1
|
Cañizo CG, Guerrero-Ramos F, Perez Escavy M, Lodewijk I, Suárez-Cabrera C, Morales L, Nunes SP, Munera-Maravilla E, Rubio C, Sánchez R, Rodriguez-Izquierdo M, Martínez de Villarreal J, Real FX, Castellano D, Martín-Arriscado C, Lora Pablos D, Rodríguez Antolín A, Dueñas M, Paramio JM, Martínez VG. Characterisation of the tumour microenvironment and PD-L1 granularity reveals the prognostic value of cancer-associated myofibroblasts in non-invasive bladder cancer. Oncoimmunology 2025; 14:2438291. [PMID: 39698899 DOI: 10.1080/2162402x.2024.2438291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/29/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
High-risk non-muscle-invasive bladder cancer (NMIBC) presents high recurrence and progression rates. Despite the use of Bacillus Calmette-Guérin gold-standard immunotherapy and the recent irruption of anti-PD-1/PD-L1 drugs, we are missing a comprehensive understanding of the tumor microenvironment (TME) that may help us find biomarkers associated to treatment outcome. Here, we prospectively analyzed TME composition and PD-L1 expression of tumor and non-tumoral tissue biopsies from 73 NMIBC patients and used scRNA-seq, transcriptomic cohorts and tissue micro-array to validate the prognostic value of cell types of interest. Compared to non-tumoral tissue, NMIBC presented microvascular alterations, increased cancer-associated fibroblast (CAF) and myofibroblast (myoCAF) presence, and varied immune cell distribution, such as increased macrophage infiltration. Heterogeneous PD-L1 expression was observed across subsets, with macrophages showing the highest expression levels, but cancer cells as the primary potential anti-PD-L1 binding targets. Unbiased analysis revealed that myoCAF and M2-like macrophages are specifically enriched in high-grade NMIBC tumors. The topological distribution of these two cell types changed as NMIBC progresses, as shown by immunofluorescence. Only myoCAFs were associated with higher rates of progression and recurrence in three independent cohorts (888 total patients), reaching prediction values comparable to transcriptomic classes, which we further validated using tissue micro-array. Our study provides a roadmap to establish the landscape of the NMIBC TME, highlighting myoCAFs as potential prognostic markers.
Collapse
Affiliation(s)
- Carmen G Cañizo
- Urology Department, University Hospital '12 de Octubre', Madrid, Spain
| | | | - Mercedes Perez Escavy
- Molecular and Translational Oncology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Institute of Biomedical Research, University Hospital '12 de Octubre', Madrid, Spain
| | - Iris Lodewijk
- Molecular and Translational Oncology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Institute of Biomedical Research, University Hospital '12 de Octubre', Madrid, Spain
| | - Cristian Suárez-Cabrera
- Molecular and Translational Oncology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Institute of Biomedical Research, University Hospital '12 de Octubre', Madrid, Spain
| | - Lucía Morales
- Molecular and Translational Oncology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Institute of Biomedical Research, University Hospital '12 de Octubre', Madrid, Spain
| | - Sandra P Nunes
- Molecular and Translational Oncology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Institute of Biomedical Research, University Hospital '12 de Octubre', Madrid, Spain
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network) Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
| | - Ester Munera-Maravilla
- Molecular and Translational Oncology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Institute of Biomedical Research, University Hospital '12 de Octubre', Madrid, Spain
| | - Carolina Rubio
- Molecular and Translational Oncology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Institute of Biomedical Research, University Hospital '12 de Octubre', Madrid, Spain
| | - Rebeca Sánchez
- Cell Technology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Raras (CIBERER), Madrid, Spain
| | | | - Jaime Martínez de Villarreal
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain
| | - Francisco X Real
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Epithelial Carcinogenesis Group, Spanish National Cancer Centre-CNIO, Madrid, Spain
- Departament de Medicina i Ciències de la Vida, Universitat Pompeu Fabra, Barcelona, Spain
| | - Daniel Castellano
- Oncology Department, University Hospital '12 de Octubre', Madrid, Spain
| | | | - David Lora Pablos
- Scientific Support Unit, Research Institute I+12, University Hospital 12 de Octubre, Madrid, Spain
| | | | - Marta Dueñas
- Molecular and Translational Oncology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Institute of Biomedical Research, University Hospital '12 de Octubre', Madrid, Spain
| | - Jesús M Paramio
- Molecular and Translational Oncology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Institute of Biomedical Research, University Hospital '12 de Octubre', Madrid, Spain
| | - Victor G Martínez
- Molecular and Translational Oncology Division, Biomedical Innovation Unit, CIEMAT, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
