1
|
Qiu T, Zhu X, Wu J, Hong W, Hu W, Fang T. Mechanisms of rifaximin inhibition of hepatic fibrosis in mice with metabolic dysfunction associated steatohepatitis through the TLR4/NFκB pathway. Sci Rep 2025; 15:9815. [PMID: 40118973 PMCID: PMC11928543 DOI: 10.1038/s41598-025-92282-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/26/2025] [Indexed: 03/24/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) has become a serious public health problem, posing an increasingly dangerous threat to human health owing to its increasing prevalence and accompanying intra- and extrahepatic adverse outcomes. Rifaximin is considered to have therapeutic potential for MASH; however, its efficacy remains controversial. Our study aimed to observe the ameliorative effects of rifaximin and explore its possible mechanisms at the cellular level. 1. 42 male C57BL/6J mice were divided into 3 groups, the CON group and MCD group were fed with normal feed and MCD feed for 12 weeks respectively, and the MCD + RFX group was treated with rifaximin by gavage for 4 weeks on the basis of MCD feed. Hematoxylin-eosin staining, Sirius red staining and immunohistochemical staining were used to observe the histopathological changes of liver and intestine. Differences in liver transaminases, inflammatory factors, fibrosis indexes and intestinal tight junction proteins were compared among the 3 groups of mice. 2. A MASH cell model was constructed by inducing HepG2 cells with free fatty acids to observe the effects of rifaximin on MASH in vitro. In addition, the effects of rifaximin on TLR4/NF-κB signaling pathway were explored by applying TLR4 agonist LPS and TLR4 inhibitor TAK-242. Hepatic histopathology was significantly improved in MASH mice after rifaximin treatment, and their serum alanine aminotransferase and aspartate aminotransferase levels were (72.72 ± 5.68) U/L and (222.8 ± 11.22) U/L, respectively, which were significantly lower than those in the MCD group [(293.3 ± 10.69) U/L and (414.1 ± 36.29) U/L, P < 0.05], and the levels of inflammatory factors and fibrosis indicators were reduced. Rifaximin ameliorated intestinal barrier injury with increased expression of intestinal tight junction protein ZO-1 in the MCD + RFX group of mice, and the concentration of LPS-binding proteins (4.92 ± 0.55 vs. 15.82 ± 1.71, P < 0.05) was lower than that in the MCD group. In the NASH cell model, rifaximin similarly exerted inhibitory effects on its inflammatory factors and TLR4/NF-κB signaling pathway. Application of TLR4 inhibitors weakened the inhibitory effect of rifaximin on MASH. Our study supports rifaximin as a potential treatment for MASH, with potential mechanisms related to improving intestinal barrier integrity and downregulating the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ting Qiu
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of General Practice, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Xiaodong Zhu
- Department of Gastroenterology, Quanzhou First Hospital, Quanzhou, China
| | - Jingju Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wenyuan Hong
- Anxi Maternal and Child Health Hospital, Quanzhou, China
| | - Weitao Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
2
|
Tuffs C, Dupovac M, Richter K, Holten S, Schaschinger T, Marg O, Poljo A, Tasdemir AN, Harnoss JM, Billeter A, Schneider M, Strowitzki MJ. Genetic Loss of HIF-Prolyl-Hydroxylase 1, but Not Pharmacological Inhibition, Mitigates Hepatic Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:480-493. [PMID: 39566823 DOI: 10.1016/j.ajpath.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024]
Abstract
Liver fibrosis is characterized by excessive deposition of extracellular matrix due to chronic inflammation of the liver. Hepatic stellate cells (HSCs) become activated and produce increased amounts of extracellular matrix. Loss of HIF-prolyl-hydroxylase 1 (PHD1) attenuates HSC activation and fibrotic tissue remodeling in a murine model of biliary liver fibrosis. Herein, the protective effect of PHD1 deficiency (PHD1-/-) in an additional (toxic) model of liver fibrosis was validated and the effect of dimethyloxalylglycine (DMOG), a pan-HIF-prolyl-hydroxylase inhibitor, on the development of liver fibrosis, was evaluated. Liver fibrosis was induced utilizing carbon tetrachloride in wild-type (WT) and PHD1-/- mice treated with either vehicle or DMOG. To assess fibrosis development, expression of profibrotic genes in the livers was analyzed by Sirius red staining. When compared with WT mice, PHD1-/- mice developed less-severe liver fibrosis. DMOG treatment did not prevent this liver fibrosis. PHD1-/- mice had fewer α-SMA+ cells and less macrophage infiltration compared with WT mice. Expression of profibrogenic and proinflammatory genes was reduced in livers from carbon tetrachloride-exposed PHD1-/- mice. In vitro analyses of PHD1-deficient human HSCs revealed attenuated mRNA levels of profibrotic genes, as well as impaired migration and invasion. Although PHD1 deficiency attenuated activation of HSCs, pharmacologic PHD inhibition did not ameliorate fibrosis development. These data indicate that selective PHD1 inhibitors could prove effective in preventing and treating liver fibrosis.
Collapse
Affiliation(s)
- Christopher Tuffs
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Mareen Dupovac
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Katrin Richter
- Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany; Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Sophia Holten
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Thomas Schaschinger
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Oliver Marg
- Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Adisa Poljo
- Clarunis University Digestive Healthcare Center Basel, Basel, Switzerland
| | - Ayse Nur Tasdemir
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Jonathan M Harnoss
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Adrian Billeter
- Clarunis University Digestive Healthcare Center Basel, Basel, Switzerland
| | - Martin Schneider
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Moritz J Strowitzki
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany.
| |
Collapse
|
3
|
Pan M, Li H, Shi X. A New Target for Hepatic Fibrosis Prevention and Treatment: The Warburg Effect. FRONT BIOSCI-LANDMRK 2024; 29:321. [PMID: 39344326 DOI: 10.31083/j.fbl2909321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 10/01/2024]
Abstract
Hepatic fibrosis is a major public health problem that endangers human wellbeing. In recent years, a number of studies have revealed the important impact of metabolic reprogramming on the occurrence and development of hepatic fibrosis. Among them, the Warburg effect, as an intracellular glucose metabolism reprogramming, can promote the occurrence and development of hepatic fibrosis by promoting the activation of hepatic stellate cells (HSCs) and inducing the polarization of liver macrophages (KC). Understanding the Warburg effect and its important role in the progression of hepatic fibrosis will assist in developing new strategies for the prevention and treatment of hepatic fibrosis. This review focuses on the Warburg effect and the specific mechanism by which it affects the progression of hepatic fibrosis by regulating HSCs activation and KC polarization. In addition, we also summarize and discuss the related experimental drugs and their mechanisms that inhibit the Warburg effect by targeting key proteins of glycolysis in order to improve hepatic fibrosis in the hope of providing more effective strategies for the clinical treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Meng Pan
- College of Basic Medical Sciences, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| | - Huanyu Li
- Second Clinical Medical College, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| | - Xiaoyan Shi
- College of Basic Medical Sciences, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| |
Collapse
|
4
|
Akkız H, Gieseler RK, Canbay A. Liver Fibrosis: From Basic Science towards Clinical Progress, Focusing on the Central Role of Hepatic Stellate Cells. Int J Mol Sci 2024; 25:7873. [PMID: 39063116 PMCID: PMC11277292 DOI: 10.3390/ijms25147873] [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: 05/23/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
The burden of chronic liver disease is globally increasing at an alarming rate. Chronic liver injury leads to liver inflammation and fibrosis (LF) as critical determinants of long-term outcomes such as cirrhosis, liver cancer, and mortality. LF is a wound-healing process characterized by excessive deposition of extracellular matrix (ECM) proteins due to the activation of hepatic stellate cells (HSCs). In the healthy liver, quiescent HSCs metabolize and store retinoids. Upon fibrogenic activation, quiescent HSCs transdifferentiate into myofibroblasts; lose their vitamin A; upregulate α-smooth muscle actin; and produce proinflammatory soluble mediators, collagens, and inhibitors of ECM degradation. Activated HSCs are the main effector cells during hepatic fibrogenesis. In addition, the accumulation and activation of profibrogenic macrophages in response to hepatocyte death play a critical role in the initiation of HSC activation and survival. The main source of myofibroblasts is resident HSCs. Activated HSCs migrate to the site of active fibrogenesis to initiate the formation of a fibrous scar. Single-cell technologies revealed that quiescent HSCs are highly homogenous, while activated HSCs/myofibroblasts are much more heterogeneous. The complex process of inflammation results from the response of various hepatic cells to hepatocellular death and inflammatory signals related to intrahepatic injury pathways or extrahepatic mediators. Inflammatory processes modulate fibrogenesis by activating HSCs and, in turn, drive immune mechanisms via cytokines and chemokines. Increasing evidence also suggests that cellular stress responses contribute to fibrogenesis. Recent data demonstrated that LF can revert even at advanced stages of cirrhosis if the underlying cause is eliminated, which inhibits the inflammatory and profibrogenic cells. However, despite numerous clinical studies on plausible drug candidates, an approved antifibrotic therapy still remains elusive. This state-of-the-art review presents cellular and molecular mechanisms involved in hepatic fibrogenesis and its resolution, as well as comprehensively discusses the drivers linking liver injury to chronic liver inflammation and LF.
Collapse
Affiliation(s)
- Hikmet Akkız
- Department of Gastroenterology and Hepatology, University of Bahçeşehir, Beşiktaş, Istanbul 34353, Turkey
| | - Robert K. Gieseler
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr University Bochum, In der Schornau 23–25, 44892 Bochum, Germany; (R.K.G.); (A.C.)
| | - Ali Canbay
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus, Ruhr University Bochum, In der Schornau 23–25, 44892 Bochum, Germany; (R.K.G.); (A.C.)
| |
Collapse
|
5
|
Parola M, Pinzani M. Liver fibrosis in NAFLD/NASH: from pathophysiology towards diagnostic and therapeutic strategies. Mol Aspects Med 2024; 95:101231. [PMID: 38056058 DOI: 10.1016/j.mam.2023.101231] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023]
Abstract
Liver fibrosis, as an excess deposition of extracellular matrix (ECM) components, results from chronic liver injury as well as persistent activation of inflammatory response and of fibrogenesis. Liver fibrosis is a major determinant for chronic liver disease (CLD) progression and in the last two decades our understanding on the major molecular and cellular mechanisms underlying the fibrogenic progression of CLD has dramatically improved, boosting pre-clinical studies and clinical trials designed to find novel therapeutic approaches. From these studies several critical concepts have emerged, starting to reveal the complexity of the pro-fibrotic microenvironment which involves very complex, dynamic and interrelated interactions between different hepatic and extrahepatic cell populations. This review will offer first a recapitulation of established and novel pathophysiological basic principles and concepts by intentionally focus the attention on NAFLD/NASH, a metabolic-related form of CLD with a high impact on the general population and emerging as a leading cause of CLD worldwide. NAFLD/NASH-related pro-inflammatory and profibrogenic mechanisms will be analysed as well as novel information on cells, mediators and signalling pathways which have taken advantage from novel methodological approaches and techniques (single cell genomics, imaging mass cytometry, novel in vitro two- and three-dimensional models, etc.). We will next offer an overview on recent advancement in diagnostic and prognostic tools, including serum biomarkers and polygenic scores, to support the analysis of liver biopsies. Finally, this review will provide an analysis of current and emerging therapies for the treatment of NAFLD/NASH patients.
Collapse
Affiliation(s)
- Maurizio Parola
- Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Corso Raffaello 30, 10125, Torino, Italy.
