)
- Anindya Roy
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Lei Shi
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Current Address: Encodia Inc, 5785 Oberlin Drive, San Diego, CA 92121
| | - Ashley Chang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Xianchi Dong
- Program in Cellular and Molecular Medicine, Children’s Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
- Current address: State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China; Engineering Research Center of Protein and Peptide Medicine,Ministry of Education
| | - Andres Fernandez
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - John C. Kraft
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Children’s Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - Viet Q. Le
- Program in Cellular and Molecular Medicine, Children’s Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - Rebecca Viazzo Winegar
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Gerald Maxwell Cherf
- Department of Bioengineering, Stanford University, Stanford CA 94305
- Current Address: Denali Therapeutics, South San Francisco, CA, USA
| | - Dean Slocum
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - P. Daniel Poulson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Garrett E. Casper
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | | | - Jonard Corpuz Valdoz
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Marcos C. Miranda
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Current Address: Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Hua Bai
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Yakov Kipnis
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Audrey Olshefsky
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Tanu Priya
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Current Address: Department of Pharmacology, Northwestern University Feinberg School of Medicine; Chicago, IL 60611, USA
| | - Lauren Carter
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Rashmi Ravichandran
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Cameron M. Chow
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Max R. Johnson
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Suna Cheng
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - McKaela Smith
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Catherine Overed-Sayer
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
- Current Address: Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Donna K. Finch
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
- Current Address: Alchemab Therapeutics Ltd, Cambridge, United Kingdom
| | - David Lowe
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
- Current Address: Evox Therapeutics Limited, Oxford Science Park, Medawar Centre, East Building, Robert Robinson Avenue, Oxford, OX4 4HG
| | - Asim K. Bera
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Gustavo Matute-Bello
- Center for Lung Biology, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington
| | - Timothy P Birkland
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Frank DiMaio
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Ganesh Raghu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | | | - Lance J. Stewart
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Melody G. Campbell
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Pam M. Van Ry
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Timothy Springer
- Program in Cellular and Molecular Medicine, Children’s Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - David Baker
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
13
|
Cai X, Tacke F, Guillot A, Liu H. Cholangiokines: undervalued modulators in the hepatic microenvironment. Front Immunol 2023; 14:1192840. [PMID: 37261338 PMCID: PMC10229055 DOI: 10.3389/fimmu.2023.1192840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/02/2023] [Indexed: 06/02/2023] Open
Abstract
The biliary epithelial cells, also known as cholangiocytes, line the intra- and extrahepatic bile ducts, forming a barrier between intra- and extra-ductal environments. Cholangiocytes are mostly known to modulate bile composition and transportation. In hepatobiliary diseases, bile duct injury leads to drastic alterations in cholangiocyte phenotypes and their release of soluble mediators, which can vary depending on the original insult and cellular states (quiescence, senescence, or proliferation). The cholangiocyte-secreted cytokines (also termed cholangiokines) drive ductular cell proliferation, portal inflammation and fibrosis, and carcinogenesis. Hence, despite the previous consensus that cholangiocytes are bystanders in liver diseases, their diverse secretome plays critical roles in modulating the intrahepatic microenvironment. This review summarizes recent insights into the cholangiokines under both physiological and pathological conditions, especially as they occur during liver injury-regeneration, inflammation, fibrosis and malignant transformation processes.
Collapse
Affiliation(s)
- Xiurong Cai
- Department of Hematology, Oncology and Tumor Immunology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Hanyang Liu
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
- Center of Gastrointestinal Diseases, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
14
|
Hernandez JC, Chen CL, Machida T, Uthaya Kumar DB, Tahara SM, Montana J, Sher L, Liang J, Jung JU, Tsukamoto H, Machida K. LIN28 and histone H3K4 methylase induce TLR4 to generate tumor-initiating stem-like cells. iScience 2023; 26:106254. [PMID: 36949755 PMCID: PMC10025994 DOI: 10.1016/j.isci.2023.106254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/09/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Chemoresistance and plasticity of tumor-initiating stem-like cells (TICs) promote tumor recurrence and metastasis. The gut-originating endotoxin-TLR4-NANOG oncogenic axis is responsible for the genesis of TICs. This study investigated mechanisms as to how TICs arise through transcriptional, epigenetic, and post-transcriptional activation of oncogenic TLR4 pathways. Here, we expressed constitutively active TLR4 (caTLR4) in mice carrying pLAP-tTA or pAlb-tTA, under a tetracycline withdrawal-inducible system. Liver progenitor cell induction accelerated liver tumor development in caTLR4-expressing mice. Lentiviral shRNA library screening identified histone H3K4 methylase SETD7 as central to activation of TLR4. SETD7 combined with hypoxia induced TLR4 through HIF2 and NOTCH. LIN28 post-transcriptionally stabilized TLR4 mRNA via de-repression of let-7 microRNA. These results supported a LIN28-TLR4 pathway for the development of HCCs in a hypoxic microenvironment. These findings not only advance our understanding of molecular mechanisms responsible for TIC generation in HCC, but also represent new therapeutic targets for the treatment of HCC.
Collapse
Affiliation(s)
- Juan Carlos Hernandez
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
- MS Biotechnology Program, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Chia-Lin Chen
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
- Department of Life Sciences & Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 110, Taiwan
| | - Tatsuya Machida
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Dinesh Babu Uthaya Kumar
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Stanley M. Tahara
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Jared Montana
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Linda Sher
- Department of Surgery, University of Southern California, Los Angeles, CA 90033, USA
| | | | - Jae U. Jung
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Hidekazu Tsukamoto
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA
- Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Keigo Machida
- Departments of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, CA, USA
| |
Collapse
|
15
|
Abstract
AIM Fibrosis is a common pathological feature of most types of chronic liver injuries. There is no specific treatment for liver fibrosis at present. The liver microenvironment, which fosters the survival and activity of liver cells, plays an important role in maintaining the normal structure and physiological function of the liver. The aim of this review is to deeply understand the role of the liver microenvironment in the dynamic and complicated development of liver fibrosis. METHODS After searching in Elsevier ScienceDirect, PubMed and Web of Science databases using 'liver fibrosis' and 'microenvironment' as keywords, studies related to microenvironment in liver fibrosis was compiled and examined. RESULTS The homeostasis of the liver microenvironment is disrupted during the development of liver fibrosis, affecting liver cell function, causing various types of cell reactions, and changing the cell-cell and cell-matrix interactions, eventually affecting fibrosis formation. CONCLUSION Liver microenvironment may be important for identifying potential therapeutic targets, and restoring microenvironment homeostasis may be an important strategy for promoting the reversal of liver fibrosis.KEY MESSAGESThe homeostasis of the liver microenvironment is disrupted in liver fibrosis;A pro-fibrotic microenvironment is formed during the development of liver fibrosis;Restoring microenvironment homeostasis may be an important strategy for promoting the reversal of liver fibrosis.
Collapse
Affiliation(s)
- Ying Meng
- Department of General Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Tong Zhao
- Department of Orthopedics, Lanzhou University First Hospital, Lanzhou, Gansu, China
| | - Zhengyi Zhang
- Department of General Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Dekui Zhang
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
16
|
Canesin G, Feldbrügge L, Wei G, Janovicova L, Janikova M, Csizmadia E, Ariffin J, Hedblom A, Herbert ZT, Robson SC, Celec P, Swanson KD, Nasser I, Popov YV, Wegiel B. Heme oxygenase-1 mitigates liver injury and fibrosis via modulation of LNX1/Notch1 pathway in myeloid cells. iScience 2022; 25:104983. [PMID: 36093061 PMCID: PMC9450142 DOI: 10.1016/j.isci.2022.104983] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/01/2022] [Accepted: 08/16/2022] [Indexed: 01/12/2023] Open
Abstract
Activation of resident macrophages (Mϕ) and hepatic stellate cells is a key event in chronic liver injury. Mice with heme oxygenase-1 (HO-1; Hmox1)-deficient Mϕ (LysM-Cre:Hmox1 flfl ) exhibit increased inflammation, periportal ductular reaction, and liver fibrosis following bile duct ligation (BDL)-induced liver injury and increased pericellular fibrosis in NASH model. RiboTag-based RNA-sequencing profiling of hepatic HO-1-deficient Mϕ revealed dysregulation of multiple genes involved in lipid and amino acid metabolism, regulation of oxidative stress, and extracellular matrix turnover. Among these genes, ligand of numb-protein X1 (LNX1) expression is strongly suppressed in HO-1-deficient Mϕ. Importantly, HO-1 and LNX1 were expressed by hepatic Mϕ in human biliary and nonbiliary end-stage cirrhosis. We found that Notch1 expression, a downstream target of LNX1, was increased in LysM-Cre:Hmox1 flfl mice. In HO-1-deficient Mϕ treated with heme, transient overexpression of LNX1 drives M2-like Mϕ polarization. In summary, we identified LNX1/Notch1 pathway as a downstream target of HO-1 in liver fibrosis.
Collapse
Affiliation(s)
- Giacomo Canesin
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Linda Feldbrügge
- Charité – Universitätsmedizin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, 13353 Berlin, Germany,Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Guangyan Wei
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Department of Radiation Oncology, First Affiliated Hospital, Sun Yat-sen University, 510080 Guangzhou, China
| | - Lubica Janovicova
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Institute of Molecular Biomedicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia
| | - Monika Janikova
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Institute of Molecular Biomedicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia
| | - Eva Csizmadia
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Juliana Ariffin
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andreas Hedblom
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Zachary T. Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Simon C. Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Peter Celec
- Institute of Molecular Biomedicine, Comenius University in Bratislava, 811 08 Bratislava, Slovakia
| | - Kenneth D. Swanson
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Imad Nasser
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yury V. Popov
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Corresponding author
| | - Barbara Wegiel
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA,Corresponding author
| |
Collapse
|
17
|
Delacroix C, Hulot JS. [Integrins in cardiac fibrosis]. Med Sci (Paris) 2022; 38:438-444. [PMID: 35608466 DOI: 10.1051/medsci/2022055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
For the last 20 years, integrins have been a therapeutic target of interest in the treatment of fibrotic diseases, particularly regarding the integrins of the αV family. Initially developed as anti-cancer drugs but with modest benefits, inhibitors of integrins (such as the anti-αV cilengitide) have shown interesting anti-fibrotic effects in different organs including the heart. Cardiac fibrosis is defined as an accumulation of stiff extracellular matrix in the myocardium, and ultimately leads to heart failure, one of the leading causes of mortality worldwide. Understanding the determinants of cardiac fibrosis and the involvement of integrins is a major matter of public health. This review presents the current knowledge on the different types of cardiac fibrosis and their etiologies, and report on first data supporting specific integrin inhibition therapy as a novel anti-fibrotic strategy, in particular to treat cardiac fibrosis.
