1
|
Garg A, Bandyopadhyay S. Role of an interdependent Wnt, GSK3-β/β-catenin and HB-EGF/EGFR mechanism in arsenic-induced hippocampal neurotoxicity in adult mice. CHEMOSPHERE 2024; 352:141375. [PMID: 38325618 DOI: 10.1016/j.chemosphere.2024.141375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/17/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
We previously reported the neurotoxic effects of arsenic in the hippocampus. Here, we explored the involvement of Wnt pathway, which contributes to neuronal functions. Administering environmentally relevant arsenic concentrations to postnatal day-60 (PND60) mice demonstrated a dose-dependent increase in hippocampal Wnt3a and its components, Frizzled, phospho-LRP6, Dishevelled and Axin1 at PND90 and PND120. However, p-GSK3-β(Ser9) and β-catenin levels although elevated at PND90, decreased at PND120. Additionally, treatment with Wnt-inhibitor, rDkk1, reduced p-GSK3-β(Ser9) and β-catenin at PND90, but failed to affect their levels at PND120, indicating a time-dependent link with Wnt. To explore other underlying factors, we assessed epidermal growth factor receptor (EGFR) pathway, which interacts with GSK3-β and appears relevant to neuronal functions. We primarily found that arsenic reduced hippocampal phosphorylated-EGFR and its ligand, Heparin-binding EGF-like growth factor (HB-EGF), at both PND90 and PND120. Moreover, treatment with HB-EGF rescued p-GSK3-β(Ser9) and β-catenin levels at PND120, suggesting their HB-EGF/EGFR-dependent regulation at this time point. Additionally, rDkk1, LiCl (GSK3-β-activity inhibitor), or β-catenin protein treatments induced a time-dependent recovery in HB-EGF, indicating potential inter-dependent mechanism between hippocampal Wnt/β-catenin and HB-EGF/EGFR following arsenic exposure. Fluorescence immunolabeling then validated these findings in hippocampal neurons. Further exploration of hippocampal neuronal survival and apoptosis demonstrated that treatment with rDkk1, LiCl, β-catenin and HB-EGF improved Nissl staining and NeuN levels, and reduced cleaved-caspase-3 levels in arsenic-treated mice. Supportively, we detected improved Y-Maze and Passive Avoidance performances for learning-memory functions in these mice. Overall, our study provides novel insights into Wnt/β-catenin and HB-EGF/EGFR pathway interaction in arsenic-induced hippocampal neurotoxicity.
Collapse
Affiliation(s)
- Asmita Garg
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sanghamitra Bandyopadhyay
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
2
|
Ding R, Chen Y, Shi X, Li Y, Yu Y, Sun Z, Duan J. Size-dependent toxicity of polystyrene microplastics on the gastrointestinal tract: Oxidative stress related-DNA damage and potential carcinogenicity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169514. [PMID: 38135073 DOI: 10.1016/j.scitotenv.2023.169514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/16/2023] [Accepted: 12/17/2023] [Indexed: 12/24/2023]
Abstract
Microplastics (MPs) and nanoplastics (NPs) have been generally regarded as emerging pollutants and received worldwide attention in recent years. Water and food consumption are the primary pathways for human exposure to MPs/NPs, thus gastrointestinal tracts may be susceptible to their toxicity. Although the recent report has indicated the presence of MPs/NPs in multiple human organs, little is known about their gastric effects. Therefore, this study focused on the adverse effects of polystyrene microplastics (PS-MPs) on gastric epithelium in vivo and in vitro. Surface-enhanced Raman spectroscopy (SERS) revealed the distribution of PS-MPs was associated with their particle sizes, and predominantly concentrated in gastric tissues. Gastric barrier injury and mitochondrial damage were observed in rats after exposure to PS-MPs. Compared with the larger ones, polystyrene nanoplastics (PS-NPs) more significantly reduced the activity of antioxidant enzymes while enhancing the level of MDA, 8-OhdG and γ-H2AX. Meanwhile, PS-MPs caused upregulation of β-catenin/YAP through redox-dependent regulation of nucleoredoxin (NXN) and dishevelled (Dvl). These findings supported the size-dependent effects of PS-MPs on oxidative stress and DNA damage. Moreover, the redox-dependent activation of the β-catenin/YAP cascade suggested a novel toxic mechanism for PS-MPs and implied the potential carcinogenic effects.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Xuemin Shi
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Yang Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
3
|
Zhang Y, Zuo D, Qiu J, Li K, Niu Y, Yuan Y, Qiu Y, Qiao L, He W, Wang C, Yuan Y, Li B. NXN suppresses metastasis of hepatocellular carcinoma by promoting degradation of Snail through binding to DUB3. Cell Death Dis 2022; 13:676. [PMID: 35927236 PMCID: PMC9352874 DOI: 10.1038/s41419-022-05135-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 07/04/2022] [Accepted: 07/25/2022] [Indexed: 01/21/2023]
Abstract
The poor prognosis of hepatocellular carcinoma (HCC) could be attributed to its high metastasis rate. Here, we report the role of nucleoredoxin (NXN), a multifunctional redox-active protein, in HCC metastasis. The expression of NXN in HCC tissues was measured by immunohistochemistry. The role of NXN on HCC proliferation was determined by CCK-8, EdU and colony formation assays in vitro and subcutaneous tumor formation model in vivo. Transwell and wound healing assays and tail vein injection model were performed to assess the function of NXN on HCC metastasis. Co-immunoprecipitation assay was performed to examine the interaction among NXN, Snail and DUB3. Our results showed that NXN was downregulated in HCC tissues compared to adjacent liver tissues. Patients with low NXN expression had shorter overall survival (OS) time (P < 0.001) than those with high NXN expression. Biologically, ectopic expression of NXN significantly inhibited the proliferation and metastasis of HCC cells both in vitro and in vivo by suppressing epithelial-mesenchymal transition (EMT). Mechanistically, NXN promoted the ubiquitin-proteasome-mediated degradation of Snail through interaction with DUB3. Further, depletion of Snail abolished NXN-inhibited cell proliferation and metastasis. In summary, NXN suppressed the proliferation and metastasis of HCC by inhibiting DUB3-mediated deubiquitylation of Snail protein. Our study demonstrates that NXN, DUB3 and Snail complex functioned as an important regulatory mechanism of HCC progression and indicates a potential therapeutic approach for the treatment of HCC metastasis.
