1
|
Hu Y, Chen H, Yang M, Xu J, Liu J, He Q, Xu X, Ji Z, Yang Y, Yan M, Zhang H. Hepatocyte growth factor facilitates the repair of spinal cord injuries by driving the chemotactic migration of mesenchymal stem cells through the β-catenin/TCF4/Nedd9 signaling pathway. Stem Cells 2024; 42:957-975. [PMID: 39269318 DOI: 10.1093/stmcls/sxae055] [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: 03/01/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024]
Abstract
Transplanted mesenchymal stem cells (MSCs) can significantly aid in repairing spinal cord injuries (SCIs) by migrating to and settling at the injury site. However, this process is typically inefficient, as only a small fraction of MSCs successfully reach the target lesion area. During SCI, the increased expression and secretion of hepatocyte growth factor (HGF) act as a chemoattractant that guides MSC migration. Nonetheless, the precise mechanisms by which HGF influences MSC migration are not fully understood. This study focused on unraveling the molecular pathways that drive MSC migration toward the SCI site in response to HGF. It was found that HGF can activate β-catenin signaling in MSCs by either phosphorylating LRP6, suppressing GSK3β phosphorylation through the AKT and ERK1/2 pathways, or enhancing the expression and nuclear translocation of TCF4. This activation leads to elevated Nedd9 expression, which promotes focal adhesion formation and F-actin polymerization, facilitating chemotactic migration. Transplanting MSCs during peak HGF expression in injured tissues substantially improves nerve regeneration, reduces scarring, and enhances hind limb mobility. Additionally, prolonging HGF release can further boost MSC migration and engraftment, thereby amplifying regenerative outcomes. However, inhibiting HGF/Met or interfering with β-catenin or Nedd9 signaling significantly impairs MSC engraftment, obstructing tissue repair and functional recovery. Together, these findings provide a theoretical basis and practical strategy for MSC transplantation therapy in SCI, highlighting the specific molecular mechanisms by which HGF regulates β-catenin signaling in MSCs, ultimately triggering their chemotactic migration.
Collapse
Affiliation(s)
- Ya'nan Hu
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, People's Republic of China
| | - Huanhuan Chen
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, People's Republic of China
- Clinical Medicine Research Center, The Suqian Clinical College of Xuzhou Medical University, Suqian 223800, People's Republic of China
| | - Min Yang
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, People's Republic of China
| | - Jianwei Xu
- Department of Pharmacology, School of Basic Medicine, Guizhou Medical University, Guiyang 550004, People's Republic of China
| | - Jinming Liu
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, People's Republic of China
| | - Qisheng He
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, People's Republic of China
| | - Xiaojing Xu
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, People's Republic of China
| | - Zhongqing Ji
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Ying Yang
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, People's Republic of China
| | - Mengwen Yan
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, People's Republic of China
| | - Huanxiang Zhang
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, People's Republic of China
| |
Collapse
|
2
|
Sang Y, Qiao L. Lung epithelial-endothelial-mesenchymal signaling network with hepatocyte growth factor as a hub is involved in bronchopulmonary dysplasia. Front Cell Dev Biol 2024; 12:1462841. [PMID: 39291265 PMCID: PMC11405311 DOI: 10.3389/fcell.2024.1462841] [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: 07/10/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is fundamentally characterized by the arrest of lung development and abnormal repair mechanisms, which result in impaired development of the alveoli and microvasculature. Hepatocyte growth factor (HGF), secreted by pulmonary mesenchymal and endothelial cells, plays a pivotal role in the promotion of epithelial and endothelial cell proliferation, branching morphogenesis, angiogenesis, and alveolarization. HGF exerts its beneficial effects on pulmonary vascular development and alveolar simplification primarily through two pivotal pathways: the stimulation of neovascularization, thereby enriching the pulmonary microvascular network, and the inhibition of the epithelial-mesenchymal transition (EMT), which is crucial for maintaining the integrity of the alveolar structure. We discuss HGF and its receptor c-Met, interact with various growth factors throughout the process of lung development and BPD, and form a signaling network with HGF as a hub, which plays the pivotal role in orchestrating and integrating epithelial, endothelial and mesenchymal.
Collapse
Affiliation(s)
- Yating Sang
- Pediatric Intensive Care Unit, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Lina Qiao
- Pediatric Intensive Care Unit, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Chen H, Luo Y, Zhu Y, Ye Y, Chen D, Song X, Xiao Z, Liu M, Li S. Enhanced secretion of hepatocyte growth factor in human umbilical cord mesenchymal stem cells ameliorates pulmonary fibrosis induced by bleomycin in rats. Front Pharmacol 2023; 13:1070736. [PMID: 36726784 PMCID: PMC9885268 DOI: 10.3389/fphar.2022.1070736] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/29/2022] [Indexed: 01/09/2023] Open
Abstract
Umbilical cord mesenchymal stem cells (UCMSCs) are a reportedly promising choice in the treatment of irreversible pulmonary fibrosis and lethal interstitial lung disease with limited drug treatment options. In this study, we investigated the therapeutic efficacy of UCMSCs overexpressing hepatocyte growth factor (HGF), which is considered one of the main anti-fibrotic factors secreted by MSCs. Adenovirus vector carrying the HGF gene was transfected into UCMSCs to produce HGF-modified UCMSCs (HGF-UCMSCs). Transfection promoted the proliferation of UCMSCs and did not change the morphology, and differentiation ability, or biomarkers. Rats were injected with HGF-UCMSCs on days 7 and 11 after intratracheal administration of bleomycin (10 mg/kg). We performed an analysis of histopathology and lung function to evaluate the anti-fibrotic effect. The results showed that HGF-UCMSCs decreased the Ashcroft scores in hematoxylin and eosin-stained sections, the percentage positive area in Masson trichrome-stained sections, and the hydroxyproline level in lungs. Forced expiratory volume in the first 300 m/forced vital capacity was also improved by HGF-UCMSCs. To explore the possible therapeutic mechanism of HGF-UCMSCs, we detected inflammatory factors in the lungs and performed mRNA sequencing in UCMSCs and HGF-UCMSCs. The data indicated that inhibition of interleukin-17 in the lung may be related to the anti-fibrosis of HGF-UCMSCs, and overexpressed HGF probably played a primary role in the treatment. Collectively, our study findings suggested that the overexpression of HGF may improve the anti-fibrotic effect of UCMSCs through directly or indirectly interacting with interleukin-17-producing cells in fibrotic lungs.
