1
|
Peng K, Wang M, Wang J, Wang Q, Li D, Sun X, Yang Y, Yang D. Nuclear receptor subfamily 1 group D member 1 suppresses the proliferation, migration of adventitial fibroblasts, and vascular intimal hyperplasia via mammalian target of rapamycin complex 1/β-catenin pathway. Clin Exp Hypertens 2023; 45:2178659. [PMID: 36794491 DOI: 10.1080/10641963.2023.2178659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
BACKGROUND In-stent restenosis hardly limits the therapeutic effect of the percutaneous vascular intervention. Although the restenosis is significantly ameliorated after the application of new drug-eluting stents, the incidence of restenosis remains at a high level. OBJECTIVE Vascular adventitial fibroblasts (AFs) play an important role in intimal hyperplasia and subsequent restenosis. The current study was aimed to investigate the role of nuclear receptor subfamily 1, group D, member 1 (NR1D1) in the vascular intimal hyperplasia. METHODS AND RESULTS We observed increased expression of NR1D1 after the transduction of adenovirus carrying Nr1d1 gene (Ad-Nr1d1) in AFs. Ad-Nr1d1 transduction significantly reduced the numbers of total AFs, Ki-67-positive AFs, and the migration rate of AFs. NR1D1 overexpression decreased the expression level of β-catenin and attenuated the phosphorylation of the effectors of mammalian target of rapamycin complex 1 (mTORC1), including mammalian target of rapamycin (mTOR) and 4E binding protein 1 (4EBP1). Restoration of β-catenin by SKL2001 abolished the inhibitory effects of NR1D1 overexpression on the proliferation and migration of AFs. Surprisingly, the restoration of mTORC1 activity by insulin could also reverse the decreased expression of β-catenin, attenuated proliferation, and migration in AFs induced by NR1D1 overexpression. In vivo, we found that SR9009 (an agonist of NR1D1) ameliorated the intimal hyperplasia at days 28 after injury of carotid artery. We further observed that SR9009 attenuated the increased Ki-67-positive AFs, an essential part of vascular restenosis at days 7 after injury to the carotid artery. CONCLUSION These data suggest that NR1D1 inhibits intimal hyperplasia by suppressing the proliferation and migration of AFs in a mTORC1/β-catenin-dependent manner.
Collapse
Affiliation(s)
- Ke Peng
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Mingliang Wang
- Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Jun Wang
- Central Sterile Supply Department, General Hospital of Western Theater Command, Chengdu, China
| | - Qiang Wang
- Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - De Li
- Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Xiongshan Sun
- Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Yongjian Yang
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Dachun Yang
- Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
2
|
Abstract
Matrix metalloproteinases (MMPs) are a class of endopeptidases that are dependent on zinc and facilitate the degradation of extracellular matrix (ECM) proteins, thereby playing pivotal parts in human physiology and pathology. MMPs regulate normal tissue and cellular functions, including tissue development, remodeling, angiogenesis, bone formation, and wound healing. Several diseases, including cancer, inflammation, cardiovascular diseases, and nervous system disorders, have been linked to dysregulated expression of specific MMP subtypes, which can promote tumor progression, metastasis, and inflammation. Various MMP-responsive drug delivery and release systems have been developed by harnessing cleavage activities and overexpression of MMPs in affected regions. Herein, we review the structure, substrates, and physiological and pathological functions of various MMPs and highlight the strategies for designing MMP-responsive nanoparticles to improve the targeting efficiency, penetration, and protection of therapeutic payloads.
Collapse
Affiliation(s)
- Chenyun Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
3
|
Waseem A, Rashid S, Rashid K, Khan MA, Khan R, Haque R, Seth P, Raza SS. Insight into the transcription factors regulating Ischemic Stroke and Glioma in Response to Shared Stimuli. Semin Cancer Biol 2023; 92:102-127. [PMID: 37054904 DOI: 10.1016/j.semcancer.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/28/2023] [Accepted: 04/09/2023] [Indexed: 04/15/2023]
Abstract
Cerebral ischemic stroke and glioma are the two leading causes of patient mortality globally. Despite physiological variations, 1 in 10 people who have an ischemic stroke go on to develop brain cancer, most notably gliomas. In addition, glioma treatments have also been shown to increase the risk of ischemic strokes. Stroke occurs more frequently in cancer patients than in the general population, according to traditional literature. Unbelievably, these events share multiple pathways, but the precise mechanism underlying their co-occurrence remains unknown. Transcription factors (TFs), the main components of gene expression programmes, finally determine the fate of cells and homeostasis. Both ischemic stroke and glioma exhibit aberrant expression of a large number of TFs, which are strongly linked to the pathophysiology and progression of both diseases. The precise genomic binding locations of TFs and how TF binding ultimately relates to transcriptional regulation remain elusive despite a strong interest in understanding how TFs regulate gene expression in both stroke and glioma. As a result, the importance of continuing efforts to understand TF-mediated gene regulation is highlighted in this review, along with some of the primary shared events in stroke and glioma.