Collapse
Affiliation(s)
- Yuanping Zhang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Dinglan Zuo
- grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jiliang Qiu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Kai Li
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yi Niu
- grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yichuan Yuan
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yuxiong Qiu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Liang Qiao
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wei He
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chenwei Wang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yunfei Yuan
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Binkui Li
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China ,grid.488530.20000 0004 1803 6191Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
4
|
Idelfonso-García OG, Alarcón-Sánchez BR, Vásquez-Garzón VR, Baltiérrez-Hoyos R, Villa-Treviño S, Muriel P, Serrano H, Pérez-Carreón JI, Arellanes-Robledo J. Is Nucleoredoxin a Master Regulator of Cellular Redox Homeostasis? Its Implication in Different Pathologies. Antioxidants (Basel) 2022; 11:antiox11040670. [PMID: 35453355 PMCID: PMC9030443 DOI: 10.3390/antiox11040670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Nucleoredoxin (NXN), an oxidoreductase enzyme, contributes to cellular redox homeostasis by regulating different signaling pathways in a redox-dependent manner. By interacting with seven proteins so far, namely disheveled (DVL), protein phosphatase 2A (PP2A), phosphofructokinase-1 (PFK1), translocation protein SEC63 homolog (SEC63), myeloid differentiation primary response gene-88 (MYD88), flightless-I (FLII), and calcium/calmodulin-dependent protein kinase II type alpha (CAMK2A), NXN is involved in the regulation of several key cellular processes, including proliferation, organogenesis, cell cycle progression, glycolysis, innate immunity and inflammation, motility, contraction, protein transport into the endoplasmic reticulum, neuronal plasticity, among others; as a result, NXN has been implicated in different pathologies, such as cancer, alcoholic and polycystic liver disease, liver fibrogenesis, obesity, Robinow syndrome, diabetes mellitus, Alzheimer’s disease, and retinitis pigmentosa. Together, this evidence places NXN as a strong candidate to be a master redox regulator of cell physiology and as the hub of different redox-sensitive signaling pathways and associated pathologies. This review summarizes and discusses the current insights on NXN-dependent redox regulation and its implication in different pathologies.
Collapse
Affiliation(s)
- Osiris Germán Idelfonso-García
- Laboratory of Liver Diseases, National Institute of Genomic Medicine–INMEGEN, Mexico City 14610, Mexico; (O.G.I.-G.); (B.R.A.-S.); (J.I.P.-C.)
- Department of Health Sciences, Metropolitan Autonomous University-Iztapalapa Campus, Mexico City 09340, Mexico;
| | - Brisa Rodope Alarcón-Sánchez
- Laboratory of Liver Diseases, National Institute of Genomic Medicine–INMEGEN, Mexico City 14610, Mexico; (O.G.I.-G.); (B.R.A.-S.); (J.I.P.-C.)
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute–CINVESTAV-IPN, Mexico City 07360, Mexico;
| | - Verónica Rocío Vásquez-Garzón
- Laboratory of Fibrosis and Cancer, Faculty of Medicine and Surgery, ‘Benito Juárez’ Autonomous University of Oaxaca–UABJO, Oaxaca 68020, Mexico; (V.R.V.-G.); (R.B.-H.)
- Directorate of Cátedras, National Council of Science and Technology–CONACYT, Mexico City 03940, Mexico
| | - Rafael Baltiérrez-Hoyos
- Laboratory of Fibrosis and Cancer, Faculty of Medicine and Surgery, ‘Benito Juárez’ Autonomous University of Oaxaca–UABJO, Oaxaca 68020, Mexico; (V.R.V.-G.); (R.B.-H.)
- Directorate of Cátedras, National Council of Science and Technology–CONACYT, Mexico City 03940, Mexico
| | - Saúl Villa-Treviño
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute–CINVESTAV-IPN, Mexico City 07360, Mexico;
| | - Pablo Muriel
- Laboratory of Experimental Hepatology, Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute–CINVESTAV-IPN, Mexico City 07360, Mexico;
| | - Héctor Serrano
- Department of Health Sciences, Metropolitan Autonomous University-Iztapalapa Campus, Mexico City 09340, Mexico;
| | - Julio Isael Pérez-Carreón
- Laboratory of Liver Diseases, National Institute of Genomic Medicine–INMEGEN, Mexico City 14610, Mexico; (O.G.I.-G.); (B.R.A.-S.); (J.I.P.-C.)
| | - Jaime Arellanes-Robledo
- Laboratory of Liver Diseases, National Institute of Genomic Medicine–INMEGEN, Mexico City 14610, Mexico; (O.G.I.-G.); (B.R.A.-S.); (J.I.P.-C.)
- Directorate of Cátedras, National Council of Science and Technology–CONACYT, Mexico City 03940, Mexico
- Correspondence: ; Tel.: +52-55-5350-1900 (ext. 1218)
| |
Collapse
|
5
|
Aoki T, Nishida N, Kudo M. Current Perspectives on the Immunosuppressive Niche and Role of Fibrosis in Hepatocellular Carcinoma and the Development of Antitumor Immunity. J Histochem Cytochem 2022; 70:53-81. [PMID: 34751050 PMCID: PMC8721576 DOI: 10.1369/00221554211056853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Immune checkpoint inhibitors have become the mainstay of treatment for hepatocellular carcinoma (HCC). However, they are ineffective in some cases. Previous studies have reported that genetic alterations in oncogenic pathways such as Wnt/β-catenin are the important triggers in HCC for primary refractoriness. T-cell exhaustion has been reported in various tumors and is likely to play a prominent role in the emergence of HCC due to chronic inflammation and cirrhosis-associated immune dysfunction. Immunosuppressive cells including regulatory T-cells and tumor-associated macrophages infiltrating the tumor are associated with hyperprogressive disease in the early stages of immune checkpoint inhibitor treatment. In addition, stellate cells and tumor-associated fibroblasts create an abundant desmoplastic environment by producing extracellular matrix. This strongly contributes to epithelial to mesenchymal transition via signaling activities including transforming growth factor beta, Wnt/β-catenin, and Hippo pathway. The abundant desmoplastic environment has been demonstrated in pancreatic ductal adenocarcinoma and cholangiocarcinoma to suppress cytotoxic T-cell infiltration, PD-L1 expression, and neoantigen expression, resulting in a highly immunosuppressive niche. It is possible that a similar immunosuppressive environment is created in HCC with advanced fibrosis in the background liver. Although sufficient understanding is required for the establishment of immune therapies of HCC, further investigations are still required in this field.
Collapse
Affiliation(s)
- Tomoko Aoki
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, Osaka-Sayama, Japan
| | - Naoshi Nishida
- Naoshi Nishida, Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, 377-2 Ohno-higashi, Osaka-Sayama 589-8511, Japan. E-mail:
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, Osaka-Sayama, Japan
| |
Collapse
|
6
|
Hyun J, Han J, Lee C, Yoon M, Jung Y. Pathophysiological Aspects of Alcohol Metabolism in the Liver. Int J Mol Sci 2021; 22:5717. [PMID: 34071962 PMCID: PMC8197869 DOI: 10.3390/ijms22115717] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
Alcoholic liver disease (ALD) is a globally prevalent chronic liver disease caused by chronic or binge consumption of alcohol. The liver is the major organ that metabolizes alcohol; therefore, it is particularly sensitive to alcohol intake. Metabolites and byproducts generated during alcohol metabolism cause liver damage, leading to ALD via several mechanisms, such as impairing lipid metabolism, intensifying inflammatory reactions, and inducing fibrosis. Despite the severity of ALD, the development of novel treatments has been hampered by the lack of animal models that fully mimic human ALD. To overcome the current limitations of ALD studies and therapy development, it is necessary to understand the molecular mechanisms underlying alcohol-induced liver injury. Hence, to provide insights into the progression of ALD, this review examines previous studies conducted on alcohol metabolism in the liver. There is a particular focus on the occurrence of ALD caused by hepatotoxicity originating from alcohol metabolism.