Collapse
Affiliation(s)
- Clément Delacroix
- Paris Centre de recherche cardiovasculaire (PARCC), Inserm U.970, 56 rue Leblanc, 75015 Paris, France
| | - Jean-Sébastien Hulot
- Paris Centre de recherche cardiovasculaire (PARCC), Inserm U.970, 56 rue Leblanc, 75015 Paris, France
| |
Collapse
|
18
|
Rahman SR, Roper JA, Grove JI, Aithal GP, Pun KT, Bennett AJ. Integrins as a drug target in liver fibrosis. Liver Int 2022; 42:507-521. [PMID: 35048542 DOI: 10.1111/liv.15157] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/30/2021] [Indexed: 02/06/2023]
Abstract
As the worldwide prevalence of chronic liver diseases is high and continuing to increase, there is an urgent need for treatment to prevent cirrhosis-related morbidity and mortality. Integrins are heterodimeric cell-surface proteins that are promising targets for therapeutic intervention. αv integrins are central in the development of fibrosis as they activate latent TGFβ, a known profibrogenic cytokine. The αv subunit can form heterodimers with β1, β3, β5, β6 or β8 subunits and one or more of these integrins are central to the development of liver fibrosis, however, their relative importance is not understood. This review summarises the current knowledge of αv integrins and their respective β subunits in different organs, with a focus on liver fibrosis and the emerging preclinical and clinical data with regards to αv integrin inhibitors.
Collapse
Affiliation(s)
- Syedia R Rahman
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK.,FRAME Alternatives Laboratory, Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK.,Nottingham Digestive Diseases Centre, Translational Medical Sciences, Medicine, University of Nottingham, Nottingham, UK
| | - James A Roper
- Novel Human Genetics Research Unit, GlaxoSmithKline, Stevenage, UK
| | - Jane I Grove
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK.,Nottingham Digestive Diseases Centre, Translational Medical Sciences, Medicine, University of Nottingham, Nottingham, UK
| | - Guruprasad P Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK.,Nottingham Digestive Diseases Centre, Translational Medical Sciences, Medicine, University of Nottingham, Nottingham, UK
| | - K Tao Pun
- Novel Human Genetics Research Unit, GlaxoSmithKline, Stevenage, UK
| | - Andrew J Bennett
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK.,FRAME Alternatives Laboratory, Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK.,Nottingham Digestive Diseases Centre, Translational Medical Sciences, Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
19
|
Li Y, Fan W, Link F, Wang S, Dooley S. Transforming growth factor β latency: A mechanism of cytokine storage and signalling regulation in liver homeostasis and disease. JHEP REPORTS : INNOVATION IN HEPATOLOGY 2022; 4:100397. [PMID: 35059619 PMCID: PMC8760520 DOI: 10.1016/j.jhepr.2021.100397] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022]
Abstract
Transforming growth factor-β (TGF-β) is a potent effector in the liver, which is involved in a plethora of processes initiated upon liver injury. TGF-β affects parenchymal, non-parenchymal, and inflammatory cells in a highly context-dependent manner. Its bioavailability is critical for a fast response to various insults. In the liver – and probably in other organs – this is made possible by the deposition of a large portion of TGF-β in the extracellular matrix as an inactivated precursor form termed latent TGF-β (L-TGF-β). Several matrisomal proteins participate in matrix deposition, latent complex stabilisation, and activation of L-TGF-β. Extracellular matrix protein 1 (ECM1) was recently identified as a critical factor in maintaining the latency of deposited L-TGF-β in the healthy liver. Indeed, its depletion causes spontaneous TGF-β signalling activation with deleterious effects on liver architecture and function. This review article presents the current knowledge on intracellular L-TGF-β complex formation, secretion, matrix deposition, and activation and describes the proteins and processes involved. Further, we emphasise the therapeutic potential of toning down L-TGF-β activation in liver fibrosis and liver cancer.
Collapse
Affiliation(s)
- Yujia Li
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Weiguo Fan
- Division of Gastroenterology and Hepatology, Stanford University, Stanford CA, USA
| | - Frederik Link
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sai Wang
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Tel.: 06213835595.
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Corresponding authors. Addresses: Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Tel.: 06213833768;
| |
Collapse
|
20
|
Airik M, McCourt B, Ozturk TT, Huynh AB, Zhang X, Tometich JT, Topaloglu R, Ozen H, Orhan D, Nejak-Bowen K, Monga SP, Hand TW, Ozaltin F, Airik R. Mitigation of portal fibrosis and cholestatic liver disease in ANKS6-deficient livers by macrophage depletion. FASEB J 2022; 36:e22157. [PMID: 35032404 PMCID: PMC8852242 DOI: 10.1096/fj.202101387r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/19/2021] [Accepted: 12/27/2021] [Indexed: 02/03/2023]
Abstract
Congenital hepatic fibrosis (CHF) is a developmental liver disease that is caused by mutations in genes that encode ciliary proteins and is characterized by bile duct dysplasia and portal fibrosis. Recent work has demonstrated that mutations in ANKS6 can cause CHF due to its role in bile duct development. Here, we report a novel ANKS6 mutation, which was identified in an infant presenting with neonatal jaundice due to underlying biliary abnormalities and liver fibrosis. Molecular analysis revealed that ANKS6 liver pathology is associated with the infiltration of inflammatory macrophages to the periportal fibrotic tissue and ductal epithelium. To further investigate the role of macrophages in CHF pathophysiology, we generated a novel liver-specific Anks6 knockout mouse model. The mutant mice develop biliary abnormalities and rapidly progressing periportal fibrosis reminiscent of human CHF. The development of portal fibrosis in Anks6 KO mice coincided with the accumulation of inflammatory monocytes and macrophages in the mutant liver. Gene expression and flow cytometric analysis demonstrated the preponderance of M1- over M2-like macrophages at the onset of fibrosis. A critical role for macrophages in promoting peribiliary fibrosis was demonstrated by depleting the macrophages with clodronate liposomes which effectively reduced inflammatory gene expression and fibrosis, and ameliorated tissue histology and biliary function in Anks6 KO livers. Together, this study demonstrates that macrophages play an important role in the initiation of liver fibrosis in ANKS6-deficient livers and their therapeutic elimination may provide an avenue to mitigate CHF in patients.
Collapse
Affiliation(s)
- Merlin Airik
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Blake McCourt
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tugba Tastemel Ozturk
- Division of Pediatric Nephrology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Amy B Huynh
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoyi Zhang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Justin T Tometich
- R.K. Mellon Institute for Pediatric Research, Department of Pediatrics, Division of Infectious Disease, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, 15224
| | - Rezan Topaloglu
- Division of Pediatric Nephrology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Hasan Ozen
- Division of Gastroenterology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Diclehan Orhan
- Pediatric Pathology Unit, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Kari Nejak-Bowen
- Department of Pathology and Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Satdarshan P Monga
- Department of Pathology and Pittsburgh Liver Research Center, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Timothy W Hand
- R.K. Mellon Institute for Pediatric Research, Department of Pediatrics, Division of Infectious Disease, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, 15224
| | - Fatih Ozaltin
- Division of Pediatric Nephrology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
- Nephrogenetics Laboratory, Division of Pediatric Nephrology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Rannar Airik
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
21
|
Salvati A, Poelstra K. Drug Targeting and Nanomedicine: Lessons Learned from Liver Targeting and Opportunities for Drug Innovation. Pharmaceutics 2022; 14:217. [PMID: 35057111 PMCID: PMC8777931 DOI: 10.3390/pharmaceutics14010217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 02/08/2023] Open
Abstract
Drug targeting and nanomedicine are different strategies for improving the delivery of drugs to their target. Several antibodies, immuno-drug conjugates and nanomedicines are already approved and used in clinics, demonstrating the potential of such approaches, including the recent examples of the DNA- and RNA-based vaccines against COVID-19 infections. Nevertheless, targeting remains a major challenge in drug delivery and different aspects of how these objects are processed at organism and cell level still remain unclear, hampering the further development of efficient targeted drugs. In this review, we compare properties and advantages of smaller targeted drug constructs on the one hand, and larger nanomedicines carrying higher drug payload on the other hand. With examples from ongoing research in our Department and experiences from drug delivery to liver fibrosis, we illustrate opportunities in drug targeting and nanomedicine and current challenges that the field needs to address in order to further improve their success.
Collapse
Affiliation(s)
- Anna Salvati
- Correspondence: (A.S.); (K.P.); Tel.: +31-503639831 (A.S.); +31-503633287 (K.P.)
| | - Klaas Poelstra
- Correspondence: (A.S.); (K.P.); Tel.: +31-503639831 (A.S.); +31-503633287 (K.P.)
| |
Collapse
|
22
|
Ong CH, Tham CL, Harith HH, Firdaus N, Israf DA. TGF-β-induced fibrosis: A review on the underlying mechanism and potential therapeutic strategies. Eur J Pharmacol 2021; 911:174510. [PMID: 34560077 DOI: 10.1016/j.ejphar.2021.174510] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/10/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022]
Abstract
Transforming growth factor-beta (TGF-β) plays multiple homeostatic roles in the regulation of inflammation, proliferation, differentiation and would healing of various tissues. Many studies have demonstrated that TGF-β stimulates activation and proliferation of fibroblasts, which result in extracellular matrix deposition. Its increased expression can result in many fibrotic diseases, and the level of expression is often correlated with disease severity. On this basis, inhibition of TGF-β and its activity has great therapeutic potential for the treatment of various fibrotic diseases such as pulmonary fibrosis, renal fibrosis, systemic sclerosis and etc. By understanding the molecular mechanism of TGF-β signaling and activity, researchers were able to develop different strategies in order to modulate the activity of TGF-β. Antisense oligonucleotide was developed to target the mRNA of TGF-β to inhibit its expression. There are also neutralizing monoclonal antibodies that can target the TGF-β ligands or αvβ6 integrin to prevent binding to receptor or activation of latent TGF-β respectively. Soluble TGF-β receptors act as ligand traps that competitively bind to the TGF-β ligands. Many small molecule inhibitors have been developed to inhibit the TGF-β receptor at its cytoplasmic domain and also intracellular signaling molecules. Peptide aptamer technology has been used to target downstream TGF-β signaling. Here, we summarize the underlying mechanism of TGF-β-induced fibrosis and also review various strategies of inhibiting TGF-β in both preclinical and clinical studies.