Collapse
Affiliation(s)
- Arshi Waseem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, India
| | - Sumaiya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Khalid Rashid
- Department of Cancer Biology, Vontz Center for Molecular Studies, Cincinnati, OH 45267-0521
| | | | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City,Mohali, Punjab 140306, India
| | - Rizwanul Haque
- Department of Biotechnology, Central University of South Bihar, Gaya -824236, India
| | - Pankaj Seth
- Molecular and Cellular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Haryana-122052, India
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, India; Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College Hospital, Era University, Sarfarazganj, Lucknow-226003, India
| |
Collapse
|
4
|
Lin S, Lin R, Zhang H, Xu Q, He Y. Peripheral vascular remodeling during ischemia. Front Pharmacol 2022; 13:1078047. [DOI: 10.3389/fphar.2022.1078047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
About 230 million people worldwide suffer from peripheral arterial disease (PAD), and the prevalence is increasing year by year. Multiple risk factors, including smoking, dyslipidemia, diabetes, and hypertension, can contribute to the development of PAD. PAD is typically characterized by intermittent claudication and resting pain, and there is a risk of severe limb ischemia, leading to major adverse limb events, such as amputation. Currently, a major progress in the research field of the pathogenesis of vascular remodeling, including atherosclerosis and neointima hyperplasia has been made. For example, the molecular mechanisms of endothelial dysfunction and smooth muscle phenotype switching have been described. Interestingly, a series of focused studies on fibroblasts of the vessel wall has demonstrated their impact on smooth muscle proliferation and even endothelial function via cell-cell communications. In this review, we aim to focus on the functional changes of peripheral arterial cells and the mechanisms of the pathogenesis of PAD. At the same time, we summarize the progress of the current clinical treatment and potential therapeutic methods for PAD and shine a light on future perspectives.
Collapse
|
5
|
Teng M, Hu C, Yang B, Xiao W, Zhou Q, Li Y, Li Z. Salvianolic acid B targets mortalin and inhibits the migration and invasion of hepatocellular carcinoma via the RECK/STAT3 pathway. Cancer Cell Int 2021; 21:654. [PMID: 34876128 PMCID: PMC8650508 DOI: 10.1186/s12935-021-02367-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/24/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Tumor migration and invasion is a complex and diverse process that involves the epithelial-mesenchymal transition (EMT) of tumor cells and degradation of the extracellular matrix by matrix metalloproteases (MMPs). Mortalin is an important oncogene. It has been reported to play an important role in tumor migration and invasion through various signaling pathways, but the underlying mechanism is not fully understood. METHODS Here, we investigated the role of mortalin in the migration of the hepatocellular carcinoma (HCC) cell lines HepG2 and HCCLM3. RESULTS The overexpression of mortalin in HepG2 cells decreased the protein level of reversion-inducing cysteine-rich protein with Kazal motifs (RECK) and activated the phosphorylation and acetylation of STAT3, thereby up-regulating matrix metalloproteinase 9 (MMP9) and promoting cell migration and invasion. In contrast, in HCCLM3 cells, mortalin knockdown increased the expression of RECK, inhibited the STAT3 pathway and the activity of MMP9, and inhibited cell migration and invasion. Furthermore, we found that salvianolic acid B, a caffeic acid phenethyl ester analog, specifically bound to mortalin and increased the degradation of mortalin proteasomes through ubiquitination, thereby up-regulating RECK, inhibiting STAT3, and finally inhibiting the migration and invasion of HCC cells. CONCLUSION Our work suggested that mortalin is a potential therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Mengying Teng
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chunyan Hu
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Bingmo Yang
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wei Xiao
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Qian Zhou
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yuan Li
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Zhong Li
- The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
6
|
Bian Z, Ji W, Xu B, Huo Z, Huang H, Huang J, Jiao J, Shao J, Zhang X. Noncoding RNAs involved in the STAT3 pathway in glioma. Cancer Cell Int 2021; 21:445. [PMID: 34425834 PMCID: PMC8381529 DOI: 10.1186/s12935-021-02144-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/11/2021] [Indexed: 01/03/2023] Open
Abstract
Glioma is the most common malignant primary brain tumour in adults. Despite improvements in neurosurgery and radiotherapy, the prognosis of glioma patients remains poor. One of the main limitations is that there are no proper clinical therapeutic targets for glioma. Therefore, it is crucial to find one or more effective targets. Signal transducer and activator of transcription 3 (STAT3) is a member of the STAT family of genes. Abnormal expression of STAT3 is involved in the process of cell proliferation, migration, invasion, immunosuppression, angiogenesis, dryness maintenance, and resistance to radiotherapy and chemotherapy in glioma. Therefore, STAT3 has been considered an ideal therapeutic target in glioma. Noncoding RNAs (ncRNAs) are a group of genes with limited or no protein-coding capacity that can regulate gene expression at the epigenetic, transcriptional and posttranscriptional level. In this review, we summarized the ncRNAs that are correlated with the ectopic expression of STAT3 in glioma.