- Institute of Biomedical Research, University Hospital '12 de Octubre', Madrid, Spain
| |
Collapse
|
2
|
Han Q, Yu J, Gao Z, Li S, Wang Z, Wu J, Huang F. Polypyrrole-modified gelatin-based hydrogel: A dressing for intestinal perforation treatment with enhanced wound healing and anti-adhesion properties. Int J Biol Macromol 2025; 309:142738. [PMID: 40180106 DOI: 10.1016/j.ijbiomac.2025.142738] [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: 05/18/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Intestinal perforation is a serious medical emergency, and traditional surgery often causes adhesion and other complications. Innovative hydrogels improve postoperative care and rehabilitation with their anti-adhesion, antibacterial, and hemostatic properties. We have developed an advanced anti-adhesion hydrogel, AA-A30, composed of polypyrrole-modified gelatin (PPy-GelMA), carboxymethyl chitosan (CMCS), and NHS-functionalized polyethylene glycol (PEG-NHS). This hydrogel is specifically tailored for intestinal perforations. Upon hydrolysis, PEG-NHS forms a protective barrier that effectively prevents adhesion to surrounding normal tissues. Furthermore, the integration of PPy-GelMA significantly extends the degradation duration of the hydrogel, from 24 to 48 h. In a mouse model of intestinal perforation, the AA-A30 hydrogel demonstrated remarkable efficacy in inhibiting inflammation and preventing tissue adhesion by modulating the expression of both inflammatory and tissue adhesion-related factors, such as IL-1β, TNF-α, and the ratio of tPA to PAI-1. These findings underscore the considerable potential of AA-A30 for the therapeutic management of intestinal perforations.
Collapse
Affiliation(s)
- Qingyue Han
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Jingrong Yu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Zhengkun Gao
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Sitong Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Zi Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Jie Wu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China
| | - Fengjie Huang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, Jiangsu Province, PR China.
| |
Collapse
|
3
|
Delrue C, Eisenga MF, Delanghe JR, Speeckaert MM. Personalized Antifibrotic Therapy in CKD Progression. J Pers Med 2024; 14:1141. [PMID: 39728054 DOI: 10.3390/jpm14121141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Chronic kidney disease (CKD) is a chronic disorder characterized by kidney fibrosis and extracellular matrix accumulation that can lead to end-stage kidney disease. Epithelial-to-mesenchymal transition, inflammatory cytokines, the TGF-β pathway, Wnt/β-catenin signaling, the Notch pathway, and the NF-κB pathway all play crucial roles in the progression of fibrosis. Current medications, such as renin-angiotensin-aldosterone system inhibitors, try to delay disease development but do not stop or reverse fibrosis. This review emphasizes the growing need for tailored antifibrotic medications for CKD treatment. Precision medicine, which combines proteomic, metabolomic, and genetic data, provides a practical way to personalize treatment regimens. Proteomic signatures, such as CKD273, and genetic markers, such as APOL1 and COL4A5, help in patient stratification and focused therapy development. Two recently developed antifibrotic medications, nintedanib and pirfenidone, have been proven to diminish fibrosis in preclinical animals. Additionally, research is being conducted on the efficacy of investigational drugs targeting CTGF and galectin-3 in the treatment of kidney fibrosis.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Michele F Eisenga
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9712 CP Groningen, The Netherlands
| | - Joris R Delanghe
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium
- Research Foundation-Flanders (FWO), 1000 Brussels, Belgium
| |
Collapse
|
4
|
Bakalenko N, Kuznetsova E, Malashicheva A. The Complex Interplay of TGF-β and Notch Signaling in the Pathogenesis of Fibrosis. Int J Mol Sci 2024; 25:10803. [PMID: 39409132 PMCID: PMC11477142 DOI: 10.3390/ijms251910803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Fibrosis is a major medical challenge, as it leads to irreversible tissue remodeling and organ dysfunction. Its progression contributes significantly to morbidity and mortality worldwide, with limited therapeutic options available. Extensive research on the molecular mechanisms of fibrosis has revealed numerous factors and signaling pathways involved. However, the interactions between these pathways remain unclear. A comprehensive understanding of the entire signaling network that drives fibrosis is still missing. The TGF-β and Notch signaling pathways play a key role in fibrogenesis, and this review focuses on their functional interplay and molecular mechanisms. Studies have shown synergy between TGF-β and Notch cascades in fibrosis, but antagonistic interactions can also occur, especially in cardiac fibrosis. The molecular mechanisms of these interactions vary depending on the cell context. Understanding these complex and context-dependent interactions is crucial for developing effective strategies for treating fibrosis.