Collapse
Affiliation(s)
- Jeongeun Hyun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea;
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan 31116, Korea
| | - Jinsol Han
- Department of Integrated Biological Science, Pusan National University, Pusan 46241, Korea; (J.H.); (C.L.)
| | - Chanbin Lee
- Department of Integrated Biological Science, Pusan National University, Pusan 46241, Korea; (J.H.); (C.L.)
| | - Myunghee Yoon
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Biomedical Research Institute, Pusan National University, Pusan 46241, Korea;
| | - Youngmi Jung
- Department of Integrated Biological Science, Pusan National University, Pusan 46241, Korea; (J.H.); (C.L.)
- Department of Biological Sciences, Pusan National University, Pusan 46241, Korea
| |
Collapse
|
7
|
Arellanes-Robledo J, Ibrahim J, Reyes-Gordillo K, Shah R, Leckey L, Lakshman MR. Flightless-I is a potential biomarker for the early detection of alcoholic liver disease. Biochem Pharmacol 2021; 183:114323. [PMID: 33166508 PMCID: PMC8614159 DOI: 10.1016/j.bcp.2020.114323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023]
Abstract
Alcoholic liver disease (ALD) is closely linked to oxidative stress induction. Antioxidant enzymes balance oxidative stress and function as intermediary signaling regulators. Nucleoredoxin (NXN), an antioxidant enzyme, regulates physiological processes through redox-sensitive interactions. NXN interacts with myeloid differentiation primary response gene-88 (MYD88) and flightless-I (FLII) to regulate toll-like receptor 4 (TLR4)/MYD88 pathway activation, but FLII also regulates key cell processes and is secreted into the bloodstream. However, the effects of chronic ethanol consumption recapitulated by either ethanol alone or in combination with lipopolysaccharides (LPS), as a two-hit ALD model, on FLII/NXN/MYD88 complex and FLII secretion have not been explored yet. In this study, we have demonstrated that ethanol feeding increased FLII protein levels, its nuclear translocation and plasma secretion, and modified its tissue distribution both in vivo and in vitro ALD models. Ethanol increased MYD88/FLII interaction ratio, and decreased NXN/MYD88 interaction ratio but this was partially reverted by two-hit model. While ethanol and two-hit model increased MYD88/TLR4 interaction ratio, two-hit model significantly decreased FLII nuclear translocation and its plasma secretion. Ethanol and LPS provoked similar effects in vitro; however, NXN overexpression partially reverted these alterations, and ethanol alone increased FLII secretion into culture medium. In summary, by analyzing the response of FLII/NXN/MYD88 complex during ALD early progression both in vivo and in vitro, we have discovered that the effects of chronic ethanol consumption disrupt this complex and identified FLII as a candidate non-invasive plasma biomarker for the early detection of ALD.
Collapse
Affiliation(s)
- Jaime Arellanes-Robledo
- Lipid Research Laboratory, VA Medical Center, Washington, D.C., USA; Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA; Laboratory of Hepatic Diseases, National Institute of Genomic Medicine - INMEGEN, CDMX, Mexico; Directorate of Cátedras, National Council of Science and Technology - CONACYT, CDMX, Mexico.
| | - Joseph Ibrahim
- Lipid Research Laboratory, VA Medical Center, Washington, D.C., USA; Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA
| | - Karina Reyes-Gordillo
- Lipid Research Laboratory, VA Medical Center, Washington, D.C., USA; Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA
| | - Ruchi Shah
- Lipid Research Laboratory, VA Medical Center, Washington, D.C., USA; Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA; Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Leslie Leckey
- Lipid Research Laboratory, VA Medical Center, Washington, D.C., USA; Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA
| | - M Raj Lakshman
- Lipid Research Laboratory, VA Medical Center, Washington, D.C., USA; Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA
| |
Collapse
|
8
|
Zheng H, Yang Z, Xin Z, Yang Y, Yu Y, Cui J, Liu H, Chen F. Glycogen synthase kinase-3β: a promising candidate in the fight against fibrosis. Theranostics 2020; 10:11737-11753. [PMID: 33052244 PMCID: PMC7545984 DOI: 10.7150/thno.47717] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023] Open
Abstract
Fibrosis exists in almost all organs/tissues of the human body, plays an important role in the occurrence and development of diseases and is also a hallmark of the aging process. However, there is no effective prevention or therapeutic method for fibrogenesis. As a serine/threonine (Ser/Thr)-protein kinase, glycogen synthase kinase-3β (GSK-3β) is a vital signaling mediator that participates in a variety of biological events and can inhibit extracellular matrix (ECM) accumulation and the epithelial-mesenchymal transition (EMT) process, thereby exerting its protective role against the fibrosis of various organs/tissues, including the heart, lung, liver, and kidney. Moreover, we further present the upstream regulators and downstream effectors of the GSK-3β pathway during fibrosis and comprehensively summarize the roles of GSK-3β in the regulation of fibrosis and provide several potential targets for research. Collectively, the information reviewed here highlights recent advances vital for experimental research and clinical development, illuminating the possibility of GSK-3β as a novel therapeutic target for the management of tissue fibrosis in the future.