Collapse
Affiliation(s)
- Huanjie Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yulong Luo
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiping Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yongshun Ye
- Huizhou Municipal Central Hospital, Guangzhou, Guangdong, China
| | - Difei Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xinyu Song
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhulin Xiao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ming Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China,*Correspondence: Ming Liu, ; Shiyue Li,
| | - Shiyue Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China,*Correspondence: Ming Liu, ; Shiyue Li,
| |
Collapse
|
4
|
Antifibrotic therapy by sustained release of low molecular weight heparin from poly(lactic-co-glycolic acid) microparticles on bleomycin-induced pulmonary fibrosis in mice. Sci Rep 2020; 10:19019. [PMID: 33149192 PMCID: PMC7642430 DOI: 10.1038/s41598-020-76034-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/20/2020] [Indexed: 12/01/2022] Open
Abstract
Heparin and low molecular weight heparin (LMWH) have recently been considered useful treatment tools for inflammation. Heparin has antifibrotic activity, mediated by cellular secretion of hepatocyte growth factor (HGF). HGF has antifibrotic properties demonstrated in experimental models of lung, kidney, heart, skin, and liver fibrosis. The ability of LMWH for HGF secretion is similar to that of normal heparin. Poly (lactic-co-glycolic acid) (PLGA) is widely used for sustained drug release, because of its biocompatibility and low toxicity. LMWH-loaded PLGA microparticles are prepared by a conventional water-in-oil-in-water emulsion method. Interstitial pneumonia is a life-threatening pathological condition that causes respiratory failure when it progresses. In the present study, we investigated the therapeutic effect of LMWH-loaded PLGA microparticles in a mouse model of bleomycin-induced lung fibrosis. The ratios of fibrotic area to total area were significantly lower in mice administered LMWH-loaded microparticles than in mice administered bleomycin alone. The microparticle administration did not further enhance the gene expression for inflammatory cytokines. In a cell culture study, HGF secretion by mouse and human lung fibroblasts was significantly increased by LMWH addition. We conclude that LMWH showed anti-inflammatory activity, through the effects of LMWH-loaded PLGA microparticles on cells at sites of inflammation.
Collapse
|
5
|
Li J, Liu X, Sha M, Li Y. The balance between HGF and TGF-β1 acts as a switch in the tissue remodeling of chronic rhinosinusitis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:933-940. [PMID: 31933903 PMCID: PMC6945174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/21/2018] [Indexed: 06/10/2023]
Abstract
OBJECTIVE The remodeling patterns in different types of chronic rhinosinusitis (CRS) are controversial. This study aimed to investigate the roles of transforming growth factor-β1 (TGF-β1) and hepatocyte growth factor (HGF) in the CRS remodeling process. METHODS Surgical samples were obtained from CRSwNP patients (n=29), CRSsNP patients (n=34), and controls (n=21). Collagen deposition was detected via Masson trichrome (MT) staining. Immunohistochemical staining was performed to examine the protein expression of α-smooth muscle actin (α-SMA). The expression of TGF-β1 and HGF was measured by ELISA. The relationship between the rate of TGF-β1/HGF and the expression of Collagen III and α-SMA was analyzed using a Pearson correlation test. Primary nasal epithelial cells (NECs) were cultured and divided into four groups. Collagen III secretion was measured in the supernatants by ELISA. The expression of α-SMA was studied by immunofluorescence. RESULTS Reduced collagen deposition and α-SMA expression were detected in the CRSwNP group (P=0.033). The expression of collagen deposition and α-SMA was increased in the CRSwNP group (P=0.001). The ELISA tests indicated that TGF-β1 levels were significantly increased in the CRSsNP compared with the controls. The expression of HGF was higher in the CRSwNP than in the other two groups. The ratio of TGF-β1/HGF was upregulated in CRSsNP and was correlated positively with collagen and α-SMA expression (P<0.05, R=0.762). TGF-β1 can increase collagen and α-SMA expression in NECs, and HGF can antagonize the remodeling action of TGF-β1. CONCLUSION Distinct remodeling patterns are revealed for different types of CRS. The balance of TGF-β1 and HGF is important in the CRS remodeling process.
Collapse
Affiliation(s)
- Jing Li
- Department of Otolaryngology, Affiliated Hangzhou First People’s Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, China
| | - Xiang Liu
- Department of Otolaryngology, Affiliated Hangzhou First People’s Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, China
| | - Min Sha
- Department of Otolaryngology, Hangzhou Children’s HospitalHangzhou, Zhejiang Province, China
| | - Yong Li
- Department of Otolaryngology, Affiliated Hangzhou First People’s Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, China
| |
Collapse
|
6
|
Atanelishvili I, Shirai Y, Akter T, Noguchi A, Ash KT, Misra S, Ghatak S, Silver RM, Bogatkevich GS. D1398G Variant of MET Is Associated with Impaired Signaling of Hepatocyte Growth Factor in Alveolar Epithelial Cells and Lung Fibroblasts. PLoS One 2016; 11:e0162357. [PMID: 27584154 PMCID: PMC5008815 DOI: 10.1371/journal.pone.0162357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 08/22/2016] [Indexed: 02/07/2023] Open
Abstract
Pulmonary fibrosis represents the terminal stage of a diverse group of lung diseases including scleroderma associated interstitial lung disease. The molecular mechanisms underlying the pathogenesis of lung fibrosis are not well understood and there is a great need for more effective treatment for this lethal disease. We recently discovered a small fragment of hepatocyte growth factor (HGF) receptor MET as a peptide designated “M10,” with strong antifibrotic properties. Furthermore, we showed that aspartic acid at position 1398 of MET is essential for M10 generation. The current study was undertaken to investigate the D1398G variant of MET in which aspartic acid at position 1398 was mutated to glycine resulting in loss of M10. We demonstrate that lung fibroblasts, A549, and primary alveolar epithelial cells (AEC) expressing D1398G MET exhibit reduced auto-phosphorylation on tyrosine residues and reduced activation of Ras and MAPK. HGF treatment of scleroderma lung fibroblasts as well as HGF treatment of TGFβ-treated normal lung fibroblasts transfected with wild type MET is associated with decreased collagen, connective tissue growth factor (CTGF, CCN2) and smooth muscle α-actin (SMA). However, HGF has no such effects in cells transfected with MET D1398G. Cisplatin- and FasL-induced apoptosis is significantly reduced in AEC transfected with MET wild type, but not in AEC transfected with MET D1398G. We conclude that the D1398G variant of MET is associated with compromised phosphorylation and impaired HGF signaling in lung fibroblasts and AEC, two cell types implicated in the pathogenesis of pulmonary fibrosis associated with scleroderma. Ongoing studies will explore the frequency of this variant and its relationship to pulmonary outcomes in scleroderma patients.
Collapse
Affiliation(s)
- Ilia Atanelishvili
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Yuichiro Shirai
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Allergy and Rheumatology, Nippon Medical School, Tokyo, Japan
| | - Tanjina Akter
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Atsushi Noguchi
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Kurt T. Ash
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Suniti Misra
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Sibnath Ghatak
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Richard M. Silver
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Galina S. Bogatkevich
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
7
|
Mungunsukh O, Lee YH, Bottaro DP, Day RM. The hepatocyte growth factor isoform NK2 activates motogenesis and survival but not proliferation due to lack of Akt activation. Cell Signal 2016; 28:1114-23. [PMID: 27224506 DOI: 10.1016/j.cellsig.2016.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/16/2016] [Indexed: 12/14/2022]
Abstract
Hepatocyte growth factor (HGF) is a pleiotrophic factor involved in cellular proliferation, migration and morphogenesis. HGF is required for normal tissue and organ development during embryogenesis, but in the adult HGF has been demonstrated to drive normal tissue repair and inhibit fibrotic remodeling. HGF has two naturally occurring human isoforms as a result of alternative splicing, NK1 and NK2. While NK1 has been defined as an agonist for HGF receptor, Met, NK2 is defined as a partial Met antagonist. Furthermore, under conditions of fibrotic remodeling, NK2 is still expressed while full length HGF is suppressed. Furthermore, the mechanism by which NK2 partially signals through Met is not completely understood. Here, we investigated the mitogenic, motogenic, and anti-apoptotic activities of NK2 compared with full length HGF in primary human bronchial epithelial cells (BEpC) and bovine pulmonary artery endothelial cells (PAEC). In human BEpC, NK2 partial activated Met, inducing Met phosphorylation at Y1234/1235 in the tyrosine-kinase domain but not at Y1349 site in the multifunctional docking domain. Partial phosphorylation of Met by NK2 resulted in activation of MAPK and STAT3, but not AKT. This correlated with motogenesis and survival in a MAPK-dependent manner, but not cell proliferation. Overexpression of a constitutively active AKT complemented NK2 signaling, allowing NK2 to induce cell proliferation. These data indicate that NK2 and HGF drive motogenic and anti-apoptotic signaling but only HGF drives cell proliferation by activating AKT-pathway signaling. These results have implications for the biological consequences of differential regulation of the two isoforms under pro-fibrotic conditions.