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Division of Medicine - Royal Free Hospital, London, NW32PF, United Kingdom.
| |
Collapse
|
6
|
Li Z, Zhu J, Ouyang H. Research progress of traditional Chinese medicine in improving hepatic fibrosis based on inhibiting pathological angiogenesis. Front Pharmacol 2023; 14:1303012. [PMID: 38155904 PMCID: PMC10754536 DOI: 10.3389/fphar.2023.1303012] [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: 09/27/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
Hepatic fibrosis is the formation of scar tissue in the liver. This scar tissue replaces healthy liver tissue and can lead to liver dysfunction and failure if left untreated. It is usually caused by chronic liver disease, such as hepatitis B or C, alcohol abuse, or non-alcoholic fatty liver disease. Pathological angiogenesis plays a crucial role in the development of hepatic fibrosis by promoting the growth of new blood vessels in the liver. These new vessels increase blood flow to the damaged areas of the liver, which triggers the activation of hepatic stellate cells (HSCs). HSCs are responsible for producing excess collagen and other extracellular matrix proteins that contribute to the development of fibrosis. Pathological angiogenesis plays a crucial role in the development of hepatic fibrosis by promoting the growth of new blood vessels in the liver. These new vessels increase blood flow to the damaged areas of the liver, which triggers the activation of HSCs. HSCs are responsible for producing excess collagen and other extracellular matrix proteins that contribute to the development of fibrosis. Traditional Chinese medicine (TCM) has been found to target pathological angiogenesis, thereby providing a potential treatment option for hepatic fibrosis. Several studies have demonstrated that TCM exhibits anti-angiogenic effects by inhibiting the production of pro-angiogenic factors, such as vascular endothelial growth factor and angiopoietin-2, and by reducing the proliferation of endothelial cells. Reviewing and highlighting the unique TCM recognition of treating hepatic fibrosis by targeting pathological angiogenesis may shed light on future hepatic fibrosis research.
Collapse
|
7
|
Friedman SL, Pinzani M. Hepatic fibrosis 2022: Unmet needs and a blueprint for the future. Hepatology 2022; 75:473-488. [PMID: 34923653 DOI: 10.1002/hep.32285] [Citation(s) in RCA: 250] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022]
Abstract
Steady progress over four decades toward understanding the pathogenesis and clinical consequences of hepatic fibrosis has led to the expectation of effective antifibrotic drugs, yet none has been approved. Thus, an assessment of the field is timely, to clarify priorities and accelerate progress. Here, we highlight the successes to date but, more importantly, identify gaps and unmet needs, both experimentally and clinically. These include the need to better define cell-cell interactions and etiology-specific elements of fibrogenesis and their link to disease-specific drivers of portal hypertension. Success in treating viral hepatitis has revealed the remarkable capacity of the liver to degrade scar in reversing fibrosis, yet we know little of the mechanisms underlying this response. Thus, there is an exigent need to clarify the cellular and molecular mechanisms of fibrosis regression in order for therapeutics to mimic the liver's endogenous capacity. Better refined and more predictive in vitro and animal models will hasten drug development. From a clinical perspective, current diagnostics are improving but not always biologically plausible or sufficiently accurate to supplant biopsy. More urgently, digital pathology methods that leverage machine learning and artificial intelligence must be validated in order to capture more prognostic information from liver biopsies and better quantify the response to therapies. For more refined treatment of NASH, orthogonal approaches that integrate genetic, clinical, and pathological data sets may yield treatments for specific subphenotypes of the disease. Collectively, these and other advances will strengthen and streamline clinical trials and better link histologic responses to clinical outcomes.
Collapse
Affiliation(s)
- Scott L Friedman
- Division of Liver DiseasesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Massimo Pinzani
- Institute for Liver and Digestive HealthUniversity College LondonLondonUK
| |
Collapse
|
8
|
Wang J, Cao X, Deng S, Wang B, Feng J, Meng F, Xu H, Wang S, Zao X, Li H, Ye Y. Effect of liver cirrhosis on erectile function in rats: A study combining bioinformatics analysis and experimental research. Andrologia 2021; 54:e14352. [PMID: 34921688 DOI: 10.1111/and.14352] [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: 10/27/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022] Open
Abstract
To explore the mechanism through which liver cirrhosis (LC) causes erectile dysfunction (ED). Bioinformatic analysis was used to predict the potential signalling pathways in LC-induced ED, and N-nitrosodiethylamine was used to establish a rat model of LC. H&E staining, Western blotting and RT-qPCR were used to detect pathological tissue damage and changes in mRNA and protein expression levels. In addition, the expression levels of sex hormones such as estradiol (E2), prolactin (PRL) and testosterone (T) were measured. The hypoxia-inducible factor (HIF) pathway was an important pathway in our bioinformatics prediction. Pathological damages were detected in the liver and penile tissues of the model rats. Compared with the normal group's serum hormone levels, E2 and PRL were increased in LC rats, while T was decreased (p < 0.01). The mRNA and protein expression results from penis tissues showed that endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS) were both downregulated, and HIF-1α was upregulated in the model group compared to the normal group (p < 0.01). These data suggest that LC hinders erectile function and causes histopathological changes in the penis by affecting the expression of HIF-1α, eNOS, iNOS, E2, PRL and T.
Collapse
Affiliation(s)
- Jisheng Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Department of Andrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xu Cao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Department of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Sheng Deng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Department of Andrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Bin Wang
- Department of Andrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junlong Feng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Department of Andrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fanchao Meng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Department of Andrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hongsheng Xu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Department of Andrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shizhen Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Department of Andrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaobin Zao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Dongzhimen Hospital, Beijing University of Chinese Medicine, Ministry of Education/Beijing Key Laboratory of Internal Medicine of Traditional Chinese Medicine, China
| | - Haisong Li
- Department of Andrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yongan Ye
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Department of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Institute of Liver Diseases, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Ezhilarasan D. Mitochondria: A critical hub for hepatic stellate cells activation during chronic liver diseases. Hepatobiliary Pancreat Dis Int 2021; 20:315-322. [PMID: 33975780 DOI: 10.1016/j.hbpd.2021.04.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 04/19/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Upon liver injury, quiescent hepatic stellate cells (qHSCs), reside in the perisinusoidal space, phenotypically transdifferentiate into myofibroblast-like cells (MFBs). The qHSCs in the normal liver are less fibrogenic, migratory, and also have less proliferative potential. However, activated HSCs (aHSCs) are more fibrogenic and have a high migratory and proliferative MFBs phenotype. HSCs activation is a highly energetic process that needs abundant intracellular energy in the form of adenosine triphosphate (ATP) for the synthesis of extracellular matrix (ECM) in the injured liver to substantiate the injury. DATA SOURCES The articles were collected through PubMed and EMBASE using search terms "mitochondria and hepatic stellate cells", "mitochondria and HSCs", "mitochondria and hepatic fibrosis", "mitochondria and liver diseases", and "mitochondria and chronic liver disease", and relevant publications published before September 31, 2020 were included in this review. RESULTS Mitochondria homeostasis is affected during HSCs activation. Mitochondria in aHSCs are highly energetic and are in a high metabolically active state exhibiting increased activity such as glycolysis and respiration. aHSCs have high glycolytic enzymes expression and glycolytic activity induced by Hedgehog (Hh) signaling from injured hepatocytes. Increased glycolysis and aerobic glycolysis (Warburg effect) end-products in aHSCs consequently activate the ECM-related gene expressions. Increased Hh signaling from injured hepatocytes downregulates peroxisome proliferator-activated receptor-γ expression and decreases lipogenesis in aHSCs. Glutaminolysis and tricarboxylic acid cycle liberate ATPs that fuel HSCs to proliferate and produce ECM during their activation. CONCLUSIONS Available studies suggest that mitochondria functions can increase in parallel with HSCs activation. Therefore, mitochondrial modulators should be tested in an elaborate manner to control or prevent the HSCs activation during liver injury to subsequently regress hepatic fibrosis.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, the Blue Lab, Molecular Medicine and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600077, Tamil Nadu, India.
| |
Collapse
|
10
|
Hypoxia, Hypoxia-Inducible Factors and Liver Fibrosis. Cells 2021; 10:cells10071764. [PMID: 34359934 PMCID: PMC8305108 DOI: 10.3390/cells10071764] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
Liver fibrosis is a potentially reversible pathophysiological event, leading to excess deposition of extracellular matrix (ECM) components and taking place as the net result of liver fibrogenesis, a dynamic and highly integrated process occurring during chronic liver injury of any etiology. Liver fibrogenesis and fibrosis, together with chronic inflammatory response, are primarily involved in the progression of chronic liver diseases (CLD). As is well known, a major role in fibrogenesis and fibrosis is played by activated myofibroblasts (MFs), as well as by macrophages and other hepatic cell populations involved in CLD progression. In the present review, we will focus the attention on the emerging pathogenic role of hypoxia, hypoxia-inducible factors (HIFs) and related mediators in the fibrogenic progression of CLD.
Collapse
|
11
|
Role of Angiogenesis in the Pathogenesis of NAFLD. J Clin Med 2021; 10:jcm10071338. [PMID: 33804956 PMCID: PMC8037441 DOI: 10.3390/jcm10071338] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/19/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the leading cause of chronic liver disease, exposing to the risk of liver fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Angio-genesis is a complex process leading to the development of new vessels from pre-existing vessels. Angiogenesis is triggered by hypoxia and inflammation and is driven by the action of proangiogenic cytokines, mainly vascular endothelial growth factor (VEGF). In this review, we focus on liver angiogenesis associated with NAFLD and analyze the evidence of liver angiogenesis in animal models of NAFLD and in NAFLD patients. We also report the data explaining the role of angiogenesis in the progression of NAFLD and discuss the potential of targeting angiogenesis, notably VEGF, to treat NAFLD.
Collapse
|
12
|
Sun J, Shi L, Xiao T, Xue J, Li J, Wang P, Wu L, Dai X, Ni X, Liu Q. microRNA-21, via the HIF-1α/VEGF signaling pathway, is involved in arsenite-induced hepatic fibrosis through aberrant cross-talk of hepatocytes and hepatic stellate cells. CHEMOSPHERE 2021; 266:129177. [PMID: 33310519 DOI: 10.1016/j.chemosphere.2020.129177] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/17/2020] [Accepted: 11/29/2020] [Indexed: 06/12/2023]
Abstract
Long-term exposure to arsenic, a widely distributed environmental toxicant, may result in damage to various organs, including the liver. Mice exposed chronically to arsenite developed hepatic damage, inflammation, and fibrosis, as well as increased levels of microRNA-21 (miR-21) and hypoxia-inducible factor (HIF)-1α. The levels of miR-21 and HIF-1α were also enhanced in primary hepatocytes and L-02 cells exposed to arsenite. The culture media from these cells induced the activation of hepatic stellate cells (HSCs), as demonstrated by up-regulation of the protein levels of α-smooth muscle actin (α-SMA) and collagen1A2 (COL1A2) and by increased activity in gel contractility assays. For L-02 cells, knockdown of miR-21 blocked the arsenite-induced up-regulation of HIF-1α and vascular endothelial growth factor (VEGF), which prevented the activation of LX-2 cells induced by medium from arsenite-exposed L-02 cells. However, these effects were reversed by down-regulation of von Hippel Lindau protein (pVHL). In arsenite-treated L-02 cells, miR-21 knockdown elevated the levels of ubiquitination and accelerated the degradation of HIF-1α via pVHL. In the livers of miR-21-/- mice exposed chronically to arsenite, there were less hepatic damage, lower fibrosis, lower levels of HIF-1α and VEGF, and higher levels of pVHL than for wild-type mice. In summary, we propose that miR-21, acting via the HIF-1α/VEGF signaling pathway, is involved in arsenite-induced hepatic fibrosis through mediating aberrant cross-talk of hepatocytes and HSCs. The findings provide evidence relating to the pathogenesis of hepatic fibrosis induced by exposure to arsenic.
Collapse
Affiliation(s)
- Jing Sun
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Le Shi
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Tian Xiao
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Junchao Xue
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Junjie Li
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Peiwen Wang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Lu Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Xiangyu Dai
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Xinye Ni
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, 213003, Jiangsu, People's Republic of China.
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
| |
Collapse
|
13
|
Rosso C, Caviglia GP, Younes R, Ribaldone DG, Fagoonee S, Pellicano R, Bugianesi E. Molecular mechanisms of hepatic fibrosis in chronic liver diseases. MINERVA BIOTECNOL 2020; 32. [DOI: 10.23736/s1120-4826.20.02619-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Novo E, Bocca C, Foglia B, Protopapa F, Maggiora M, Parola M, Cannito S. Liver fibrogenesis: un update on established and emerging basic concepts. Arch Biochem Biophys 2020; 689:108445. [PMID: 32524998 DOI: 10.1016/j.abb.2020.108445] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/20/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023]
Abstract
Liver fibrogenesis is defined as a dynamic and highly integrated process occurring during chronic injury to liver parenchyma that can result in excess deposition of extracellular matrix (ECM) components (i.e., liver fibrosis). Liver fibrogenesis, together with chronic inflammatory response, is then primarily involved in the progression of chronic liver diseases (CLD) irrespective of the specific etiology. In the present review we will first offer a synthetic and updated overview of major basic concepts in relation to the role of myofibroblasts (MFs), macrophages and other hepatic cell populations involved in CLD to then offer an overview of established and emerging issues and mechanisms that have been proposed to favor and/or promote CLD progression. A special focus will be dedicated to selected issues that include emerging features in the field of cholangiopathies, the emerging role of genetic and epigenetic factors as well as of hypoxia, hypoxia-inducible factors (HIFs) and related mediators.