Collapse
Affiliation(s)
- Zheng Bian
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Wei Ji
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Bin Xu
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Zhengyuan Huo
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Hui Huang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Jin Huang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Jiantong Jiao
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Junfei Shao
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China.
| | - Xiaolu Zhang
- Department of Neurosurgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China.
| |
Collapse
|
7
|
Wang A, Cao S, Aboelkassem Y, Valdez-Jasso D. Quantification of uncertainty in a new network model of pulmonary arterial adventitial fibroblast pro-fibrotic signalling. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2020; 378:20190338. [PMID: 32448066 PMCID: PMC7287331 DOI: 10.1098/rsta.2019.0338] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/16/2020] [Indexed: 05/21/2023]
Abstract
Here, we present a novel network model of the pulmonary arterial adventitial fibroblast (PAAF) that represents seven signalling pathways, confirmed to be important in pulmonary arterial fibrosis, as 92 reactions and 64 state variables. Without optimizing parameters, the model correctly predicted 80% of 39 results of input-output and inhibition experiments reported in 20 independent papers not used to formulate the original network. Parameter uncertainty quantification (UQ) showed that this measure of model accuracy is robust to changes in input weights and half-maximal activation levels (EC50), but is more affected by uncertainty in the Hill coefficient (n), which governs the biochemical cooperativity or steepness of the sigmoidal activation function of each state variable. Epistemic uncertainty in model structure, due to the reliance of some network components and interactions on experiments using non-PAAF cell types, suggested that this source of uncertainty had a smaller impact on model accuracy than the alternative of reducing the network to only those interactions reported in PAAFs. UQ highlighted model parameters that can be optimized to improve prediction accuracy and network modules where there is the greatest need for new experiments. This article is part of the theme issue 'Uncertainty quantification in cardiac and cardiovascular modelling and simulation'.
Collapse
Affiliation(s)
| | | | | | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92092, USA
| |
Collapse
|
8
|
Wang C, Qu Y, Suo R, Zhu Y. Long non-coding RNA MALAT1 regulates angiogenesis following oxygen-glucose deprivation/reoxygenation. J Cell Mol Med 2019; 23:2970-2983. [PMID: 30784209 PMCID: PMC6433728 DOI: 10.1111/jcmm.14204] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/17/2018] [Accepted: 01/06/2019] [Indexed: 12/30/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been identified as playing critical roles in multiple diseases. However, little is known regarding their roles and mechanisms in post-stroke angiogenesis. Our studies focused on deciphering the functional roles and the underlying mechanisms of the lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in the process of angiogenesis following oxygen-glucose deprivation/reoxygenation (OGD/R). We characterized the up-regulation of MALAT1 expression in the process of angiogenesis after hypoxic injury in vivo and in vitro. We further showed that compared with the empty vector, MALAT1 knockdown had significantly reduced the capacity for angiogenesis, which was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT), scratching, cell cycle and immunofluorescent staining. Thus, our findings suggest that MALAT1 may mediate proangiogenic function in OGD/R. To further explore the potential mechanisms, we used lentiviruses expressing shMALAT1 and empty vector; the results revealed that shMALAT1 reduced the expression of 15-lipoxygenase 1 (15-LOX1), vascular endothelial growth factor (VEGF) and the phosphorylation of signal transducers and activators of transcription 3 (pSTAT3). Taken together, our results are the first to propose that MALAT1 may regulate angiogenesis through the 15-LOX1/STAT3 signalling pathway, and they may provide a critical target for the treatment of hypoxic injury and an avenue for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Chengya Wang
- Department of NeurologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Youyang Qu
- Department of NeurologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Rui Suo
- Department of NeurologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Yulan Zhu
- Department of NeurologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| |
Collapse
|
9
|
Ben P, Hu M, Wu H, Zhang Z, Gao Y, Luo L, Yin Z. L-Theanine Down-Regulates the JAK/STAT3 Pathway to Attenuate the Proliferation and Migration of Vascular Smooth Muscle Cells Induced by Angiotensin II. Biol Pharm Bull 2018; 41:1678-1684. [DOI: 10.1248/bpb.b18-00387] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Peiling Ben
- Department of Medicine, Chuzhou City Vocation College
| | - Monong Hu
- Department of Medicine, Chuzhou City Vocation College
| | - Huizhen Wu
- Department of Medicine, Chuzhou City Vocation College
| | - Zhengping Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University
| | - Yanhong Gao
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University
| |
Collapse
|
10
|