Collapse
Affiliation(s)
| | | | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, St-Petersburg 194064, Russia; (N.B.); (E.K.)
| |
Collapse
|
5
|
Lahane GP, Dhar A, Bhat A. Therapeutic approaches and novel antifibrotic agents in renal fibrosis: A comprehensive review. J Biochem Mol Toxicol 2024; 38:e23795. [PMID: 39132761 DOI: 10.1002/jbt.23795] [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: 01/25/2024] [Revised: 06/20/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
Renal fibrosis (RF) is one of the underlying pathological conditions leading to progressive loss of renal function and end-stage renal disease (ESRD). Over the years, various therapeutic approaches have been explored to combat RF and prevent ESRD. Despite significant advances in understanding the underlying molecular mechanism(s), effective therapeutic interventions for RF are limited. Current therapeutic strategies primarily target these underlying mechanisms to halt or reverse fibrotic progression. Inhibition of transforming growth factor-β (TGF-β) signaling, a pivotal mediator of RF has emerged as a central strategy to manage RF. Small molecules, peptides, and monoclonal antibodies that target TGF-β receptors or downstream effectors have demonstrated potential in preclinical models. Modulating the renin-angiotensin system and targeting the endothelin system also provide established approaches for controlling fibrosis-related hemodynamic changes. Complementary to pharmacological strategies, lifestyle modifications, and dietary interventions contribute to holistic management. This comprehensive review aims to summarize the underlying mechanisms of RF and provide an overview of the therapeutic strategies and novel antifibrotic agents that hold promise in its treatment.
Collapse
Affiliation(s)
- Ganesh Panditrao Lahane
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad, Telangana, India
| | - Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad, Telangana, India
| | - Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir, India
| |
Collapse
|
6
|
Shi L, Hu Y, Zeng H, Shi H, Xu W, Sun Y, Chu H, Ji C, Qian H. Mesenchymal stem cell-derived extracellular vesicles ameliorate renal interstitial fibrosis via the miR-13474/ADAM17 axis. Sci Rep 2024; 14:17703. [PMID: 39085289 PMCID: PMC11291924 DOI: 10.1038/s41598-024-67339-5] [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: 04/23/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Renal interstitial fibrosis (RIF) is a prevalent consequence of chronic renal diseases, characterized by excessive extracellular matrix (ECM) deposition. A Disintegrin and Metalloprotease 17 (ADAM17), a transmembrane metalloproteinase, plays a central role in driving renal fibrosis progression by activating Notch 1 protein and the downstream TGF-β signaling pathway. Our study investigated potential therapeutic interventions for renal fibrosis, focusing on human umbilical cord mesenchymal stem cell-derived extracellular vesicles (hucMSC-EVs). We found that hucMSC-EVs inhibit ADAM17, thereby impeding renal fibrosis progression. Analysis of hucMSC-EVs miRNA profiles revealed significant enrichment of miR-13474, which effectively targeted and inhibited ADAM17 mRNA expression, subsequently suppressing Notch1 activation, TGF-β signaling, and collagen deposition. Overexpression of miR-13474 enhanced hucMSC-EVs' inhibitory effect on renal fibrosis, while its downregulation abolished this protective effect. Our findings highlight the efficacy of hucMSC-EVs overexpressing miR-13474 in mitigating renal fibrosis via ADAM17 targeting. These insights offer potential therapeutic strategies for managing renal fibrosis.
Collapse
Affiliation(s)
- Linru Shi
- Center for Molecular & Imageology of Jiangsu University, Division of Nephrology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214200, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Yuyan Hu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
- Shaoxing Central Hospital Medical Alliance General Hospital, The Department of Laboratory, Shaoxing, 312030, Zhejiang, China
| | - Houcheng Zeng
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Hui Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Yaoxiang Sun
- Center for Molecular & Imageology of Jiangsu University, Division of Nephrology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214200, Jiangsu, China
| | - Hong Chu
- Center for Molecular & Imageology of Jiangsu University, Division of Nephrology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214200, Jiangsu, China.
| | - Cheng Ji
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| | - Hui Qian
- Center for Molecular & Imageology of Jiangsu University, Division of Nephrology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, 214200, Jiangsu, China.
