1
|
Du L, Zhu X, Jiang Z, Wang W, Liu P, Zhu L, Zhang F. Resveratrol inhibits ferroptosis in the lung tissues of heat stroke-induced rats via the Nrf2 pathway. BMC Pharmacol Toxicol 2024; 25:88. [PMID: 39563478 DOI: 10.1186/s40360-024-00810-1] [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: 01/19/2024] [Accepted: 10/30/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Heat stroke (HS) can lead to the development of pulmonary ferroptosis. The inhibition of pulmonary ferroptosis during HS improves patient prognosis. The aim of this study was to investigate the effects of resveratrol (RES) on heat stress at an ambient temperature of 42 °C. METHODS Heat stress was induced in Beas-2B cells and lung injury was induced in HS rats at an ambient temperature of 42 °C. The anti-oxidative stress and anti-ferroptotic effects of RES were confirmed through tail vein injection of nuclear factor-2 associated factor (Nrf2) shRNA recombinant adeno-associated virus 6 (AAV6-shNrf2). RESULTS RES treatment attenuated the upregulation of reactive oxygen species (ROS) and malondialdehyde (MDA) levels and alleviated glutathione inhibition in HS. In addition, RES treatment reduced the accumulation of Fe2+ in heat-stressed Beas-2B cells and increased the ferroptosis resistance-related proteins FTH1, GPX4, and SLC7A11 as well as the anti-oxidative stress pathway proteins Nrf2, NQO1, and HO-1. The antioxidant and anti-ferroptotic effects of RES in heat-stressed Beas-2B cells were effectively reversed upon treatment with Nrf2-IN-1, an Nrf2 pathway inhibitor. In the HS rat model, the antioxidant and anti-ferroptotic effects of RES were reversed by an ambient temperature of 42 °C and relative humidity of 60 ± 5%. CONCLUSIONS RES effectively protected HS rats from lung injury, inhibited the accumulation of Fe2+, ROS, and MDA in the lung, and upregulated FTH1, GPX4, SLC7A11, Nrf2, NQO1, and HO-1.
Collapse
Affiliation(s)
- Liwen Du
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China
| | - Xueqi Zhu
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China
| | - Zhenluo Jiang
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China
| | - Weidong Wang
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China
| | - Peng Liu
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China
| | - Leilei Zhu
- Department of Emergency, Ningbo No.2 Hospital. No. 41, Northwest Street, Haishu District, Ningbo, 315010, China.
| | - Fangqi Zhang
- Department of Pulmonary and Critical Care Medicine, The 987th Hospital of Joint Logistics Support Force of Chinese PLA, Baoji, 721000, China
| |
Collapse
|
2
|
Wang HT, Han JT, Hu DH. [Research advances on the role of acid fibroblast growth factor in promotion of wound healing]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2022; 38:859-863. [PMID: 36177591 DOI: 10.3760/cma.j.cn501120-20210811-00276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Acid fibroblast growth factor (aFGF) is a member of fibroblast growth factors (FGF) family, widely promoting embryonic development, wound healing, vascular regeneration, nerve injury repair, as well as regulating immune metabolism. Many pathophysiological processes, such as inflammation, neovascularization, proliferation and migration of repair cells, and deposition of collagen and other extracellular matrix are involved in the process of wound healing. Based on the relevant literature in recent years, this article mainly reviews the research progresses on the roles and mechanism of aFGF in biological signal transduction, regulation of cell growth, and involvement in tissue repair, and discusses the current research hot spots as well as the prospective future direction of clinical applications of aFGF in the aspect of clinical pharmacokinetics and safety.