Collapse
Affiliation(s)
- Hanxue Zheng
- Lab of Tissue Engineering, Faculty of Life Sciences, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Zhi Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Zhenlong Xin
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yuan Yu
- Lab of Tissue Engineering, Faculty of Life Sciences, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Jihong Cui
- Lab of Tissue Engineering, Faculty of Life Sciences, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Hongbo Liu
- Lab of Tissue Engineering, Faculty of Life Sciences, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Fulin Chen
- Lab of Tissue Engineering, Faculty of Life Sciences, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, 229 TaiBai North Road, Xi'an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| |
Collapse
|
9
|
Alarcón‐Sánchez BR, Guerrero‐Escalera D, Rosas‐Madrigal S, Ivette Aparicio‐Bautista D, Reyes‐Gordillo K, Lakshman MR, Ortiz‐Fernández A, Quezada H, Medina‐Contreras Ó, Villa‐Treviño S, Isael Pérez‐Carreón J, Arellanes‐Robledo J. Nucleoredoxin interaction with flightless‐I/actin complex is differentially altered in alcoholic liver disease. Basic Clin Pharmacol Toxicol 2020; 127:389-404. [DOI: 10.1111/bcpt.13451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/30/2020] [Accepted: 06/03/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Brisa Rodope Alarcón‐Sánchez
- Laboratory of Liver Diseases National Institute of Genomic Medicine CDMX Mexico
- Departament of Cell Biology Center for Research and Advanced Studies of the National Polytechnic Institute CDMX Mexico
| | | | - Sandra Rosas‐Madrigal
- Laboratory of Cardiovascular Diseases National Institute of Genomic Medicine CDMX Mexico
| | | | - Karina Reyes‐Gordillo
- Lipid Research Laboratory VA Medical Center Washington DC USA
- Department of Biochemistry and Molecular Medicine The George Washington University Medical Center Washington DC USA
| | - M. Raj Lakshman
- Lipid Research Laboratory VA Medical Center Washington DC USA
- Department of Biochemistry and Molecular Medicine The George Washington University Medical Center Washington DC USA
| | - Arturo Ortiz‐Fernández
- Departament of Cell Biology Center for Research and Advanced Studies of the National Polytechnic Institute CDMX Mexico
| | - Héctor Quezada
- Research Laboratory in Immunology and Proteomics Children's Hospital of Mexico "Federico Gómez” CDMX Mexico
| | - Óscar Medina‐Contreras
- Research Department in Community Health Children's Hospital of Mexico "Federico Gómez" CDMX Mexico
| | - Saúl Villa‐Treviño
- Departament of Cell Biology Center for Research and Advanced Studies of the National Polytechnic Institute CDMX Mexico
| | | | - Jaime Arellanes‐Robledo
- Laboratory of Liver Diseases National Institute of Genomic Medicine CDMX Mexico
- Directorate of Cátedras National Council of Science and Technology CDMX Mexico
| |
Collapse
|
10
|
Heme Oxygenase-1 Suppresses Wnt Signaling Pathway in Nonalcoholic Steatohepatitis-Related Liver Fibrosis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4910601. [PMID: 32461992 PMCID: PMC7212281 DOI: 10.1155/2020/4910601] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/29/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022]
Abstract
Methods Mice were fed with a methionine-choline-deficient (MCD) diet for 8 weeks to induce steatohepatitis-related liver fibrosis and were treated with HO-1 inducer Hemin and inhibitor ZnPP. Mouse sera were collected for the biochemical analysis, and livers were obtained for further histological observation and gene expression analysis. HSC-T6 cells were cultured for the in vitro study and were administrated with Hemin and si-HO-1 to induce or inhibit the expression of HO-1. qPCR and Western blot were used to assess the mRNA and protein levels of genes. Results MCD-fed mice developed marked macrovesicular steatosis, focal necrosis, and inflammatory infiltration and pericellular fibrosis in liver sections. Administration of Hemin could significantly ameliorate the severity of steatosis, inflammation, and fibrosis and also could decrease the serum ALT and AST. We demonstrated that HO-1 induction was able to downregulate the key regulator of the canonical Wnt pathway Wnt1 and the noncanonical Wnt pathway Wnt5a. The downstream factors of the Wnt pathway β-catenin and NFAT5 were inhibited by Hemin, but GSK-3β was upregulated compared to the MCD group, which were consistent with the in vitro study. Hemin markedly inhibited the TGF-β1/Smad signaling pathway in both in vivo and in vitro studies. Conclusion Our study demonstrated that HO-1 inhibited the activation of canonical and noncanonical Wnt signaling pathways in NASH-related liver fibrosis. Thus, these results may suggest a new therapeutic strategy for NASH-related liver fibrosis.
Collapse
|
11
|
Dai X, Chen C, Xue J, Xiao T, Mostofa G, Wang D, Chen X, Xu H, Sun Q, Li J, Wei Y, Chen F, Quamruzzaman Q, Zhang A, Liu Q. Exosomal MALAT1 derived from hepatic cells is involved in the activation of hepatic stellate cells via miRNA-26b in fibrosis induced by arsenite. Toxicol Lett 2019; 316:73-84. [PMID: 31513886 DOI: 10.1016/j.toxlet.2019.09.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/28/2019] [Accepted: 09/08/2019] [Indexed: 12/15/2022]
Abstract
In the liver microenvironment, interactions among diverse types of hepatic cells are involved in liver fibrosis. In fibrotic tissues, exosomes act as transporters in intercellular communication. Long non-coding RNAs (lncRNAs) are involved in the activation of hepatic stellate cells (HSCs), which are participants in liver fibrosis. However, the functions of exosomal lncRNAs in liver fibrosis induced by arsenite are undefined. The purposes of the present study were (a) to determine if lncRNAs secreted from human hepatic (L-02) cells exposed to arsenite are shuttled to hepatic stellate LX-2 cells and (b) to establish their effects on LX-2 cells. In mice, MALAT1 was overexpressed in the progression of liver fibrosis induced by arsenite as well as in L-02 cells exposed to arsenite. Co-cultures with arsenite-treated L-02 cells induced the activation of LX-2 cells and overexpression of MALAT1. Arsenite-treated L-02 cells transported MALAT1 into LX-2 cells. Downregulation of MALAT1, which reduced the MALAT1 levels in exosomes derived from arsenite-treated L-02 cells, inhibited the activation of LX-2 cells. Additionally, exosomal MALAT1 derived from arsenite-treated L-02 cells promoted the activation of LX-2 cells via microRNA-26b regulation of COL1A2. Furthermore, circulating exosomal MALAT1 was up-regulated in people exposed to arsenite. In sum, exosomes derived from arsenite-treated hepatic cells transferred MALAT1 to HSCs, which induced their activation. These findings support the concept that, during liver fibrosis induced by arsenite, exosomal lncRNAs are involved in cell-cell communication.