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
7
|
Gowripriya T, Meharaj Afrin K, Paurna M, Yashwanth R, Bhaskar JP, Suresh R, Balamurugan K. Regulation of miR-61 and col-19 via TGF-β and Notch signalling in Caenorhabditis elegans against Klebsiella aerogenes infection. Microb Pathog 2024; 186:106505. [PMID: 38122874 DOI: 10.1016/j.micpath.2023.106505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/06/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Klebsiella aerogenes, previously known as Enterobacter aerogenes, is a gram-negative bacterium typically present in the gastrointestinal tract. While numerous studies reported the pathogenicity and drug resistance of this bacterium there remains a lack of comprehensive research on K. aerogenes induced alterations in the host cellular mechanisms. In this study, we identify a previously uncharacterized C. elegans miR-61 that defines an evolutionarily conserved miRNA important for development and innate immunity regulation through Notch and TGF-β signaling pathway. We employed C. elegans wild-type (N2) as well as mutant strains, such as TGF-β (sma-6) and notch-signaling pathway mutants (adm-4 and mir-61). Our results have demonstrated that the K. aerogenes infected mutants exhibited significantly reduced survival rate, reduced pharyngeal pumping, altered swimming and chemotactic behavior. Moreover, K. aerogenes affects the healthspan by increasing ROS level in the mutants. The gene expression analysis revealed that K. aerogenes upregulated egl-30, tph-1 and sod-1 in adm-4, mir-61 mutants not in sma-6. The in-silico analysis indicated an interaction between mir-61 and col-19, which was confirmed by the upregulation of miR-61 expression and the downregulation of col-19 in sma-6, adm-4, and wild-type strains. These findings suggest that C. elegans activates mir-61 and col-19 regulation through the Notch and TGF-β signaling pathway against K. aerogenes infection.
Collapse
Affiliation(s)
- Thirumugam Gowripriya
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, 630 003, India.
| | | | - Manikandan Paurna
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, 630 003, India.
| | - Radhakrishnan Yashwanth
- ITC Life Sciences and Technology Centre, Peenya Industrial Area, Bangalore, 560 058, Karnataka, India.
| | - James Prabhanand Bhaskar
- ITC Life Sciences and Technology Centre, Peenya Industrial Area, Bangalore, 560 058, Karnataka, India.
| | - Ramamurthi Suresh
- ITC Life Sciences and Technology Centre, Peenya Industrial Area, Bangalore, 560 058, Karnataka, India.
| | | |
Collapse
|
8
|
Liao T, Xiong L, Wang X, Yang S, Liang Z. Mitochondrial disorders as a mechanism for the development of obese Sarcopenia. Diabetol Metab Syndr 2023; 15:224. [PMID: 37926816 PMCID: PMC10626707 DOI: 10.1186/s13098-023-01192-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023] Open
Abstract
Obese sarcopenia is a severe and prevalent disease in an aging society. Compared to sarcopenia alone, the development and advanced stage of obesity sarcopenia is faster and more severe. Diagnosis of the cause of adipocyte accumulation is also more complicated; however, no effective pharmacological treatment is available. Chronic inflammation is one of the causes of sarcopenia, and obese patients, who are more likely to develop chronic inflammation, may simultaneously suffer from obesity and sarcopenia. Mitochondrial metabolic disorders have been more easily observed in the tissue cells of patients with obesity and sarcopenia. Mitochondrial metabolic disorders include abnormal mtDNA release, mitochondrial autophagy, and dynamic mitochondrial disorders. Therefore, this review will reveal the mechanism of development of obesity myasthenia gravis from the perspective of mitochondria and discuss the currently existing small-molecule drugs.
Collapse
Affiliation(s)
- Tingfeng Liao
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
- Department of Geriatrics, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
| | - Lijiao Xiong
- Department of Geriatrics, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
| | - Xiaohao Wang
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
- Department of Geriatrics, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
| | - Shu Yang
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China.
- Department of Geriatrics, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.
| | - Zhen Liang
- School of Medicine, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China.
- Department of Geriatrics, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
9
|
Huang R, Fu P, Ma L. Kidney fibrosis: from mechanisms to therapeutic medicines. Signal Transduct Target Ther 2023; 8:129. [PMID: 36932062 PMCID: PMC10023808 DOI: 10.1038/s41392-023-01379-7] [Citation(s) in RCA: 230] [Impact Index Per Article: 115.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/12/2023] [Accepted: 02/20/2023] [Indexed: 03/19/2023] Open
Abstract
Chronic kidney disease (CKD) is estimated to affect 10-14% of global population. Kidney fibrosis, characterized by excessive extracellular matrix deposition leading to scarring, is a hallmark manifestation in different progressive CKD; However, at present no antifibrotic therapies against CKD exist. Kidney fibrosis is identified by tubule atrophy, interstitial chronic inflammation and fibrogenesis, glomerulosclerosis, and vascular rarefaction. Fibrotic niche, where organ fibrosis initiates, is a complex interplay between injured parenchyma (like tubular cells) and multiple non-parenchymal cell lineages (immune and mesenchymal cells) located spatially within scarring areas. Although the mechanisms of kidney fibrosis are complicated due to the kinds of cells involved, with the help of single-cell technology, many key questions have been explored, such as what kind of renal tubules are profibrotic, where myofibroblasts originate, which immune cells are involved, and how cells communicate with each other. In addition, genetics and epigenetics are deeper mechanisms that regulate kidney fibrosis. And the reversible nature of epigenetic changes including DNA methylation, RNA interference, and chromatin remodeling, gives an opportunity to stop or reverse kidney fibrosis by therapeutic strategies. More marketed (e.g., RAS blockage, SGLT2 inhibitors) have been developed to delay CKD progression in recent years. Furthermore, a better understanding of renal fibrosis is also favored to discover biomarkers of fibrotic injury. In the review, we update recent advances in the mechanism of renal fibrosis and summarize novel biomarkers and antifibrotic treatment for CKD.