Collapse
Affiliation(s)
- Chun Hao Ong
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43300, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43300, Malaysia
| | - Hanis Hazeera Harith
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43300, Malaysia
| | - Nazmi Firdaus
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43300, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43300, Malaysia.
| |
Collapse
|
23
|
Wilkinson AL, John AE, Barrett JW, Gower E, Morrison VS, Man Y, Pun KT, Roper JA, Luckett JC, Borthwick LA, Barksby BS, Burgoyne RA, Barnes R, Fisher AJ, Procopiou PA, Hatley RJD, Barrett TN, Marshall RP, Macdonald SJF, Jenkins RG, Slack RJ. Pharmacological characterisation of GSK3335103, an oral αvβ6 integrin small molecule RGD-mimetic inhibitor for the treatment of fibrotic disease. Eur J Pharmacol 2021; 913:174618. [PMID: 34762934 DOI: 10.1016/j.ejphar.2021.174618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/20/2021] [Accepted: 11/03/2021] [Indexed: 11/18/2022]
Abstract
Fibrosis is the formation of scar tissue due to injury or long-term inflammation and is a leading cause of morbidity and mortality. Activation of the pro-fibrotic cytokine transforming growth factor-β (TGFβ) via the alpha-V beta-6 (αvβ6) integrin has been identified as playing a key role in the development of fibrosis. Therefore, a drug discovery programme to identify an orally bioavailable small molecule αvβ6 arginyl-glycinyl-aspartic acid (RGD)-mimetic was initiated. As part of a medicinal chemistry programme GSK3335103 was identified and profiled in a range of pre-clinical in vitro and in vivo systems. GSK3335103 was shown to bind to the αvβ6 with high affinity and demonstrated fast binding kinetics. In primary human lung epithelial cells, GSK3335103-induced concentration- and time-dependent internalisation of αvβ6 with a rapid return of integrin to the cell surface observed after washout. Following sustained engagement of the αvβ6 integrin in vitro, lysosomal degradation was induced by GSK3335103. GSK3335103 was shown to engage with the αvβ6 integrin and inhibit the activation of TGFβ in both ex vivo IPF tissue and in a murine model of bleomycin-induced lung fibrosis, as measured by αvβ6 engagement, TGFβ signalling and collagen deposition, with a prolonged duration of action observed in vivo. In summary, GSK3335103 is a potent αvβ6 inhibitor that attenuates TGFβ signalling in vitro and in vivo with a well-defined pharmacokinetic/pharmacodynamic relationship. This translates to a significant reduction of collagen deposition in vivo and therefore GSK3335103 represents a potential novel oral therapy for fibrotic disorders.
Collapse
Affiliation(s)
- Alex L Wilkinson
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Alison E John
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, UK
| | - John W Barrett
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - E Gower
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Valerie S Morrison
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Yim Man
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - K Tao Pun
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - James A Roper
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Jeni C Luckett
- Nottingham Respiratory Research Unit, University of Nottingham, Nottingham, UK
| | - Lee A Borthwick
- Fibrosis Research Group, Newcastle University Biosciences Institute, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Ben S Barksby
- Fibrosis Research Group, Newcastle University Biosciences Institute, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Rachel A Burgoyne
- Fibrosis Research Group, Newcastle University Biosciences Institute, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Rory Barnes
- Fibrosis Research Group, Newcastle University Biosciences Institute, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Andrew J Fisher
- Fibrosis Research Group, Newcastle University Biosciences Institute, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK; Institute of Transplantation, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS, Foundation Trust, Newcastle Upon Tyne, UK
| | | | - Richard J D Hatley
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Tim N Barrett
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Richard P Marshall
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Simon J F Macdonald
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - R Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, Guy Scadding Building, Cale Street, London, UK
| | - Robert J Slack
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK.
| |
Collapse
|
24
|
Yeh CF, Chou C, Yang KC. Mechanotransduction in fibrosis: Mechanisms and treatment targets. CURRENT TOPICS IN MEMBRANES 2021; 87:279-314. [PMID: 34696888 DOI: 10.1016/bs.ctm.2021.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
To perceive and integrate the environmental cues, cells and tissues sense and interpret various physical forces like shear, tensile, and compression stress. Mechanotransduction involves the sensing and translation of mechanical forces into biochemical and mechanical signals to guide cell fate and achieve tissue homeostasis. Disruption of this mechanical homeostasis by tissue injury elicits multiple cellular responses leading to pathological matrix deposition and tissue stiffening, and consequent evolution toward pro-inflammatory/pro-fibrotic phenotypes, leading to tissue/organ fibrosis. This review focuses on the molecular mechanisms linking mechanotransduction to fibrosis and uncovers the potential therapeutic targets to halt or resolve fibrosis.
Collapse
Affiliation(s)
- Chih-Fan Yeh
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan; Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Caroline Chou
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan; Washington University in St. Louis, St. Louis, MO, United States
| | - Kai-Chien Yang
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan; Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan; Research Center for Developmental Biology & Regenerative Medicine, National Taiwan University, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
25
|
Yang A, Yan X, Xu H, Fan X, Zhang M, Huang T, Li W, Chen W, Jia J, You H. Selective depletion of hepatic stellate cells-specific LOXL1 alleviates liver fibrosis. FASEB J 2021; 35:e21918. [PMID: 34569648 DOI: 10.1096/fj.202100374r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/06/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022]
Abstract
The role of LOXL1 in fibrosis via mediating ECM crosslinking and stabilization is well established; however, the role of hepatic stellate cells (HSCs)-specific LOXL1 in the development of fibrosis remains unknown. We generated HSCs-specific Loxl1-depleted mice (Loxl1Gfap-cre mice) to investigate the HSCs-specific contribution of LOXL1 in the pathogenesis of fibrosis. Loxl1fl/fl mice were used as the control. Furthermore, we used RNA sequencing to explore the underlying changes in the transcriptome. Results of the sirius red staining, type I collagen immunolabeling, and hydroxyproline content analysis, coupled with the reduced expression of profibrogenic genes revealed that Loxl1Gfap-cre mice with CCl4 -induced fibrosis exhibited decreased hepatic fibrosis. In addition, Loxl1Gfap-cre mice exhibited reduced macrophage tissue infiltration by CD68-positive cells and decreased expression of inflammatory genes compared with the controls. RNA sequencing identified integrin α8 (ITGA8) as a key modulator of LOXL1-mediated liver fibrosis. Functional analyses showed that siRNA silencing of Itga8 in cultured fibroblasts led to a decline in the LOXL1 expression and inhibition of fibroblast activation. Mechanistic analyses indicated that LOXL1 activated the FAK/PI3K/AKT/HIF1a signaling pathway, and the addition of inhibitors of FAK or PI3K reversed these results via downregulation of LOXL1. Furthermore, HIF1a directly interacted with LOXL1 and upregulated its expression, indicating that LOXL1 can positively self-regulate by forming a positive feedback loop with the FAK/PI3K/AKT/HIF1a pathway. We demonstrated that HSCs-specific Loxl1 deficiency prevented fibrosis, inflammation and that ITGA8/FAK/PI3K/AKT/HIF1a was essential for the function and expression of LOXL1. Knowledge of this approach can provide novel mechanisms and targets to treat fibrosis in the future.
Collapse
Affiliation(s)
- Aiting Yang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China
| | - Xuzhen Yan
- National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Hufeng Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China
| | - Xu Fan
- National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Mengyang Zhang
- National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Tao Huang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China
| | - Weiyu Li
- National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Wei Chen
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China
| | - Jidong Jia
- Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| | - Hong You
- Beijing Clinical Medicine Institute, Beijing, P.R. China.,National Clinical Research Center of Digestive Diseases, Beijing, P.R. China.,Liver Research Center, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
26
|
Slack RJ, Macdonald SJF, Roper JA, Jenkins RG, Hatley RJD. Emerging therapeutic opportunities for integrin inhibitors. Nat Rev Drug Discov 2021; 21:60-78. [PMID: 34535788 PMCID: PMC8446727 DOI: 10.1038/s41573-021-00284-4] [Citation(s) in RCA: 304] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
Integrins are cell adhesion and signalling proteins crucial to a wide range of biological functions. Effective marketed treatments have successfully targeted integrins αIIbβ3, α4β7/α4β1 and αLβ2 for cardiovascular diseases, inflammatory bowel disease/multiple sclerosis and dry eye disease, respectively. Yet, clinical development of others, notably within the RGD-binding subfamily of αv integrins, including αvβ3, have faced significant challenges in the fields of cancer, ophthalmology and osteoporosis. New inhibitors of the related integrins αvβ6 and αvβ1 have recently come to the fore and are being investigated clinically for the treatment of fibrotic diseases, including idiopathic pulmonary fibrosis and nonalcoholic steatohepatitis. The design of integrin drugs may now be at a turning point, with opportunities to learn from previous clinical trials, to explore new modalities and to incorporate new findings in pharmacological and structural biology. This Review intertwines research from biological, clinical and medicinal chemistry disciplines to discuss historical and current RGD-binding integrin drug discovery, with an emphasis on small-molecule inhibitors of the αv integrins. Integrins are key signalling molecules that are present on the surface of subsets of cells and are therefore good potential therapeutic targets. In this Review, Hatley and colleagues discuss the development of integrin inhibitors, particularly the challenges in developing inhibitors for integrins that contain an αv-subunit, and suggest how these challenges could be addressed.
Collapse
Affiliation(s)
| | | | | | - R G Jenkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
27
|
He ZB, Niu WB, Peng C, Gao C, Gao HJ, Niu J. The relationship between integrin avß6 and HBV infection in patients with liver cirrhosis and hepatocellular carcinoma: a preliminary report. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2021; 112:462-466. [PMID: 32450701 DOI: 10.17235/reed.2020.6607/2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE the aim of this study was to investigate the expression of integrin αvβ6 in normal, hepatitis B, HBV-associated cirrhosis and HBV-associated HCC liver tissues. METHODS immunohistochemistry and real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) were used to study the expression of integrin αvβ6 in HBV-associated cirrhosis (n = 88), chronic hepatitis B ( n= 11), HBV-associated HCC (n = 84) and normal (n = 10) human liver tissues. RESULTS the expression of integrin αvβ6 was significantly upregulated in HBV-associated liver cirrhosis and the expression increased with an increase in severity of cirrhosis. Furthermore, it was moderately or weakly expressed in chronic hepatitis B and HBV-associated HCC liver tissues when compared to normal liver tissue. CONCLUSION integrin αvβ6 could be a predictive marker for the progression of liver cirrhosis associated with HBV infection. Further studies are needed to determine the association between the expression of integrin αvβ6 in hepatitis B and HBV-associated HCC liver tissues.
Collapse
Affiliation(s)
- Zhao-Bin He
- Hepatobiliary Medicine, Qilu Hospital. Shandong University
| | - Wei-Bo Niu
- Hepatobiliary Medicine, Qilu Hospital. Shandong University
| | - Cheng Peng
- Hepatobiliary Medicine, Qilu Hospital. Shandong University
| | - Chao Gao
- Hepatobiliary Medicine, Qilu Hospital. Shandong University
| | - Hui-Jie Gao
- Hepatobiliary Medicine, Qilu Hospital. Shandong University
| | - Jun Niu
- Hepatobiliary Medicine, Qilu Hospital. Shandong University, China
| |
Collapse
|
28
|
Kyrönlahti A, Godbole N, Akinrinade O, Soini T, Nyholm I, Andersson N, Hukkinen M, Lohi J, Wilson DB, Pihlajoki M, Pakarinen MP, Heikinheimo M. Evolving Up-regulation of Biliary Fibrosis-Related Extracellular Matrix Molecules After Successful Portoenterostomy. Hepatol Commun 2021; 5:1036-1050. [PMID: 34141988 PMCID: PMC8183171 DOI: 10.1002/hep4.1684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Successful portoenterostomy (SPE) improves the short-term outcome of patients with biliary atresia (BA) by relieving cholestasis and extending survival with native liver. Despite SPE, hepatic fibrosis progresses in most patients, leading to cirrhosis and a deterioration of liver function. The goal of this study was to characterize the effects of SPE on the BA liver transcriptome. We used messenger RNA sequencing to analyze global gene-expression patterns in liver biopsies obtained at the time of portoenterostomy (n = 13) and 1 year after SPE (n = 8). Biopsies from pediatric (n = 2) and adult (n = 2) organ donors and other neonatal cholestatic conditions (n = 5) served as controls. SPE was accompanied by attenuation of inflammation and concomitant up-regulation of key extracellular matrix (ECM) genes. Highly overexpressed genes promoting biliary fibrosis and bile duct integrity, such as integrin subunit beta 6 and previously unreported laminin subunit alpha 3, emerged as candidates to control liver fibrosis after SPE. At a cellular level, the relative abundance of activated hepatic stellate cells and liver macrophages decreased following SPE, whereas portal fibroblasts (PFs) and cholangiocytes persisted. Conclusion: The attenuation of inflammation following SPE coincides with emergence of an ECM molecular fingerprint, a set of profibrotic molecules mechanistically connected to biliary fibrosis. The persistence of activated PFs and cholangiocytes after SPE suggests a central role for these cell types in the progression of biliary fibrosis.