Collapse
Affiliation(s)
- Xiangyu Dai
- Center for Global Health, China International Cooperation Center for Environment and Human Health, 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
| | - Chao Chen
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan, people's Republic of China
| | - Junchao Xue
- Center for Global Health, China International Cooperation Center for Environment and Human Health, 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
| | - Tian Xiao
- Center for Global Health, China International Cooperation Center for Environment and Human Health, 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
| | - Golam Mostofa
- Dhaka Community Hospital Trust, Dhaka 1217, Bangladesh
| | - Dapeng Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Xiong Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Hui Xu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, 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
| | - Qian Sun
- Center for Global Health, China International Cooperation Center for Environment and Human Health, 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, China International Cooperation Center for Environment and Human Health, 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
| | - Yongyue Wei
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Feng Chen
- Center for Global Health, China International Cooperation Center for Environment and Human Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | | | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, People's Republic of China
| | - Qizhan Liu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, 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
|
12
|
Suryadevara V, Huang L, Kim SJ, Cheresh P, Shaaya M, Bandela M, Fu P, Feghali-Bostwick C, Di Paolo G, Kamp DW, Natarajan V. Role of phospholipase D in bleomycin-induced mitochondrial reactive oxygen species generation, mitochondrial DNA damage, and pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2019; 317:L175-L187. [PMID: 31090437 DOI: 10.1152/ajplung.00320.2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a pernicious lung disease characterized by alveolar epithelial apoptosis, dysregulated repair of epithelial injury, scar formation, and respiratory failure. In this study, we identified phospholipase D (PLD)-generated phosphatidic acid (PA) signaling in the development of pulmonary fibrosis (PF). Of the PLD isoenzymes, the protein expression of PLD2, but not PLD1, was upregulated in lung tissues from IPF patients and bleomycin challenged mice. Both PLD1 (Pld1-/-)- and PLD2 (Pld2-/-)-deficient mice were protected against bleomycin-induced lung inflammation and fibrosis, thereby establishing the role of PLD in fibrogenesis. The role of PLD1 and PLD2 in bleomycin-induced lung epithelial injury was investigated by infecting bronchial airway epithelial cells (Beas2B) with catalytically inactive mutants of PLD (hPLD1-K898R or mPld2-K758R) or downregulation of expression of PLD1 or PLD2 with siRNA. Bleomycin stimulated mitochondrial (mt) superoxide production, mtDNA damage, and apoptosis in Beas2B cells, which was attenuated by the catalytically inactive mutants of PLD or PLD2 siRNA. These results show a role for PLD1 and PLD2 in bleomycin-induced generation of mt reactive oxygen species, mt DNA damage, and apoptosis of lung epithelial cells in mice. Thus, PLD may be a novel therapeutic target in ameliorating experimental PF in mice.
Collapse
Affiliation(s)
- Vidyani Suryadevara
- Department of Bioengineering, University of Illinois at Chicago , Chicago, Illinois
| | - Longshuang Huang
- Department of Pharmacology, University of Illinois at Chicago , Chicago, Illinois
| | - Seok-Jo Kim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine and the Jesse Brown VA Medical Center , Chicago, Illinois
| | - Paul Cheresh
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine and the Jesse Brown VA Medical Center , Chicago, Illinois
| | - Mark Shaaya
- Department of Pharmacology, University of Illinois at Chicago , Chicago, Illinois
| | - Mounica Bandela
- Department of Bioengineering, University of Illinois at Chicago , Chicago, Illinois
| | - Panfeng Fu
- Department of Pharmacology, University of Illinois at Chicago , Chicago, Illinois
| | | | - Gilbert Di Paolo
- Department of Pathology and Cell Biology, Columbia University Medical Center , New York, New York
| | - David W Kamp
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine and the Jesse Brown VA Medical Center , Chicago, Illinois
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois at Chicago , Chicago, Illinois.,Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| |
Collapse
|
13
|
Arellanes-Robledo J, Reyes-Gordillo K, Ibrahim J, Leckey L, Shah R, Lakshman MR. Ethanol targets nucleoredoxin/dishevelled interactions and stimulates phosphatidylinositol 4-phosphate production in vivo and in vitro. Biochem Pharmacol 2018; 156:135-146. [PMID: 30125555 PMCID: PMC6297114 DOI: 10.1016/j.bcp.2018.08.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/16/2018] [Indexed: 02/07/2023]
Abstract
Nucleoredoxin (NXN) is a redox-regulating protein potentially targeted by reactive oxygen species (ROS). It regulates molecular pathways that participate in several key cellular processes. However, the role of NXN in the alcohol liver disease (ALD) redox regulation has not been fully understood. Here, we investigated the effects of ethanol and ethanol plus lipopolysaccharide, a two-hit liver injury model (Ethanol/LPS), on NXN/dishevelled (DVL) interaction and on DVL-dependent phosphoinositides production both in mouse liver and in a co-culture system consisting of human hepatic stellate cells (HSC) and ethanol metabolizing-VL17A human hepatocyte cells. Ethanol and two-hit model increased Nxn protein and mRNA expression, and 4-hydroxynonenal adducts. Two-hit model promoted Nxn nuclear translocation and Dvl/Phosphatidylinositol 4-kinase type-IIα (Pi4k2a) interaction ratio but surprisingly decreased Dvl protein and mRNA levels and reverted ethanol-induced Nxn/Dvl and Dvl/frizzled (Fzd) interaction ratios. Ethanol resulted in a significant increase of Dvl protein and mRNA expression, and decreased Nxn/Dvl interaction ratio but promoted the interaction of Dvl with Fzd and Pi4k2a; formation of this complex induced phosphatidylinositol 4-phosphate [PI(4)P] production. Ethanol and LPS treatments provoked similar alterations on NXN/DVL interaction and its downstream effect in HSC/VL17A co-culture system. Interestingly, ROS and glutathione levels as well as most of ethanol-induced alterations were modified by NXN overexpression in the co-culture system. In conclusion, two-hit model of ethanol exposure disrupts NXN/DVL homeostatic status to allow DVL/FZD/PI4K2A complex formation and stimulates PI(4)P production. These results provide a new mechanism showing that NXN also participates in the regulation of phosphoinositides production that is altered by ethanol during alcoholic liver disease progression.
Collapse
Affiliation(s)
- Jaime Arellanes-Robledo
- Lipid Research Laboratory, VA Medical Center, Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA; Laboratory of Hepatic Diseases, National Institute of Genomic Medicine - INMEGEN, CDMX, Mexico; National Council of Science and Technology - CONACYT, CDMX, Mexico.
| | - Karina Reyes-Gordillo
- Lipid Research Laboratory, VA Medical Center, Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA.
| | - Joseph Ibrahim
- Lipid Research Laboratory, VA Medical Center, Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA
| | - Leslie Leckey
- Lipid Research Laboratory, VA Medical Center, Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA
| | - Ruchi Shah
- Lipid Research Laboratory, VA Medical Center, Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA
| | - M Raj Lakshman
- Lipid Research Laboratory, VA Medical Center, Department of Biochemistry and Molecular Medicine, The George Washington University Medical Center, Washington, D.C., USA
| |
Collapse
|
14
|
Emerging role and therapeutic implication of Wnt signaling pathways in liver fibrosis. Gene 2018; 674:57-69. [PMID: 29944952 DOI: 10.1016/j.gene.2018.06.053] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/14/2018] [Accepted: 06/16/2018] [Indexed: 02/08/2023]
Abstract
Activation of hepatic stellate cells (HSCs) is a pivotal cellular event in liver fibrosis. Therefore, improving our understanding of the molecular pathways that are involved in these processes is essential to generate new therapies for liver fibrosis. Greater knowledge of the role of the Wnt signaling pathway in liver fibrosis could improve understanding of the liver fibrosis pathogenesis. The aim of this review is to describe the present knowledge about the Wnt signaling pathway, which significantly participates in liver fibrosis and HSC activation, and look ahead on new perspectives of Wnt signaling pathway research. Moreover, we will discuss the different interactions with Wnt signaling pathway-regulated liver fibrosis. The Wnt signaling pathway modulates several important aspects of function, including cell proliferation, activation and differentiation. Targeting the Wnt signaling pathway can be a promising direction in liver fibrosis treatment. We discuss new perspectives of Wnt signaling pathway activation in liver fibrosis. For example, antagonist to Wnt and Wnt ligands could inhibit liver fibrosis by regulating Wnt/β-catenin signaling pathway. These findings identify the Wnt signaling pathway as a potentially important for therapeutic targets in liver fibrosis. Future studies are needed in order to find safer and more effective Wnt-based drugs.