Collapse
Affiliation(s)
- H T Wang
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - J T Han
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - D H Hu
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| |
Collapse
|
3
|
Zhu D, Zhang Q, Li Q, Wang G, Guo Z. Inhibition of AHNAK nucleoprotein 2 alleviates pulmonary fibrosis by downregulating the TGF-β1/Smad3 signaling pathway. J Gene Med 2022; 24:e3442. [PMID: 35882062 DOI: 10.1002/jgm.3442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/22/2022] [Accepted: 06/16/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic and advanced interstitial lung disease with poor prognosis. AHNAK nucleoprotein 2 (AHNAK2) is a macromolecular protein that is important for cell migration and muscle membrane repair. The protein acts via epithelial-mesenchymal transition (EMT), which is a key mechanism in the pathogenesis of IPF. However, very few studies have elucidated the effect of AHNAK2 in the development of IPF. Therefore, we aimed to determine the role of AHNAK2 in IPF development. METHODS C57BL/6 mice were induced with bleomycin (BLM), while A549 and Beas-2b pulmonary epithelial cell lines were treated with TGF-β1 to induce IPF model. The expression of AHNAK2 was detected using immunohistochemistry (IHC) staining in vivo, and real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting (WB) in vitro. C57BL/6 mice were injected with adeno-associated virus (AAV)-sh NC or AAV-sh AHNAK2; the pulmonary function and EMT markers expression were measured in mice. The migratory abilities of the two transforming growth factor beta 1 (TGF-β1)-induced cell lines were examined using wound-healing and Transwell assays after transfection with si-NC, si-AHNAK2-1 and -2. EMT markers expression was detected using RT-qPCR and WB. Smad3 and phosphorylated-Smad3 of the two cells were examined using WB. Following Smad3 inhibition by Smad3 phosphorylation inhibitor (SIS3), TGF-β1-induced cell migration and EMT markers expression were evaluated again after different transfections. RESULTS AHNAK2 expression was higher in the IPF model than in the normal model in vivo and in vitro. Partial inhibition of AHNAK2 suppressed the EMT process and improved pulmonary ventilation and compliance in the mouse model of IPF. Similarly, knockdown of AHNAK2 suppressed the migration of pulmonary epithelial cells and reversed EMT. Furthermore, Smad3 of the two TGF-β1-induced cell lines was not activated when AHNAK2 was inhibited. When SIS3 inhibited the activation of Smad3, the suppression of AHNAK2 had no effect on A549 and Beas-2b, regardless of TGF-β1 induction. CONCLUSIONS Inhibition of AHNAK2 alleviates pulmonary fibrosis and partially reverses EMT by inhibiting the TGF-β1/Smad3 signaling pathway. Therefore, AHNAK2 is a potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Dongyi Zhu
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qian Zhang
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qinchuan Li
- Department of Cardiothoracic Surgery, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guangxue Wang
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongliang Guo
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Roque Barboza W. Heat shock proteins in pulmonary fibrosis: pawns of cell homeostasis. Am J Physiol Cell Physiol 2022; 322:C1105-C1109. [PMID: 35508189 DOI: 10.1152/ajpcell.00073.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Idiopathic lung fibrosis (IPF) is a fatal disease that primarily affects the elderly. Up to date, the specific pathophysiology of IPF remains unknown. However, it is theorized to be caused by chronic repetitive injuries to the alveolar epithelium, eventually exhausting the stem cell capacity and activating pathological pathways. Heat shock proteins (HSPs), a category of stress response proteins, are also suggested to contribute to IPF pathophysiology. Furthermore, HSPs are key components in the regulation of cell homeostasis and act as chaperones for a multitude of new proteins. This review thoroughly evaluates the roles that specific HSPs, HSP90, HSP70, and HSP47, have in the fibrotic process. A close look into the roles of these HSPs in IPF pathophysiology will give valuable insight into the future of IPF treatment and prevention.