Collapse
Affiliation(s)
- Erica Novo
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| | - Claudia Bocca
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| | - Beatrice Foglia
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| | - Francesca Protopapa
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| | - Marina Maggiora
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| | - Maurizio Parola
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy.
| | - Stefania Cannito
- University of Torino, Dept. Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, Corso Raffaello 30, 10125, Torino, Italy
| |
Collapse
|
15
|
Luo X, Gao ZX, Lin SW, Tong ML, Liu LL, Lin LR, Ke WJ, Yang TC. Recombinant Treponema pallidum protein Tp0136 promotes fibroblast migration by modulating MCP-1/CCR2 through TLR4. J Eur Acad Dermatol Venereol 2020; 34:862-872. [PMID: 31856347 DOI: 10.1111/jdv.16162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/12/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Chancre self-healing is an important clinical feature in the early stages of syphilis infection. Wound healing may involve an important mechanism by the migration of fibroblasts filling the injured lesion. However, the specific mechanism underlying this process is still unknown. OBJECTIVES We aimed to analyse the role of Tp0136 in the migration of fibroblasts and the related mechanism. METHODS The migration ability of fibroblasts was detected by a wound-healing assay. RT-PCR and ELISA detected the expression of MCP-1, IL-6 and MMP-9. TLR4 expression was detected by RT-PCR. The protein levels of CCR2 and relevant signalling pathway molecules were measured by Western blotting. RESULTS Tp0136 significantly promoted fibroblast migration. Subsequently, the levels of MCP-1 and its receptor CCR2 were increased in this process. The migration of fibroblasts was significantly inhibited by an anti-MCP-1 neutralizing antibody or CCR2 inhibitors. Furthermore, studies demonstrated that Tp0136 could activate the ERK/JNK/PI3K/NF-κB signalling pathways through TLR4 activity and that signalling pathways inhibitors could weaken MCP-1 secretion and fibroblast migration. CONCLUSIONS These findings demonstrate that Tp0136 promotes the migration of fibroblasts by inducing MCP-1/CCR2 expression through signalling involving the TLR4, ERK, JNK, PI3K and NF-κB signalling pathways, which could contribute to the mechanism of chancre self-healing in syphilis.
Collapse
Affiliation(s)
- X Luo
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Z-X Gao
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - S-W Lin
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - M-L Tong
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China.,Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China
| | - L-L Liu
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China.,Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China
| | - L-R Lin
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China.,Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China
| | - W-J Ke
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - T-C Yang
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China.,Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
16
|
Lacin S, Yalcin S. The Prognostic Value of Circulating VEGF-A Level in Patients With Hepatocellular Cancer. Technol Cancer Res Treat 2020; 19:1533033820971677. [PMID: 33234055 PMCID: PMC7705781 DOI: 10.1177/1533033820971677] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Neovascularization plays a crucial pathogenic role in tumor development and
vascular endothelial growth factor (VEGF-A) is a key signaling element that
drives angiogenesis, thereby facilitating hepatocellular cancer (HCC) growth
and metastasis. We aimed to define the relationship between serum VEGF-A
levels and clinical outcomes in a cohort of Turkish patients with HCC. Methods: We enrolled and prospectively followed 84 patients with HCC in our study.
Serum VEGF-A levels were measured and we assessed the association between
VEGF-A levels and clinical features. Results: Forty-eight patients had cirrhosis while 35 patients were noncirrhotic. Serum
VEGF-A levels were significantly lower in HCC patients with cirrhosis
compared to non-cirrhotic HCC patients (p = 0.03).In terms of viral
hepatitis subtype, 36 (%42.8) of patients were hepatitis B virus (HBV)
positive and 8 (%9.5) of patients were hepatitis C virus (HCV) positive.
Patients with serum VEGF-A levels ≥100 pg/mL had significantly lower OS
rates than patients with serum VEGF-A level <100 pg/mL (p = 0.01). The OS
rates were 5.8 and 14.2 months, respectively (p = 0.02). The median OS was
7.38 months (95% CI: 5.89-13.79 months). We observed a significant
relationship between serum VEGF-A level and tumor size. Patients with tumor
size ≤ 5cm had lower VEGF-A levels than patients with VEGF-A levels <5
cm. The VEGF-A levels were 132.7 and 342.1 pg/mL, respectively (p <
0.001). The median follow-up was 32 months. Conclusions: Serum VEGF-A level, a biological marker of angiogenesis, is an independent
predictor of survival in patients with HCC. Serum VEGF-A levels may be
utilized to predict response to treatment targeting serum VEGF-A in patients
with HCC.
Collapse
Affiliation(s)
- Sahin Lacin
- Department of Medical Oncology, Faculty of Medicine, 64173Yeditepe University, İstanbul, Turkey
| | - Suayib Yalcin
- Department of Medical Oncology, 64005Hacettepe University, Hacettepe Cancer Institute, Ankara, Turkey
| |
Collapse
|
17
|
Yu F, Dong B, Dong P, He Y, Zheng J, Xu P. Hypoxia induces the activation of hepatic stellate cells through the PVT1-miR-152-ATG14 signaling pathway. Mol Cell Biochem 2019; 465:115-123. [PMID: 31893334 DOI: 10.1007/s11010-019-03672-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 12/01/2019] [Indexed: 02/07/2023]
Abstract
Increasing studies have indicated that hypoxia serves as a pivotal microenvironmental factor that facilitates activation of hepatic stellate cells (HSCs). However, the mechanism by which hypoxia activates HSCs is not clear. Here, we demonstrated that plasmacytoma variant translocation 1 (PVT1) and autophagy were overexpressed in liver fibrotic specimens. In primary mouse HSCs, both PVT1 and autophagy were induced by hypoxia. Further study showed that hypoxia-induced autophagy depended on expression of PVT1 and miR-152 in HSCs. Luciferase reporter assay indicated that autophagy-related gene 14 (ATG14) was a direct target of miR-152. In addition, inhibition of autophagy by 3-methyladenine and Beclin-1 siRNA impeded activation of HSCs cultured in 1% O2. Taken together, autophagy induction via the PVT1-miR-152-ATG14 signaling pathway contributes to activation of HSCs under hypoxia condition.
Collapse
Affiliation(s)
- Fujun Yu
- Department of Gastroenterology, Shanghai Songjiang District Central Hospital, Shanghai, China
- Department of Gastroenterology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (Preparatory Stage), Shanghai, China
- Department of Gastroenterology, Shanghai Songjiang Clinical Medical College of Nanjing Medical University, Shanghai, China
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Buyuan Dong
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peihong Dong
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanghuan He
- Department of Gastroenterology, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Jianjian Zheng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, China.
| | - Ping Xu
- Department of Gastroenterology, Shanghai Songjiang District Central Hospital, Shanghai, China.
- Department of Gastroenterology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (Preparatory Stage), Shanghai, China.
- Department of Gastroenterology, Shanghai Songjiang Clinical Medical College of Nanjing Medical University, Shanghai, China.
| |
Collapse
|
18
|
Foglia B, Sutti S, Pedicini D, Cannito S, Bocca C, Maggiora M, Bevacqua MR, Rosso C, Bugianesi E, Albano E, Novo E, Parola M. MicroRNA-Based Combinatorial Cancer Therapy: Effects of MicroRNAs on the Efficacy of Anti-Cancer Therapies. Cells 2019; 9:29. [PMID: 31861937 PMCID: PMC7017087 DOI: 10.3390/cells9010028] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023] Open
Abstract
The susceptibility of cancer cells to different types of treatments can be restricted by intrinsic and acquired therapeutic resistance, leading to the failure of cancer regression and remission. To overcome this problem, a combination therapy has been proposed as a fundamental strategy to improve therapeutic responses; however, resistance is still unavoidable. MicroRNA (miRNAs) are associated with cancer therapeutic resistance. The modulation of dysregulated miRNA levels through miRNA-based therapy comprising a replacement or inhibition approach has been proposed to sensitize cancer cells to other anti-cancer therapies. The combination of miRNA-based therapy with other anti-cancer therapies (miRNA-based combinatorial cancer therapy) is attractive, due to the ability of miRNAs to target multiple genes associated with the signaling pathways controlling therapeutic resistance. In this article, we present an overview of recent findings on the role of therapeutic resistance-related miRNAs in different types of cancer. We review the feasibility of utilizing dysregulated miRNAs in cancer cells and extracellular vesicles as potential candidates for miRNA-based combinatorial cancer therapy. We also discuss innate properties of miRNAs that need to be considered for more effective combinatorial cancer therapy.
Collapse
Affiliation(s)
- Beatrice Foglia
- Department Clinical and Biological Science, Unit of Experimental Medicine and Clinical Pathology, University of Torino, 10125 Torino, Italy; (B.F.); (S.C.); (C.B.); (M.M.); (M.R.B.); (E.N.)
| | - Salvatore Sutti
- Department of Health Science, University of East Piedmont, 28100 Novara, Italy; (S.S.); (E.A.)
| | - Dario Pedicini
- IRCC, Istituto per la Ricerca e la Cura del Cancro, Candiolo, 10060 Torino, Italy;
| | - Stefania Cannito
- Department Clinical and Biological Science, Unit of Experimental Medicine and Clinical Pathology, University of Torino, 10125 Torino, Italy; (B.F.); (S.C.); (C.B.); (M.M.); (M.R.B.); (E.N.)
| | - Claudia Bocca
- Department Clinical and Biological Science, Unit of Experimental Medicine and Clinical Pathology, University of Torino, 10125 Torino, Italy; (B.F.); (S.C.); (C.B.); (M.M.); (M.R.B.); (E.N.)
| | - Marina Maggiora
- Department Clinical and Biological Science, Unit of Experimental Medicine and Clinical Pathology, University of Torino, 10125 Torino, Italy; (B.F.); (S.C.); (C.B.); (M.M.); (M.R.B.); (E.N.)
| | - Maria Rosaria Bevacqua
- Department Clinical and Biological Science, Unit of Experimental Medicine and Clinical Pathology, University of Torino, 10125 Torino, Italy; (B.F.); (S.C.); (C.B.); (M.M.); (M.R.B.); (E.N.)
| | - Chiara Rosso
- Division of Gastroenterology and Hepatology, Department of Medical Science, University of Torino, 10154 Torino, Italy; (C.R.); (E.B.)
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Science, University of Torino, 10154 Torino, Italy; (C.R.); (E.B.)
| | - Emanuele Albano
- Department of Health Science, University of East Piedmont, 28100 Novara, Italy; (S.S.); (E.A.)
| | - Erica Novo
- Department Clinical and Biological Science, Unit of Experimental Medicine and Clinical Pathology, University of Torino, 10125 Torino, Italy; (B.F.); (S.C.); (C.B.); (M.M.); (M.R.B.); (E.N.)
| | - Maurizio Parola
- Department Clinical and Biological Science, Unit of Experimental Medicine and Clinical Pathology, University of Torino, 10125 Torino, Italy; (B.F.); (S.C.); (C.B.); (M.M.); (M.R.B.); (E.N.)
| |
Collapse
|
19
|
Delogu W, Caligiuri A, Provenzano A, Rosso C, Bugianesi E, Coratti A, Macias-Barragan J, Galastri S, Di Maira G, Marra F. Myostatin regulates the fibrogenic phenotype of hepatic stellate cells via c-jun N-terminal kinase activation. Dig Liver Dis 2019; 51:1400-1408. [PMID: 31005555 DOI: 10.1016/j.dld.2019.03.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/06/2019] [Accepted: 03/06/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Myostatin is mainly expressed in skeletal muscle, where it negatively regulates trophism. This myokine is implicated in the pathophysiology of nonalcoholic steatohepatitis, an emerging cause of liver fibrosis. In this study we explored the effects of myostatin on the biology of hepatic stellate cells. METHODS The effects of myostatin were assessed both in LX-2 and in human primary stellate cells. Cell migration was determined in Boyden chambers. Activation of intracellular pathways was evaluated by Western blotting. Procollagen type 1 secretion was measured by enzyme immunoassay. The role of c-Jun N-terminal kinase was assessed by pharmacologic and genetic inhibition. RESULTS Activin receptor-2B was up-regulated in livers of mice with experimental fibrosis, and detectable in human stellate cells. Serum myostatin levels increased in a model of acute liver injury. Myostatin reduced HSC proliferation, induced cell migration, and increased expression of procollagen type1, tissue inhibitor of metalloproteinase-1, and transforming growth factor-β1. Myostatin activated different signaling pathways, including c-Jun N-terminal kinase and Smad3. Genetic and/or pharmacologic inhibition of c-Jun N-terminal kinase activity significantly reduced cell migration and procollagen secretion in response to myostatin. CONCLUSIONS Activation of activin receptor-2B by myostatin modulates the fibrogenic phenotype of human stellate cells, indicating that a myokine may be implicated in the pathogenesis of hepatic fibrosis.