Collapse
Affiliation(s)
- Antti Kyrönlahti
- Pediatric Research CenterChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Nimish Godbole
- Pediatric Research CenterChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Oyediran Akinrinade
- Pediatric Research CenterChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Tea Soini
- Pediatric Research CenterChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland.,Center for Infectious MedicineDepartment of MedicineKarolinska InstitutetStockholmSweden
| | - Iiris Nyholm
- Pediatric Research CenterChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland.,Pediatric SurgeryPediatric Liver and Gut Research GroupChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Noora Andersson
- Pediatric Research CenterChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Maria Hukkinen
- Pediatric SurgeryPediatric Liver and Gut Research GroupChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Jouko Lohi
- Department of PathologyHelsinki University HospitalHelsinkiFinland
| | - David B Wilson
- Department of PediatricsSt. Louis Children's HospitalWashington University School of MedicineSt. LouisMOUSA
| | - Marjut Pihlajoki
- Pediatric Research CenterChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland.,Center for Infectious MedicineDepartment of MedicineKarolinska InstitutetStockholmSweden
| | - Mikko P Pakarinen
- Pediatric Research CenterChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland.,Pediatric SurgeryPediatric Liver and Gut Research GroupChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Markku Heikinheimo
- Pediatric Research CenterChildren's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland.,Department of PediatricsSt. Louis Children's HospitalWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
29
|
Wei G, Cao J, Huang P, An P, Badlani D, Vaid KA, Zhao S, Wang DQH, Zhuo J, Yin L, Frassetto A, Markel A, Presnyak V, Gandham S, Hua S, Lukacs C, Finn PF, Giangrande PH, Martini PGV, Popov YV. Synthetic human ABCB4 mRNA therapy rescues severe liver disease phenotype in a BALB/c.Abcb4 -/- mouse model of PFIC3. J Hepatol 2021; 74:1416-1428. [PMID: 33340584 PMCID: PMC8188846 DOI: 10.1016/j.jhep.2020.12.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Progressive familial intrahepatic cholestasis type 3 (PFIC3) is a rare lethal autosomal recessive liver disorder caused by loss-of-function variations of the ABCB4 gene, encoding a phosphatidylcholine transporter (ABCB4/MDR3). Currently, no effective treatment exists for PFIC3 outside of liver transplantation. METHODS We have produced and screened chemically and genetically modified mRNA variants encoding human ABCB4 (hABCB4 mRNA) encapsulated in lipid nanoparticles (LNPs). We examined their pharmacological effects in a cell-based model and in a new in vivo mouse model resembling human PFIC3 as a result of homozygous disruption of the Abcb4 gene in fibrosis-susceptible BALB/c.Abcb4-/- mice. RESULTS We show that treatment with liver-targeted hABCB4 mRNA resulted in de novo expression of functional hABCB4 protein and restored phospholipid transport in cultured cells and in PFIC3 mouse livers. Importantly, repeated injections of the hABCB4 mRNA effectively rescued the severe disease phenotype in young Abcb4-/- mice, with rapid and dramatic normalisation of all clinically relevant parameters such as inflammation, ductular reaction, and liver fibrosis. Synthetic mRNA therapy also promoted favourable hepatocyte-driven liver regeneration to restore normal homeostasis, including liver weight, body weight, liver enzymes, and portal vein blood pressure. CONCLUSIONS Our data provide strong preclinical proof-of-concept for hABCB4 mRNA therapy as a potential treatment option for patients with PFIC3. LAY SUMMARY This report describes the development of an innovative mRNA therapy as a potential treatment for PFIC3, a devastating rare paediatric liver disease with no treatment options except liver transplantation. We show that administration of our mRNA construct completely rescues severe liver disease in a genetic model of PFIC3 in mice.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/administration & dosage
- ATP Binding Cassette Transporter, Subfamily B/deficiency
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- Animals
- Cholestasis, Intrahepatic/drug therapy
- Cholestasis, Intrahepatic/genetics
- Cholestasis, Intrahepatic/metabolism
- Disease Models, Animal
- Gene Deletion
- HEK293 Cells
- Homozygote
- Humans
- Liposomes/chemistry
- Liver/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Nanoparticle Drug Delivery System/chemistry
- Nanoparticles/chemistry
- Phenotype
- RNA, Messenger/administration & dosage
- RNA, Messenger/genetics
- Transfection
- Treatment Outcome
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
- Guangyan Wei
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | - Pinzhu Huang
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ping An
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Division of Gastroenterology and Hepatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Disha Badlani
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kahini A Vaid
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shuangshuang Zhao
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David Q-H Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jenny Zhuo
- Rare Diseases, Moderna Inc, Cambridge, MA, USA
| | - Ling Yin
- Rare Diseases, Moderna Inc, Cambridge, MA, USA
| | | | - Arianna Markel
- Rare Diseases, Moderna Inc, Cambridge, MA, USA; Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | | | | | - Serenus Hua
- Analytical Development, Moderna Inc, Cambridge, MA, USA
| | | | | | | | | | - Yury V Popov
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Hatipoglu OF, Uctepe E, Opoku G, Wake H, Ikemura K, Ohtsuki T, Inagaki J, Gunduz M, Gunduz E, Watanabe S, Nishinaka T, Takahashi H, Hirohata S. Osteopontin silencing attenuates bleomycin-induced murine pulmonary fibrosis by regulating epithelial-mesenchymal transition. Biomed Pharmacother 2021; 139:111633. [PMID: 34243624 DOI: 10.1016/j.biopha.2021.111633] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/22/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and most deadly form of interstitial lung disease. Osteopontin (OPN), a matricellular protein with proinflammatory and profibrotic properties, plays a major role in several fibrotic diseases, including IPF; OPN is highly upregulated in patients' lung samples. In this study, we knocked down OPN in a bleomycin (BLM)-induced pulmonary fibrosis (PF) mouse model using small interfering RNA (siRNA) to determine whether the use of OPN siRNA is an effective therapeutic strategy for IPF. We found that fibrosing areas were significantly smaller in specimens from OPN siRNA-treated mice. The number of alveolar macrophages, neutrophils, and lymphocytes in bronchoalveolar lavage fluid was also reduced in OPN siRNA-treated mice. Regarding the expression of epithelial-mesenchymal transition (EMT)-related proteins, the administration of OPN-siRNA to BLM-treated mice upregulated E-cadherin expression and downregulated vimentin expression. Moreover, in vitro, we incubated the human alveolar adenocarcinoma cell line A549 with transforming growth factor (TGF)-β1 and subsequently transfected the cells with OPN siRNA. We found a significant upregulation of Col1A1, fibronectin, and vimentin after TGF-β1 stimulation in A549 cells. In contrast, a downregulation of Col1A1, fibronectin, and vimentin mRNA levels was observed in TGF-β1-stimulated OPN knockdown A549 cells. Therefore, the downregulation of OPN effectively reduced pulmonary fibrotic and EMT changes both in vitro and in vivo. Altogether, our results indicate that OPN siRNA exerts a protective effect on BLM-induced PF in mice. Our results provide a basis for the development of novel targeted therapeutic strategies for IPF.
Collapse
Affiliation(s)
| | - Eyyup Uctepe
- Acıbadem Labmed Ankara Tissue Typing Laboratory, Turkey
| | - Gabriel Opoku
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Japan
| | - Hidenori Wake
- Department of Pharmacology, Faculty of Medicine, Kindai University, Japan
| | - Kentaro Ikemura
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Japan
| | - Takashi Ohtsuki
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Japan
| | - Junko Inagaki
- Department of Cell Chemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Japan
| | - Mehmet Gunduz
- Department of Otolaryngology, Moriya Keiyu Hospital, Japan
| | - Esra Gunduz
- Department of Otolaryngology, Moriya Keiyu Hospital, Japan
| | - Shogo Watanabe
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Japan
| | - Takashi Nishinaka
- Department of Pharmacology, Faculty of Medicine, Kindai University, Japan
| | - Hideo Takahashi
- Department of Pharmacology, Faculty of Medicine, Kindai University, Japan
| | - Satoshi Hirohata
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Japan.
| |
Collapse
|
31
|
Wei T, Hao W, Tang L, Wu H, Huang S, Yang Y, Qian N, Liu J, Yang W, Duan X. Comprehensive RNA-Seq Analysis of Potential Therapeutic Targets of Gan-Dou-Fu-Mu Decoction for Treatment of Wilson Disease Using a Toxic Milk Mouse Model. Front Pharmacol 2021; 12:622268. [PMID: 33935715 PMCID: PMC8082393 DOI: 10.3389/fphar.2021.622268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/19/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Gan–Dou–Fu–Mu decoction (GDFMD) improves liver fibrosis in experimental and clinical studies including those on toxic mouse model of Wilson disease (Model). However, the mechanisms underlying the effect of GDFMD have not been characterized. Herein, we deciphered the potential therapeutic targets of GDFMD using transcriptome analysis. Methods: We constructed a tx-j Wilson disease (WD) mouse model, and assessed the effect of GDFMD on the liver of model mice by hematoxylin and eosin, Masson, and immunohistochemical staining. Subsequently, we identified differentially expressed genes (DEGs) that were upregulated in the Model (Model vs. control) and those that were downregulated upon GDFMD treatment (compared to the Model) using RNA-sequencing (RNA-Seq). Biological functions and signaling pathways in which the DEGs were involved were determined by gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway analyses. A protein–protein interaction (PPI) network was constructed using the STRING database, and the modules were identified using MCODE plugin with the Cytoscape software. Several genes identified in the RNA-Seq analysis were validated by real-time quantitative PCR. Results: Total of 2124 DEGs were screened through the Model vs. control and Model vs. GDFMD comparisons, and dozens of GO and KEGG pathway terms modulated by GDFMD were identified. Dozens of pathways involved in metabolism (including metabolic processes for organic acids, carboxylic acids, monocarboxylic acids, lipids, fatty acids, cellular lipids, steroids, alcohols, eicosanoids, long-chain fatty acids), immune and inflammatory response (such as complement and coagulation cascades, cytokine–cytokine receptor interaction, inflammatory mediator regulation of TRP channels, antigen processing and presentation, T-cell receptor signaling pathway), liver fibrosis (such as ECM-receptor interactions), and cell death (PI3K-Akt signaling pathway, apoptosis, TGF-beta signaling pathway, etc.) were identified as potential targets of GDFMD in the Model. Some hub genes and four modules were identified in the PPI network. The results of real-time quantitative PCR analysis were consistent with those of RNA-Seq analysis. Conclusions: We performed gene expression profiling of GDFMD-treated WD model mice using RNA-Seq analysis and found the genes, pathways, and processes effected by the treatment. Our study provides a theoretical basis to prevent liver fibrosis resulting from WD using GDFMD.