Collapse
Affiliation(s)
- Willy Roque Barboza
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
5
|
Chen K, Rao Z, Dong S, Chen Y, Wang X, Luo Y, Gong F, Li X. Roles of the fibroblast growth factor signal transduction system in tissue injury repair. BURNS & TRAUMA 2022; 10:tkac005. [PMID: 35350443 PMCID: PMC8946634 DOI: 10.1093/burnst/tkac005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Following injury, tissue autonomously initiates a complex repair process, resulting in either partial recovery or regeneration of tissue architecture and function in most organisms. Both the repair and regeneration processes are highly coordinated by a hierarchy of interplay among signal transduction pathways initiated by different growth factors, cytokines and other signaling molecules under normal conditions. However, under chronic traumatic or pathological conditions, the reparative or regenerative process of most tissues in different organs can lose control to different extents, leading to random, incomplete or even flawed cell and tissue reconstitution and thus often partial restoration of the original structure and function, accompanied by the development of fibrosis, scarring or even pathogenesis that could cause organ failure and death of the organism. Ample evidence suggests that the various combinatorial fibroblast growth factor (FGF) and receptor signal transduction systems play prominent roles in injury repair and the remodeling of adult tissues in addition to embryonic development and regulation of metabolic homeostasis. In this review, we attempt to provide a brief update on our current understanding of the roles, the underlying mechanisms and clinical application of FGFs in tissue injury repair.
Collapse
Affiliation(s)
| | | | - Siyang Dong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of breast surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yajing Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xulan Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongde Luo
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Fanghua Gong
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Xiaokun Li
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| |
Collapse
|
6
|
Prudovsky I. Cellular Mechanisms of FGF-Stimulated Tissue Repair. Cells 2021; 10:cells10071830. [PMID: 34360000 PMCID: PMC8304273 DOI: 10.3390/cells10071830] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/10/2023] Open
Abstract
Growth factors belonging to the FGF family play important roles in tissue and organ repair after trauma. In this review, I discuss the regulation by FGFs of the aspects of cellular behavior important for reparative processes. In particular, I focus on the FGF-dependent regulation of cell proliferation, cell stemness, de-differentiation, inflammation, angiogenesis, cell senescence, cell death, and the production of proteases. In addition, I review the available literature on the enhancement of FGF expression and secretion in damaged tissues resulting in the increased FGF supply required for tissue repair.
Collapse
Affiliation(s)
- Igor Prudovsky
- Maine Medical Center Research Institute, 81 Research Dr., Scarborough, ME 04074, USA
| |
Collapse
|
7
|
Lipocalin 10 as a New Prognostic Biomarker in Sepsis-Induced Myocardial Dysfunction and Mortality: A Pilot Study. Mediators Inflamm 2021; 2021:6616270. [PMID: 34121925 PMCID: PMC8166480 DOI: 10.1155/2021/6616270] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/23/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction Sepsis-induced myocardial dysfunction (SIMD) is the most common complications of sepsis and septic shock with extremely high incidence and mortality. Lipocalin 10 (Lcn10) has recently been identified as a potential biomarker for heart failure, yet its relation to sepsis has not been investigated. The purpose of this study was to explore whether circulating Lcn10 could be used as a prognostic tool in patients with SIMD. Methods In this single-center observational pilot study, seventy-five sepsis patients were enrolled after sepsis diagnosis or ICU admission (45.3% female, median age 60 years), and 35 patients (46.7%) developed myocardial dysfunction. Serum Lcn10 levels of septic patients were measured using the enzyme-linked immunosorbent assay (ELISA) at the time of admission. Other biomarkers of cardiac function and Lcn10 concentration were compared between SIMD and non-SIMD groups. Results We observed that the median Lcn10 levels were 2.780 ng/mL in patients with SIMD and 2.075 ng/mL in patients without SIMD (P < 0.05). The area under the receiver operating characteristic (ROC) curve for the diagnosis of SIMD was 0.797 (P < 0.05). In addition, elevated serum Lcn10 levels at the time of admission were positively associated with 28-day mortality in septic patients. Conclusions Our study indicates that circulating Lcn10 levels may serve as a novel biomarker for the diagnosis and prognosis of myocardial dysfunction induced by sepsis. An additional large multicenter study may be warranted to confirm the findings of this study.