Collapse
Affiliation(s)
- Wanda Delogu
- Dipartimento di Medicina Sperimentale Clinica, University of Florence, Florence, Italy
| | - Alessandra Caligiuri
- Dipartimento di Medicina Sperimentale Clinica, University of Florence, Florence, Italy
| | - Angela Provenzano
- Dipartimento di Medicina Sperimentale Clinica, University of Florence, Florence, Italy
| | - Chiara Rosso
- Dipartimento di Scienze Mediche, University of Turin, Turin, Italy
| | | | - Andrea Coratti
- SOD Chirurgia Oncologia a indirizzo robotico, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Jose Macias-Barragan
- Dipartimento di Medicina Sperimentale Clinica, University of Florence, Florence, Italy
| | - Sara Galastri
- Dipartimento di Medicina Sperimentale Clinica, University of Florence, Florence, Italy
| | - Giovanni Di Maira
- Dipartimento di Medicina Sperimentale Clinica, University of Florence, Florence, Italy
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale Clinica, University of Florence, Florence, Italy; Research Center Denothe, University of Florence, Florence, Italy.
| |
Collapse
|
20
|
ERK Pathway in Activated, Myofibroblast-Like, Hepatic Stellate Cells: A Critical Signaling Crossroad Sustaining Liver Fibrosis. Int J Mol Sci 2019; 20:ijms20112700. [PMID: 31159366 PMCID: PMC6600376 DOI: 10.3390/ijms20112700] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/21/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022] Open
Abstract
Fibrogenic progression of chronic liver disease, whatever the etiology, is characterized by persistent chronic parenchymal injury, chronic activation of inflammatory response, and sustained activation of liver fibrogenesis, and of pathological wound healing response. A critical role in liver fibrogenesis is played by hepatic myofibroblasts (MFs), a heterogeneous population of α smooth-muscle actin—positive cells that originate from various precursor cells through a process of activation and transdifferentiation. In this review, we focus the attention on the role of extracellular signal-regulated kinase (ERK) signaling pathway as a critical one in modulating selected profibrogenic phenotypic responses operated by liver MFs. We will also analyze major therapeutic antifibrotic strategies developed in the last two decades in preclinical studies, some translated to clinical conditions, designed to interfere directly or indirectly with the Ras/Raf/MEK/ERK signaling pathway in activated hepatic MFs, but that also significantly increased our knowledge on the biology and pathobiology of these fascinating profibrogenic cells.
Collapse
|
21
|
Liver Zonation in Health and Disease: Hypoxia and Hypoxia-Inducible Transcription Factors as Concert Masters. Int J Mol Sci 2019; 20:ijms20092347. [PMID: 31083568 PMCID: PMC6540308 DOI: 10.3390/ijms20092347] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 02/06/2023] Open
Abstract
The liver and its zonation contribute to whole body homeostasis. Acute and chronic, not always liver, diseases impair proper metabolic zonation. Various underlying pathways, such as β-catenin, hedgehog signaling, and the Hippo pathway, along with the physiologically occurring oxygen gradient, appear to be contributors. Interestingly, hypoxia and hypoxia-inducible transcription factors can orchestrate those pathways. In the current review, we connect novel findings of liver zonation in health and disease and provide a view about the dynamic interplay between these different pathways and cell-types to drive liver zonation and systemic homeostasis.
Collapse
|
22
|
Shoukry H, Taher M, Enany A, Ahmed T, Hassan I. Combination of Caffeine and Liver Albumin Plus Protects against Smoking-Induced Liver Injury in Rats. DUBAI MEDICAL JOURNAL 2019. [DOI: 10.1159/000497078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
<b><i>Background:</i></b> A number of studies have revealed the hepatoprotective effect of coffee and tea. However, the role of caffeine on smoking-induced liver injury is not well elucidated. Liver Albumin Plus (LAP) is a liver supplement given in different liver diseases; to our knowledge, its role in smoking-induced liver injury is not clear. <b><i>Objectives:</i></b> This study aimed to find out the protective effect of caffeine and LAP alone and in combination in attenuation of smoking-induced liver injury. <b><i>Methods:</i></b> Thirty male albino rats were divided into a control group and a smoking group; the smoking group was then subdivided into a smoking group, a smoking + caffeine group, a smoking + LAP group, and a smoking + caffeine + LAP group. At the end of the experimental study, blood samples were collected for assessment of liver enzymes, alpha-fetoprotein (AFP), and interleukin 6, and livers were excised. Biochemical analysis of hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>), superoxide dismutase (SOD), and hypoxia-inducible factor (HIF) as well as histological examination were done. <b><i>Results:</i></b> The results showed that smoking elevated liver enzymes, AFP, H<sub>2</sub>O<sub>2</sub>, and HIF and decreased SOD; histologically, deterioration of the liver was observed. On administration of caffeine, significant (<i>p</i> < 0.05) improvement in all measured parameters and preserved liver histological structure were observed, while intake of LAP alone showed some improvement. In combination, all liver parameters were improved and histological structure was preserved in contrast to each drug alone. <b><i>Conclusion:</i></b> It is better to give a combination of caffeine and LAP with cigarettes smoking to attenuate smoking-induced liver injury.
Collapse
|
23
|
Hypoxia-Inducible Factor Prolyl 4-Hydroxylases and Metabolism. Trends Mol Med 2018; 24:1021-1035. [DOI: 10.1016/j.molmed.2018.10.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 12/17/2022]
|
24
|
Liver fibrosis: Pathophysiology, pathogenetic targets and clinical issues. Mol Aspects Med 2018; 65:37-55. [PMID: 30213667 DOI: 10.1016/j.mam.2018.09.002] [Citation(s) in RCA: 757] [Impact Index Per Article: 108.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 02/06/2023]
Abstract
The progression of chronic liver diseases (CLD), irrespective of etiology, involves chronic parenchymal injury, persistent activation of inflammatory response as well as sustained activation of liver fibrogenesis and wound healing response. Liver fibrogenesis, is a dynamic, highly integrated molecular, cellular and tissue process responsible for driving the excess accumulation of extracellular matrix (ECM) components (i.e., liver fibrosis) sustained by an eterogeneous population of hepatic myofibroblasts (MFs). The process of liver fibrogenesis recognizes a number of common and etiology-independent mechanisms and events but it is also significantly influenced by the specific etiology, as also reflected by peculiar morphological patterns of liver fibrosis development. In this review we will analyze the most relevant established and/or emerging pathophysiological issues underlying CLD progression with a focus on the role of critical hepatic cell populations, mechanisms and signaling pathways involved, as they represent potential therapeutic targets, to finally analyze selected and relevant clinical issues.
Collapse
|
25
|
Garbuzenko DV, Arefyev NO, Kazachkov EL. Antiangiogenic therapy for portal hypertension in liver cirrhosis: Current progress and perspectives. World J Gastroenterol 2018; 24:3738-3748. [PMID: 30197479 PMCID: PMC6127663 DOI: 10.3748/wjg.v24.i33.3738] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 02/06/2023] Open
Abstract
Developing medicines for hemodynamic disorders that are characteristic of cirrhosis of the liver is a relevant problem in modern hepatology. The increase in hepatic vascular resistance to portal blood flow and subsequent hyperdynamic circulation underlie portal hypertension (PH) and promote its progression, despite the formation of portosystemic collaterals. Angiogenesis and vascular bed restructurization play an important role in PH pathogenesis as well. In this regard, strategic directions in the therapy for PH in cirrhosis include selectively decreasing hepatic vascular resistance while preserving or increasing portal blood flow, and correcting hyperdynamic circulation and pathological angiogenesis. The aim of this review is to describe the mechanisms of angiogenesis in PH and the methods of antiangiogenic therapy. The PubMed database, the Google Scholar retrieval system, and the reference lists from related articles were used to search for relevant publications. Articles corresponding to the aim of the review were selected for 2000-2017 using the keywords: "liver cirrhosis", "portal hypertension", "pathogenesis", "angiogenesis", and "antiangiogenic therapy". Antiangiogenic therapy for PH was the inclusion criterion. In this review, we have described angiogenesis inhibitors and their mechanism of action in relation to PH. Although most of them were studied only in animal experiments, this selective therapy for abnormally growing newly formed vessels is pathogenetically reasonable to treat PH and associated complications.
Collapse
Affiliation(s)
| | - Nikolay Olegovich Arefyev
- Department of Pathological Anatomy and Forensic Medicine, South Ural State Medical University, Chelyabinsk 454092, Russia
| | - Evgeniy Leonidovich Kazachkov
- Department of Pathological Anatomy and Forensic Medicine, South Ural State Medical University, Chelyabinsk 454092, Russia
| |
Collapse
|
26
|
Hu Y, Hu D, Yu H, Xu W, Fu R. Hypoxia‑inducible factor 1α and ROCK1 regulate proliferation and collagen synthesis in hepatic stellate cells under hypoxia. Mol Med Rep 2018; 18:3997-4003. [PMID: 30132575 DOI: 10.3892/mmr.2018.9397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 02/27/2018] [Indexed: 11/06/2022] Open
Abstract
Hypoxia serves a critical role in the pathogenesis of liver fibrosis. Hypoxia‑inducible factor 1α (HIF1‑α) is induced when cells are exposed to low O2 concentrations. Recently, it has been suggested that Rho‑associated coiled‑coil‑forming kinase 1 (ROCK1) may be an important HIF1‑α regulator. In the present study, it was analyzed whether crosstalk between HIF1‑α and ROCK1 regulates cell proliferation and collagen synthesis in hepatic stellate cells (HSCs) under hypoxic conditions. For this purpose, a rat hepatic HSC line (HSC‑T6) was cultured under hypoxic or normoxic conditions, and HIF1‑α and ROCK1 expression was measured at different time points. Additionally, HSC‑T6 cells were transfected with HIF1‑α small interfering RNA (siHIF1‑α), and measured protein expression and mRNA transcript levels of α‑smooth muscle actin, collagen 1A1 and ROCK1. Collagen 3A1 secretion was also measured by ELISA. Cell proliferation was assessed by the MTT assay under these hypoxic conditions. The results indicated that a specific ROCK inhibitor, Y‑27632, increased HIF1‑α and ROCK1 expression over time in HSC‑T6 cells in response to hypoxia. In addition, knockdown of HIF1‑α inhibited HSC‑T6 proliferation, suppressed collagen 1A1 expression, decreased collagen 3A1 secretion and attenuated ROCK1 expression. Notably, ROCK1 inhibition caused HSC‑T6 quiescence, suppressed collagen secretion and downregulated HIF1‑α expression. Collectively, these findings indicated that the interplay between HIF1‑α and ROCK1 may be a critical factor that regulates cell proliferation and collagen synthesis in rat HSCs under hypoxia.