Collapse
Affiliation(s)
- Ognoon Mungunsukh
- The Uniformed Services University of the Health Sciences, Department of Pharmacology, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Young H Lee
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Donald P Bottaro
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Regina M Day
- The Uniformed Services University of the Health Sciences, Department of Pharmacology, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| |
Collapse
|
8
|
Xia Y, Xia Y, Lv Q, Yue M, Qiao S, Yang Y, Wei Z, Dai Y. Madecassoside ameliorates bleomycin-induced pulmonary fibrosis in mice through promoting the generation of hepatocyte growth factor via PPAR-γ in colon. Br J Pharmacol 2016; 173:1219-35. [PMID: 26750154 PMCID: PMC5341335 DOI: 10.1111/bph.13421] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 01/04/2016] [Accepted: 01/07/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Madecassoside has potent anti-pulmonary fibrosis (PF) effects when administered p.o., despite having extremely low oral bioavailability. Herein, we explored the mechanism of this anti-PF effect with regard to gut hormones. EXPERIMENTAL APPROACH A PF model was established in mice by intratracheal instillation of bleomycin. Haematoxylin and eosin stain and Masson's trichrome stain were used to assess histological changes in the lung. Quantitative-PCR and Western blot detected mRNA and protein levels, respectively, and cytokines were measured by ELISA. Small interfering RNA was used for gene-silencing. EMSA was applied to detect DNA-binding activity. KEY RESULTS Administration of madecassoside, p.o., but not its main metabolite madecassic acid, exhibited a direct anti-PF effect in mice. However, i.p. madecassoside had no anti-PF effect. Madecassoside increased the expression of hepatocyte growth factor (HGF) in colon tissues, and HGF receptor antagonists attenuated its anti-PF effect. Madecassoside facilitated the secretion of HGF from colonic epithelial cells by activating the PPAR-γ pathway, as shown by an up-regulation of PPAR-γ mRNA expression, nuclear translocation and DNA-binding activity both in vitro and in vivo. Also GW9662, a selective PPAR-γ antagonist, almost completely prevented the madecassoside-induced increased expression of HGF and amelioration of PF. CONCLUSIONS AND IMPLICATIONS The potent anti-PF effects induced by p.o. madecassoside in mice are not mediated by its metabolites or itself after absorption into blood. Instead, madecassoside increases the activity of PPAR-γ, which subsequently increases HGF expression in colonic epithelial cells. HGF then enters into the circulation and lung tissue to exert an anti-PF effect.
Collapse
Affiliation(s)
- Ying Xia
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Department of Pharmacology of Chinese Materia MedicaChina Pharmaceutical University24 Tong Jia XiangNanjing210009China
| | - Yu‐Feng Xia
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Department of Pharmacology of Chinese Materia MedicaChina Pharmaceutical University24 Tong Jia XiangNanjing210009China
| | - Qi Lv
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Department of Pharmacology of Chinese Materia MedicaChina Pharmaceutical University24 Tong Jia XiangNanjing210009China
| | - Meng‐Fan Yue
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Department of Pharmacology of Chinese Materia MedicaChina Pharmaceutical University24 Tong Jia XiangNanjing210009China
| | - Si‐Miao Qiao
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Department of Pharmacology of Chinese Materia MedicaChina Pharmaceutical University24 Tong Jia XiangNanjing210009China
| | - Yan Yang
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Department of Pharmacology of Chinese Materia MedicaChina Pharmaceutical University24 Tong Jia XiangNanjing210009China
| | - Zhi‐Feng Wei
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Department of Pharmacology of Chinese Materia MedicaChina Pharmaceutical University24 Tong Jia XiangNanjing210009China
| | - Yue Dai
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Department of Pharmacology of Chinese Materia MedicaChina Pharmaceutical University24 Tong Jia XiangNanjing210009China
| |
Collapse
|
9
|
Li F, Qu H, Cao HC, Li MH, Chen C, Chen XF, Yu B, Yu L, Zheng LM, Zhang W. Both FOXO3a and FOXO1 are involved in the HGF-protective pathway against apoptosis in endothelial cells. Cell Biol Int 2015; 39:1131-7. [PMID: 25952685 DOI: 10.1002/cbin.10486] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/29/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Fang Li
- Biomedical Research Institute; Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center; Shenzhen Guangdong China
| | - Huan Qu
- Department of Cardiovascularology; Peking University Shenzhen Hospital; Shenzhen Guangdong China
| | - Heng-Chang Cao
- Department of Emergency Surgery; Peking University Shenzhen Hospital; Shenzhen Guangdong China
| | - Mei-Hong Li
- Department of Cardiovascularology; Peking University Shenzhen Hospital; Shenzhen Guangdong China
| | - Chen Chen
- Department of Cardiovascularology; Peking University Shenzhen Hospital; Shenzhen Guangdong China
| | - Xiao-Fan Chen
- Biomedical Research Institute; Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center; Shenzhen Guangdong China
| | - Bo Yu
- Department of Dermatology; Peking University Shenzhen Hospital; Shenzhen Guangdong China
| | - Lin Yu
- Department of obstetrics and gynaecology; Peking University Shenzhen Hospital; Shenzhen Guangdong China
| | - Le-Min Zheng
- Department of Cardiovascular Sciences; Peking University Health Science Center; Peking University; Peking China
| | - Wei Zhang
- Biomedical Research Institute; Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center; Shenzhen Guangdong China
- Department of Dermatology; Peking University Shenzhen Hospital; Shenzhen Guangdong China
| |
Collapse
|
10
|
Co-targeting c-Met and COX-2 leads to enhanced inhibition of lung tumorigenesis in a murine model with heightened airway HGF. J Thorac Oncol 2015; 9:1285-93. [PMID: 25057941 DOI: 10.1097/jto.0000000000000245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND The hepatocyte growth factor (HGF)/c-Met pathway is often dysregulated in non-small-cell lung cancer (NSCLC). HGF activation of c-Met induces cyclooxygenase-2 (COX-2), resulting in downstream stimulation by prostaglandin E2 of additional pathways. Targeting both c-Met and COX-2 might lead to enhanced antitumor effects by blocking signaling upstream and downstream of c-Met. METHODS Effects of crizotinib or celecoxib alone or in combination were tested in NSCLC cells in vitro and in mice transgenic for airway expression of human HGF. RESULTS Proliferation and invasion of NSCLC cells treated with a combination of crizotinib and celecoxib were significantly lower compared with single treatments. Transgenic mice showed enhanced COX-2 expression localized to preneoplastic areas following exposure to the tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, which was not present without carcinogen exposure. This shows that COX-2 activity is present during lung tumor development in a high HGF environment. After 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone treatment, a significant decrease in the number of lung tumors per animal was observed after 13-week treatments of crizotinib, celecoxib, or the combination compared with placebo (p < 0.001). With combination treatment, the number of tumors was also significantly lower than single agent treatment (p < 0.001). In the resulting lung tumors, P-c-Met, COX-2, prostaglandin E2, and P-MAPK were significantly downmodulated by combination treatment compared with single treatment. Expression of the epithelial-mesenchymal transition markers E-cadherin and snail was also modulated by combination treatment. CONCLUSIONS In the presence of high HGF, dual inhibition of c-Met and COX-2 may enhance antitumor effects. This combination may have clinical potential in NSCLCs with high HGF/c-Met expression or epithelial-mesenchymal transition phenotype.