Collapse
Affiliation(s)
- Rongshuang Huang
- Kidney Research Institute, Division of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Fu
- Kidney Research Institute, Division of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Liang Ma
- Kidney Research Institute, Division of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Shen Qi Wan-Containing Serum Alleviates Renal Interstitial Fibrosis via Restraining Notch1-Mediated Epithelial-Mesenchymal Transition. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:3352353. [PMID: 36793762 PMCID: PMC9925256 DOI: 10.1155/2023/3352353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/04/2022] [Accepted: 11/15/2022] [Indexed: 02/09/2023]
Abstract
Objective Shen Qi Wan (SQW) is the most classic prescription for the clinical therapy of chronic kidney disease in China. Nevertheless, the function of SQW in renal interstitial fibrosis (RIF) has not been clearly clarified. Our purpose was to explore the protective function of SQW on RIF. Methods After intervention with SQW-containing serum alone at increasing concentrations (2.5, 5, and 10%) or in combination with siNotch1, the transforming growth factor-beta (TGF-β)-induced HK-2 cell viability, extracellular matrix (ECM)-, epithelial-mesenchymal transition (EMT), and Notch1 pathway-associated protein expressions were assessed by cell counting kit-8, qRT-PCR, western blot, and immunofluorescence assays. Results SQW-containing serum intensified the viability of TGF-β-mediated HK-2 cells. Besides, it augmented the collagen II and E-cadherin levels, and weakened the fibronectin, α-SMA, vimentin, N-cadherin, and collagen I levels in HK-2 cells triggered by TGF-β. Moreover, it is found that TGF-β led to the upregulation of Notch1, Jag1, HEY1, HES1, and TGF-β in HK-2 cells, which was partially offset by SQW-containing serum. Furthermore, cotreatment of SQW-containing serum and Notch1 knockdown further apparently alleviated the Notch1, vimentin, N-cadherin, collagen I, and fibronectin levels in HK-2 cells induced by TGF-β. Conclusion Collectively, these findings elucidated that SQW-containing serum attenuated RIF via restraining EMT through the repression of the Notch1 pathway.
Collapse
|
11
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
12
|
Wu Y, Liang M, Huang F, Cheng OH, Xiao X, Lee TH, Truong L, Cheng J. Notch Blockade Specifically in Bone Marrow-Derived FSP-1-Positive Cells Ameliorates Renal Fibrosis. Cells 2023; 12:cells12020214. [PMID: 36672147 PMCID: PMC9856686 DOI: 10.3390/cells12020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The infiltration of inflammatory cells during a kidney injury stimulates myofibroblast activation leading to kidney fibrosis. Fibroblast-specific protein 1 (FSP-1) positive cells have been reported as either myofibroblasts or monocytes during tissue fibrosis. The functions of FSP-1+ cells that are associated with the development of renal fibrosis and the signaling pathways that regulate FSP-1+ cell activation have not been well defined. METHODS In mice with unilateral ureteral obstruction (UUO), we characterized FSP-1+ cells and determined the role of the Notch signaling pathway in the activation of bone marrow-derived FSP-1+ cells during kidney fibrosis. RESULTS In kidneys from mice with UUO, the FSP-1+ cells accumulated significantly in the tubulointerstitial area. By using immunostaining and FSP-1 reporter mice, we found that FSP-1 was co-stained with inflammatory cell markers, but not myofibroblast markers. Results from mice with bone marrow transplantations showed that FSP-1+ cells in obstructed kidneys represent a bone marrow-derived population of inflammatory cells. In cultured FSP-1+ cells, the inhibition of Notch signaling suppressed the activation and cytokine secretion of FSP-1+ cells that were induced by LPS but not by IL-4. The specific KO or blockade of Notch signaling in bone marrow-derived FSP-1+ cells suppressed UUO-induced ECM deposition, the infiltration of FSP-1+ inflammatory cells, and cytokine production. These responses ameliorated myofibroblast accumulation and renal fibrosis in obstructed kidneys. CONCLUSION Our study reveals that most FSP-1+ cells in obstructed kidneys are activated macrophages that are derived from bone marrow and that Notch signaling activates the production of M1 cytokines in FSP-1+ monocytes/macrophages, which is important for renal inflammation and fibrosis.