Collapse
Affiliation(s)
- Yibing Hu
- Department of Infectious Diseases, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325200, P.R. China
| | - Danping Hu
- Department of Infectious Diseases, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325200, P.R. China
| | - Huanhuan Yu
- Department of Infectious Diseases, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325200, P.R. China
| | - Wangwang Xu
- Department of Infectious Diseases, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325200, P.R. China
| | - Rongquan Fu
- Department of Infectious Diseases, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325200, P.R. China
| |
Collapse
|
27
|
p300/CBP as a Key Nutritional Sensor for Hepatic Energy Homeostasis and Liver Fibrosis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8168791. [PMID: 29862292 PMCID: PMC5976926 DOI: 10.1155/2018/8168791] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/14/2018] [Accepted: 04/12/2018] [Indexed: 12/23/2022]
Abstract
The overwhelming frequency of metabolic diseases such as obesity and diabetes are closely related to liver diseases, which might share common pathogenic signaling processes. These metabolic disorders in the presence of inflammatory response seem to be triggered by and to reside in the liver, which is the central metabolic organ that plays primary roles in regulating lipid and glucose homeostasis upon alterations of metabolic conditions. Recently, abundant emerging researches suggested that p300 and CREB binding protein (CBP) are crucial regulators of energy homeostasis and liver fibrosis through both their acetyltransferase activities and transcriptional coactivators. Plenty of recent findings demonstrated the potential roles of p300/CBP in mammalian metabolic homeostasis in response to nutrients. This review is focused on the different targets and functions of p300/CBP in physiological and pathological processes, including lipogenesis, lipid export, gluconeogenesis, and liver fibrosis, also provided some nutrients as the regulator of p300/CBP for nutritional therapeutic approaches to treat liver diseases.
Collapse
|
28
|
Mazza G, Al-Akkad W, Rombouts K. Engineering in vitro models of hepatofibrogenesis. Adv Drug Deliv Rev 2017; 121:147-157. [PMID: 28578016 DOI: 10.1016/j.addr.2017.05.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/17/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023]
Abstract
Chronic liver disease is a major cause of morbidity and mortality worldwide marked by chronic inflammation and fibrosis/scarring, resulting in end-stage liver disease and its complications. Hepatic stellate cells (HSCs) are a dominant contributor to liver fibrosis by producing excessive extracellular matrix (ECM), irrespective of the underlying disease aetiologies, and for many decades research has focused on the development of a number of anti-fibrotic strategies targeting this cell. Despite major improvements in two-dimensional systems (2D) by using a variety of cell culture models of different complexity, an efficient anti-fibrogenic therapy has yet to be developed. The development of well-defined three-dimensional (3D) in vitro models, which mimic ECM structures as found in vivo, have demonstrated the importance of cell-matrix bio-mechanics, the complex interactions between HSCs and hepatocytes and other non-parenchymal cells, and this to improve and promote liver cell-specific functions. Henceforth, refinement of these 3D in vitro models, which reproduce the liver microenvironment, will lead to new objectives and to a possible new era in the search for antifibrogenic compounds.
Collapse
|
29
|
Cannito S, Novo E, Parola M. Therapeutic pro-fibrogenic signaling pathways in fibroblasts. Adv Drug Deliv Rev 2017; 121:57-84. [PMID: 28578015 DOI: 10.1016/j.addr.2017.05.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/28/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023]
Abstract
Myofibroblasts (MFs) play a critical role in the progression of chronic inflammatory and fibroproliferative diseases in different tissues/organs, whatever the etiology. Fibrosis is preceded and sustained by persistent injury and inflammatory response in a profibrogenic scenario involving mutual interactions, operated by several mediators and pathways, of MFs and related precursor cells with innate immunity cells and virtually any cell type in a defined tissue. These interactions, mediators and related signaling pathways are critical in initiating and perpetuating the differentiation of precursor cells into MFs that in different tissues share peculiar traits and phenotypic responses, including the ability to proliferate, produce ECM components, migrate and contribute to the modulation of inflammatory response and tissue angiogenesis. Literature studies related to liver, lung and kidney fibrosis have outlined a number of MF-related core regulatory fibrogenic signaling pathways conserved across these different organs and potentially targetable in order to develop effective antifibrotic therapeutic strategies.
Collapse
|
30
|
Kawaratani H, Moriya K, Namisaki T, Uejima M, Kitade M, Takeda K, Okura Y, Kaji K, Takaya H, Nishimura N, Sato S, Sawada Y, Seki K, Kubo T, Mitoro A, Yamao J, Yoshiji H. Therapeutic strategies for alcoholic liver disease: Focusing on inflammation and fibrosis (Review). Int J Mol Med 2017; 40:263-270. [PMID: 28627645 DOI: 10.3892/ijmm.2017.3015] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/30/2017] [Indexed: 11/06/2022] Open
Abstract
Excessive alcohol consumption is the most common cause of liver disease in the world. Chronic alcohol abuse leads to liver damage, liver inflammation, fibrosis and hepatocellular carcinoma. Inflammatory cytokines, such as tumor necrosis factor-α and interferon-γ, induce liver injury, which leads to the develo-pment of alcoholic liver disease (ALD). Hepatoprotective cytokines, such as interleukin (IL)-6 and IL-10, are also associated with ALD. IL-6 improves ALD via the activation of STAT3 and the subsequent induction of a variety of hepatoprotective genes in hepatocytes. Alcohol consumption promotes liver inflammation by incre-asing the translocation of gut-derived endotoxins to the portal circulation and by activating Kupffer cells through the lipopolysaccharide/Toll-like receptor 4 pathways. Oxidative stress and microflora products are also associated with ALD. Hepatic stellate cells play an important role in angiogenesis and liver fibrosis. Anti-angiogenic therapy has been found to be effective in the prevention of fibrosis. This suggests that blocking angiogenesis could be a promising therapeutic option for patients with advanced fibrosis. This review discusses the main pathways associated with liver inflammation and liver fibrosis as well as new therapeutic strategies.
Collapse
Affiliation(s)
- Hideto Kawaratani
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Kei Moriya
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Tadashi Namisaki
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Masakazu Uejima
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Mitsuteru Kitade
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Kousuke Takeda
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Yasushi Okura
- Department of Endoscopy and Ultrasound, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Kousuke Kaji
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Hiroaki Takaya
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Norihisa Nishimura
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Shinya Sato
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Yasuhiko Sawada
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Kenichiro Seki
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Takuya Kubo
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Akira Mitoro
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Junichi Yamao
- Department of Endoscopy and Ultrasound, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Hitoshi Yoshiji
- The Third Department of Internal Medicine, Nara Medical University, Kashihara, Nara 634-8522, Japan
| |
Collapse
|
31
|
Novo E, Villano G, Turato C, Cannito S, Paternostro C, Busletta C, Biasiolo A, Quarta S, Morello E, Bocca C, Miglietta A, David E, Sutti S, Plebani M, Albano E, Parola M, Pontisso P. SerpinB3 Promotes Pro-fibrogenic Responses in Activated Hepatic Stellate Cells. Sci Rep 2017; 7:3420. [PMID: 28611447 PMCID: PMC5469760 DOI: 10.1038/s41598-017-03744-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 05/05/2017] [Indexed: 01/15/2023] Open
Abstract
SerpinB3 is a hypoxia- and hypoxia-inducible factor-2α-dependent cystein protease inhibitor that is up-regulated in hepatocellular carcinoma and in parenchymal cells during chronic liver diseases (CLD). SerpinB3 up-regulation in CLD patients has been reported to correlate with the extent of liver fibrosis and the production of transforming growth factor-β1, but the actual role of SerpinB3 in hepatic fibrogenesis is still poorly characterized. In the present study we analyzed the pro-fibrogenic action of SerpinB3 in cell cultures and in two different murine models of liver fibrosis. "In vitro" experiments revealed that SerpinB3 addition to either primary cultures of human activated myofibroblast-like hepatic stellate cells (HSC/MFs) or human stellate cell line (LX2 cells) strongly up-regulated the expression of genes involved in fibrogenesis and promoted oriented migration, but not cell proliferation. Chronic liver injury by CCl4 administration or by feeding a methionine/choline deficient diet to transgenic mice over-expressing human SerpinB3 in hepatocytes confirmed that SerpinB3 over-expression significantly increased the mRNA levels of pro-fibrogenic genes, collagen deposition and αSMA-positive HSC/MFs as compared to wild-type mice, without affecting parenchymal damage. The present study provides for the first time evidence that hepatocyte release of SerpinB3 during CLD can contribute to liver fibrogenesis by acting on HSC/MFs.
Collapse
Affiliation(s)
- Erica Novo
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | | | | | - Stefania Cannito
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Claudia Paternostro
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Chiara Busletta
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | | | - Santina Quarta
- Department of Medicine, University of Padova, Padova, Italy
| | - Elisabetta Morello
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Claudia Bocca
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Antonella Miglietta
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy
| | - Ezio David
- Pathology Unit, S. Giovanni Battista Hospital, Torino, Italy
| | - Salvatore Sutti
- Department of Health Sciences, University "A. Avogadro" of East Piedmont, Novara, Italy
| | - Mario Plebani
- Department of Medicine, University of Padova, Padova, Italy
| | - Emanuele Albano
- Department of Health Sciences, University "A. Avogadro" of East Piedmont, Novara, Italy
| | - Maurizio Parola
- Department Clinical and Biological Sciences, Unit of Experimental Medicine and Clinical Pathology, University of Torino, Torino, Italy.
| | | |
Collapse
|
32
|
Cannito S, Morello E, Bocca C, Foglia B, Benetti E, Novo E, Chiazza F, Rogazzo M, Fantozzi R, Povero D, Sutti S, Bugianesi E, Feldstein AE, Albano E, Collino M, Parola M. Microvesicles released from fat-laden cells promote activation of hepatocellular NLRP3 inflammasome: A pro-inflammatory link between lipotoxicity and non-alcoholic steatohepatitis. PLoS One 2017; 12:e0172575. [PMID: 28249038 PMCID: PMC5331985 DOI: 10.1371/journal.pone.0172575] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 02/07/2017] [Indexed: 02/06/2023] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is a major form of chronic liver disease in the general population in relation to its high prevalence among overweight/obese individuals and patients with diabetes type II or metabolic syndrome. NAFLD can progress to steatohepatitis (NASH), fibrosis and cirrhosis and end-stage of liver disease but mechanisms involved are still incompletely characterized. Within the mechanisms proposed to mediate the progression of NAFLD, lipotoxicity is believed to play a major role. In the present study we provide data suggesting that microvesicles (MVs) released by fat-laden cells undergoing lipotoxicity can activate NLRP3 inflammasome following internalization by either cells of hepatocellular origin or macrophages. Inflammasome activation involves NF-kB-mediated up-regulation of NLRP3, pro-caspase-1 and pro-Interleukin-1, then inflammasome complex formation and Caspase-1 activation leading finally to an increased release of IL-1β. Since the release of MVs from lipotoxic cells and the activation of NLRP3 inflammasome have been reported to occur in vivo in either clinical or experimental NASH, these data suggest a novel rational link between lipotoxicity and increased inflammatory response.