Collapse
|
11
|
Dong LH, Jiang YY, Liu YJ, Cui S, Xia CC, Qu C, Jiang X, Qu YQ, Chang PY, Liu F. The anti-fibrotic effects of mesenchymal stem cells on irradiated lungs via stimulating endogenous secretion of HGF and PGE2. Sci Rep 2015; 5:8713. [PMID: 25736907 PMCID: PMC4348621 DOI: 10.1038/srep08713] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 02/02/2015] [Indexed: 12/11/2022] Open
Abstract
Radiation-induced pulmonary fibrosis is a common disease and has a poor prognosis owing to the progressive breakdown of gas exchange regions in the lung. Recently, a novel strategy of administering mesenchymal stem cells for pulmonary fibrosis has achieved high therapeutic efficacy. In the present study, we attempted to use human adipose tissue-derived mesenchymal stem cells to prevent disease in Sprague-Dawley rats that received semi-thoracic irradiation (15 Gy). To investigate the specific roles of mesenchymal stem cells in ameliorating radiation-induced pulmonary fibrosis, we treated control groups of irradiated rats with human skin fibroblasts or phosphate-buffered saline. After mesenchymal stem cells were infused, host secretions of hepatocyte growth factor (HGF) and prostaglandin E2 (PGE2) were elevated compared with those of the controls. In contrast, tumour necrosis factor-alpha (TNF-α) and transforming growth factor-beta1 (TGF-β1) levels were decreased after infusion of mesenchymal stem cells. Consequently, the architecture of the irradiated lungs was preserved without marked activation of fibroblasts or collagen deposition within the injured sites. Moreover, mesenchymal stem cells were able to prevent the irradiated type II alveolar epithelial cells from undergoing epithelial-mesenchymal transition. Collectively, these data confirmed that mesenchymal stem cells have the potential to limit pulmonary fibrosis after exposure to ionising irradiation.
Collapse
Affiliation(s)
- Li-Hua Dong
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Yi-Yao Jiang
- Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Tianjin 300000, China
| | - Yong-Jun Liu
- Tianjin Allian Stem Cell Techonology CO., LTD, Tianjin 300308, China
| | - Shuang Cui
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Cheng-Cheng Xia
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Chao Qu
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Xin Jiang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Ya-Qin Qu
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Peng-Yu Chang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Feng Liu
- Nephrology department, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
12
|
Liu Y, Liu JH, Chai K, Tashiro SI, Onodera S, Ikejima T. Inhibition of c-Met promoted apoptosis, autophagy and loss of the mitochondrial transmembrane potential in oridonin-induced A549 lung cancer cells. ACTA ACUST UNITED AC 2013; 65:1622-42. [PMID: 24102522 DOI: 10.1111/jphp.12140] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 08/02/2013] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Herein, inhibition of hepatocyte growth factor receptor, c-Met, significantly increased cytochrome c release and Bax/Bcl-2 ratio, indicating that c-Met played an anti-apoptotic role. The following experiments are to elucidate this anti-apoptotic mechanism, then the effect of c-Met on autophagy has also been discussed. METHODS Investigated was the influence of c-Met on apoptosis, autophagy and loss of mitochondrial transmembrane potential (Δψm), and the relevant proteins were examined. KEY FINDINGS First, we found that activation of extracellular signal-regulated kinase (ERK), p53 was promoted by c-Met interference. Subsequent studies indicated that ERK was the upstream effector of p53, and this ERK-p53 pathway mediated release of cytochrome c and up-regulation of Bax/Bcl-2 ratio. Secondly, the inhibition of c-Met augmented oridonin-induced loss of mitochondrial transmembrane potential (Δψm), resulting apoptosis. Finally, the inhibition of c-Met increased oridonin-induced A549 cell autophagy accompanied by Beclin-1 activation and conversion from microtubule-associated protein light chain 3 (LC3)-I to LC3-II. Activation of ERK-p53 was also detected in autophagy process and could be augmented by inhibition of c-Met. Moreover, suppression of autophagy by 3-methyladenine (3-MA) or small interfering RNA against Beclin-1 or Atg5 decreased oridonin-induced apoptosis. Inhibition of apoptosis by pan-caspase inhibitor (z-VAD-fmk) decreased oridonin-induced autophagy as well and Loss of Δψm also occurred during autophagic process. CONCLUSION Thus, inhibiting c-Met enhanced oridonin-induced apoptosis, autophagy and loss of Δψm in A549 cells.
Collapse
Affiliation(s)
- Ying Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China; China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, China
| | | | | | | | | | | |
Collapse
|
13
|
Rampanelli E, Rouschop K, Teske GJD, Claessen N, Leemans JC, Florquin S. CD44v3-v10 reduces the profibrotic effects of TGF-β1 and attenuates tubular injury in the early stage of chronic obstructive nephropathy. Am J Physiol Renal Physiol 2013; 305:F1445-54. [PMID: 24026183 DOI: 10.1152/ajprenal.00340.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
CD44 family members are cell surface glycoproteins, which are expressed on tubular epithelial cells (TEC) solely upon kidney injury and are involved in renal fibrosis development. Renal interstitial fibrosis is the final manifestation of chronic kidney diseases and is regulated by a complex network of cytokines, including the profibrotic factor transforming growth factor-β1 (TGF-β1) and the two antifibrotic cytokines bone morphogenic protein-7 (BMP-7) and hepatocyte growth factor (HGF). The present study investigates the potential role of CD44 standard (CD44s) and CD44v3-v10 (CD44v3) isoforms as modulators of the balance between TGF-β1 and HGF/BMP-7. CD44s is the shortest and most common isoform. CD44v3-v10 (CD44v3) has heparan sulfate moieties, which enable the binding to HGF/BMP-7, and hence, might exert renoprotective effects. Using transgenic mice overexpressing either CD44s or CD44v3 specifically on proximal TEC, we found that in vitro the overexpression of CD44v3 on primary TEC renders cells less susceptible to TGF-β1 profibrotic actions and more sensitive to BMP-7 and HGF compared with TEC overexpressing CD44s. One day after unilateral ureteric obstruction, obstructed kidneys from CD44v3 transgenic mice showed less tubular damage and myofibroblasts accumulation, which was associated with decreased TGF-β1 signaling and increased BMP-7 synthesis and signaling compared with kidneys from wild-type and CD44s transgenic mice. These data suggest that CD44v3 plays a renoprotective role in early stage of chronic obstructive nephropathy.