Jayatilaka H, Umanzor FG, Shah V, Meirson T, Russo G, Starich B, Tyle P, Lee JSH, Khatau S, Gil-Henn H, Wirtz D. Tumor cell density regulates matrix metalloproteinases for enhanced migration. Oncotarget 2018; 9:32556-32569. [PMID: 30220965 PMCID: PMC6135685 DOI: 10.18632/oncotarget.25863] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/18/2018] [Indexed: 02/01/2023] Open
Abstract
Matrix metalloproteinases (MMPs) may play a critical role in metastatic cancers, yet multiple human clinical trials targeting MMPs have surprisingly failed. Cancer cell density changes dramatically during the early growth of a primary tumor and during the early seeding steps of secondary tumors and has been implicated in playing an important role in regulating metastasis and drug resistance. This study reveals that the expression of MMPs is tightly regulated by local tumor cell density through the synergistic signaling mechanism of Interleukin 6 (IL-6) and Interleukin 8 (IL-8) via the JAK2/STAT3 complex. Local tumor cell density also plays a role in the responsiveness of cells to matrix metalloproteinases inhibitors (MMPI), such as Batimastat, Marimastat, Bryostatin I, and Cipemastat, where different migratory phenotypes are observed in low and high cell density conditions. Cell density-dependent MMP regulation can be directly targeted by the simultaneous inhibition of IL-6 and IL-8 receptors via Tocilizumab and Reparixin to significantly decrease the expression of MMPs in mouse xenograft models and decrease effective metastasis. This study reveals a new strategy to decrease MMP expression through pharmacological intervention of the cognate receptors of IL-6 and IL-8 to decrease metastatic capacity of tumor cells.
Collapse
Affiliation(s)
- Hasini Jayatilaka
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA.,Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD, USA.,Department of Pediatrics, Bass Center for Childhood Cancer, Stanford University, Stanford, CA, USA
| | - Fatima G Umanzor
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA.,Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD, USA
| | - Vishwesh Shah
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Tomer Meirson
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Gabriella Russo
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Bartholomew Starich
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Pranay Tyle
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Jerry S H Lee
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA.,Center for Strategic Scientific Initiatives, National Cancer Institute, Bethesda, MD, USA.,Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Medicine/Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shyam Khatau
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Hava Gil-Henn
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA.,Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology and Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Liu Y, Zhang H, Yan L, Du W, Zhang M, Chen H, Zhang L, Li G, Li J, Dong Y, Zhu D. MMP-2 and MMP-9 contribute to the angiogenic effect produced by hypoxia/15-HETE in pulmonary endothelial cells. J Mol Cell Cardiol 2018; 121:36-50. [PMID: 29913136 DOI: 10.1016/j.yjmcc.2018.06.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 06/06/2018] [Accepted: 06/13/2018] [Indexed: 11/15/2022]
Abstract
Matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) are the predominant gelatinases in the developing lung. Studies have shown that the expression of MMP-2 and MMP-9 is upregulated in hypoxic fibroblasts, 15-hydroxyeicosatetraenoic acid (15-HETE) regulated fibroblasts migration via modulating MMP-2 or MMP-9, and that hypoxia/15-HETE is a predominant contributor to the development of pulmonary arterial hypertension (PAH) through increased angiogenesis. However, the roles of MMP-2 and MMP-9 in pulmonary arterial endothelial cells (PAECs) angiogenesis as well as the molecular mechanism of hypoxia-regulated MMP-2 and MMP-9 expression have not been identified. The aim of this study was to investigate the role of MMP-2 and MMP-9 in PAEC proliferation and vascular angiogenesis and to determine the effects of hypoxia-induced 15-HETE on the expression of MMP-2 and MMP-9. Western blot, immunofluorescence, and real-time PCR were used to measure the expression of MMP-2 and MMP-9 in hypoxic PAECs. Immunohistochemical staining, flow cytometry, and tube formation as well as cell proliferation, viability, scratch-wound, and Boyden chamber migration assays were used to identify the roles and relationships between MMP-2, MMP-9, and 15-HETE in hypoxic PAECs. We found that hypoxia increased MMP-2 and MMP-9 expression in pulmonary artery endothelium both in vivo and in vitro in a time-dependent pattern. Moreover, administration of the MMP-2 and MMP-9 inhibitor MMI-166 significantly reversed hypoxia-induced increases in right ventricular systemic pressure (RVSP), right ventricular function, and thickening of the tunica media. Furthermore, up-regulation of MMP-2 and MMP-9 expression was induced by 15-HETE, which regulates PAEC proliferation, migration, and cell cycle transition that eventually leads to angiogenesis. Our study demonstrated that hypoxia increases the expression of MMP-2 and MMP-9 through the 15-lipoxygenase/15-HETE pathway, and that MMP-2 and MMP-9 promote PAEC angiogenesis. These findings suggest that MMP-2 and MMP-9 may serve as new potential therapeutic targets for the treatment of PAH.