Collapse
|
8
|
Li C, Deng C, Zhou T, Hu J, Dai B, Yi F, Tian N, Jiang L, Dong X, Zhu Q, Zhang S, Cui H, Cao L, Shang Y. MicroRNA-370 carried by M2 macrophage-derived exosomes alleviates asthma progression through inhibiting the FGF1/MAPK/STAT1 axis. Int J Biol Sci 2021; 17:1795-1807. [PMID: 33994863 PMCID: PMC8120458 DOI: 10.7150/ijbs.59715] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/12/2021] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence has suggested the functions of exosomes in allergic diseases including asthma. By using a mouse model with asthma induced by ovalbumin (OVA), we explored the roles of M2 macrophage-derived exosomes (M2Φ-Exos) in asthma progression. M2Φ-Exos significantly alleviated OVA-induced fibrosis and inflammatory responses in mouse lung tissues, as well as inhibited abnormal proliferation, invasion, and fibrosis-related protein production in platelet derived growth factor (PDGF-BB) treated primary mouse airway smooth muscle cells (ASMCs). The OVA administration in mice or the PDGF-BB treatment in ASMCs reduced the expression of miR-370, which was detected in M2Φ-Exos by miRNA sequencing. However, treating the mice or ASMCs with M2Φ-Exos reversed the inhibitory effect of OVA or PDGF-BB on miR-370 expression. We identified that the target of miR-370 was fibroblast growth factor 1 (FGF1). Downregulation of miR-370 by Lv-miR-370 inhibitor or overexpression of FGF1 by Lv-FGF1 blocked the protective roles of M2Φ-Exos in asthma-like mouse and cell models. M2Φ-Exos were found to inactivate the MAPK signaling pathway, which was recovered by miR-370 inhibition or FGF1 overexpression. Collectively, we conclude that M2Φ-Exos carry miR-370 to alleviate asthma progression through downregulating FGF1 expression and the MAPK/STAT1 signaling pathway. Our study may offer a novel insight into asthma treatment.
Collapse
Affiliation(s)
- Chunlu Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Chengsi Deng
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Tingting Zhou
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Jiapeng Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Bing Dai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Fei Yi
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Na Tian
- Jilin Tuohua Biotechnology Co., Ltd. Changchun, Jilin 13000, China
| | - Lijun Jiang
- Jilin Tuohua Biotechnology Co., Ltd. Changchun, Jilin 13000, China
| | - Xiang Dong
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Qingfeng Zhu
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Siyi Zhang
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Hongyan Cui
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Liu Cao
- College of Basic Medicine Science, China Medical University, Shenyang 110122, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
9
|
Li C, Dai B, Hu J, Shang Y. WITHDRAWN: M2 macrophage-derived exosomes carry microRNA-370 to alleviate asthma progression through inhibiting the FGF1/MAPK/STAT1 axis. Exp Cell Res 2020:112285. [PMID: 32941809 DOI: 10.1016/j.yexcr.2020.112285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 01/16/2023]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Chunlu Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, P.R. China
| | - Bing Dai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, P.R. China
| | - Jiapeng Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, P.R. China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, P.R. China
| |
Collapse
|
10
|
Chen L, Yang Y, Peng X, Yan H, Zhang X, Yin L, Yu H. Transcription factor YY1 inhibits the expression of THY1 to promote interstitial pulmonary fibrosis by activating the HSF1/miR-214 axis. Aging (Albany NY) 2020; 12:8339-8351. [PMID: 32396525 PMCID: PMC7244040 DOI: 10.18632/aging.103142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/31/2020] [Indexed: 04/07/2023]
Abstract
Interstitial pulmonary fibrosis (IPF) is a progressive disease of diverse etiology manifesting with proliferation of lung fibroblasts and accumulation of extracellular matrix deposition in pulmonary interstitium. Recent studies show aberrant expression of mRNAs and microRNAs (miRNAs) in human embryonic pulmonary fibroblasts (HEPFs). In this study, we investigated effects of the YY1/HSF1/miR-214/THY1 axis on the functions of HEPFs and IPF. Loss- and gain-of-function tests were conducted to identify roles of YY1, HSF1, miR-214, and THY1 in IPF. As determined by RT-qPCR or western blot assay, silencing YY1 down-regulated HSF1 expression and attenuated the expression of pro-proliferative and fibrosis markers in HEPFs. Meanwhile, viability of HEPFs was impeded by YY1 knockdown. The binding relationship between miR-214 and THY1 was verified using dual-luciferase reporter assay. In HEPFs, down-regulation of HSF1 reduced miR-214 expression to repress proliferation and fibrogenic transformation of HEPFs, while inhibition of miR-214 expression could restrain the fibrogenic transformation property of HEPFs by up-regulating THY1. Subsequently, IPF model in mice was induced by bleomycin treatment. These animal experiments validated the protective effects of YY1 knockdown against IPF-induced lung pathological manifestations, which could be reversed by THY1 knockdown. Our study demonstrates the important involvement of YY1/HSF1/miR-214/THY1 axis in the development of IPF.