Collapse
Affiliation(s)
- Yongdong Wu
- Department of Nephrology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ming Liang
- Department of Nephrology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: (M.L.); (J.C.); Tel.: +1-713-798-2698 (J.C.); Fax: +1-713-798-5010 (J.C.)
| | - Fengzhang Huang
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Owen H. Cheng
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiaoguang Xiao
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tae Hoon Lee
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Luan Truong
- Department of Pathology, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Jizhong Cheng
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: (M.L.); (J.C.); Tel.: +1-713-798-2698 (J.C.); Fax: +1-713-798-5010 (J.C.)
| |
Collapse
|
13
|
Yang T, Wu E, Zhu X, Leng Y, Ye S, Dong R, Liu J, Zhong J, Zheng Y, Xu W, Luo J, Kong L, Zhang H. TKF, a mexicanolide-type limonoid derivative, suppressed hepatic stellate cells activation and liver fibrosis through inhibition of the YAP/Notch3 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154466. [PMID: 36182796 DOI: 10.1016/j.phymed.2022.154466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/02/2022] [Accepted: 09/18/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Liver fibrosis is a common scarring response and may ultimately lead to liver cancer, unfortunately, there is currently no effective antifibrotic drug approved for human use. Limonoids exhibit a broad spectrum of biological activities; however, the potential role of limonoids against fibrosis is largely unknown. PURPOSE This study investigates the antifibrotic activities and potential mechanisms of TKF (3-tigloyl-khasenegasin F), a natural mexicanolide-type limonoid derivative. STUDY DESIGN/METHODS Two well-established mouse models (CCl4 challenge and bile duct ligation) were used to assess anti-fibrotic effects of TKF in vivo. Human hepatic stellate cell (HSC) line LX-2 and mouse primary hepatic stellate cells (pHSCs) also served as in vitro liver fibrosis models. RESULT TKF administration significantly attenuated hepatic histopathological injury and collagen accumulation and suppressed fibrogenesis-associated gene expression including Col1a1, Acta2, and Timp1. In LX-2 cells and mouse pHSCs, TKF dose-dependently suppressed HSC activation and the expression levels of fibrogenic markers. Mechanistic studies showed that TKF inhibited Notch3-Hes1 and YAP signalings in vivo and in vitro. Furthermore, YAP inhibition or knockdown downregulated the Notch3 expression; however, Notch3 inhibition or knockdown did not affect the level of YAP in activated HSC. We revealed that TKF inhibited Notch3-Hes1 activation and downregulated hepatic fibrogenic gene expression via inhibiting YAP. CONCLUSION The therapeutic benefit of TKF against liver fibrosis results from inhibition of YAP and Notch3-Hes1 pathways, indicating that TKF may be a novel therapeutic candidate for liver fibrosis.
Collapse
Affiliation(s)
- Ting Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Enyi Wu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaoyun Zhu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yingrong Leng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shengtao Ye
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ruirui Dong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiaman Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiawen Zhong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ying Zheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenjun Xu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Luo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Hao Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
14
|
Li CW, Yu K, Shyh-Chang N, Jiang Z, Liu T, Ma S, Luo L, Guang L, Liang K, Ma W, Miao H, Cao W, Liu R, Jiang LJ, Yu SL, Li C, Liu HJ, Xu LY, Liu RJ, Zhang XY, Liu GS. Pathogenesis of sarcopenia and the relationship with fat mass: descriptive review. J Cachexia Sarcopenia Muscle 2022; 13:781-794. [PMID: 35106971 PMCID: PMC8977978 DOI: 10.1002/jcsm.12901] [Citation(s) in RCA: 255] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/26/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023] Open
Abstract
Age-associated obesity and muscle atrophy (sarcopenia) are intimately connected and are reciprocally regulated by adipose tissue and skeletal muscle dysfunction. During ageing, adipose inflammation leads to the redistribution of fat to the intra-abdominal area (visceral fat) and fatty infiltrations in skeletal muscles, resulting in decreased overall strength and functionality. Lipids and their derivatives accumulate both within and between muscle cells, inducing mitochondrial dysfunction, disturbing β-oxidation of fatty acids, and enhancing reactive oxygen species (ROS) production, leading to lipotoxicity and insulin resistance, as well as enhanced secretion of some pro-inflammatory cytokines. In turn, these muscle-secreted cytokines may exacerbate adipose tissue atrophy, support chronic low-grade inflammation, and establish a vicious cycle of local hyperlipidaemia, insulin resistance, and inflammation that spreads systemically, thus promoting the development of sarcopenic obesity (SO). We call this the metabaging cycle. Patients with SO show an increased risk of systemic insulin resistance, systemic inflammation, associated chronic diseases, and the subsequent progression to full-blown sarcopenia and even cachexia. Meanwhile in many cardiometabolic diseases, the ostensibly protective effect of obesity in extremely elderly subjects, also known as the 'obesity paradox', could possibly be explained by our theory that many elderly subjects with normal body mass index might actually harbour SO to various degrees, before it progresses to full-blown severe sarcopenia. Our review outlines current knowledge concerning the possible chain of causation between sarcopenia and obesity, proposes a solution to the obesity paradox, and the role of fat mass in ageing.