Collapse
Affiliation(s)
- Stefania Cannito
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Elisabetta Morello
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Claudia Bocca
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Beatrice Foglia
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Elisa Benetti
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Erica Novo
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Fausto Chiazza
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Mara Rogazzo
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Roberto Fantozzi
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Davide Povero
- Department of Pediatrics, University of California San Diego (UCSD), La Jolla, CA, United States of America
| | - Salvatore Sutti
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune Diseases, University “Amedeo Avogadro” of East Piedmont, Novara, Italy
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology, University of Torino, Torino, Italy
| | - Ariel E. Feldstein
- Department of Pediatrics, University of California San Diego (UCSD), La Jolla, CA, United States of America
| | - Emanuele Albano
- Department of Health Sciences and Interdisciplinary Research Center for Autoimmune Diseases, University “Amedeo Avogadro” of East Piedmont, Novara, Italy
| | - Massimo Collino
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Maurizio Parola
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| |
Collapse
|
33
|
Zhang F, Hao M, Jin H, Yao Z, Lian N, Wu L, Shao J, Chen A, Zheng S. Canonical hedgehog signalling regulates hepatic stellate cell-mediated angiogenesis in liver fibrosis. Br J Pharmacol 2017; 174:409-423. [PMID: 28052321 DOI: 10.1111/bph.13701] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 12/18/2016] [Accepted: 12/23/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Hepatic stellate cells (HSCs) are liver-specific pericytes regulating angiogenesis during liver fibrosis. We aimed to elucidate the mechanisms by which hedgehog signalling regulated HSC angiogenic properties and to validate the therapeutic implications. EXPERIMENTAL APPROACH Rats and mice were treated with carbon tetrachloride for in vivo evaluation of hepatic angiogenesis and fibrotic injury. Diversified molecular approaches including real-time PCR, Western blot, luciferase reporter assay, chromatin immunoprecipitation, electrophoretic mobility shift assay and co-immunoprecipitation were used to investigate the underlying mechanisms in vitro. KEY RESULTS Angiogenesis was concomitant with up-regulation of Smoothened (SMO) and hypoxia inducible factor-1α (HIF-1α) in rat fibrotic liver. The SMO inhibitor cyclopamine and Gli1 inhibitor GANT-58 reduced expression of VEGF and angiopoietin 1 in HSCs and suppressed HSC tubulogenesis capacity. HIF-1α inhibitor PX-478 suppressed HSC angiogenic behaviour, and inhibition of hedgehog decreased HIF-1α expression. Furthermore, heat shock protein 90 (HSP90) was characterized as a direct target gene of canonical hedgehog signalling in HSCs. HSP90 inhibitor 17-AAG reduced HSP90 binding to HIF-1α, down-regulated HIF-1α protein abundance and decreased HIF-1α binding to DNA. 17-AAG also abolished 1-stearoyl-2-arachidonoyl-sn-glycerol (SAG) (a SMO agonist)-enhanced HSC angiogenic properties. Finally, the natural compound ligustrazine was found to inhibit canonical hedgehog signalling leading to suppressed angiogenic properties of HSCs in vitro and ameliorated liver fibrosis and sinusoidal angiogenesis in mice. CONCLUSION AND IMPLICATIONS We have provided evidence that the canonical hedgehog pathway controlled HSC-mediated liver angiogenesis. Selective inhibition of HSC hedgehog signalling could be a promising therapeutic approach for hepatic fibrosis.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meng Hao
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huanhuan Jin
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhen Yao
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Naqi Lian
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Wu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiangjuan Shao
- Department of Pharmacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, Saint Louis, MO, USA
| | - Shizhong Zheng
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
34
|
Abstract
In patients with advanced liver disease with portal hypertension, portal-systemic collaterals contribute to circulatory disturbance, gastrointestinal hemorrhage, hepatic encephalopathy, ascites, hepatopulmonary syndrome and portopulmonary hypertension. Angiogenesis has a pivotal role in the formation of portal-systemic shunts. Recent research has defined many of the mediators and mechanisms involved in this angiogenic process, linking the central roles of hepatic stellate cells and endothelial cells. Studies of animal models have demonstrated the potential therapeutic impact of drugs to inhibit angiogenesis in cirrhosis. For example, inhibition of VEGF reduces portal pressure, hyperdynamic splanchnic circulation, portosystemic collateralization and liver fibrosis. An improved understanding of the role of other angiogenic factors provides hope for a novel targeted therapy for portal hypertension with a tolerable adverse effect profile.
Collapse
Affiliation(s)
- Juan Cristóbal Gana
- Department of Pediatric Gastroenterology & Nutrition, Division of Pediatrics, Escuela de Medicina, Pontificia Universidad Católica de Chile. Chile
| | - Carolina A Serrano
- Department of Pediatric Gastroenterology & Nutrition, Division of Pediatrics, Escuela de Medicina, Pontificia Universidad Católica de Chile. Chile
| | - Simon C Ling
- Division of Gastroenterology, Hepatology & Nutrition, Department of Paediatrics, University of Toronto, and The Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
35
|
Longato L, Andreola F, Davies SS, Roberts JL, Fusai G, Pinzani M, Moore K, Rombouts K. Reactive gamma-ketoaldehydes as novel activators of hepatic stellate cells in vitro. Free Radic Biol Med 2017; 102:162-173. [PMID: 27890721 DOI: 10.1016/j.freeradbiomed.2016.11.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 11/28/2022]
Abstract
AIMS Products of lipid oxidation, such as 4-hydroxynonenal (4-HNE), are key activators of hepatic stellate cells (HSC) to a pro-fibrogenic phenotype. Isolevuglandins (IsoLG) are a family of acyclic γ-ketoaldehydes formed through oxidation of arachidonic acid or as by-products of the cyclooxygenase pathway. IsoLGs are highly reactive aldehydes which are efficient at forming protein adducts and cross-links at concentrations 100-fold lower than 4-hydroxynonenal. Since the contribution of IsoLGs to liver injury has not been studied, we synthesized 15-E2-IsoLG and used it to investigate whether IsoLG could induce activation of HSC. RESULTS Primary human HSC were exposed to 15-E2-IsoLG for up to 48h. Exposure to 5μM 15-E2-IsoLG in HSCs promoted cytotoxicity and apoptosis. At non-cytotoxic doses (50 pM-500nM) 15-E2-IsoLG promoted HSC activation, indicated by increased expression of α-SMA, sustained activation of ERK and JNK signaling pathways, and increased mRNA and/or protein expression of cytokines and chemokines, which was blocked by inhibitors of JNK and NF-kB. In addition, IsoLG promoted formation of reactive oxygen species, and induced an early activation of ER stress, followed by autophagy. Inhibition of autophagy partially reduced the pro-inflammatory effects of IsoLG, suggesting that it might serve as a cytoprotective response. INNOVATION This study is the first to describe the biological effects of IsoLG in primary HSC, the main drivers of hepatic fibrosis. CONCLUSIONS IsoLGs represent a newly identified class of activators of HSC in vitro, which are biologically active at concentrations as low as 500 pM, and are particularly effective at promoting a pro-inflammatory response and autophagy.
Collapse
Affiliation(s)
- Lisa Longato
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK
| | - Fausto Andreola
- Liver Failure Group, Institute for Liver & Digestive Health, University College of London, Royal Free, London, UK
| | - Sean S Davies
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Jackson L Roberts
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Giuseppe Fusai
- Division of Surgery, University College London, Royal Free, London, UK
| | - Massimo Pinzani
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK
| | - Kevin Moore
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK
| | - Krista Rombouts
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK.
| |
Collapse
|
36
|
Sansoè G, Aragno M, Mastrocola R, Mengozzi G, Novo E, Parola M. Role of Chymase in the Development of Liver Cirrhosis and Its Complications: Experimental and Human Data. PLoS One 2016; 11:e0162644. [PMID: 27637026 PMCID: PMC5026361 DOI: 10.1371/journal.pone.0162644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 08/23/2016] [Indexed: 12/15/2022] Open
Abstract
Background Tissue Angiotensin II (Ang-II), produced through local non ACE-dependent pathways, stimulates liver fibrogenesis, renal vasoconstriction and sodium retention. Aim To highlight chymase-dependent pathway of Ang-II production in liver and kidney during cirrhosis development. Methods Liver histology, portal pressure, liver and kidney function, and hormonal status were investigated in rat liver cirrhosis induced through 13 weeks of CCl4, with or without chymase inhibitor SF2809E, administered between 4th and 13th CCl4 weeks; liver and kidney chymase immunolocation and Ang-II content were assessed. Chymase immunohistochemistry was also assessed in normal and cirrhotic human liver, and chymase mRNA transcripts were measured in human HepG2 cells and activated hepatic stellate cells (HSC/MFs) in vitro. Results Rats receiving both CCl4 and SF2809E showed liver fibrotic septa focally linking portal tracts but no cirrhosis, as compared to ascitic cirrhotic rats receiving CCl4. SF2809E reduced portal pressure, plasma bilirubin, tissue content of Ang-II, plasma renin activity, norepinephrine and vasopressin, and increased glomerular filtration rate, water clearance, urinary sodium excretion. Chymase tissue content was increased and detected in α-SMA-positive liver myofibroblasts and in kidney tubular cells of cirrhotic rats. In human cirrhosis, chymase was located in hepatocytes of regenerative nodules. Human HepG2 cells and HSC/MFs responded to TGF-β1 by up-regulating chymase mRNA transcription. Conclusions Chymase, through synthesis of Ang-II and other mediators, plays a role in the derangement of liver and kidney function in chronic liver diseases. In human cirrhosis, chymase is well-represented and apt to become a future target of pharmacological treatment.
Collapse
Affiliation(s)
- Giovanni Sansoè
- Division of Gastroenterology, Humanitas Gradenigo Hospital, Torino, Italy
- * E-mail:
| | - Manuela Aragno
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Raffaella Mastrocola
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Giulio Mengozzi
- Clinical Biochemistry Laboratory, San Giovanni Battista Hospital, Torino, Italy
| | - Erica Novo
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Maurizio Parola
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| |
Collapse
|
37
|
Darby IA, Hewitson TD. Hypoxia in tissue repair and fibrosis. Cell Tissue Res 2016; 365:553-62. [PMID: 27423661 DOI: 10.1007/s00441-016-2461-3] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 06/23/2016] [Indexed: 12/23/2022]
|
38
|
Garbuzenko DV, Arefyev NO, Belov DV. Mechanisms of adaptation of the hepatic vasculature to the deteriorating conditions of blood circulation in liver cirrhosis. World J Hepatol 2016; 8:665-672. [PMID: 27326313 PMCID: PMC4909428 DOI: 10.4254/wjh.v8.i16.665] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 04/25/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023] Open
Abstract
PubMed, EMBASE, Orphanet, MIDLINE, Google Scholar and Cochrane Library were searched for articles published between 1983 and 2015. Relevant articles were selected by using the following terms: "Liver cirrhosis", "Endothelial dysfunction", "Sinusoidal remodeling", "Intrahepatic angiogenesis" and "Pathogenesis of portal hypertension". Then the reference lists of identified articles were searched for other relevant publications as well. Besides gross hepatic structural disorders related to diffuse fibrosis and formation of regenerative nodules, the complex morphofunctional rearrangement of the hepatic microvascular bed and intrahepatic angiogenesis also play important roles in hemodynamic disturbances in liver cirrhosis. It is characterized by endothelial dysfunction and impaired paracrine interaction between activated stellate hepatocytes and sinusoidal endotheliocytes, sinusoidal remodeling and capillarization, as well as development of the collateral microcirculation. In spite of the fact that complex morphofunctional rearrangement of the hepatic microvascular bed and intrahepatic angiogenesis in liver cirrhosis are the compensatory-adaptive reaction to the deteriorating conditions of blood circulation, they contribute to progression of disease and development of serious complications, in particular, related to portal hypertension.