Collapse
Affiliation(s)
- Lin Chen
- Department of Respiratory and Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, P.R. China
| | - Yang Yang
- Department of Respiratory and Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, P.R. China
| | - Xiaying Peng
- Department of Respiratory and Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, P.R. China
| | - Haiying Yan
- Department of Respiratory and Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, P.R. China
| | - Xin Zhang
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, P.R. China
| | - Lin Yin
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, P.R. China
| | - Hua Yu
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, P.R. China
| |
Collapse
|
11
|
Ni S, Song M, Guo W, Guo T, Shen Q, Peng H. Biomarkers and their potential functions in idiopathic pulmonary fibrosis. Expert Rev Respir Med 2020; 14:593-602. [PMID: 32187497 DOI: 10.1080/17476348.2020.1745066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Idiopathic pulmonary fibrosis (IPF) is a chronic, devastating, and progressive lung disease that is characterized by fibrosis and respiratory failure. IPF holds high morbidity and poor prognosis and still faces considerable problems of reliable diagnosis and valid prognosis. A growing body of literature have reported changes in the level of various biomarkers in IPF patients, which means that they are expected to become a new tool for the clinical practice of IPF.Areas covered: We reviewed the recent literature about biomarkers and focus on the role they play in IPF. We systematically searched Medline/PubMed through February 2020. Many works of literature have shown that a variety of biomolecules and genomics played multiple roles in the diagnosis or differential diagnosis, prognosis, and indication of acute deterioration of IPF and so on.Expert opinion: Significant advances have been made in the role of biomarkers for IPF these years; however, current data indicate that a single biomarker is unlikely to have a transformative effect on clinical practice; therefore, the combined effect of various biomarkers can be considered to improve the accuracy of diagnosis and prognosis. Further research of biomarkers may provide new insights for the diagnosis, prognosis, and even therapy of IPF.
Collapse
Affiliation(s)
- Shanshan Ni
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| | - Min Song
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| | - Wei Guo
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| | - Ting Guo
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| | - Qinxue Shen
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| | - Hong Peng
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
12
|
Lou Q, Li Y, Hou B, Liu Y, Zhang Y, Hao J, Ma Y. Heat shock transcription factor 1 affects kidney tubular cell migration by regulating the TGF‑β1‑Smad2/3 signaling pathway. Mol Med Rep 2019; 20:4323-4330. [PMID: 31545442 DOI: 10.3892/mmr.2019.10689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 08/23/2019] [Indexed: 11/06/2022] Open
Abstract
Cell migration is important for renal recovery from tubular cell injury. Heat shock transcription factor 1 (HSF1) is a well‑studied regulatory factor that is active during acute kidney injury. HSF1 is also involved in the migration process during tumor metastasis. Therefore, we hypothesized that HSF1 may promote the recovery of renal function by affecting kidney tubular cell migration. A wound healing assay was used to examine the cell migration rate. The results demonstrated that the migration of rat kidney proximal tubular cells (RPTCs) was increased following knockdown of HSF1. In addition, the invasion ability of HSF1 knockdown RPTCs was also significantly upregulated. The present study also identified that transforming growth factor‑β1 (TGF‑β1) was highly expressed at the edge of the wound in control cells, and its expression was further increased upon knockdown of HSF1. Inhibition of TGF‑β1 signaling prevented RPTC HSF1 knockdown cell migration, suggesting that HSF1‑regulated RPTC cell migration was dependent on the TGF‑β1 signaling pathway. Furthermore, phosphorylation of TGF‑β1 and Smad2/3 was induced in HSF1 knockdown cells. Together, these results suggest that HSF1 may suppress RPTC migration by inhibiting the activation of the TGF‑β1‑Smad2/3 signaling pathway.