Collapse
Affiliation(s)
- Ying Liu
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Hongyue Zhang
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Lixin Yan
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Wei Du
- School of Pharmacy, Harbin University of Commerce, Harbin, Heilongjiang Province, China
| | - Min Zhang
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - He Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lixin Zhang
- Department of Immunology, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, Heilongjiang Province, China
| | - Guangqun Li
- Medical Laboratory Technology, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Jijin Li
- Medical Laboratory Technology, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Yinchu Dong
- Medical Laboratory Technology, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Daling Zhu
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China.
| |
Collapse
|
12
|
Chai X, Sun D, Han Q, Yi L, Wu Y, Liu X. Hypoxia induces pulmonary arterial fibroblast proliferation, migration, differentiation and vascular remodeling via the PI3K/Akt/p70S6K signaling pathway. Int J Mol Med 2018; 41:2461-2472. [PMID: 29436587 PMCID: PMC5846667 DOI: 10.3892/ijmm.2018.3462] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 11/29/2017] [Indexed: 12/20/2022] Open
Abstract
The present study was designed to examine whether hypoxia induces the proliferation, migration and differentiation of pulmonary arterial fibroblasts (PAFs) via the PI3K/Akt/p70S6K signaling pathway. PAFs were subjected to normoxia (21% O2) or hypoxia (1% O2). The proliferation, migration, differentiation and cellular p110α, p-Akt, and p-p70S6K expression levels of the PAFs were examined in vitro. In addition, rats were maintained under hypoxic conditions, and the right ventricular systolic pressure (RVSP), right ventricular hypertrophy index (RVHI) and right ventricular weight/body weight ratio (RV/BW) were examined. The expression levels of p110α, p-Akt, p70S6K, fibronectin and α-SMA in the rat pulmonary vessels were also examined. Hypoxia significantly elevated the proliferation, migration and differentiation of rat PAFs. It also strongly elevated the expression of p110α, p-Akt and p-p70S6K in PAFs in vitro. NVP-BEZ235 was revealed to significantly reduce the hypoxia-induced proliferation, migration and differentiation. In vivo experiments demonstrated that hypoxia significantly induced the elevation of RVSP, RVHI, RV/BW, medial thickening, adventitious thickening, and fibronectin and collagen deposition around pulmonary artery walls. The expression of p110α, p-Akt and p70S6K was evident in the pulmonary arteries of the hypoxic rats. NVP-BEZ235 significantly reduced the hypoxia-induced hypoxic pulmonary vascular remodeling, as well as fibronectin and collagen deposition in the pulmonary arteries. Therefore, hypoxia was demonstrated to induce the proliferation, migration and differentiation of PAFs and the hypoxic pulmonary vascular remodeling of rats via the PI3K/Akt/p70S6K signaling pathway.
Collapse
Affiliation(s)
- Xiaoyu Chai
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| | - Dan Sun
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| | - Qian Han
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| | - Liang Yi
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| | - Yanping Wu
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| | - Xinmin Liu
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| |
Collapse
|
13
|
Scott RA, Kharkar PM, Kiick KL, Akins RE. Aortic adventitial fibroblast sensitivity to mitogen activated protein kinase inhibitors depends on substrate stiffness. Biomaterials 2017; 137:1-10. [PMID: 28527302 DOI: 10.1016/j.biomaterials.2017.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/25/2017] [Accepted: 05/07/2017] [Indexed: 11/18/2022]
Abstract
Adventitial fibroblasts (AFs) are key determinants of arterial function and critical mediators of arterial disease progression. The effects of altered stiffness, particularly those observed across individuals during normal vascular function, and the mechanisms by which AFs respond to altered stiffness, are not well understood. To study the effects of matrix stiffness on AF phenotype, cytokine production, and the regulatory pathways utilized to interpret basic cell-matrix interactions, human aortic AFs were grown in 5%, 7.5%, and 10% (w/v%) PEG-based hydrogels with Young's moduli of 1.2, 3.3, and 9.6 kPa, respectively. In 5% gels, AFs had higher proliferation rates, elevated monocyte chemoattractant protein-1 secretion, and enhanced monocyte recruitment. Significantly more AFs were α-smooth muscle actin positive in 7.5% gels, indicating myofibroblast development. AFs in 10% gels had low proliferation rates but produced high levels of interleukin-6 and vascular endothelial growth factor-A. Importantly, these modulus-dependent changes in AF phenotype were accompanied by alterations in the mitogen-activated protein kinase (MAPK) pathways contributing to the production of cytokines. These data indicate that complex cell regulatory changes occur with altered tissue stiffness and suggest that therapeutics affecting MAPK pathways may have altered effects on AFs depending on substrate stiffness.