Collapse
Affiliation(s)
- Elena Rampanelli
- Dept. of Pathology, Rm. L2-112, Academic Medical Center, P.O. Box 22660, 1100 AZ, Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
14
|
Chakraborty S, Chopra P, Hak A, Dastidar SG, Ray A. Hepatocyte growth factor is an attractive target for the treatment of pulmonary fibrosis. Expert Opin Investig Drugs 2013; 22:499-515. [PMID: 23484858 DOI: 10.1517/13543784.2013.778972] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Pulmonary fibrosis (PF) is a progressive fatal disorder and is characterized by alveolar epithelial injury, myofibroblast proliferation, and extracellular matrix remodeling, resulting in irreversible distortion of lung's architecture. Available therapies are associated with side effects and show restricted efficacy. Therefore, there is an urgent need to find a therapeutic solution to PF. Therapeutic strategies interfering myofibroblast expansion, apoptosis of epithelial and endothelial cells might be beneficial for treatment of PF. Hepatocyte growth factor (HGF), a pleiotropic growth factor, plays an important role in lung development, inflammation, repair, and regeneration. In animal model of PF, administration of recombinant HGF protein or ectopic HGF expression ameliorates fibrosis. AREAS COVERED The focus of this review is to highlight HGF as a promising therapeutic approach for the treatment of PF. The review discusses the currently available treatment option for PF as well as highlights the possible beneficial effect of HGF as a drug target. EXPERT OPINION HGF with its anti-fibrotic effect provides a promising new therapeutic approach by protecting lung from fibrotic remodeling and also promoting normal regeneration of lung. The development of HGF mimetics may provide a potential attractive therapy for treatment of this devastating and complex disease.
Collapse
Affiliation(s)
- Sushmita Chakraborty
- Daiichi Sankyo Life Science Research Centre in India (RCI), Department of Biology, Haryana, India
| | | | | | | | | |
Collapse
|
15
|
Sakai N, Tager AM. Fibrosis of two: Epithelial cell-fibroblast interactions in pulmonary fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1832:911-21. [PMID: 23499992 PMCID: PMC4041487 DOI: 10.1016/j.bbadis.2013.03.001] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 03/03/2013] [Accepted: 03/04/2013] [Indexed: 12/12/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by the progressive and ultimately fatal accumulation of fibroblasts and extracellular matrix in the lung that distorts its architecture and compromises its function. IPF is now thought to result from wound-healing processes that, although initiated to protect the host from injurious environmental stimuli, lead to pathological fibrosis due to these processes becoming aberrant or over-exuberant. Although the environmental stimuli that trigger IPF remain to be identified, recent evidence suggests that they initially injure the alveolar epithelium. Repetitive cycles of epithelial injury and resultant alveolar epithelial cell death provoke the migration, proliferation, activation and myofibroblast differentiation of fibroblasts, causing the accumulation of these cells and the extracellular matrix that they synthesize. In turn, these activated fibroblasts induce further alveolar epithelial cell injury and death, thereby creating a vicious cycle of pro-fibrotic epithelial cell-fibroblast interactions. Though other cell types certainly make important contributions, we focus here on the "pas de deux" (steps of two), or perhaps more appropriate to IPF pathogenesis, the "folie à deux" (madness of two) of epithelial cells and fibroblasts that drives the progression of pulmonary fibrosis. We describe the signaling molecules that mediate the interactions of these cell types in their "fibrosis of two", including transforming growth factor-β, connective tissue growth factor, sonic hedgehog, prostaglandin E2, angiotensin II and reactive oxygen species. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Norihiko Sakai
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114
| | - Andrew M. Tager
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114
- Pulmonary and Critical Care Unit, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114
| |
Collapse
|
16
|
Abstract
Idiopathic pulmonary fibrosis is currently believed to be driven by alveolar epithelial cells, with abnormally activated alveolar epithelial cells accumulating in an attempt to repair injured alveolar epithelium (1). Thus, targeting the alveolar epithelium to prevent or inhibit the development of pulmonary fibrosis might be an interesting therapeutic option in this disease. Hepatocyte growth factor (HGF) is a growth factor for epithelial and endothelial cells, which is secreted by different cell types, especially fibroblasts and neutrophils. HGF has mitogenic, motogenic, and morphogenic properties and exerts an antiapoptotic action on epithelial and endothelial cells. HGF has also proangiogenic effect. In vitro, HGF inhibits epithelial-to-mesenchymal cell transition and promotes myofibroblast apoptosis. In vivo, HGF has antifibrotic properties demonstrated in experimental models of lung, kidney, heart, skin, and liver fibrosis. Hence, the modulation of HGF may be an attractive target for the treatment of lung fibrosis.
Collapse
|
17
|
Liu Y, Shi QF, Qi M, Tashiro SI, Onodera S, Ikejima T. Interruption of Hepatocyte Growth Factor Signaling Augmented Oridonin-Induced Death in Human Non-small Cell Lung Cancer A549 Cells via c-Met-Nuclear Factor-κB-Cyclooxygenase-2 and c-Met-Bcl-2-Caspase-3 Pathways. Biol Pharm Bull 2012; 35:1150-8. [DOI: 10.1248/bpb.b12-00197] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Ying Liu
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University
| | - Qi-Feng Shi
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University
| | - Min Qi
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University
| | - Shin-Ichi Tashiro
- Department of Clinical and Biomedical Sciences, Showa Pharmaceutical University
| | - Satoshi Onodera
- Department of Clinical and Biomedical Sciences, Showa Pharmaceutical University
| | - Takashi Ikejima
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University
| |
Collapse
|
18
|
Baarsma HA, Meurs H, Halayko AJ, Menzen MH, Schmidt M, Kerstjens HAM, Gosens R. Glycogen synthase kinase-3 regulates cigarette smoke extract- and IL-1β-induced cytokine secretion by airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2011; 300:L910-9. [PMID: 21421749 DOI: 10.1152/ajplung.00232.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is a constitutively active kinase that regulates multiple signaling proteins and transcription factors involved in inflammation. Its role in inflammatory lung diseases, including chronic obstructive pulmonary disease (COPD), is largely unknown. We investigated the role of GSK-3 in the secretion of chemokines and growth factors by human airway smooth muscle cells after exposure to cigarette smoke extract (CSE) or interleukin-1β (IL-1β), important factors involved in the development of COPD. Cultured human airway smooth muscle cells were exposed to increasing concentrations of CSE (1-15%) and IL-1β (0.01-1.0 ng/ml), which induced the secretion of VEGF-A and IL-8, whereas eotaxin secretion was induced by IL-1β only. Inhibition of GSK-3 by the selective inhibitor SB216763 or CHIR/CT99021 attenuated the cytokine and growth factor release induced by CSE and/or IL-1β, without affecting the basal release. Secretion of the cytokines by airway smooth muscle partially depends on NF-κB signaling, and GSK-3 has been implicated in regulating multiple steps in activating the NF-κB signaling pathway. IL-1β treatment induced degradation of the NF-κB inhibitory protein Iκ-Bα followed by nuclear translocation and DNA binding of p65 NF-κB, which were unaffected by inhibition of GSK-3. However, induction of NF-κB-dependent transcriptional activity by IL-1β and CSE was largely reduced upon GSK-3 inhibition by SB216763. Collectively, we demonstrate that CSE and IL-1β activate airway smooth muscle cells to secrete the proinflammatory cytokines IL-8, eotaxin, and VEGF-A. Furthermore, we show that GSK-3 regulates the release of these cytokines induced by CSE and IL-1β by promoting NF-κB-dependent gene transcription.