Collapse
|
15
|
Wang Q, Zang W, Han L, Yang L, Ye S, Ouyang J, Zhang C, Bi Y, Zhang C, Bian H. Wenyang Huazhuo Tongluo formula inhibits fibrosis via suppressing Wnt/β-catenin signaling pathway in a Bleomycin-induced systemic sclerosis mouse model. Chin Med 2018; 13:17. [PMID: 29599817 PMCID: PMC5870182 DOI: 10.1186/s13020-018-0175-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 03/20/2018] [Indexed: 02/06/2023] Open
Abstract
Background Systemic sclerosis (SSc) is an autoimmune disease characterized by fibrosis of the skin and internal organs. So far, no Western medicine treatment can completely inhibit or reverse the progress of SSc, while at the same time, our previous series of studies have shown that the treatment of SSc by the Wenyang Huazhuo Tongluo formula (WYHZTL), a Chinese herbal decoction, shows a delightful prospect. The aim of this study is to further investigate the mechanism of anti-fibrosis of WYHZTL formula in SSc mouse model. Methods The Bleomycin-induced SSc mouse model was treated with saline (BLM), high-dosage of WYHZTL formula (WYHZTL-H), medium-dosage of WYHZTL formula (WYHZTL-M), low-dosage of WYHZTL formula (WYHZTL-L) and XAV-939, a small molecule inhibitor of Wnt/β-catenin signaling pathway, by the intragastric administration and intraperitoneal injection, respectively. The mRNA and protein levels of Wnt/β-catenin signaling pathway associated genes, fibrosis markers and histopathology were detected by reverse transcription-quantitative polymerase chain reaction, Western blotting and hematoxylin/eosin-staining. The levels of Wnt1, CTGF and DKK1 protein in serum were detected by enzyme-linked immunosorbent assay. Results Compared with BLM group, the WYHZTL formula and XAV-939 could significantly inhibit the thickness of the skin tissue of the SSc mouse model. The mRNA expression levels of GSK3β and DKK1 in the WYHZTL formula and XAV-939-treated group were significantly higher than those in the BLM group, while Wnt1, β-catenin, TCF4, cyclin D1, survivin, VEGF, CTGF, FN1, collagen I/III were decreased. Compared with BLM group, the protein expression levels of GSK3β and DKK1 in the WYHZTL formula and XAV-939-treated group were upregulated, while Wnt1, β-catenin, cyclin D1, survivin, CTGF, FN1, collagen I/III were downregulated. WYHZTL formula and XAV-939 could inhibit expression of Wnt1 and CTGF, but promoted DKK1 in serum. Furthermore, WYHZTL-H seemed more effective than WYHZTL-M and/or XAV-939 on regulating Wnt1, β-catenin, TCF4, GSK3β, DKK1, cyclin D1, survivin, VEGF, CTGF, FN1 and collagen I/III. Conclusion This present study demonstrates that WYHZTL formula has anti-fibrosis effect in Bleomycin-induced SSc mouse model in a dosage-dependent manner, and the molecular mechanism may be related to the inhibition of Wnt/β-catenin signaling pathway. Electronic supplementary material The online version of this article (10.1186/s13020-018-0175-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qian Wang
- 1Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China.,2Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan China
| | - Wenhua Zang
- 1Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China.,2Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan China
| | - Li Han
- 1Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China.,2Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan China
| | - Lei Yang
- 1Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China.,2Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan China
| | - Songshan Ye
- 1Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China.,2Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan China
| | - Jingfeng Ouyang
- 3Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chaoyun Zhang
- 1Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Yuefeng Bi
- 4School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan China
| | - Cuiyue Zhang
- 1Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Hua Bian
- 1Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China.,2Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan China
| |
Collapse
|
16
|
Zhou F, Wang A, Li D, Wang Y, Lin L. Pinocembrin from Penthorum chinense Pursh suppresses hepatic stellate cells activation through a unified SIRT3-TGF-β-Smad signaling pathway. Toxicol Appl Pharmacol 2018; 341:38-50. [DOI: 10.1016/j.taap.2018.01.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/03/2018] [Accepted: 01/13/2018] [Indexed: 01/18/2023]
|
17
|
Park JW, Jung KH, Lee JH, Moon SH, Cho YS, Lee KH. Inhibition of aldehyde dehydrogenase 1 enhances the cytotoxic effect of retinaldehyde on A549 cancer cells. Oncotarget 2017; 8:99382-99393. [PMID: 29245909 PMCID: PMC5725100 DOI: 10.18632/oncotarget.19544] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 06/26/2017] [Indexed: 01/05/2023] Open
Abstract
We hypothesized that aldehyde dehydrogenase1 (ALDH1) protects cancer cells from retinaldehyde-induced cytotoxicity, and that targeting this enzyme would enhance the therapeutic effect of retinaldehyde. ALDEFLUOR™ assays showed high ALDH activity in A549 and H522 cancer cells and low activity in H1666 and T47D cancer cells. Immunoblots showed that expression of ALDH1A1 and ALDH1A3 was high in A549 and H522 cells, but low in H1666 cells. HPLC confirmed that N, N-diethylaminobenzaldehyde (DEAB) inhibits ALDH-mediated disposal of retinaldehyde in A549 cells and lysates. Treatment of A549 cells with retinaldehyde in the presence of DEAB augmented reactive oxygen species production and decreased glucose uptake and oxygen consumption. Importantly, DEAB substantially potentiated the ability of retinaldehyde to dose-dependently suppress the survival of A549 and H522 cells, whereas the added effect of DEAB was minor in H1666 and T47D cells. Gene silencing with specific siRNA revealed that ALDH1A1 contributed to protection of A549 cells against retinaldehyde toxicity. These results demonstrate that ALDH1 confers protection against retinaldehyde toxicity in cancer cells.