Collapse
Affiliation(s)
- Rebecca A Scott
- Nemours - Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Materials Science & Engineering, University of Delaware, Newark, DE 19716, USA; Delaware Biotechnology Institute, Newark, DE 19711, USA
| | - Prathamesh M Kharkar
- Department of Materials Science & Engineering, University of Delaware, Newark, DE 19716, USA
| | - Kristi L Kiick
- Department of Materials Science & Engineering, University of Delaware, Newark, DE 19716, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA; Delaware Biotechnology Institute, Newark, DE 19711, USA
| | - Robert E Akins
- Nemours - Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Materials Science & Engineering, University of Delaware, Newark, DE 19716, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
14
|
Li Y, Zhang L, Wang X, Chen M, Liu Y, Xing Y, Wang X, Gao S, Zhu D. Elk-1-mediated 15-lipoxygenase expression is required for hypoxia-induced pulmonary vascular adventitial fibroblast dynamics. Acta Physiol (Oxf) 2016; 218:276-289. [PMID: 27174674 DOI: 10.1111/apha.12711] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 04/26/2016] [Accepted: 05/09/2016] [Indexed: 12/20/2022]
Abstract
AIM 15-Lipoxygenase (15-LO) is an important factor in the pathogenesis of pulmonary artery hypertension (PAH). However, the role of 15-LO in the adventitia of the pulmonary arterial wall is unclear. The aim of this study was to explore the role of 15-LO in the modulation of pulmonary adventitial fibroblast (PAF) dynamics. METHODS Rats were exposed to normoxic or hypoxic (fraction of inspired O2 = 0.12) treatments for 7 days. PAF proliferation and cell cycle alterations were measured by MTT assay, cell immunofluorescence, flow cytometry and Western blot analysis. The 15-LO promoter was analysed by luciferase reporter and ChIP assays. RESULTS Our results showed that hypoxia induced 15-LO expression in PAFs both in vivo and in vitro. In addition, hypoxia stimulated JNK phosphorylation in PAFs. Blocking 15-LO or JNK suppressed 15-LO-induced PAF proliferation and cell cycle alterations. The inhibition of p27kipl by gene silencing attenuated 15-LO-induced PAF proliferation and cell cycle alterations. Furthermore, JNK inhibition or Elk-1 knockdown suppressed hypoxia-induced 15-LO expression in PAFs. Luciferase reporter and ChIP assays revealed that the 15-LO promoter contains Elk-1-binding sites and also that Elk-1 increased the hypoxia-induced activity of the 15-LO promoter. CONCLUSION These results suggest that hypoxia promotes changes in the cellular dynamics of PAFs by inducing 15-LO expression, which leads to vascular adventitial remodelling. The modulation of p27kipl expression by 15-LO enhances PAF proliferation and cell cycle alterations. Furthermore, the JNK-dependent increase in Elk-1 signalling is required for hypoxia-induced 15-LO expression in PAFs.