Collapse
Affiliation(s)
- Hoeke A Baarsma
- Department of Molecular Pharmacology, University of Groningen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
19
|
Barton-Pai A, Feleder C, Johnson A. Tumor necrosis factor-α induces increased lung vascular permeability: a role for GSK3α/β. Eur J Pharmacol 2011; 657:159-66. [PMID: 21316358 DOI: 10.1016/j.ejphar.2011.01.060] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2010] [Revised: 01/24/2011] [Accepted: 01/27/2011] [Indexed: 12/20/2022]
Abstract
We tested the hypothesis that glycogen synthase kinase 3α/β (GSK3α/β) modulates tumor necrosis factor-a (TNF) induced increased lung vascular permeability. Rats were treated with TNF (i.v., ~100ng/ml) or vehicle 0.5h, 4.0h and 24.0h prior to lung isolation. Rats were co-treated with the GSK3α/β inhibitors SB216763 (0.6mg/kg) or TDZD-8 (1.0mg/kg). After TNF, the isolated lung was assessed for hemodynamics, wet-dry/dry weight (W-D/D) and extravascular albumin. Extravascular albumin significantly increased at TNF-24h compared to Control. In the GSK3α/β-inhibited+TNF groups, extravascular albumin was similar to the Control and respective SB216763 and TDZD-8 groups. In separate studies, to assess GSK3α/β-activity, lung lysate was assessed for phospho-GSK3α/β-Ser(21/9), total GSK3α/β, un-phospho-β-catenin-Ser(33/37) and total β-catenin. In the TNF-4.0h group, there was no change in GSK3α/phospho-GSK3α-Ser(21) but there was an increase in GSK3β/GSK3β-Ser(9) compared to Control, indicating GSK3β activation at TNF-4.0h. GSK3β activation was verified because there was a decrease in un-phospho-β-catenin-Ser(33/37)/β-catenin in the TNF-4.0 group, a specific outcome for GSK3β activation. In the SB216763+TNF group, un-phospho-β-catenin-Ser(33/37) was similar to Control, indicating prevention of TNF-induced GSK3β activation. In the TNF-24h group, there were increases in the biomarkers of inflammation phospho-eNOS-Ser (1117) and oxidized protein, which did not occur in the SB216763+TNF-24h and TDZD-8+TNF-24h groups. In the SB216763+TNF-24h and TDZD-8+TNF-24h groups, un-phospho-β-catenin-Ser(33/37) was greater than in the Control, indicating continued inhibition of GSK3β. The data indicates that pharmacologic inhibition of GSK3β inhibits TNF induced increased endothelial permeability associated with lung inflammation.
Collapse
Affiliation(s)
- Amy Barton-Pai
- Pharmacy Practice, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, United States.
| | | | | |
Collapse
|
20
|
Panganiban RAM, Day RM. Hepatocyte growth factor in lung repair and pulmonary fibrosis. Acta Pharmacol Sin 2011; 32:12-20. [PMID: 21131996 DOI: 10.1038/aps.2010.90] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pulmonary remodeling is characterized by the permanent and progressive loss of the normal alveolar architecture, especially the loss of alveolar epithelial and endothelial cells, persistent proliferation of activated fibroblasts, or myofibroblasts, and alteration of extracellular matrix. Hepatocyte growth factor (HGF) is a pleiotropic factor, which induces cellular motility, survival, proliferation, and morphogenesis, depending upon the cell type. In the adult, HGF has been demonstrated to play a critical role in tissue repair, including in the lung. Administration of HGF protein or ectopic expression of HGF has been demonstrated in animal models of pulmonary fibrosis to induce normal tissue repair and to prevent fibrotic remodeling. HGF-induced inhibition of fibrotic remodeling may occur via multiple direct and indirect mechanisms including the induction of cell survival and proliferation of pulmonary epithelial and endothelial cells, and the reduction of myofibroblast accumulation.
Collapse
|
21
|
Panganiban RAM, Day RM. Hepatocyte growth factor in lung repair and pulmonary fibrosis. Int J Radiat Biol 2010; 89:656-67. [PMID: 21131996 DOI: 10.3109/09553002.2012.711502] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pulmonary remodeling is characterized by the permanent and progressive loss of the normal alveolar architecture, especially the loss of alveolar epithelial and endothelial cells, persistent proliferation of activated fibroblasts, or myofibroblasts, and alteration of extracellular matrix. Hepatocyte growth factor (HGF) is a pleiotropic factor, which induces cellular motility, survival, proliferation, and morphogenesis, depending upon the cell type. In the adult, HGF has been demonstrated to play a critical role in tissue repair, including in the lung. Administration of HGF protein or ectopic expression of HGF has been demonstrated in animal models of pulmonary fibrosis to induce normal tissue repair and to prevent fibrotic remodeling. HGF-induced inhibition of fibrotic remodeling may occur via multiple direct and indirect mechanisms including the induction of cell survival and proliferation of pulmonary epithelial and endothelial cells, and the reduction of myofibroblast accumulation.
Collapse
Affiliation(s)
- Ronald Allan M Panganiban
- Department of Pharmacology, Uniformed Services University of Health Sciences, Bethesda, MD 20852, USA
| | | |
Collapse
|
22
|
Lee YH, Marquez AP, Mungunsukh O, Day RM. Hepatocyte growth factor inhibits apoptosis by the profibrotic factor angiotensin II via extracellular signal-regulated kinase 1/2 in endothelial cells and tissue explants. Mol Biol Cell 2010; 21:4240-50. [PMID: 20926686 PMCID: PMC2993751 DOI: 10.1091/mbc.e10-04-0341] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 09/03/2010] [Accepted: 09/23/2010] [Indexed: 01/07/2023] Open
Abstract
Hepatocyte growth factor (HGF), an endogenous tissue repair factor, attenuates apoptosis in many primary cell types, but the mechanism is not completely understood. Our laboratory demonstrated that angiotensin (Ang) II activates the intrinsic apoptotic pathway in primary endothelial cells (ECs) via reduction of the antiapoptotic protein Bcl-x(L). Ang II decreased Bcl-x(L) mRNA half-life by reducing its binding to nucleolin, a protein that normally binds a 3' AU-rich region and stabilizes Bcl-x(L) mRNA. We hypothesized HGF may block apoptosis induced by Ang II. We used primary EC and ex vivo cultures of rat lung tissue to investigate HGF inhibition of Ang II-induced apoptosis. Our data indicated HGF abrogated Ang II-induced apoptosis by inhibiting cytochrome c release, caspase-3 activation, and DNA fragmentation. RNA-immunoprecipitation experiments demonstrated that HGF stabilized Bcl-x(L) mRNA by increasing nucleolin binding to the 3'-untranslated region that was associated with cytoplasmic localization of nucleolin. Cytoplasmic localization of nucleolin and Bcl-x(L) mRNA stabilization required HGF activation of extracellular signal-regulated kinase (ERK)1/2, but not phosphatidylinositol 3-kinase. HGF also blocked Ang II-induced caspase-3 activation and lactate dehydrogenase release in tissue explants in an ERK-dependent manner.