Collapse
Affiliation(s)
- Jin Won Park
- Department of Nuclear Medicine, Samsung Medical Center, Seoul, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung-Ho Jung
- Department of Nuclear Medicine, Samsung Medical Center, Seoul, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Hee Lee
- Department of Nuclear Medicine, Samsung Medical Center, Seoul, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Seoul, Korea
| | - Young Seok Cho
- Department of Nuclear Medicine, Samsung Medical Center, Seoul, Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Seoul, Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Wu X, Wang Y, Wang S, Xu R, Lv X. Purinergic P2X7 receptor mediates acetaldehyde-induced hepatic stellate cells activation via PKC-dependent GSK3β pathway. Int Immunopharmacol 2017; 43:164-171. [PMID: 28061416 DOI: 10.1016/j.intimp.2016.12.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 11/22/2016] [Accepted: 12/12/2016] [Indexed: 12/31/2022]
Abstract
The activation of hepatic stellate cells (HSCs) is an essential part in the development of alcoholic liver fibrosis (ALF). In this study, stimulated HSCs with 200μM acetaldehyde for 48h was used to imitate alcoholic liver fibrosis in vitro. The western blot and qRT-PCR results showed that P2X7R expression was significantly increased in the activation of HSCs after acetaldehyde treatment. Interestingly, activation of P2X7R by stimulating with P2X7R agonist BzATP significantly promoted acetaldehyde-induced CyclinD1 expression, cell proportion in S phase, inflammatory response, and the protein and mRNA levels of α-SMA, collagen I. In contrast, blockage of P2X7R by stimulating with the inhibitor A438079 or transfecting with specific siRNA dramatically suppressed acetaldehyde-induced HSCs activation. Furthermore, PKC activation treated with PMA could obviously up-regulate the expression of α-SMA and collagen I and the phosphorylation of GSK3β, while inhibition of PKC significantly reduced GSK3β activation. Moreover, GSK3β inhibition harvested a dramatic decrease of the mRNA and protein levels of α-SMA and collagen I by suppressing GSK3β phosphorylation. Taken together, these results suggested that purinergic P2X7R mediated acetaldehyde-induced activation of HSCs via PKC-dependent GSK3β pathway, which maybe a novel target for limiting HSCs activation.
Collapse
Affiliation(s)
- Xiaojuan Wu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Yuhui Wang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Sheng Wang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Rixiang Xu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Xiongwen Lv
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
19
|
Boye A, Zou YH, Yang Y. Metabolic derivatives of alcohol and the molecular culprits of fibro-hepatocarcinogenesis: Allies or enemies? World J Gastroenterol 2016; 22:50-71. [PMID: 26755860 PMCID: PMC4698508 DOI: 10.3748/wjg.v22.i1.50] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 10/12/2015] [Accepted: 11/19/2015] [Indexed: 02/06/2023] Open
Abstract
Chronic intake of alcohol undoubtedly overwhelms the structural and functional capacity of the liver by initiating complex pathological events characterized by steatosis, steatohepatitis, hepatic fibrosis and cirrhosis. Subsequently, these initial pathological events are sustained and ushered into a more complex and progressive liver disease, increasing the risk of fibro-hepatocarcinogenesis. These coordinated pathological events mainly result from buildup of toxic metabolic derivatives of alcohol including but not limited to acetaldehyde (AA), malondialdehyde (MDA), CYP2E1-generated reactive oxygen species, alcohol-induced gut-derived lipopolysaccharide, AA/MDA protein and DNA adducts. The metabolic derivatives of alcohol together with other comorbidity factors, including hepatitis B and C viral infections, dysregulated iron metabolism, abuse of antibiotics, schistosomiasis, toxic drug metabolites, autoimmune disease and other non-specific factors, have been shown to underlie liver diseases. In view of the multiple etiology of liver diseases, attempts to delineate the mechanism by which each etiological factor causes liver disease has always proved cumbersome if not impossible. In the case of alcoholic liver disease (ALD), it is even more cumbersome and complicated as a result of the many toxic metabolic derivatives of alcohol with their varying liver-specific toxicities. In spite of all these hurdles, researchers and experts in hepatology have strived to expand knowledge and scientific discourse, particularly on ALD and its associated complications through the medium of scientific research, reviews and commentaries. Nonetheless, the molecular mechanisms underpinning ALD, particularly those underlying toxic effects of metabolic derivatives of alcohol on parenchymal and non-parenchymal hepatic cells leading to increased risk of alcohol-induced fibro-hepatocarcinogenesis, are still incompletely elucidated. In this review, we examined published scientific findings on how alcohol and its metabolic derivatives mount cellular attack on each hepatic cell and the underlying molecular mechanisms leading to disruption of core hepatic homeostatic functions which probably set the stage for the initiation and progression of ALD to fibro-hepatocarcinogenesis. We also brought to sharp focus, the complex and integrative role of transforming growth factor beta/small mothers against decapentaplegic/plasminogen activator inhibitor-1 and the mitogen activated protein kinase signaling nexus as well as their cross-signaling with toll-like receptor-mediated gut-dependent signaling pathways implicated in ALD and fibro-hepatocarcinogenesis. Looking into the future, it is hoped that these deliberations may stimulate new research directions on this topic and shape not only therapeutic approaches but also models for studying ALD and fibro-hepatocarcinogenesis.
Collapse
|
20
|
Li W, Zhu C, Wu Y, Wang Z, Zhu C. Increased Pygo2 expression in liver of patients with hepatitis B virus-related fibrosis. Liver Int 2015; 35:2522-9. [PMID: 26013055 DOI: 10.1111/liv.12877] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 05/18/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS It has been reported that Wnt/β-catenin signalling pathway played a key role in liver fibrosis and that Pygo2 was an important mediator in β-catenin induced pathway. However, the role of Pygo2 in liver fibrogenesis was unknown. Our study was to investigate the expression of Pygo2 and its diagnostic value in patients with HBV-related liver fibrosis. METHODS Hundred and sixty-four patients with HBV infection underwent liver biopsy and liver stiffness measurement (LSM) by transient elastography (Fibroscan(®) ; Echosens). Liver function was tested by routine biochemical examinations. Liver condition was assessed by haematoxylin and eosin (H&E) and Masson's trichrome staining. The expression of Pygo2 in liver tissue was measured by Real-time PCR and immunohistochemistry, respectively, while the serum levels of Pygo2 were detected by ELISA. The relationship between degree of liver fibrosis and Pygo2 expression was assessed by correlation analysis. Receiver operating characteristics (ROC) analysis was used to evaluate diagnostic accuracy of serum Pygo2, LSM and their combination. RESULTS The mRNA and protein levels of Pygo2 in HBV-infected patients were all higher than in normal persons (P < 0.05 respectively). Moreover, Pygo2 expression increased along with the progression of liver fibrosis (P < 0.05 respectively). The trend of serum Pygo2 agreed with its expression in liver tissue. The combination of serum Pygo2 and LSM had a significantly higher area under the curve than Pygo2 or LSM alone (P < 0.05 respectively). CONCLUSIONS This study suggested that Pygo2 was involved in HBV-induced liver fibrogenesis. Pygo2 is a valuable biomarker for the evaluation of fibrosis in HBV-infected patients.