Collapse
Affiliation(s)
- Y. Li
- Department of Pharmacology; Harbin Medical University-Daqing; Daqing Heilongjiang China
- Biopharmaceutical Institute of the Heilongjiang Academy of Medical Sciences; Harbin Heilongjiang China
| | - L. Zhang
- Department of Pharmacology; Harbin Medical University-Daqing; Daqing Heilongjiang China
| | - X. Wang
- Department of Pharmacology; Harbin Medical University-Daqing; Daqing Heilongjiang China
| | - M. Chen
- Department of Pharmacology; Harbin Medical University-Daqing; Daqing Heilongjiang China
| | - Y. Liu
- Department of Pharmacology; Harbin Medical University-Daqing; Daqing Heilongjiang China
| | - Y. Xing
- Department of Pharmacology; Harbin Medical University-Daqing; Daqing Heilongjiang China
| | - X. Wang
- Department of Pharmacology; Harbin Medical University-Daqing; Daqing Heilongjiang China
| | - S. Gao
- Biopharmaceutical Institute of the Heilongjiang Academy of Medical Sciences; Harbin Heilongjiang China
| | - D. Zhu
- Department of Pharmacology; Harbin Medical University-Daqing; Daqing Heilongjiang China
- Biopharmaceutical Institute of the Heilongjiang Academy of Medical Sciences; Harbin Heilongjiang China
| |
Collapse
|
15
|
Wang X, Wang Y, Zhang J, Guan X, Chen M, Li Y, Zhang L. Galectin-3 contributes to vascular fibrosis in monocrotaline-induced pulmonary arterial hypertension rat model. J Biochem Mol Toxicol 2016; 31. [PMID: 27870162 DOI: 10.1002/jbt.21879] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/13/2016] [Accepted: 10/20/2016] [Indexed: 01/31/2023]
Abstract
Galectin-3 (Gal-3) plays a critical role in vascular inflammation and fibrosis. The role of TGF-β1 in mediating pulmonary vascular fibrosis is well documented; thus, we suspected that Gal-3 could be an important factor in TGF-β1-induced fibrosis in pulmonary adventitial fibroblasts (PAFs). We treated rats with monocrotaline (MCT) and cultured PAFs with TGF-β1 to stimulate fibrosis. We found that MCT injection induced vessel thickening and extracellular matrix deposition in vivo. TGF-β1 stimulated the production of collagen and fibronectin (Fn) protein in vitro. TGF-β1 promoted the expression of Gal-3 and its translocation, while silencing Gal-3 reduced Col-1a deposition. Blockage of STAT3 decreased the expression of Gal-3 induced by TGF-β1. Gal-3 increased Col-1a accumulation and downregulated matrix metallopeptidase 9 (MMP-9) expression in PAFs, but it did not affect Fn expression. These findings demonstrate that Gal-3 is required for TGF-β1-stimulated vascular fibrosis via a STAT3 signaling cascade and that MMP-9 is also involved in TGF-β1/Gal-3-induced vascular fibrosis.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China.,Department of Physiology, Harbin Medical University-Daqing, Daqing, Heilongjiang Province, 163319, People's Republic of China
| | - Yanli Wang
- Department of Cardiology, Kailuan General Hospital, Tangshan, Hebei Province, 063000, People's Republic of China
| | - Jinbo Zhang
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China
| | - Xue Guan
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China
| | - Minggang Chen
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China
| | - Yumei Li
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China.,Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang Province, 163319, China
| | - Li Zhang
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China.,Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang Province, 163319, China
| |
Collapse
|
16
|
Zhang L, Ma C, Zhang C, Ma M, Zhang F, Zhang L, Chen Y, Cao F, Li S, Zhu D. Reactive oxygen species effect PASMCs apoptosis via regulation of dynamin-related protein 1 in hypoxic pulmonary hypertension. Histochem Cell Biol 2016; 146:71-84. [DOI: 10.1007/s00418-016-1424-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2016] [Indexed: 01/27/2023]
|
17
|
Wang X, Chen L, Liu J, Yan T, Wu G, Xia Y, Zong G, Li F. In vivo treatment of rat arterial adventitia with interleukin‑1β induces intimal proliferation via the JAK2/STAT3 signaling pathway. Mol Med Rep 2016; 13:3451-8. [PMID: 26955959 PMCID: PMC4805072 DOI: 10.3892/mmr.2016.4982] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 02/09/2016] [Indexed: 11/09/2022] Open
Abstract