Collapse
Affiliation(s)
- Taohua Wei
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China.,Graduate School, Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Hefei, China
| | - Wenjie Hao
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China.,Graduate School, Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Hefei, China
| | - Lulu Tang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Hefei, China
| | - Huan Wu
- Scientific Research and Experiment Center, Anhui University of Chinese Medicine, Hefei, China
| | - Shi Huang
- Graduate School, Anhui University of Chinese Medicine, Hefei, China
| | - Yue Yang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China.,Graduate School, Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Hefei, China
| | - Nannan Qian
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China.,Graduate School, Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Hefei, China
| | - Jie Liu
- Institute for Medical Virology, Goethe University Frankfurt Am Main, Frankfurt, Germany
| | - Wenming Yang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China.,Graduate School, Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Xin'An Medicine, Ministry of Education, Hefei, China
| | - Xianchun Duan
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
32
|
Modulation of α Vβ 6 integrin in osteoarthritis-related synovitis and the interaction with VTN (381-397 a.a.) competing for TGF-β1 activation. Exp Mol Med 2021; 53:210-222. [PMID: 33526813 PMCID: PMC8080589 DOI: 10.1038/s12276-021-00558-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis is characterized by structural alteration of joints. Fibrosis of the synovial tissue is often detected and considered one of the main causes of joint stiffness and pain. In our earlier proteomic study, increased levels of vitronectin (VTN) fragment (amino acids 381–397) were observed in the serum of osteoarthritis patients. In this work, the affinity of this fragment for integrins and its putative role in TGF-β1 activation were investigated. A competition study determined the interaction of VTN(381–397 a.a.) with αVβ6 integrin. Subsequently, the presence of αVβ6 integrin was substantiated on primary human fibroblast-like synoviocytes (FLSs) by western blot and flow cytometry. By immunohistochemistry, β6 was detected in synovial membranes, and its expression showed a correlation with tissue fibrosis. Moreover, β6 expression was increased under TGF-β1 stimulation; hence, a TGF-β bioassay was applied. We observed that αVβ6 could mediate TGF-β1 bioavailability and that VTN(381–397 a.a.) could prevent TGF-β1 activation by interacting with αVβ6 in human FLSs and increased α-SMA. Finally, we analyzed serum samples from healthy controls and patients with osteoarthritis and other rheumatic diseases by nano-LC/Chip MS–MS, confirming the increased expression of VTN(381–397 a.a.) in osteoarthritis as well as in lupus erythematosus and systemic sclerosis. These findings corroborate our previous observations concerning the overexpression of VTN(381–397 a.a.) in osteoarthritis but also in other rheumatic diseases. This fragment interacts with αVβ6 integrin, a receptor whose expression is increased in FLSs from the osteoarthritic synovial membrane and that can mediate the activation of the TGF-β1 precursor in human FLSs. Insights into a mechanism underlying the formation of fibrotic tissue within joints in osteoarthritis may also prove relevant to other rheumatological disorders. The general mechanisms underlying fibrosis are reasonably well understood, but it remains unclear what triggers these processes in osteoarthitis. Researchers of the University of Liège in Belgium have uncovered a possible explanation based on experiments with cultured primary synovial fibroblasts from patients. Osteoarthitis is characterized by increased levels of a fragment of the protein vitronectin, and the researchers demonstrated that this in turn binds to a protein called αVβ6, potentially promoting initiation of fibrosis. They also observed elevated levels of the same vitronectin fragment in two other rheumatoid disorders, lupus and systemic sclerosis, and concluded that further research is needed to characterize this protein’s role in inflammation and fibrosis.
Collapse
|
33
|
Extracellular Matrix Remodeling in Chronic Liver Disease. CURRENT TISSUE MICROENVIRONMENT REPORTS 2021; 2:41-52. [PMID: 34337431 PMCID: PMC8300084 DOI: 10.1007/s43152-021-00030-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/09/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF THE REVIEW This review aims to summarize the current knowledge of the extracellular matrix remodeling during hepatic fibrosis. We discuss the diverse interactions of the extracellular matrix with hepatic cells and the surrounding matrix in liver fibrosis, with the focus on the molecular pathways and the mechanisms that regulate extracellular matrix remodeling. RECENT FINDINGS The extracellular matrix not only provides structure and support for the cells, but also controls cell behavior by providing adhesion signals and by acting as a reservoir of growth factors and cytokines. SUMMARY Hepatic fibrosis is characterized by an excessive accumulation of extracellular matrix. During fibrogenesis, the natural remodeling process of the extracellular matrix varies, resulting in the excessive accumulation of its components, mainly collagens. Signals released by the extracellular matrix induce the activation of hepatic stellate cells, which are the major source of extracellular matrix and most abundant myofibroblasts in the liver. GRAPHICAL ABSTRACT
Collapse
|
34
|
Zhou S, Liang X, Wang N, Shao L, Yu W, Ding N, Zhang K, Liu M. Association of human platelet antigens polymorphisms with the levels of serum fibrosis marks in chronic hepatitis C patients. Transfus Apher Sci 2020; 60:102967. [PMID: 33059995 DOI: 10.1016/j.transci.2020.102967] [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: 04/02/2020] [Revised: 08/24/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Host genetic polymorphisms influence the fibrosis progression of chronic hepatitis C (CHC) patients. Previous studies have shown the association of human platelet antigens (HPAs) polymorphisms with CHC. However, little is known regarding the association of HPAs polymorphisms with the fibrosis progression of CHC. The aim of this study was to determine the association of HPA -2, -3, -5 and -15 polymorphisms with the levels of serum fibrosis marks in CHC patients. METHODS The HPA -2, -3, -5 and -15 were genotyped by 5'-nuclease assay in 211 CHC patients, while the serum concentration of hyaluronic acid (HA), collagen IV (CIV), amino-terminal pro-peptide of type III procollagen (PIIINP), and laminin (LN) from the same samples were measured by time resolved fluorescence immunoassay. RESULTS The level of serum LN was significantly lower in CHC patients with HPA-15aa genotype compared to those with HPA-15ab/bb (P = 0.032) but did not differ among HPA-2, -3 and -5 genotypes. There were no difference in HA, CIV and PIIINP levels among HPA-2, -3,-5 and -15 genotypes. CONCLUSIONS This study demonstrates that HPA-15 aa polymorphism is associated lower serum LN in CHC, which suggests that HPA -15 aa may be involved in the fibrosis progression of CHC.
Collapse
Affiliation(s)
- Shihang Zhou
- Department of Blood Group, Dalian Blood Center, Dalian, China
| | - Xiaohua Liang
- Department of Blood Group, Dalian Blood Center, Dalian, China
| | - Ni Wang
- Department of Blood Group, Dalian Blood Center, Dalian, China
| | - Linnan Shao
- Department of Blood Group, Dalian Blood Center, Dalian, China
| | - Weijian Yu
- Department of Blood Group, Dalian Blood Center, Dalian, China
| | - Nan Ding
- Dalian Sixth People's Hospital Affiliated of Dalian Medical University, Dalian, China
| | - Kaili Zhang
- Department of Cell Biology, Dalian Medical University, Dalian, China
| | - Ming Liu
- Department of Cell Biology, Dalian Medical University, Dalian, China.
| |
Collapse
|
35
|
John AE, Graves RH, Pun KT, Vitulli G, Forty EJ, Mercer PF, Morrell JL, Barrett JW, Rogers RF, Hafeji M, Bibby LI, Gower E, Morrison VS, Man Y, Roper JA, Luckett JC, Borthwick LA, Barksby BS, Burgoyne RA, Barnes R, Le J, Flint DJ, Pyne S, Habgood A, Organ LA, Joseph C, Edwards-Pritchard RC, Maher TM, Fisher AJ, Gudmann NS, Leeming DJ, Chambers RC, Lukey PT, Marshall RP, Macdonald SJF, Jenkins RG, Slack RJ. Translational pharmacology of an inhaled small molecule αvβ6 integrin inhibitor for idiopathic pulmonary fibrosis. Nat Commun 2020; 11:4659. [PMID: 32938936 PMCID: PMC7494911 DOI: 10.1038/s41467-020-18397-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 08/17/2020] [Indexed: 12/16/2022] Open
Abstract
The αvβ6 integrin plays a key role in the activation of transforming growth factor-β (TGFβ), a pro-fibrotic mediator that is pivotal to the development of idiopathic pulmonary fibrosis (IPF). We identified a selective small molecule αvβ6 RGD-mimetic, GSK3008348, and profiled it in a range of disease relevant pre-clinical systems. To understand the relationship between target engagement and inhibition of fibrosis, we measured pharmacodynamic and disease-related end points. Here, we report, GSK3008348 binds to αvβ6 with high affinity in human IPF lung and reduces downstream pro-fibrotic TGFβ signaling to normal levels. In human lung epithelial cells, GSK3008348 induces rapid internalization and lysosomal degradation of the αvβ6 integrin. In the murine bleomycin-induced lung fibrosis model, GSK3008348 engages αvβ6, induces prolonged inhibition of TGFβ signaling and reduces lung collagen deposition and serum C3M, a marker of IPF disease progression. These studies highlight the potential of inhaled GSK3008348 as an anti-fibrotic therapy. The αvβ6 integrin is key in activating the pro-fibrotic cytokine TGFβ in idiopathic pulmonary fibrosis. Here, the authors show an inhaled small molecule αvβ6 inhibitor GSK3008348 induces prolonged inhibition of TGFβ signaling pathways in human and murine models of lung fibrosis via αvβ6 degradation.