Collapse
Affiliation(s)
- Wenting Li
- Department of Infectious Diseases, Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Chuanlong Zhu
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yuanbo Wu
- Department of Neurology, Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zheng Wang
- Department of Respiratory and Critical Medicine, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chuanwu Zhu
- Department of Hepatology, The Fifth People's Hospital of Suzhou, Suzhou, Jiangsu Province, China
| |
Collapse
|
21
|
Wang XR, Lu YH, Zheng X, Xu WY, Chen YH. Effect of Yinshao powder on expression of transforming growth factor-β1 and β-catenin in hepatic fibrosis in rats. Shijie Huaren Xiaohua Zazhi 2015; 23:2697-2706. [DOI: 10.11569/wcjd.v23.i17.2697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of traditional Chinese medicine Yinshao powder on the expression of transforming growth factor-β1 (TGF-β1) and β-catenin in hepatic fibrosis in rats to clarify the molecular mechanism of clearing heat, expelling damp and promoting blood circulation.
METHODS: Sixty adult male Wistar rats were divided randomly into six groups: a normal control group, a model group, low-, medium- and high-dose Yinshao powder groups and a colchicine group. Hepatic fibrosis was induced by giving subcutaneous injection of carbon tetrachloride, a high-fat diet and 30 mL/L alcohol for 16 wk. At the fifth week, Yinshao powder or colchicine was intragastrically administered. After 12 wk of treatment, the rats were sacrificed to measure serum levels of hyaluronidase (HA), type Ⅳ collagen, procollagen type Ⅲ (PCⅢ), and laminin (LN), observe hepatic pathological changes by HE staining and Masson staining, and determine the mRNA and protein expression levels of TGF-β1 and β-catenin by RT-PCR and Western blot.
RESULTS: Compared with the colchicine group and model group, serum levels of HA, type Ⅳ collagen, PCⅢ, and LN declined significantly in the other groups. The degree of hepatic fibrosis in the Yinshao powder groups was less than that in the colchicine group and model group. The mRNA and protein expression levels of TGF-β1 and β-catenin were significantly lower in the Yinshao powder groups than in the model group and colchicine group.
CONCLUSION: Clearing heat, expelling damp and promoting blood circulation with Yinshao powder for intervention of hepatic fibrosis may be via molecular mechanisms associated with inhibiting or down-regulating the expression of TGF-β1 and β-catenin and blocking the activation of TGF-β1 signaling pathway and Wnt/β-catenin signaling pathway.
Collapse
|
22
|
The WNT signaling pathway contributes to dectin-1-dependent inhibition of Toll-like receptor-induced inflammatory signature. Mol Cell Biol 2014; 34:4301-14. [PMID: 25246634 DOI: 10.1128/mcb.00641-14] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Macrophages regulate cell fate decisions during microbial challenges by carefully titrating signaling events activated by innate receptors such as dectin-1 or Toll-like receptors (TLRs). Here, we demonstrate that dectin-1 activation robustly dampens TLR-induced proinflammatory signature in macrophages. Dectin-1 induced the stabilization of β-catenin via spleen tyrosine kinase (Syk)-reactive oxygen species (ROS) signals, contributing to the expression of WNT5A. Subsequently, WNT5A-responsive protein inhibitors of activated STAT (PIAS-1) and suppressor of cytokine signaling 1 (SOCS-1) mediate the downregulation of IRAK-1, IRAK-4, and MyD88, resulting in decreased expression of interleukin 12 (IL-12), IL-1β, and tumor necrosis factor alpha (TNF-α). In vivo activation of dectin-1 with pathogenic fungi or ligand resulted in an increased bacterial burden of Mycobacteria, Klebsiella, Staphylococcus, or Escherichia, with a concomitant decrease in TLR-triggered proinflammatory cytokines. All together, our study establishes a new role for dectin-1-responsive inhibitory mechanisms employed by virulent fungi to limit the proinflammatory environment of the host.
Collapse
|
23
|
Ma ZG, Chen L, Zhan LL, Lv XP. Regulation of activation and proliferation of hepatic stellate cells through the Wnt signaling pathway: Implications for treatment of liver fibrosis. Shijie Huaren Xiaohua Zazhi 2014; 22:3048-3055. [DOI: 10.11569/wcjd.v22.i21.3048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis, a reversible pathological condition occurring after chronic liver injury, is closely associated with the activation and proliferation of hepatic stellate cells (HSCs). Several studies have demonstrated that the Wnt signaling pathway is involved in the activation of HSCs, and then contributes to the occurrence and development of liver fibrosis. Thus, the Wnt signaling pathway and HSCs are considered to be the valid targets for the treatment of liver fibrosis. In recent years, domestic and international scholars have carried on many studies on the regulation of the Wnt signaling pathway and HSCs, trying to find out effective antifibrotic therapies. A large number of studies have shown that regulating HSCs can play a positive role in the treatment of liver fibrosis through the Wnt signaling pathway. On the basis of the relationship among the Wnt signaling pathway, HSCs and liver fibrosis, this review mainly summarizes the latest progress in the study of strategies for regulating HSCs through the Wnt signaling pathway and discusses the implications for the treatment of liver fibrosis.
Collapse
|
24
|
Acetaldehyde targets superoxide dismutase 2 in liver cancer cells inducing transient enzyme impairment and a rapid transcriptional recovery. Food Chem Toxicol 2014; 69:102-8. [DOI: 10.1016/j.fct.2014.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/18/2014] [Accepted: 04/02/2014] [Indexed: 01/03/2023]
|
25
|
Cui L, Jia X, Zhou Q, Zhai X, Zhou Y, Zhu H. Curcumin affects β-catenin pathway in hepatic stellate cell in vitro and in vivo. ACTA ACUST UNITED AC 2014; 66:1615-22. [PMID: 24945564 DOI: 10.1111/jphp.12283] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 05/15/2014] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Emerging evidence indicates that Wnt/β-catenin pathway is linked to the fibrosis of different organs including liver fibrosis. β-Catenin promotes hepatic stellate cells (HSCs) activation, a key event in the development of liver fibrosis, and has emerged as a novel mediator of fibrosis. Curcumin, a natural active ingredient derived from turmeric, possesses an inhibitory effect on liver fibrosis. This study is aimed to examine whether curcumin affects β-catenin expression/activity in HSCs and explores the underlying mechanisms. METHODS The researchers used Western blot, real-time PCR, transfection assay and electrophoretic mobility shift assay and employed cultured HSCs and rat model of liver injury. KEY FINDINGS Results showed that curcumin could reduce β-catenin protein level in HSCs in vitro and in vivo. Both β-catenin transactivation activity and DNA-binding activity were suppressed by curcumin. Moreover, nuclear β-catenin protein level was decreased by curcumin treatment. Further experiments suggested that delta-like homologue 1 contributed to curcumin inhibition of β-catenin transactivation activity in cultured HSCs. CONCLUSIONS Curcumin affects β-catenin pathway in HSCs and might suggest a possible new explanation for the effects of curcumin on HSC activation and liver fibrosis.
Collapse
Affiliation(s)
- Lei Cui
- Department of Radiology, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | | | | | | | | | | |
Collapse
|