Collapse
Affiliation(s)
- Chun-Wei Li
- Department of Clinical Nutrition & Health Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kang Yu
- Department of Clinical Nutrition & Health Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ng Shyh-Chang
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zongmin Jiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Taoyan Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shilin Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lanfang Luo
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lu Guang
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Kun Liang
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenwu Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hefan Miao
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenhua Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ruirui Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ling-Juan Jiang
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Song-Lin Yu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Li
- Department of General Surgery, Tianjin Union Medical Center, The Affiliated Hospital of Nankai University, China (Tianjin Union Medical Center, Tianjin, China
| | - Hui-Jun Liu
- Department of nursing & Clinical Nutrition, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Long-Yu Xu
- Department of Sport Physiatry, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rong-Ji Liu
- Department of Pharmacy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Yuan Zhang
- Department of stomatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gao-Shan Liu
- Department of Health Education, Shijingshan Center for Disease Prevention and Control, Beijing, China
| |
Collapse
|
15
|
Gao J, Hao Y, Piao X, Gu X. Aldehyde Dehydrogenase 2 as a Therapeutic Target in Oxidative Stress-Related Diseases: Post-Translational Modifications Deserve More Attention. Int J Mol Sci 2022; 23:ijms23052682. [PMID: 35269824 PMCID: PMC8910853 DOI: 10.3390/ijms23052682] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023] Open
Abstract
Aldehyde dehydrogenase 2 (ALDH2) has both dehydrogenase and esterase activity; its dehydrogenase activity is closely related to the metabolism of aldehydes produced under oxidative stress (OS). In this review, we recapitulate the enzyme activity of ALDH2 in combination with its protein structure, summarize and show the main mechanisms of ALDH2 participating in metabolism of aldehydes in vivo as comprehensively as possible; we also integrate the key regulatory mechanisms of ALDH2 participating in a variety of physiological and pathological processes related to OS, including tissue and organ fibrosis, apoptosis, aging, and nerve injury-related diseases. On this basis, the regulatory effects and application prospects of activators, inhibitors, and protein post-translational modifications (PTMs, such as phosphorylation, acetylation, S-nitrosylation, nitration, ubiquitination, and glycosylation) on ALDH2 are discussed and prospected. Herein, we aimed to lay a foundation for further research into the mechanism of ALDH2 in oxidative stress-related disease and provide a basis for better use of the ALDH2 function in research and the clinic.
Collapse
Affiliation(s)
- Jie Gao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (J.G.); (Y.H.)
| | - Yue Hao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (J.G.); (Y.H.)
| | - Xiangshu Piao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Xianhong Gu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (J.G.); (Y.H.)
- Correspondence:
| |
Collapse
|
16
|
Huang S, Fu D, Wan Z, Li M, Li H, Chong T. Effects of a gamma-secretase inhibitor of notch signalling on transforming growth factor β1-induced urethral fibrosis. J Cell Mol Med 2021; 25:8796-8808. [PMID: 34363303 PMCID: PMC8435429 DOI: 10.1111/jcmm.16837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/10/2021] [Accepted: 07/24/2021] [Indexed: 11/30/2022] Open
Abstract
Urethral stricture (US) is a common disorder of the lower urinary tract in men caused by fibrosis. The recurrence rate of US is high; however, there are no effective therapies to prevent or treat urethral fibrosis. The pathogenesis of urethral fibrosis involves myofibroblast activation and excessive extracellular matrix (ECM) deposition. The molecular mechanisms underlying this pathological activation are not completely understood. It has been demonstrated that Notch signalling contributes to the development of fibrosis and inflammation. However, whether this contributes to urethral fibrosis remains unclear. In this study, activation of Notch signalling was observed in patients with US. Additionally, it was noted that activation of Notch signalling promoted ECM production and myofibroblast activation in human urethral scar fibroblasts (HUSFs) treated with transforming growth factor (TGF) β1. However, the Notch inhibitor N‐[N‐(3,5‐difluorophenacetyl)‐L‐alanyl]‐S‐phenylglycine t‐butyl ester (DAPT) suppressed activation of Notch signalling as well as proliferation and migration of the TGFβ1‐treated HUSFs. Additionally, DAPT ameliorated TGFβ1‐induced urethral fibrosis in Sprague Dawley rats by suppressing ECM production, myofibroblast activation and the TGFβ signalling pathway. These findings demonstrate that Notch signalling may be a promising and potential target in the prevention or treatment of urethral fibrosis.