Collapse
Affiliation(s)
- Qiang Lou
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yuanyuan Li
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Beibei Hou
- International Office of Henan University, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yonglian Liu
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yan Zhang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Jielu Hao
- Department of Nephrology, Changzheng Hospital Affiliated to Second Military Medical University, Shanghai 200003, P.R. China
| | - Yuanfang Ma
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| |
Collapse
|
13
|
Potla R, Tulapurkar ME, Luzina IG, Atamas SP, Singh IS, Hasday JD. Exposure to febrile-range hyperthermia potentiates Wnt signalling and epithelial-mesenchymal transition gene expression in lung epithelium. Int J Hyperthermia 2017; 34:1-10. [PMID: 28540808 DOI: 10.1080/02656736.2017.1316875] [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: 10/19/2022] Open
Abstract
BACKGROUND As environmental and body temperatures vary, lung epithelial cells experience temperatures significantly different from normal core temperature. Our previous studies in human lung epithelium showed that: (i) heat shock accelerates wound healing and activates profibrotic gene expression through heat shock factor-1 (HSF1); (ii) HSF1 is activated at febrile temperatures (38-41 °C) and (iii) hypothermia (32 °C) activates and hyperthermia (39.5 °C) reduces expression of a subset of miRNAs that target protein kinase-Cα (PKCα) and enhance proliferation. METHODS We analysed the effect of hypo- and hyperthermia exposure on Wnt signalling by exposing human small airway epithelial cells (SAECs) and HEK293T cells to 32, 37 or 39.5 °C for 24 h, then analysing Wnt-3a-induced epithelial-mesenchymal transition (EMT) gene expression by qRT-PCR and TOPFlash reporter plasmid activity. Effects of miRNA mimics and inhibitors and the HSF1 inhibitor, KNK437, were evaluated. RESULTS Exposure to 39.5 °C for 24 h increased subsequent Wnt-3a-induced EMT gene expression in SAECs and Wnt-3a-induced TOPFlash activity in HEK293T cells. Increased Wnt responsiveness was associated with HSF1 activation and blocked by KNK437. Overexpressing temperature-responsive miRNA mimics reduced Wnt responsiveness in 39.5 °C-exposed HEK293T cells, but inhibitors of the same miRNAs failed to restore Wnt responsiveness in 32 °C-exposed HEK293T cells. CONCLUSIONS Wnt responsiveness, including expression of genes associated with EMT, increases after exposure to febrile-range temperature through an HSF1-dependent mechanism that is independent of previously identified temperature-dependent miRNAs. This process may be relevant to febrile fibrosing lung diseases, including the fibroproliferative phase of acute respiratory distress syndrome (ARDS) and exacerbations of idiopathic pulmonary fibrosis (IPF).
Collapse
Affiliation(s)
- Ratnakar Potla
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Mohan E Tulapurkar
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Irina G Luzina
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA.,b Medicine and Research Services, Baltimore Veterans Affairs Medical Care System , Baltimore , MD , USA
| | - Sergei P Atamas
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA.,b Medicine and Research Services, Baltimore Veterans Affairs Medical Care System , Baltimore , MD , USA
| | - Ishwar S Singh
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA
| | - Jeffrey D Hasday
- a Department of Medicine , University of Maryland School of Medicine , Baltimore , MD , USA.,b Medicine and Research Services, Baltimore Veterans Affairs Medical Care System , Baltimore , MD , USA
| |
Collapse
|