Previous studies have indicated that adventitial inflammation is involved in the development of atherosclerosis. The aim of this study was to investigate the effect of arterial adventitia inflammation induced by interleukin (IL)-1β on intimal proliferation and the mechanisms involved in this process. The left common carotid artery adventitia of male rats in the experimental and control groups (25 rats/group) was wrapped with agar containing or without a sustained-release suspension of 2.5 µg IL-1β, respectively. Five animals in each group were randomly selected for sacrifice at 2 h, 8 h, 24 h, 48 h, and 1 week post-treatment. Hematoxylin and eosin staining was performed for to analyze the morphology of the adventitia. The expression of janus kinase (JAK)2, signal transducer and activator of transcription (STAT)3, phosphorylated (p-)JAK2 and p-STAT3 were detected by western blot analysis or immunohistochemistry staining. A model of adventitial inflammation was successfully created by wrapping IL-1β around the rat carotid artery. IL-1β treatment induced vascular smooth muscle cell proliferation and migration as well as intimal proliferation. In addition, the expression of p-JAK2 and p-STAT3 increased after IL-1β treatment. Furthermore, an inhibitor of JAK2/STAT3 pathway, AG490, suppressed IL-1β-induced intimal proliferation and phosphorylation of JAK2 and STAT3. Thus, the JAK2/STAT3 signaling pathway is involved in intimal proliferation caused by vascular adventitial inflammation. Inhibiting the JAK2/STAT3 signaling pathway may be a novel method for the clinical treatment of artery atherosclerosis.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Cardiovascular Diseases, The 101st Hospital of PLA, Wuxi, Jiangsu 214044, P.R. China
| | - Lihua Chen
- Department of Radiology, The 101st Hospital of PLA, Wuxi, Jiangsu 214044, P.R. China
| | - Jie Liu
- Department of Cardiovascular Diseases, The 101st Hospital of PLA, Wuxi, Jiangsu 214044, P.R. China
| | - Tao Yan
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| | - Gangyong Wu
- Department of Cardiovascular Diseases, The 101st Hospital of PLA, Wuxi, Jiangsu 214044, P.R. China
| | - Yang Xia
- Department of Cardiovascular Diseases, The 101st Hospital of PLA, Wuxi, Jiangsu 214044, P.R. China
| | - Gangjun Zong
- Department of Cardiovascular Diseases, The 101st Hospital of PLA, Wuxi, Jiangsu 214044, P.R. China
| | - Fengsheng Li
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| |
Collapse
|
18
|
Zhang L, Chen Y, Li G, Chen M, Huang W, Liu Y, Li Y. TGF-β1/FGF-2 signaling mediates the 15-HETE-induced differentiation of adventitial fibroblasts into myofibroblasts. Lipids Health Dis 2016; 15:2. [PMID: 26729053 PMCID: PMC4700586 DOI: 10.1186/s12944-015-0174-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/29/2015] [Indexed: 12/15/2022] Open
Abstract
Background Pulmonary adventitial fibroblasts (PAFs) are activated under stress stimuli leading to their differentiation into myofibroblasts, which is involved in vessel remodeling. 15-HETE is known as an important factor in vessel remodeling under hypoxia; however, the role of 15-HETE in PAF phenotypic alteration is not clear. Results The effect of 15-HETE on PAF phenotypic alterations was investigated in the present study. PAFs were treated with 15-HETE (0.5 μM) for 24 h, and the myofibroblast marker α-smooth muscle actin (α-SMA) was analyzed. The 15-HETE induced α-SMA expression and cell morphology. 15-HETE upregulated FGF-2 levels in PAFs, and knockdown FGF-2 by siRNAs blocked the enhanced α-SMA expression induced by 15-HETE. p38 kinase was activated, and blocked depressed 15-HETE-induced FGF-2 expression. The downstream of p38 pathway, Egr-1 activation, was also raised by 15-HETE treatment, and silenced Egr-1 suppressed the 15-HETE-induced upregulation of FGF-2. TGF-β1 was upregulated with FGF-2 treatment, and α-SMA expression induced by FGF-2 was inhibited after the cell was transferred with TGF-β1 siRNA. Meanwhile, FGF-2 increased α-SMA expression and improved proliferation, which was associated with p27kip1 and cyclin E variation. Conclusions The above results suggest that p38/Egr-1 pathway-mediated FGF-2 is involved in 15-HETE-induced differentiation of PAFs into myofibroblasts and cell proliferation.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Xinyang Road 39, Daqing, Heilongjiang Province, 163319, China
| | - Yan Chen
- Daqing Qil Fields General Hospital, Heilongjiang, Daqing, Heilongjiang Province, 163319, China
| | - Guixia Li
- Department of Pharmacology, Harbin Medical University-Daqing, Xinyang Road 39, Daqing, Heilongjiang Province, 163319, China
| | - Minggang Chen
- Department of Pharmacology, Harbin Medical University-Daqing, Xinyang Road 39, Daqing, Heilongjiang Province, 163319, China
| | - Wei Huang
- Department of Pharmacology, Harbin Medical University-Daqing, Xinyang Road 39, Daqing, Heilongjiang Province, 163319, China
| | - Yanrui Liu
- Department of Pharmacology, Harbin Medical University-Daqing, Xinyang Road 39, Daqing, Heilongjiang Province, 163319, China
| | - Yumei Li
- Department of Pharmacology, Harbin Medical University-Daqing, Xinyang Road 39, Daqing, Heilongjiang Province, 163319, China. .,Biopharmaceutical Institute of the Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, 150081, China.
| |
Collapse
|