Collapse
Affiliation(s)
- Alison E John
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Rebecca H Graves
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - K Tao Pun
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Giovanni Vitulli
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Ellen J Forty
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Paul F Mercer
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Josie L Morrell
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - John W Barrett
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Rebecca F Rogers
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Maryam Hafeji
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Lloyd I Bibby
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Elaine Gower
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Valerie S Morrison
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Yim Man
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - James A Roper
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Jeni C Luckett
- Radiological Sciences, University of Nottingham, Nottingham, UK
| | - Lee A Borthwick
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Ben S Barksby
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Rachel A Burgoyne
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Rory Barnes
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Joelle Le
- Drug Design and Selection - Molecular Design, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - David J Flint
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Anthony Habgood
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Louise A Organ
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Chitra Joseph
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | | | - Toby M Maher
- NIHR Respiratory Clinical Research Facility, Royal Brompton Hospital, London, UK.,Fibrosis Research Group, National Heart and Lung Institute, Imperial College, London, UK
| | - Andrew J Fisher
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK.,Institute of Transplantation, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS, Foundation Trust, Newcastle upon Tyne, UK
| | - Natasja Stæhr Gudmann
- Nordic Bioscience A/S, Biomarkers and Research, Herlev Hovedgade 205-207, Herlev, Denmark
| | - Diana J Leeming
- Nordic Bioscience A/S, Biomarkers and Research, Herlev Hovedgade 205-207, Herlev, Denmark
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Pauline T Lukey
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Richard P Marshall
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Simon J F Macdonald
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - R Gisli Jenkins
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK.
| | - Robert J Slack
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| |
Collapse
|
36
|
Fabris L, Cadamuro M, Cagnin S, Strazzabosco M, Gores GJ. Liver Matrix in Benign and Malignant Biliary Tract Disease. Semin Liver Dis 2020; 40:282-297. [PMID: 32162285 DOI: 10.1055/s-0040-1705109] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The extracellular matrix is a highly reactive scaffold formed by a wide array of multifunctional molecules, encompassing collagens and noncollagenous glycoproteins, proteoglycans, glycosaminoglycans, and polysaccharides. Besides outlining the tissue borders, the extracellular matrix profoundly regulates the behavior of resident cells by transducing mechanical signals, and by integrating multiple cues derived from the microenvironment. Evidence is mounting that changes in the biostructure of the extracellular matrix are instrumental for biliary repair. Following biliary damage and eventually, malignant transformation, the extracellular matrix undergoes several quantitative and qualitative modifications, which direct interactions among hepatic progenitor cells, reactive ductular cells, activated myofibroblasts and macrophages, to generate the ductular reaction. Herein, we will give an overview of the main molecular factors contributing to extracellular matrix remodeling in cholangiopathies. Then, we will discuss the structural alterations in terms of biochemical composition and physical stiffness featuring the "desmoplastic matrix" of cholangiocarcinoma along with their pro-oncogenic effects.
Collapse
Affiliation(s)
- Luca Fabris
- Department of Molecular Medicine, University of Padua, Padua, Italy.,Liver Center, Department of Medicine, Yale University, New Haven, Connecticut
| | | | - Silvia Cagnin
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Mario Strazzabosco
- Liver Center, Department of Medicine, Yale University, New Haven, Connecticut
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Michigan
| |
Collapse
|
37
|
Zhang N, Zhao SS, Zhang YX, Wang YC, Shao RG, Wang JX, He HW. A novel biphenyl compound IMB-S7 ameliorates hepatic fibrosis in BDL rats by suppressing Sp1-mediated integrin αv expression. Acta Pharmacol Sin 2020; 41:661-669. [PMID: 31932644 PMCID: PMC7470799 DOI: 10.1038/s41401-019-0325-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/31/2019] [Indexed: 01/15/2023]
Abstract
Chronic tissue injury with fibrosis results in the disruption of tissue architecture, organ dysfunction, and eventual organ failure. Therefore, the development of effective antifibrotic drugs is urgently required. IMB-S7 is novel biphenyl compound derived from bifendate (biphenyldicarboxylate) that is used for the treatment of chronic hepatitis in China. In the current study we investigated the potential of IMB-S7 as an antihepatic fibrosis agent. In bile duct ligation (BDL) rat model, oral administration of IMB-S7 (400 mg· kg-1· d-1, for 14 days) significantly ameliorated BDL-induced liver necrosis, bile duct proliferation, and collagen accumulation. We then showed that IMB-S7 treatment markedly suppressed the TGF-β/Smad pathway in human hepatic stellate cell line LX2 and mouse primary HSCs, as well as in liver samples of BDL rats, thus inhibiting the transcription of most fibrogenesis-associated genes, including TGF-β1, COL1A1, and ACTA2. Furthermore, IMB-S7 treatment significantly suppressed the expression of integrin αv at the mRNA and protein levels in TGF-β-treated LX2 cells and liver samples of BDL rats. Using integrin αv overexpression and silencing, we demonstrated that integrin αv activity correlated positively with the activation of TGF-β/Smad pathway. Based on dual luciferase assay and DNA affinity precipitation assay, we revealed that IMB-S7 inactivated integrin αv through competitively inhibiting the binding of Sp1, a transcription factor, to the integrin αv (ITGAV) promoter (-173/-163 bp). These results suggest that IMB-S7 inhibits HSCs activation and liver fibrosis through Sp1-integrin αv signaling, and IMB-S7 may be a promising candidate to combat hepatic fibrosis in the future.
Collapse
Affiliation(s)
- Na Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Shuang-Shuang Zhao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China
- The Joint Program in Infection and Immunity, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yi-Xuan Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Yu-Cheng Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Rong-Guang Shao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Ju-Xian Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China.
| | - Hong-Wei He
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, 100050, China.
| |
Collapse
|
38
|
Poelstra K. Innovative Nanotechnological Formulations to Reach the Hepatic Stellate Cell. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s43152-020-00004-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Abstract
Purpose of Review
Treatment of liver fibrosis benefits from hepatic stellate cell (HSC)-specific delivery. Since the description of first carrier to HSC, many developments have taken place in this area. The purpose is to give an overview of the different carriers and homing moieties that are available for HSC targeting and illustrate the opportunities and hurdles they provide.
Recent Findings
There is a growing number of homing devices to deliver drugs to HSC, and options to deliver siRNA to HSC have emerged. Other developments include controlling corona formation, development of linker technology, and design of theranostic approaches. We are on the eve of reaching the clinic with innovative HSC-specific compounds.
Summary
An overview of different core molecules is presented together with an overview of targeting strategies toward different receptors on HSC, providing a versatile toolbox. Many therapeutics, ranging from small chemical entities and proteins to RNA- or DNA-modulating substances, have already been incorporated in these constructs in the recent years.
Collapse
|
39
|
Wei G, An P, Vaid KA, Nasser I, Huang P, Tan L, Zhao S, Schuppan D, Popov YV. Comparison of murine steatohepatitis models identifies a dietary intervention with robust fibrosis, ductular reaction, and rapid progression to cirrhosis and cancer. Am J Physiol Gastrointest Liver Physiol 2020; 318:G174-G188. [PMID: 31630534 PMCID: PMC6985845 DOI: 10.1152/ajpgi.00041.2019] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Progressive fibrosis, functional liver failure, and cancer are the central liver-related outcomes of nonalcoholic steatohepatitis (NASH) but notoriously difficult to achieve in mouse models. We performed a direct, quantitative comparison of hepatic fibrosis progression in well-defined methionine- and choline-deficient (MCD) and choline-deficient, amino-acid defined (CDAA) diets with increasing fat content (10-60% by calories) in C57Bl/6J and BALB/cAnNCrl mice. In C57Bl/6J mice, MCD feeding resulted in moderate fibrosis at week 8 (up to twofold increase in total hepatic collagen content) and progressive weight loss irrespective of dietary fat. In contrast, CDAA-fed mice did not lose weight and developed progressive fibrosis starting from week 4. High dietary fat in the CDAA diet model induced the lipid metabolism genes for sterol regulatory element-binding protein and stearoyl-CoA desaturase-2 and increased ductular reaction and fibrosis in a dose-dependent manner. Longitudinal analysis of CDAA with 60% fat (HF-CDAA) feeding revealed pronounced ductular reaction and perisinusoidal bridging fibrosis, with a sevenfold increase of hepatic collagen at week 12, which showed limited spontaneous reversibility. At 24 wk, HF-CDAA mice developed signs of cirrhosis with pan-lobular "chicken wire" fibrosis, 10-fold hydroxyproline increase, regenerative nodules, portal hypertension and elevated serum bilirubin and ammonia levels; 80% of mice (8/10) developed multiple glypican-3- and/or glutamine synthetase-positive hepatocellular carcinomas (HCC). High-fat (60%) supplementation of MCD in C57Bl/6J or feeding the HF-CDAA diet fibrosis-prone BALB/cAnNCrl strain failed to result in increased fibrosis. In conclusion, HF-CDAA feeding in C57Bl/6J mice was identified as an optimal model of steatohepatitis with robust fibrosis and ductular proliferations that progress to cirrhosis and HCC within 24 wk. This robust model will aid the testing of interventions and drugs for severe NASH.NEW & NOTEWORTHY Via quantitative comparison of several dietary models, we report HF-CDAA feeding in C57Bl/6 mice as an excellent model recapitulating several key aspects of fibrotic NASH: 1) robust, poorly reversible liver fibrosis, 2) prominent ductular reaction, and 3) progression to cirrhosis, portal hypertension, and liver cancer within 24 wk. High fat dose-dependently activates SREBP2/SCD2 genes and drives liver fibrosis in e HF-CDAA model. These features qualify the model as a robust and practical tool to study mechanisms and novel treatments addressing severe human NASH.
Collapse
Affiliation(s)
- Guangyan Wei
- 1Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,2Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ping An
- 2Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts,3Division of Gastroenterology and Hepatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Kahini A. Vaid
- 2Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Imad Nasser
- 4Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Pinzhu Huang
- 2Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts,5Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Tan
- 1Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China,2Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Shuangshuang Zhao
- 2Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Detlef Schuppan
- 2Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts,6Institute of Translational Immunology, University Medical Center, Mainz, Germany
| | - Yury V. Popov
- 2Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
40
|
Cairns JT, Habgood A, Edwards-Pritchard RC, Joseph C, John AE, Wilkinson C, Stewart ID, Leslie J, Blaxall BC, Susztak K, Alberti S, Nordheim A, Oakley F, Jenkins G, Tatler AL. Loss of ELK1 has differential effects on age-dependent organ fibrosis. Int J Biochem Cell Biol 2019; 120:105668. [PMID: 31877385 DOI: 10.1016/j.biocel.2019.105668] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 01/07/2023]
Abstract
ETS domain-containing protein-1 (ELK1) is a transcription factor important in regulating αvβ6 integrin expression. αvβ6 integrins activate the profibrotic cytokine Transforming Growth Factor β1 (TGFβ1) and are increased in the alveolar epithelium in idiopathic pulmonary fibrosis (IPF). IPF is a disease associated with aging and therefore we hypothesised that aged animals lacking Elk1 globally would develop spontaneous fibrosis in organs where αvβ6 mediated TGFβ activation has been implicated. Here we identify that Elk1-knockout (Elk1-/0) mice aged to one year developed spontaneous fibrosis in the absence of injury in both the lung and the liver but not in the heart or kidneys. The lungs of Elk1-/0 aged mice demonstrated increased collagen deposition, in particular collagen 3α1, located in small fibrotic foci and thickened alveolar walls. Despite the liver having relatively low global levels of ELK1 expression, Elk1-/0 animals developed hepatosteatosis and fibrosis. The loss of Elk1 also had differential effects on Itgb1, Itgb5 and Itgb6 expression in the four organs potentially explaining the phenotypic differences in these organs. To understand the potential causes of reduced ELK1 in human disease we exposed human lung epithelial cells and murine lung slices to cigarette smoke extract, which lead to reduced ELK1 expression andmay explain the loss of ELK1 in human disease. These data support a fundamental role for ELK1 in protecting against the development of progressive fibrosis via transcriptional regulation of beta integrin subunit genes, and demonstrate that loss of ELK1 can be caused by cigarette smoke.