Collapse
Affiliation(s)
- Dmitry Victorovich Garbuzenko
- Dmitry Victorovich Garbuzenko, Nikolay Olegovich Arefyev, Department of Faculty Surgery, South Ural State Medical University, 454092 Chelyabinsk, Russia
| | - Nikolay Olegovich Arefyev
- Dmitry Victorovich Garbuzenko, Nikolay Olegovich Arefyev, Department of Faculty Surgery, South Ural State Medical University, 454092 Chelyabinsk, Russia
| | - Dmitry Vladimirovich Belov
- Dmitry Victorovich Garbuzenko, Nikolay Olegovich Arefyev, Department of Faculty Surgery, South Ural State Medical University, 454092 Chelyabinsk, Russia
| |
Collapse
|
39
|
Ju C, Colgan SP, Eltzschig HK. Hypoxia-inducible factors as molecular targets for liver diseases. J Mol Med (Berl) 2016; 94:613-27. [PMID: 27094811 PMCID: PMC4879168 DOI: 10.1007/s00109-016-1408-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/04/2016] [Accepted: 03/08/2016] [Indexed: 12/11/2022]
Abstract
Liver disease is a growing global health problem, as deaths from end-stage liver cirrhosis and cancer are rising across the world. At present, pharmacologic approaches to effectively treat or prevent liver disease are extremely limited. Hypoxia-inducible factor (HIF) is a transcription factor that regulates diverse signaling pathways enabling adaptive cellular responses to perturbations of the tissue microenvironment. HIF activation through hypoxia-dependent and hypoxia-independent signals have been reported in liver disease of diverse etiologies, from ischemia-reperfusion-induced acute liver injury to chronic liver diseases caused by viral infection, excessive alcohol consumption, or metabolic disorders. This review summarizes the evidence for HIF stabilization in liver disease, discusses the mechanistic involvement of HIFs in disease development, and explores the potential of pharmacological HIF modifiers in the treatment of liver disease.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Bevacizumab/therapeutic use
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Clinical Trials as Topic
- Fatty Liver/genetics
- Fatty Liver/metabolism
- Fatty Liver/pathology
- Fatty Liver/therapy
- Gene Expression Regulation
- Hepatitis, Viral, Human/genetics
- Hepatitis, Viral, Human/metabolism
- Hepatitis, Viral, Human/pathology
- Hepatitis, Viral, Human/therapy
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Liver Cirrhosis/genetics
- Liver Cirrhosis/metabolism
- Liver Cirrhosis/pathology
- Liver Cirrhosis/therapy
- Liver Diseases, Alcoholic/genetics
- Liver Diseases, Alcoholic/metabolism
- Liver Diseases, Alcoholic/pathology
- Liver Diseases, Alcoholic/therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Molecular Targeted Therapy
- Oligonucleotides/therapeutic use
- Signal Transduction
Collapse
Affiliation(s)
- Cynthia Ju
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado, Auroa, Colorado, 800045, USA.
| | - Sean P Colgan
- Department of Medicine and Mucosal Inflammation Program, School of Medicine, University of Colorado, Auroa, Colorado, 800045, USA
| | - Holger K Eltzschig
- Department of Anesthesiology and Organ Protection Program, School of Medicine, University of Colorado, Auroa, Colorado, 800045, USA
| |
Collapse
|
40
|
Zhu L, Chen X, Kong X, Cai YD. Investigation of the roles of trace elements during hepatitis C virus infection using protein-protein interactions and a shortest path algorithm. Biochim Biophys Acta Gen Subj 2016; 1860:2756-68. [PMID: 27208424 DOI: 10.1016/j.bbagen.2016.05.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/05/2016] [Accepted: 05/13/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hepatitis is a type of infectious disease that induces inflammation of the liver without pinpointing a particular pathogen or pathogenesis. Type C hepatitis, as a type of hepatitis, has been reported to induce cirrhosis and hepatocellular carcinoma within a very short amount of time. It is a great threat to human health. Some studies have revealed that trace elements are associated with infection with and immune rejection against hepatitis C virus (HCV). However, the mechanism underlying this phenomenon is still unclear. METHODS In this study, we aimed to expand our knowledge of this phenomenon by designing a computational method to identify genes that may be related to both HCV and trace element metabolic processes. The searching procedure included three stages. First, a shortest path algorithm was applied to a large network, constructed by protein-protein interactions, to identify potential genes of interest. Second, a permutation test was executed to exclude false discoveries. Finally, some rules based on the betweenness and associations between candidate genes and HCV and trace elements were built to select core genes among the remaining genes. RESULTS 12 lists of genes, corresponding to 12 types of trace elements, were obtained. These genes are deemed to be associated with HCV infection and trace elements metabolism. CONCLUSIONS The analyses indicate that some genes may be related to both HCV and trace element metabolic processes, further confirming the associations between HCV and trace elements. The method was further tested on another set of HCV genes, the results indicate that this method is quite robustness. GENERAL SIGNIFICANCE The newly found genes may partially reveal unknown mechanisms between HCV infection and trace element metabolism. This article is part of a Special Issue entitled "System Genetics" Guest Editor: Dr. Yudong Cai and Dr. Tao Huang.
Collapse
Affiliation(s)
- LiuCun Zhu
- School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - XiJia Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Xiangyin Kong
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, People's Republic of China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China.
| |
Collapse
|
41
|
Wang D, Wang Q, Yin J, Dong R, Wang Q, Du X, Lu J. Combined administration of propranolol + AG490 offers better effects on portal hypertensive rats with cirrhosis. J Gastroenterol Hepatol 2016; 31:1037-44. [PMID: 26487394 DOI: 10.1111/jgh.13207] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 10/07/2015] [Accepted: 10/07/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS AG490, the specific inhibitor of JAK2/STAT3 signaling, has been shown to decrease portal pressure, splanchnic hyperdynamic circulation and liver fibrosis in cirrhotic rats. Nonselective betablockers such as propranolol are the only drugs recommended in the treatment of portal hypertension. The aim of this study was to explore the combinative effect of treatment with propranolol and AG490 on portal hypertension. METHODS Rats induced by common bile duct ligation were treated with vehicle, AG490, propranolol, or AG490 + propranolol for 2 weeks. Hemodynamics parameters were assessed. Expressions of phospho-STAT3 protein and its down-regulated cytokines in splanchnic organs were detected by ELISA or western blot. Lipopolysaccharide binding protein (LBP) and IL-6 were assessed by ELISA or western blot. Characterization of liver and mesentery was performed by histological analyses. RESULTS Highly expressed phospho-STAT3 protein in cirrhotic rats could successfully be inhibited by AG490 or AG490 + propranolol treatments but not by propranolol alone. Both AG490 and propranolol significantly reduced portal pressure and hyperdynamic splanchnic circulation, and combination of AG490 and propranolol achieved an additive effect than with either drug alone. AG490, alone or in combination with propranolol, inhibited liver fibrosis, splenomegaly and splanchnic angiogenesis. Increased markers of bacterial translocation (LBP and IL6) were greatly reduced by propranolol but not by AG490. CONCLUSIONS The combination of propranolol and AG490 caused a greater improvement of portal hypertension and might therefore offer a potentially promising therapy in the portal hypertension treatment.
Collapse
Affiliation(s)
- Dong Wang
- Departments of General Surgery, TangDu Hospital, Xi'an, China
| | - Qin Wang
- Pharmacogenomics, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Jikai Yin
- Departments of General Surgery, TangDu Hospital, Xi'an, China
| | - Rui Dong
- Departments of General Surgery, TangDu Hospital, Xi'an, China
| | - Qing Wang
- Departments of General Surgery, TangDu Hospital, Xi'an, China
| | - Xilin Du
- Departments of General Surgery, TangDu Hospital, Xi'an, China
| | - Jianguo Lu
- Departments of General Surgery, TangDu Hospital, Xi'an, China
| |
Collapse
|
42
|
Cannito S, Turato C, Paternostro C, Biasiolo A, Colombatto S, Cambieri I, Quarta S, Novo E, Morello E, Villano G, Fasolato S, Musso T, David E, Tusa I, Rovida E, Autelli R, Smedile A, Cillo U, Pontisso P, Parola M. Hypoxia up-regulates SERPINB3 through HIF-2α in human liver cancer cells. Oncotarget 2016; 6:2206-21. [PMID: 25544768 PMCID: PMC4385846 DOI: 10.18632/oncotarget.2943] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 12/09/2014] [Indexed: 02/04/2023] Open
Abstract
SERPINB3 is a cysteine-proteases inhibitor up-regulated in a significant number of cirrhotic patients carrying hepatocellular carcinoma (HCC) and recently proposed as a prognostic marker for HCC early recurrence. SERPINB3 has been reported to stimulate proliferation, inhibit apoptosis and, similar to what reported for hypoxia, to trigger epithelial-to-mesenchymal transition (EMT) and increased invasiveness in liver cancer cells. This study has investigated whether SERPINB3 expression is regulated by hypoxia-related mechanisms in liver cancer cells. Exposure of HepG2 and Huh7 cells to hypoxia up-regulated SERPINB3 transcription, protein synthesis and release in the extracellular medium. Hypoxia-dependent SERPINB3 up-regulation was selective (no change detected for SERPINB4) and operated through hypoxia inducible factor (HIF)-2α (not HIF-1α) binding to SERPINB3 promoter, as confirmed by chromatin immuno-precipitation assay and silencing experiments employing specific siRNAs. HIF-2α-mediated SERPINB3 up-regulation under hypoxic conditions required intracellular generation of ROS. Immuno-histochemistry (IHC) and transcript analysis, performed in human HCC specimens, revealed co-localization of the two proteins in liver cancer cells and the existence of a positive correlation between HIF-2α and SERPINB3 transcript levels, respectively. Hypoxia, through HIF-2α-dependent and redox-sensitive mechanisms, up-regulates the transcription, synthesis and release of SERPINB3, a molecule with a high oncogenic potential.
Collapse
Affiliation(s)
- Stefania Cannito
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Interuniversity Center for Liver Pathophysiology, University of Torino, Italy
| | | | - Claudia Paternostro
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Interuniversity Center for Liver Pathophysiology, University of Torino, Italy
| | | | | | - Irene Cambieri
- Department of Plastic Surgery and Burn Unit Skin Bank, CTO Hospital, Torino, Italy
| | | | - Erica Novo
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Interuniversity Center for Liver Pathophysiology, University of Torino, Italy
| | - Elisabetta Morello
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Interuniversity Center for Liver Pathophysiology, University of Torino, Italy
| | | | | | - Tiziana Musso
- Department of Public Health and Pediatric Sciences, University of Torino, Italy
| | - Ezio David
- Pathology Unit, San Giovanni Battista Hospital, Torino, Italy
| | - Ignazia Tusa
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, Italy
| | - Elisabetta Rovida
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, Italy
| | - Riccardo Autelli
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Interuniversity Center for Liver Pathophysiology, University of Torino, Italy
| | - Antonina Smedile
- Gastroenterology and Hepatology Division, San Giovanni Battista Hospital, Torino, Italy
| | - Umberto Cillo
- Unit of Hepatobiliary Surgery and Liver Transplantation, University of Padova, Italy
| | | | - Maurizio Parola
- Department of Clinical and Biological Sciences, Unit of Experimental Medicine and Interuniversity Center for Liver Pathophysiology, University of Torino, Italy
| |
Collapse
|
43
|
Li J, Zhang YL, Chen R, Wang Y, Xiong KP, Huang JY, Han F, Liu CF. Elevated Serum Liver Enzymes in Patients with Obstructive Sleep Apnea-hypopnea Syndrome. Chin Med J (Engl) 2015; 128:2983-7. [PMID: 26608975 PMCID: PMC4795259 DOI: 10.4103/0366-6999.168943] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Obstructive sleep apnea-hypopnea syndrome (OSAS) is associated with elevated liver enzymes and fatty liver. The purpose of this study was to measure serum liver enzyme levels in patients evaluated by polysomnography (PSG) and the factors associated with liver injury in OSAS patients. METHODS All patients referred to PSG for evaluation of sleep apnea symptoms between June 2011 and November 2014 were included in this study. Demographic data and PSG parameters were recorded. Serum alanine aminotransferase, aspartate aminotransferase, and gamma-glutamyl transferase levels were systematically measured. OSAS patients were divided into mild, moderate, and severe groups according to the apnea-hypopnea index (AHI) values of 5-14 events/h, 15-29 events/h, and ≥30 events/h. RESULTS A total of 540 patients were enrolled in this study; among these patients, 386 were male. Elevated liver enzymes were present in 42.3% of OSAS patients (32.4% in mild/moderate group; 51.0% in severe group) and 28.1% patients without OSAS. Patients with OSAS had higher body mass index (BMI) (P < 0.01). In the bivariate correlation, the liver enzymes level was negatively correlated with age and the lowest arterial oxygen saturation (SaO 2 ), and was positively correlated with BMI, oxygen desaturation index, percent of total time with oxygen saturation level <90% (TS90%), AHI, total cholesterol (TC), and triglyceride (TG). In logistic regression analysis, Age, BMI, TS90%, TC, and TG were included in the regression equation. CONCLUSIONS Our data suggest that OSAS is a risk factor for elevated liver enzymes. The severity of OSAS is correlated with liver enzyme levels; we hypothesize that hypoxia is one of main causes of liver damage in patients with OSAS.