Collapse
Affiliation(s)
- Young H. Lee
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Ana P. Marquez
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Ognoon Mungunsukh
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Regina M. Day
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| |
Collapse
|
23
|
Nuclear co-localization and functional interaction of COX-2 and HIF-1α characterize bone metastasis of human breast carcinoma. Breast Cancer Res Treat 2010; 129:433-50. [PMID: 21069452 DOI: 10.1007/s10549-010-1240-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 10/21/2010] [Indexed: 12/31/2022]
Abstract
The aim of this article is to identify nuclear co-localization of COX-2 and HIF-1α in human-bone metastasis of breast cancer, index of transcriptionally activated cells and functional for gene expression. In particular, we verified whether hypoxia exerted a direct role on metastasis-gene expression or through COX-2 signaling, due to the relevance for clinical implications to individuate molecular targets for diagnosis and therapy. The experiments were performed in vitro with two metastatic clones, 1833 and MDA-231BO, and the parental MDA-MB231 cells, in vivo (1833-xenograft model), and in human-bone metastasis specimens. In 1833 cells in vitro, COX-2 signaling pathway was critical for nuclear HIF-1α-protein expression/translocation, mechanisms determining HIF-1 activity and gene expression. The data were corroborated by immunohistochemistry in human-bone metastasis specimens. COX-2 and HIF-1α showed wide co-localization in the nucleus, indicative of COX-2-nuclear import in transcriptionally activated metastatic cells and consistent with COX-2-HIF-1α functional interaction. A network of microenvironmental signals controlled COX-2 induction and HIF-1 activation downstream. In fact, hypoxia through HGF and TGF-β1 autoregulatory loops triggered a specific array of transcription factors responsible for COX-2 transactivation. The novelty was that HGF and TGF-β1 biological signals were produced by hypoxic metastatic cells and, therefore, the microenvironment seemed to be modified by metastatic-cell engraftment in the bone. In agreement, HIF-1α expression in bone marrow supportive cells occurred in metastasis-bearing animals. Altogether, the data supported the pre-metastatic-niche theory. Our observations might be useful to design therapies against bone metastasis, by affecting the phenotype changes of metastatic cells occurring at the secondary growth site through COX-2-HIF-1 interaction.
Collapse
|
24
|
Mungunsukh O, Lee YH, Marquez AP, Cecchi F, Bottaro DP, Day RM. A tandem repeat of a fragment of Listeria monocytogenes internalin B protein induces cell survival and proliferation. Am J Physiol Lung Cell Mol Physiol 2010; 299:L905-14. [PMID: 20889677 DOI: 10.1152/ajplung.00094.2010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Hepatocyte growth factor (HGF) is critical for tissue homeostasis and repair in many organs including the lung, heart, kidney, liver, nervous system, and skin. HGF is a heterodimeric protein containing 20 disulfide bonds distributed among an amino-terminal hairpin, four kringle domains, and a serine protease-like domain. Due to its complex structure, recombinant production of HGF in prokaryotes requires denaturation and refolding, processes that are impractical for large-scale manufacture. Thus, pharmaceutical quantities of HGF are not available despite its potential applications. A fragment of the Listeria monocytogenes internalin B protein from amino acids 36-321 (InlB₃₆₋₃₂₁) was demonstrated to bind to and partially activate the HGF receptor Met. InlB₃₆₋₃₂₁ has a stable β-sheet structure and is easily produced in its native conformation by Escherichia coli. We cloned InlB₃₆₋₃₂₁ (1×InlB₃₆₋₃₂₁) and engineered a head-to-tail repeat of InlB₃₆₋₃₂₁ with a linker peptide (2×InlB₃₆₋₃₂₁); 1×InlB₃₆₋₃₂₁ and 2×InlB₃₆₋₃₂₁ were purified from E. coli. Both 1× and 2×InlB₃₆₋₃₂₁ activated the Met tyrosine kinase. We subsequently compared signal transduction of the two proteins in primary lung endothelial cells. 2×InlB₃₆₋₃₂₁ activated ERK1/2, STAT3, and phosphatidylinositol 3-kinase/Akt pathways, whereas 1×InlB₃₆₋₃₂₁ activated only STAT3 and ERK1/2. The 2×InlB₃₆₋₃₂₁ promoted improved motility compared with 1×InlB₃₆₋₃₂₁ and additionally stimulated proliferation equivalent to full-length HGF. Both the 1× and 2×InlB₃₆₋₃₂₁ prevented apoptosis by the profibrotic peptide angiotensin II in cell culture and ex vivo lung slice cultures. The ease of large-scale production and capacity of 2×InlB₃₆₋₃₂₁ to mimic HGF make it a potential candidate as a pharmaceutical agent for tissue repair.
Collapse
Affiliation(s)
- Ognoon Mungunsukh
- Dept. of Pharmacology, Uniformed Services University of the Health Sciences, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Li XY, Zhan XR, Lu C, Liu XM, Wang XC. Mechanisms of hepatocyte growth factor-mediated signaling in differentiation of pancreatic ductal epithelial cells into insulin-producing cells. Biochem Biophys Res Commun 2010; 398:389-94. [DOI: 10.1016/j.bbrc.2010.06.078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 06/18/2010] [Indexed: 10/19/2022]
|
26
|
Bauman KA, Wettlaufer SH, Okunishi K, Vannella KM, Stoolman JS, Huang SK, Courey AJ, White ES, Hogaboam CM, Simon RH, Toews GB, Sisson TH, Moore BB, Peters-Golden M. The antifibrotic effects of plasminogen activation occur via prostaglandin E2 synthesis in humans and mice. J Clin Invest 2010; 120:1950-60. [PMID: 20501949 DOI: 10.1172/jci38369] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 03/17/2010] [Indexed: 02/06/2023] Open
Abstract
Plasminogen activation to plasmin protects from lung fibrosis, but the mechanism underlying this antifibrotic effect remains unclear. We found that mice lacking plasminogen activation inhibitor-1 (PAI-1), which are protected from bleomycin-induced pulmonary fibrosis, exhibit lung overproduction of the antifibrotic lipid mediator prostaglandin E2 (PGE2). Plasminogen activation upregulated PGE2 synthesis in alveolar epithelial cells, lung fibroblasts, and lung fibrocytes from saline- and bleomycin-treated mice, as well as in normal fetal and adult primary human lung fibroblasts. This response was exaggerated in cells from Pai1-/- mice. Although enhanced PGE2 formation required the generation of plasmin, it was independent of proteinase-activated receptor 1 (PAR-1) and instead reflected proteolytic activation and release of HGF with subsequent induction of COX-2. That the HGF/COX-2/PGE2 axis mediates in vivo protection from fibrosis in Pai1-/- mice was demonstrated by experiments showing that a selective inhibitor of the HGF receptor c-Met increased lung collagen to WT levels while reducing COX-2 protein and PGE2 levels. Of clinical interest, fibroblasts from patients with idiopathic pulmonary fibrosis were found to be defective in their ability to induce COX-2 and, therefore, unable to upregulate PGE2 synthesis in response to plasmin or HGF. These studies demonstrate crosstalk between plasminogen activation and PGE2 generation in the lung and provide a mechanism for the well-known antifibrotic actions of the fibrinolytic pathway.