Collapse
Affiliation(s)
- Jennifer T Cairns
- Nottingham NIHR Biomedical Research Centre, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals, City Campus, Nottingham, NG5 1PB, UK
| | - Anthony Habgood
- Nottingham NIHR Biomedical Research Centre, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals, City Campus, Nottingham, NG5 1PB, UK
| | - Rochelle C Edwards-Pritchard
- Nottingham NIHR Biomedical Research Centre, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals, City Campus, Nottingham, NG5 1PB, UK
| | - Chitra Joseph
- Nottingham NIHR Biomedical Research Centre, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals, City Campus, Nottingham, NG5 1PB, UK
| | - Alison E John
- Nottingham NIHR Biomedical Research Centre, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals, City Campus, Nottingham, NG5 1PB, UK
| | - Chloe Wilkinson
- Nottingham NIHR Biomedical Research Centre, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals, City Campus, Nottingham, NG5 1PB, UK
| | - Iain D Stewart
- Nottingham NIHR Biomedical Research Centre, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals, City Campus, Nottingham, NG5 1PB, UK
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, 4th Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Burns C Blaxall
- Department of Personalized Medicine and Pharmacogenetics, The Christ Hospital Health Network, Cincinnati, OH, USA
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Siegfried Alberti
- Interfaculty Institute of Cell Biology, Tuebingen University, Tuebingen, Germany
| | - Alfred Nordheim
- Interfaculty Institute of Cell Biology, Tuebingen University, Tuebingen, Germany; Leibniz Institute on Ageing (FLI), Jena, Germany
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, 4th Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Gisli Jenkins
- Nottingham NIHR Biomedical Research Centre, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals, City Campus, Nottingham, NG5 1PB, UK
| | - Amanda L Tatler
- Nottingham NIHR Biomedical Research Centre, Division of Respiratory Medicine, University of Nottingham, Nottingham University Hospitals, City Campus, Nottingham, NG5 1PB, UK.
| |
Collapse
|
41
|
The Many Roles of Cell Adhesion Molecules in Hepatic Fibrosis. Cells 2019; 8:cells8121503. [PMID: 31771248 PMCID: PMC6952767 DOI: 10.3390/cells8121503] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 01/09/2023] Open
Abstract
Fibrogenesis is a progressive scarring event resulting from disrupted regular wound healing due to repeated tissue injury and can end in organ failure, like in liver cirrhosis. The protagonists in this process, either liver-resident cells or patrolling leukocytes attracted to the site of tissue damage, interact with each other by soluble factors but also by direct cell–cell contact mediated by cell adhesion molecules. Since cell adhesion molecules also support binding to the extracellular matrix, they represent excellent biosensors, which allow cells to modulate their behavior based on changes in the surrounding microenvironment. In this review, we focus on selectins, cadherins, integrins and members of the immunoglobulin superfamily of adhesion molecules as well as some non-classical cell adhesion molecules in the context of hepatic fibrosis. We describe their liver-specific contributions to leukocyte recruitment, cell differentiation and survival, matrix remodeling or angiogenesis and touch on their suitability as targets in antifibrotic therapies.
Collapse
|
42
|
Gore E, Bigaeva E, Oldenburger A, Kim YO, Rippmann JF, Schuppan D, Boersema M, Olinga P. PI3K inhibition reduces murine and human liver fibrogenesis in precision-cut liver slices. Biochem Pharmacol 2019; 169:113633. [PMID: 31494146 DOI: 10.1016/j.bcp.2019.113633] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/03/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Liver fibrosis results from continuous inflammation and injury. Despite its high prevalence worldwide, no approved antifibrotic therapies exist. Omipalisib is a selective inhibitor of the PI3K/mTOR pathway that controls nutrient metabolism, growth and proliferation. It has shown antifibrotic properties in vitro. While clinical trials for idiopathic pulmonary fibrosis have been initiated, an in-depth preclinical evaluation is lacking. We evaluated omipalisib's effects on fibrogenesis using the ex vivo model of murine and human precision-cut tissue slices (PCTS). METHODS Murine and human liver and jejunum PCTS were incubated with omipalisib up to 10 μM for 48 h. PI3K pathway activation was assessed through protein kinase B (Akt) phosphorylation and antifibrotic efficacy was determined via a spectrum of fibrosis markers at the transcriptional and translational level. RESULTS During incubation of PCTS the PI3K pathway was activated and incubation with omipalisib prevented Akt phosphorylation (IC50 = 20 and 1.5 nM for mouse and human, respectively). Viability of mouse and human liver PCTS was compromised only at the high concentration of 10 and 1-5 μM, respectively. However, viability of jejunum PCTS decreased with 0.1 (mouse) and 0.01 μM (human). Spontaneously increased fibrosis related genes and proteins were significantly and similarly suppressed in mouse and in human liver PCTS. CONCLUSIONS Omipalisib has antifibrotic properties in ex vivo mouse and human liver PCTS, but higher concentrations showed toxicity in jejunum PCTS. While the PI3K/mTOR pathway appears to be a promising target for the treatment of liver fibrosis, PCTS revealed likely side effects in the intestine at higher doses.
Collapse
Affiliation(s)
- Emilia Gore
- Groningen Research Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713AV, The Netherlands
| | - Emilia Bigaeva
- Groningen Research Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713AV, The Netherlands
| | - Anouk Oldenburger
- Cardiometabolic Disease Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, Biberach an der Riss 88397, Germany
| | - Yong Ook Kim
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Obere Zahlbacherstraße 63, Mainz 55131, Germany
| | - Jörg F Rippmann
- Cardiometabolic Disease Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, Biberach an der Riss 88397, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Obere Zahlbacherstraße 63, Mainz 55131, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 330 Brookline Avenue, MA 02215, USA
| | - Miriam Boersema
- Groningen Research Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713AV, The Netherlands
| | - Peter Olinga
- Groningen Research Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713AV, The Netherlands.
| |
Collapse
|
43
|
Abstract
Integrins are heterodimeric cell surface receptors ensuring the mechanical connection between cells and the extracellular matrix. In addition to the anchorage of cells to the extracellular matrix, these receptors have critical functions in intracellular signaling, but are also taking center stage in many physiological and pathological conditions. In this review, we provide some historical, structural, and physiological notes so that the diverse functions of these receptors can be appreciated and put into the context of the emerging field of mechanobiology. We propose that the exciting journey of the exploration of these receptors will continue for at least another new generation of researchers.
Collapse
Affiliation(s)
- Michael Bachmann
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Sampo Kukkurainen
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Vesa P Hytönen
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| |
Collapse
|
44
|
Evaluation of integrin αvβ 6 cystine knot PET tracers to detect cancer and idiopathic pulmonary fibrosis. Nat Commun 2019; 10:4673. [PMID: 31611594 PMCID: PMC6791878 DOI: 10.1038/s41467-019-11863-w] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 08/06/2019] [Indexed: 12/13/2022] Open
Abstract
Advances in precision molecular imaging promise to transform our ability to detect, diagnose and treat disease. Here, we describe the engineering and validation of a new cystine knot peptide (knottin) that selectively recognizes human integrin αvβ6 with single-digit nanomolar affinity. We solve its 3D structure by NMR and x-ray crystallography and validate leads with 3 different radiolabels in pre-clinical models of cancer. We evaluate the lead tracer’s safety, biodistribution and pharmacokinetics in healthy human volunteers, and show its ability to detect multiple cancers (pancreatic, cervical and lung) in patients at two study locations. Additionally, we demonstrate that the knottin PET tracers can also detect fibrotic lung disease in idiopathic pulmonary fibrosis patients. Our results indicate that these cystine knot PET tracers may have potential utility in multiple disease states that are associated with upregulation of integrin αvβ6. Knottin is a cystine knot peptide. Here, the authors develop a knottin-based tracer for positron emission tomography and demonstrate its ability to detect cancer and idiopathic pulmonary fibrosis through selective binding to integrin αvβ6.
Collapse
|
45
|
Ghasemi A, Zadsar M, Shaiegan M, Samiei S, Namvar A, Rasouli M, Moosanejad M. Human platelet antigens polymorphisms; association to the development of liver fibrosis in patients with chronic hepatitis C. J Med Virol 2019; 92:45-52. [PMID: 30729550 DOI: 10.1002/jmv.25423] [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: 11/13/2018] [Revised: 01/20/2019] [Accepted: 02/02/2019] [Indexed: 11/09/2022]
Abstract
Recently, human platelet antigens (HPAs) polymorphisms are found to play a role in susceptibility to hepatitis C virus (HCV) infection and fibrosis progression. The aim of the current study was to evaluate the possible association between the HPAs polymorphisms with liver fibrosis progression in HCV patients. HPAs polymorphisms genotyping was performed in HCV patients (n = 71) by Sequence-specific primers-polymerase chain reaction. Fibrosis progression was evaluated using the Metavir scoring system and liver biopsy, and the patients were assigned to two groups, namely, G1 (n = 35) that included patients with F1 (portal fibrosis without septa) or F2 (few septa) and G2 (n = 36) that comprised patients with F3 (numerous septa) or F4 (cirrhosis). The data analyses were performed using Pearson's χ2 test. The genotype frequency of HPA-3ab was significantly higher in G1 patients than in G2 patients (P = 0.015). No statistically significant differences were found between the patient groups (G1 and G2) regarding the distributions of the allelic and genotypic frequencies of the HPA-1, -2, -4, -5, and -15 systems. Multivariate logistic regression showed an independent association between the genotype HPA-3aa/BB and severe fibrosis (F3-F4), when compared with genotype HPA-3ab, independent of the viral genotype, high alanine transaminase, sex, age, time of infection, diabetes, and high cholesterol as risk factors. The present study suggested that the HPA-3ab genotype could be noticed as a potential protecting factor against hepatic fibrosis. Therefore, the antigenic variation of integrins might be considered as a part of the coordinated inflammatory process involved in the progression of liver fibrosis.
Collapse
Affiliation(s)
- Ali Ghasemi
- Department of Hematology, Blood Transfusion Research Center, High Institute for Research & Education in Transfusion Medicine, Tehran, Iran
| | - Maryam Zadsar
- Department of Microbiology, Blood Transfusion Research Center, High Institute for Research & Education in Transfusion Medicine, Tehran, Iran
| | - Mojgan Shaiegan
- Department of Immunohematology, Blood Transfusion Research Center, High Institute for Research & Education in Transfusion Medicine, Tehran, Iran
| | - Shahram Samiei
- Department of Biochemistry, Blood Transfusion Research Center, High Institute for Research & Education in Transfusion Medicine, Tehran, Iran
| | - Ali Namvar
- Department of Genetics, Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Mahboobeh Rasouli
- Department of Biostatics, School of Public Health, Iran University of Medical Science, Tehran, Iran
| | - Molood Moosanejad
- Department of Clinical Consult, Blood Transfusion Research Center, High Institute for Research & Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
46
|
Hatley RJD, Barrett TN, Slack RJ, Watson ME, Baillache DJ, Gruszka A, Washio Y, Rowedder JE, Pogány P, Pal S, Macdonald SJF. The Design of Potent, Selective and Drug-Like RGD αvβ1 Small-Molecule Inhibitors Derived from non-RGD α4β1 Antagonists. ChemMedChem 2019; 14:1315-1320. [PMID: 31207080 DOI: 10.1002/cmdc.201900359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Indexed: 11/06/2022]
Abstract
Up to 45 % of deaths in developed nations can be attributed to chronic fibroproliferative diseases, highlighting the need for effective therapies. The RGD (Arg-Gly-Asp) integrin αvβ1 was recently investigated for its role in fibrotic disease, and thus warrants therapeutic targeting. Herein we describe the identification of non-RGD hit small-molecule αvβ1 inhibitors. We show that αvβ1 activity is embedded in a range of published α4β1 (VLA-4) ligands; we also demonstrate how a non-RGD integrin inhibitor (of α4β1 in this case) was converted into a potent non-zwitterionic RGD integrin inhibitor (of αvβ1 in this case). We designed urea ligands with excellent selectivity over α4β1 and the other αv integrins (αvβ3, αvβ5, αvβ6, αvβ8). In silico docking models and density functional theory (DFT) calculations aided the discovery of the lead urea series.