Collapse
Affiliation(s)
- Jie Li
- Department of Neurology and Sleep Center, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Yan-Lin Zhang
- Department of Neurology and Sleep Center, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Rui Chen
- Department of Neurology and Sleep Center, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Yi Wang
- Department of Neurology and Sleep Center, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Kang-Ping Xiong
- Department of Neurology and Sleep Center, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Jun-Ying Huang
- Department of Neurology and Sleep Center, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Fei Han
- Department of Neurology and Sleep Center, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Chun-Feng Liu
- Department of Neurology and Sleep Center, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu 215123, China
- Beijing Key Laboratory for Parkinson's Disease, Beijing 100053, China
| |
Collapse
|
44
|
Kawada N. Cytoglobin as a Marker of Hepatic Stellate Cell-derived Myofibroblasts. Front Physiol 2015; 6:329. [PMID: 26617531 PMCID: PMC4643130 DOI: 10.3389/fphys.2015.00329] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/30/2015] [Indexed: 12/13/2022] Open
Abstract
Myofibroblasts play important roles in inflammation, fibrosis and tumorigenesis in chronically inflamed liver. Liver myofibroblasts originate from hepatic stellate cells, portal fibroblasts or mesothelial cells, and they are localized in and around fibrotic septum and portal tracts. Liver myofibroblasts are the source of extracellular matrix materials, including type I collagen and multiple fibrogenic growth factors, such as transforming growth factor-β and vascular endothelial growth factor. Although a detailed characterization of the function of individual myofibroblasts has not been conducted, owing to the lack of appropriate cell markers, recent lineage-tracing technology has revealed the limited contribution of myofibroblasts that are derived from portal fibroblasts to various types of liver fibrosis, as compared with the contribution of hepatic stellate cells. In addition, cytoglobin, which is the fourth globin in mammals and function as a local gas sensor, provides a new perspective on the involvement of stellate cells in fibrosis and carcinogenesis, possibly through its anti-oxidative properties and is a promising new marker that discriminates between myofibroblasts derived from stellate cells and those from portal fibroblasts.
Collapse
Affiliation(s)
- Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka City University Osaka, Japan
| |
Collapse
|
45
|
Bocca C, Novo E, Miglietta A, Parola M. Angiogenesis and Fibrogenesis in Chronic Liver Diseases. Cell Mol Gastroenterol Hepatol 2015; 1:477-488. [PMID: 28210697 PMCID: PMC5301407 DOI: 10.1016/j.jcmgh.2015.06.011] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 06/02/2015] [Indexed: 12/12/2022]
Abstract
Pathologic angiogenesis appears to be intrinsically associated with the fibrogenic progression of chronic liver diseases, which eventually leads to the development of cirrhosis and related complications, including hepatocellular carcinoma. Several laboratories have suggested that this association is relevant for chronic liver disease progression, with angiogenesis proposed to sustain fibrogenesis. This minireview offers a synthesis of relevant findings and opinions that have emerged in the last few years relating liver angiogenesis to fibrogenesis. We discuss liver angiogenesis in normal and pathophysiologic conditions with a focus on the role of hypoxia and hypoxia-inducible factors and assess the evidence supporting a clear relationship between angiogenesis and fibrogenesis. A section is dedicated to the critical interactions between liver sinusoidal endothelial cells and either quiescent hepatic stellate cells or myofibroblast-like stellate cells. Finally, we introduce the unusual, dual (profibrogenic and proangiogenic) role of hepatic myofibroblasts and emerging evidence supporting a role for specific mediators like vasohibin and microparticles and microvesicles.
Collapse
Key Words
- ANGPTL3, angiopoietin-like-3 peptide
- Akt, protein kinase B
- Ang-1, angiopoietin-1
- CCL2, chemokine ligand 2
- CCR, chemokine receptor
- CLD, chronic liver disease
- ET-1, endothelin 1
- HCC, hepatocellular carcinoma
- HIF, hypoxia-inducible factor
- HSC, hepatic stellate cell
- HSC/MFs, myofibroblast-like cells from activated hepatic stellate cells
- Hh, Hedgehog
- Hypoxia
- LSEC, liver sinusoidal endothelial cell
- Liver Angiogenesis
- Liver Fibrogenesis
- MF, myofibroblast
- MP, microparticle
- Myofibroblasts
- NAFLD, nonalcoholic fatty liver disease
- NASH, nonalcoholic steatohepatitis
- NO, nitric oxide
- PDGF, platelet-derived growth factor
- ROS, reactive oxygen species
- VEGF, vascular endothelial growth factor
- VEGF-R2, vascular endothelial growth factor receptor type 2
- eNOS, endothelial nitric oxide synthase
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
| | | | | | - Maurizio Parola
- Unit of Experimental Medicine and Clinical Pathology, Department of Clinical and Biological Sciences, School of Medicine, University of Torino, Torino, Italy
| |
Collapse
|
46
|
Rovida E, Di Maira G, Tusa I, Cannito S, Paternostro C, Navari N, Vivoli E, Deng X, Gray NS, Esparís-Ogando A, David E, Pandiella A, Dello Sbarba P, Parola M, Marra F. The mitogen-activated protein kinase ERK5 regulates the development and growth of hepatocellular carcinoma. Gut 2015; 64:1454-65. [PMID: 25183205 DOI: 10.1136/gutjnl-2014-306761] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The extracellular signal-regulated kinase 5 (ERK5 or BMK1) is involved in tumour development. The ERK5 gene may be amplified in hepatocellular carcinoma (HCC), but its biological role has not been clarified. In this study, we explored the role of ERK5 expression and activity in HCC in vitro and in vivo. DESIGN ERK5 expression was evaluated in human liver tissue. Cultured HepG2 and Huh-7 were studied after ERK5 knockdown by siRNA or in the presence of the specific pharmacological inhibitor, XMD8-92. The role of ERK5 in vivo was assessed using mouse Huh-7 xenografts. RESULTS In tissue specimens from patients with HCC, a higher percentage of cells with nuclear ERK5 expression was found both in HCC and in the surrounding cirrhotic tissue compared with normal liver tissue. Inhibition of ERK5 decreased HCC cell proliferation and increased the proportion of cells in G0/G1 phase. These effects were associated with increased expression of p27 and p15 and decreased CCND1. Treatment with XMD8-92 or ERK5 silencing prevented cell migration induced by epidermal growth factor or hypoxia and caused cytoskeletal remodelling. In mouse xenografts, the rate of tumour appearance and the size of tumours were significantly lower when Huh-7 was silenced for ERK5. Moreover, systemic treatment with XMD8-92 of mice with established HCC xenografts markedly reduced tumour growth and decreased the expression of the proto-oncogene c-Rel. CONCLUSIONS ERK5 regulates the biology of HCC cells and modulates tumour development and growth in vivo. This pathway should be investigated as a possible therapeutic target in HCC.
Collapse
Affiliation(s)
- Elisabetta Rovida
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università di Firenze, Italy
| | - Giovanni Di Maira
- Dipartimento di Medicina Sperimentale e Clinica Università di Firenze, Italy
| | - Ignazia Tusa
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università di Firenze, Italy
| | - Stefania Cannito
- Dipartimento di Medicina e Oncologia Sperimentali, Università di Torino, Italy
| | - Claudia Paternostro
- Dipartimento di Medicina e Oncologia Sperimentali, Università di Torino, Italy
| | - Nadia Navari
- Dipartimento di Medicina Sperimentale e Clinica Università di Firenze, Italy
| | - Elisa Vivoli
- Dipartimento di Medicina Sperimentale e Clinica Università di Firenze, Italy
| | - Xianming Deng
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Nathanael S Gray
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Azucena Esparís-Ogando
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Spain
| | - Ezio David
- Pathology Unit, Ospedale S. Giovanni Battista, Torino, Italy
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Spain
| | - Persio Dello Sbarba
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università di Firenze, Italy
| | - Maurizio Parola
- Dipartimento di Medicina e Oncologia Sperimentali, Università di Torino, Italy
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica Università di Firenze, Italy
| |
Collapse
|
47
|
Strategies to prevent and reverse liver fibrosis in humans and laboratory animals. Arch Toxicol 2015; 89:1727-50. [PMID: 25963329 DOI: 10.1007/s00204-015-1525-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 04/28/2015] [Indexed: 02/07/2023]
Abstract
Liver fibrosis results from chronic damage to the liver in conjunction with various pathways and is mediated by a complex microenvironment. Based on clinical observations, it is now evident that fibrosis is a dynamic, bidirectional process with an inherent capacity for recovery and remodeling. The major mechanisms involved in liver fibrosis include the repetitive injury of hepatocytes, the activation of the inflammatory response after injury stimulation, and the activation and proliferation of hepatic stellate cells (HSCs), which represents the major extracellular matrix (ECM)-producing cells, stimulated by hepatocyte injury and inflammation. The microenvironment in the liver is synergistically regulated abnormal ECM deposition, scar formation, angiogenesis, and fibrogenesis. Moreover, recent studies have clarified novel mechanism in fibrosis such as epigenetic regulation of HSCs, the leptin and PPARγ pathways, the coagulation system, and even autophagy. Uncovering the mechanisms of liver fibrogenesis provides a basis to develop potential therapies to reverse and treat the fibrotic response, thereby improving the outcomes of patients with chronic liver disease. Although both scientific and clinical challenges remain, emerging studies attempt to reveal the ideal anti-fibrotic drug that could be easily delivered to the liver with high specificity and low toxicity. This review highlights the mechanisms, including novel pathways underlying fibrogenesis that may be translated into preventive and treatment strategies, reviews both current and novel agents that target specific pathways or multiple targets, and discusses novel drug delivery systems such as nanotechnology that can be applied in the treatment of liver fibrosis. In addition, we also discuss some current treatment strategies that are being applied in animal models and in clinical trials.
Collapse
|
48
|
Abstract
Recent data indicate that hepatic angiogenesis, regardless of the etiology, takes place in chronic liver diseases (CLDs) that are characterized by inflammation and progressive fibrosis. Because anti-angiogenic therapy has been found to be efficient in the prevention of fibrosis in experimental models of CLDs, it is suggested that blocking angiogenesis could be a promising therapeutic option in patients with advanced fibrosis. Consequently, efforts are being directed to revealing the mechanisms involved in angiogenesis during the progression of liver fibrosis. Literature evidences indicate that hepatic angiogenesis and fibrosis are closely related in both clinical and experimental conditions. Hypoxia is a major inducer of angiogenesis together with inflammation and hepatic stellate cells. These profibrogenic cells stand at the intersection between inflammation, angiogenesis and fibrosis and play also a pivotal role in angiogenesis. This review mainly focuses to give a clear view on the relevant features that communicate angiogenesis with progression of fibrosis in CLDs towards the-end point of cirrhosis that may be translated into future therapies. The pathogenesis of hepatic angiogenesis associated with portal hypertension, viral hepatitis, non-alcoholic fatty liver disease and alcoholic liver disease are also discussed to emphasize the various mechanisms involved in angiogenesis during liver fibrogenesis.
Collapse
Affiliation(s)
- Gülsüm Özlem Elpek
- Gülsüm Özlem Elpek, Department of Pathology, Akdeniz University Medical School, 07070 Antalya, Turkey
| |
Collapse
|
49
|
P2X7 receptor as a key player in oxidative stress-driven cell fate in nonalcoholic steatohepatitis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:172493. [PMID: 25815106 PMCID: PMC4359843 DOI: 10.1155/2015/172493] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/17/2015] [Indexed: 02/06/2023]
Abstract
Incidences of nonalcoholic fatty liver disease parallels increase in the global obesity epidemic. NAFLD has been shown to be associated with risks of cardiometabolic disorders and kidney disturbances. It is accompanied by insulin and leptin resistance that complicate the diagnosis and treatment of this public health menace. Though significant research is underway for understanding the molecular mechanisms of NAFLD and its subsequent inflammatory and fibrotic manifestations like nonalcoholic steatohepatitis, the role of purinergic receptors has been unclear. It is increasingly being recognized that damage associated molecular patterns like NAD and ATP that are released from injured cells via hepatocellular injury either by oxidative stress or lipotoxicity from steatosis activate the purinergic receptor. Based on evidence from inflammatory responses in the airways and vasculature and autoimmune complications in humans and rodents, it is beyond doubt that hepatocellular inflammation such as that seen in NASH can result from the activation of purinergic receptors. This event can result in the formation of inflammasomes and can be an important pathway for the progression of NASH. The present review evaluates the current knowledge of the role of oxidative stress and its signaling via P2X7 receptors in hepatocellular injury that might contribute to the NASH pathophysiology.
Collapse
|
50
|
Kawada N, Parola M. Interactions of Stellate Cells with Other Non-Parenchymal Cells. STELLATE CELLS IN HEALTH AND DISEASE 2015:185-207. [DOI: 10.1016/b978-0-12-800134-9.00012-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|