Collapse
Affiliation(s)
- Kristy A Bauman
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Crosby LM, Waters CM. Epithelial repair mechanisms in the lung. Am J Physiol Lung Cell Mol Physiol 2010; 298:L715-31. [PMID: 20363851 DOI: 10.1152/ajplung.00361.2009] [Citation(s) in RCA: 517] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The recovery of an intact epithelium following lung injury is critical for restoration of lung homeostasis. The initial processes following injury include an acute inflammatory response, recruitment of immune cells, and epithelial cell spreading and migration upon an autologously secreted provisional matrix. Injury causes the release of factors that contribute to repair mechanisms including members of the epidermal growth factor and fibroblast growth factor families (TGF-alpha, KGF, HGF), chemokines (MCP-1), interleukins (IL-1beta, IL-2, IL-4, IL-13), and prostaglandins (PGE(2)), for example. These factors coordinate processes involving integrins, matrix materials (fibronectin, collagen, laminin), matrix metalloproteinases (MMP-1, MMP-7, MMP-9), focal adhesions, and cytoskeletal structures to promote cell spreading and migration. Several key signaling pathways are important in regulating these processes, including sonic hedgehog, Rho GTPases, MAP kinase pathways, STAT3, and Wnt. Changes in mechanical forces may also affect these pathways. Both localized and distal progenitor stem cells are recruited into the injured area, and proliferation and phenotypic differentiation of these cells leads to recovery of epithelial function. Persistent injury may contribute to the pathology of diseases such as asthma, chronic obstructive pulmonary disease, and pulmonary fibrosis. For example, dysregulated repair processes involving TGF-beta and epithelial-mesenchymal transition may lead to fibrosis. This review focuses on the processes of epithelial restitution, the localization and role of epithelial progenitor stem cells, the initiating factors involved in repair, and the signaling pathways involved in these processes.
Collapse
Affiliation(s)
- Lynn M Crosby
- Departments of 1Physiology, University of Tennessee Health Science Center, Memphis, TN 38163-0001, USA
| | | |
Collapse
|
28
|
Chen W, Bai L, Wang X, Xu S, Belinsky SA, Lin Y. Acquired activation of the Akt/cyclooxygenase-2/Mcl-1 pathway renders lung cancer cells resistant to apoptosis. Mol Pharmacol 2009; 77:416-23. [PMID: 19933775 DOI: 10.1124/mol.109.061226] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Acquired apoptosis resistance plays an important role in acquired chemoresistance in cancer cells during chemotherapy. Our previous observations demonstrated that acquired tumor necrosis factor-related apoptosis-inducing ligand resistance in lung cancer cells was associated with Akt-mediated stabilization of cellular FLICE-like inhibitory protein (c-FLIP) and Mcl-1. In this report, we determined that these cells also have acquired resistance to apoptosis induced by chemotherapeutics such as cisplatin and doxorubicin (Adriamycin), which was detected in vitro in cell cultures and in vivo in xenografted tumors. We further found that cyclooxygenase-2 (COX-2) is dramatically overexpressed in cells with acquired apoptosis resistance. COX-2 seems to be a crucial mediator in acquired apoptosis resistance because suppressing COX-2 activity with a chemical inhibitor or reducing COX-2 protein expression level with COX-2 small interfering RNA dramatically alleviated resistance to therapeutic-induced apoptosis. Inhibiting Akt markedly suppressed COX-2 expression, suggesting COX-2 is a downstream effector of this cell survival kinase-mediated apoptosis resistance. Furthermore, the expression of Mcl-1 but not c-FLIP was significantly reduced when COX-2 was suppressed, and knockdown of Mcl-1 substantially sensitized the cells to apoptosis. Our results establish a novel pathway that consists of Akt, COX-2, and Mcl-1 for acquired apoptosis resistance, which could be a molecular target for circumventing acquired chemoresistance in lung cancer.
Collapse
Affiliation(s)
- Wenjie Chen
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest Dr., SE, Albuquerque, NM 87108, USA
| | | | | | | | | | | |
Collapse
|
29
|
Haribalaganesh R, Sheikpranbabu S, Elayappan B, Venkataraman D, Gurunathan S. Pigment epithelium–derived factor down regulates hyperglycemia-induced apoptosis via PI3K/Akt activation in goat retinal pericytes. Angiogenesis 2009; 12:381-9. [DOI: 10.1007/s10456-009-9159-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 10/20/2009] [Indexed: 01/01/2023]
|
30
|
Hepatocyte growth factor protects endothelial progenitor cell from damage of low-density lipoprotein cholesterol via the PI3K/Akt signaling pathway. Mol Biol Rep 2009; 37:2423-9. [DOI: 10.1007/s11033-009-9753-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 08/06/2009] [Indexed: 10/20/2022]
|
31
|
Johnson A. TNF-induced activation of pulmonary microvessel endothelial cells: a role for GSK3beta. Am J Physiol Lung Cell Mol Physiol 2009; 296:L700-9. [PMID: 19218353 DOI: 10.1152/ajplung.90566.2008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The hypothesis tested was PKCalpha mediates the phosphorylation of glycogen synthetase kinase 3beta (GSK3beta) and that the GSK3beta inhibition modulates the response to tumor necrosis factor-alpha (TNF) in rat pulmonary microvessel endothelial cells (PMEC). PMEC were treated with TNF for 4.0 h (100 ng/ml) or vehicle. First, to assess the role of PKCalpha in the phosphorylation of GSK3beta (i.e., an indicator of GSK3beta inhibition), PMEC were pretreated with 1) nonsense-RNA-PKCalpha, 2) siRNA-PKCalpha, and 3) the PKC inhibitor Gö6983. In the nonsense RNA-PKCalpha+TNF and TNF groups, there was increased phosphorylated GSK3beta-Ser9 that did not occur in the Gö6983+TNF group. In the TNF groups, there was a significant correlation between PKCalpha protein and phosphorylated GSK3beta-Ser9 that did not occur in the groups without TNF. Second, to assess the role of GSK3beta in beta-catenin activity, PMEC were pretreated with 1) wild-type (w) GSK3beta plasmid to enhance GSK3beta activity, 2) kinase dead (kd)-GSK3beta plasmid, and 3) the GSK3beta inhibitor SB-216763. In the TNF group, there was increased unphosphorylated beta-catenin-Ser37/33 compared with the control group. In the GSK3beta-inhibited groups (i.e., SB-216763 and kdGSK3beta) +/- TNF, the unphosphorylated beta-catenin-Ser37/33 was similar to the TNF group. In the GSK3beta-enhanced group +/- TNF, the unphosphorylated beta-catenin-Ser37/33 was similar to the control. Finally, PMEC were also treated with TOPflash, a beta-catenin-dependent promoter luciferase reporter, or the mutant construct FOPflash, 2 days before treatment with TNF. In the TNF group, there was an increased TOPflash/FOPflash activity ratio compared with the control group. In the GSK3beta-inhibited groups (i.e., SB-216763 and kdGSK3beta) +/- TNF, the TOPflash/FOPflash activity ratio was similar to the TNF group. In the GSK3beta-enhanced group +/- TNF, the TOPflash/FOPflash activity ratio was similar to the control. The data indicate that TNF induces endothelial activation that is modulated by a PKCalpha-dependent inhibition of GSK3beta.
Collapse
Affiliation(s)
- Arnold Johnson
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA.
| |
Collapse
|