Collapse
Affiliation(s)
- Richard J D Hatley
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Tim N Barrett
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Robert J Slack
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Morag E Watson
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Daniel J Baillache
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Anna Gruszka
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Yoshiaki Washio
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - James E Rowedder
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Peter Pogány
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Sandeep Pal
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Simon J F Macdonald
- GlaxoSmithKline (GSK), Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| |
Collapse
|
47
|
Zagory JA, Fenlon M, Dietz W, Zhao M, Nguyen MV, Trinh P, Adoumie M, Park A, Xu J, Mahdi E, Glazier A, Malkoff N, Mavila N, Wang KS. Prominin-1 Promotes Biliary Fibrosis Associated With Biliary Atresia. Hepatology 2019; 69:2586-2597. [PMID: 30723921 PMCID: PMC6541523 DOI: 10.1002/hep.30550] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/29/2019] [Indexed: 01/09/2023]
Abstract
In patients with biliary atresia (BA), the extent of intrahepatic biliary fibrosis negatively correlates with successful surgical bypass of the congenital cholangiopathy as well as subsequent transplant-free survival. We recently linked the expansion of a population of prominin-1 (Prom1)-expressing hepatic progenitor cells to biliary fibrogenesis. Herein, we hypothesized that Prom1-expressing progenitor cells play a role in BA-associated fibrosis. Rhesus rotavirus (RRV)-mediated experimental BA was induced in newborn mice homozygous for the transgene Prom1cre-ert2-nlacz , which was knocked in to the Prom1 gene locus, thus creating functional Prom1 knockout (KO) mice, and their wildtype (WT) littermates. Clinical data and tissue samples from BA infants from the Childhood Liver Disease Research Consortium were analyzed. Extrahepatic biliary obliteration was present in both WT and KO mice; there was no difference in serum total bilirubin (TBili) levels. The intrahepatic periportal expansion of the PROM1pos cell population, typically observed in RRV-induced BA, was absent in KO mice. RRV-treated KO mice demonstrated significantly fewer cytokeratin-19 (CK19)-positive ductular reactions (P = 0.0004) and significantly less periportal collagen deposition (P = 0.0001) compared with WT. RRV-treated KO mice expressed significantly less integrin-β6, which encodes a key biliary-specific subunit of a transforming growth factor (TGF) β activator (P = 0.0004). Infants with successful biliary drainage (Tbili ≤1.5 mg/dL within 3 months postoperatively), which is highly predictive of increased transplant-free survival, expressed significantly less hepatic PROM1, CK19, and COLLAGEN-1α compared with those with TBili >1.5 (P < 0.05). Conclusion: Prom1 plays an important role in biliary fibrogenesis, in part through integrin-mediated TGF pathway activation.
Collapse
Affiliation(s)
| | - Michael Fenlon
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - William Dietz
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - Menghan Zhao
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - Marie V. Nguyen
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - Pavin Trinh
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - Maeva Adoumie
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - Alex Park
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - Jiabo Xu
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - Elaa Mahdi
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - Alison Glazier
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - Nicolas Malkoff
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | - Nirmala Mavila
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA,Cedars-Sinai Medical Center, Gastroenterology, Los Angeles, CA, USA
| | - Kasper S. Wang
- Children’s Hospital Los Angeles, Surgery, Los Angeles, CA, USA
| | | |
Collapse
|
48
|
Yokoyama H, Masaki T, Inoue I, Nakamura M, Mezaki Y, Saeki C, Oikawa T, Saruta M, Takahashi H, Ikegami M, Hano H, Ikejima K, Kojima S, Matsuura T. Histological and biochemical evaluation of transforming growth factor-β activation and its clinical significance in patients with chronic liver disease. Heliyon 2019; 5:e01231. [PMID: 30815603 PMCID: PMC6378908 DOI: 10.1016/j.heliyon.2019.e01231] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/26/2019] [Accepted: 02/11/2019] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a key driver for liver fibrogenesis. TGF-β must be activated in order to function. Plasma kallikrein (PLK) is a TGF-β activator that cleaves the latency-associated protein (LAP) between arginine58 and lysine59 residues and releases active TGF-β from the latent TGF-β-LAP complex. Thus, the generation of two LAP degradation products, ending at arginine58 (R58/LAP-DPs) and beginning from lysine59 (L59/LAP-DPs), reflects PLK-dependent TGF-β activation. However, the significance and details of TGF-β activation in patients with chronic liver disease (CLD) remain uncertain. We herein examined the PLK-dependent TGF-β activation in patients by detecting R58 and L59/LAP-DPs. A total of 234 patients with CLD were included in this study. Liver biopsy specimens were used for immunostaining to detect R58/LAP-DPs, while plasma samples were subjected to an enzyme-linked immunosorbent assay to measure the L59/LAP-DP concentration. R58/LAP-DP was robustly expressed in and around the sinusoidal cells before the development of the fibrous regions. The R58/LAP-DP expression at fibrosis stage 1 was higher than at any other stages, and the relationship between the plasma L59/LAP-DP level and the stage of fibrosis also showed a similar trend. The abundance of plasma L59/LAP-DP showed no correlation with the levels of direct serum biomarkers of liver fibrosis; however, its changes during interferon-based therapy for chronic hepatitis C were significantly associated with virological responses. Our results suggest that PLK-dependent TGF-β activation occurs in the early stages of fibrosis and that its unique surrogate markers, R58 and L59/LAP-DPs, are useful for monitoring the clinical course of CLD.
Collapse
Affiliation(s)
- Hiroshi Yokoyama
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takahiro Masaki
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Ikuyo Inoue
- Liver Cancer Prevention Research Unit, RIKEN Center for Integrative Medical Sciences, Saitama, Japan
| | - Mariko Nakamura
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshihiro Mezaki
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Chisato Saeki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tsunekazu Oikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyuki Takahashi
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Masahiro Ikegami
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Hano
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kenichi Ikejima
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Soichi Kojima
- Liver Cancer Prevention Research Unit, RIKEN Center for Integrative Medical Sciences, Saitama, Japan
| | - Tomokazu Matsuura
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
49
|
Greenhalgh SN, Matchett KP, Taylor RS, Huang K, Li JT, Saeteurn K, Donnelly MC, Simpson EEM, Pollack JL, Atakilit A, Simpson KJ, Maher JJ, Iredale JP, Sheppard D, Henderson NC. Loss of Integrin αvβ8 in Murine Hepatocytes Accelerates Liver Regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:258-271. [PMID: 30448409 PMCID: PMC6360354 DOI: 10.1016/j.ajpath.2018.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/14/2018] [Accepted: 10/10/2018] [Indexed: 02/08/2023]
Abstract
Recent fate-mapping studies in mice have provided substantial evidence that mature adult hepatocytes are a major source of new hepatocytes after liver injury. In other systems, integrin αvβ8 has a major role in activating transforming growth factor (TGF)-β, a potent inhibitor of hepatocyte proliferation. We hypothesized that depletion of hepatocyte integrin αvβ8 would increase hepatocyte proliferation and accelerate liver regeneration after injury. Using Itgb8flox/flox;Alb-Cre mice to deplete hepatocyte αvβ8, after partial hepatectomy, hepatocyte proliferation and liver-to-body weight ratio were significantly increased in Itgb8flox/flox;Alb-Cre mice compared with control mice. Antibody-mediated blockade of hepatocyte αvβ8 in vitro, with assessment of TGF-β signaling pathways by real-time quantitative PCR array, supported the hypothesis that integrin αvβ8 inhibition alters hepatocyte TGF-β signaling toward a pro-regenerative phenotype. A diethylnitrosamine-induced model of hepatocellular carcinoma, used to examine the possibility that this pro-proliferative phenotype might be oncogenic, revealed no difference in either tumor number or size between Itgb8flox/flox;Alb-Cre and control mice. Immunohistochemistry for integrin αvβ8 in healthy and injured human liver demonstrated that human hepatocytes express integrin αvβ8. Depletion of hepatocyte integrin αvβ8 results in increased hepatocyte proliferation and accelerated liver regeneration after partial hepatectomy in mice. These data demonstrate that targeting integrin αvβ8 may represent a promising therapeutic strategy to drive liver regeneration in patients with a broad range of liver diseases.
Collapse
Affiliation(s)
- Stephen N Greenhalgh
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Kylie P Matchett
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard S Taylor
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Katherine Huang
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - John T Li
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Koy Saeteurn
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Mhairi C Donnelly
- Department of Hepatology, Scottish Liver Transplant Unit and University of Edinburgh, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Eilidh E M Simpson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Joshua L Pollack
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Amha Atakilit
- Department of Pathology, University of California, San Francisco, San Francisco, California
| | - Kenneth J Simpson
- Department of Hepatology, Scottish Liver Transplant Unit and University of Edinburgh, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Jacquelyn J Maher
- Liver Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - John P Iredale
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Senate House, University of Bristol, Bristol, United Kingdom
| | - Dean Sheppard
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
50
|
Manco R, Leclercq IA, Clerbaux LA. Liver Regeneration: Different Sub-Populations of Parenchymal Cells at Play Choreographed by an Injury-Specific Microenvironment. Int J Mol Sci 2018; 19:E4115. [PMID: 30567401 PMCID: PMC6321497 DOI: 10.3390/ijms19124115] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023] Open
Abstract
Liver regeneration is crucial for the maintenance of liver functional mass during homeostasis and diseases. In a disease context-dependent manner, liver regeneration is contributed to by hepatocytes or progenitor cells. As long as they are replicatively competent, hepatocytes are the main cell type responsible for supporting liver size homeostasisand regeneration. The concept that all hepatocytes within the lobule have the same proliferative capacity but are differentially recruited according to the localization of the wound, or whether a yet to be defined sub-population of hepatocytes supports regeneration is still debated. In a chronically or severely injured liver, hepatocytes may enter a state of replicative senescence. In such conditions, small biliary cells activate and expand, a process called ductular reaction (DR). Work in the last few decades has demonstrated that DR cells can differentiate into hepatocytes and thereby contribute to parenchymal reconstitution. In this study we will review the molecular mechanisms supporting these two processes to determine potential targets that would be amenable for therapeutic manipulation to enhance liver regeneration.
Collapse
Affiliation(s)
- Rita Manco
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium.
| | - Isabelle A Leclercq
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium.
| | - Laure-Alix Clerbaux
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium.
| |
Collapse
|