Collapse
Affiliation(s)
- Shanlong Huang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Delai Fu
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ziyan Wan
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Min Li
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hecheng Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tie Chong
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
17
|
Zhang Y, Jin D, Kang X, Zhou R, Sun Y, Lian F, Tong X. Signaling Pathways Involved in Diabetic Renal Fibrosis. Front Cell Dev Biol 2021; 9:696542. [PMID: 34327204 PMCID: PMC8314387 DOI: 10.3389/fcell.2021.696542] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
Diabetic kidney disease (DKD), as the most common complication of diabetes mellitus (DM), is the major cause of end-stage renal disease (ESRD). Renal interstitial fibrosis is a crucial metabolic change in the late stage of DKD, which is always considered to be complex and irreversible. In this review, we discuss the pathological mechanisms of diabetic renal fibrosis and discussed some signaling pathways that are closely related to it, such as the TGF-β, MAPK, Wnt/β-catenin, PI3K/Akt, JAK/STAT, and Notch pathways. The cross-talks among these pathways were then discussed to elucidate the complicated cascade behind the tubulointerstitial fibrosis. Finally, we summarized the new drugs with potential therapeutic effects on renal fibrosis and listed related clinical trials. The purpose of this review is to elucidate the mechanisms and related pathways of renal fibrosis in DKD and to provide novel therapeutic intervention insights for clinical research to delay the progression of renal fibrosis.
Collapse
Affiliation(s)
- Yuqing Zhang
- Endocrinology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - De Jin
- Endocrinology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaomin Kang
- Endocrinology Department, Guang'anmen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Rongrong Zhou
- Endocrinology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuting Sun
- Endocrinology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengmei Lian
- Endocrinology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolin Tong
- Endocrinology Department, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
18
|
Liu Y, Su YY, Yang Q, Zhou T. Stem cells in the treatment of renal fibrosis: a review of preclinical and clinical studies of renal fibrosis pathogenesis. Stem Cell Res Ther 2021; 12:333. [PMID: 34112221 PMCID: PMC8194041 DOI: 10.1186/s13287-021-02391-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/14/2021] [Indexed: 02/05/2023] Open
Abstract
Renal fibrosis commonly leads to glomerulosclerosis and renal interstitial fibrosis and the main pathological basis involves tubular atrophy and the abnormal increase and excessive deposition of extracellular matrix (ECM). Renal fibrosis can progress to chronic kidney disease. Stem cells have multilineage differentiation potential under appropriate conditions and are easy to obtain. At present, there have been some studies showing that stem cells can alleviate the accumulation of ECM and renal fibrosis. However, the sources of stem cells and the types of renal fibrosis or renal fibrosis models used in these studies have differed. In this review, we summarize the pathogenesis (including signaling pathways) of renal fibrosis, and the effect of stem cell therapy on renal fibrosis as described in preclinical and clinical studies. We found that stem cells from various sources have certain effects on improving renal function and alleviating renal fibrosis. However, additional clinical studies should be conducted to confirm this conclusion in the future.
Collapse
Affiliation(s)
- Yiping Liu
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China
| | - Yan-Yan Su
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou, China
| | - Qian Yang
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China.
| |
Collapse
|
19
|
Condorelli AG, El Hachem M, Zambruno G, Nystrom A, Candi E, Castiglia D. Notch-ing up knowledge on molecular mechanisms of skin fibrosis: focus on the multifaceted Notch signalling pathway. J Biomed Sci 2021; 28:36. [PMID: 33966637 PMCID: PMC8106838 DOI: 10.1186/s12929-021-00732-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Fibrosis can be defined as an excessive and deregulated deposition of extracellular matrix proteins, causing loss of physiological architecture and dysfunction of different tissues and organs. In the skin, fibrosis represents the hallmark of several acquired (e.g. systemic sclerosis and hypertrophic scars) and inherited (i.e. dystrophic epidermolysis bullosa) diseases. A complex series of interactions among a variety of cellular types and a wide range of molecular players drive the fibrogenic process, often in a context-dependent manner. However, the pathogenetic mechanisms leading to skin fibrosis are not completely elucidated. In this scenario, an increasing body of evidence has recently disclosed the involvement of Notch signalling cascade in fibrosis of the skin and other organs. Despite its apparent simplicity, Notch represents one of the most multifaceted, strictly regulated and intricate pathways with still unknown features both in health and disease conditions. Starting from the most recent advances in Notch activation and regulation, this review focuses on the pro-fibrotic function of Notch pathway in fibroproliferative skin disorders describing molecular networks, interplay with other pro-fibrotic molecules and pathways, including the transforming growth factor-β1, and therapeutic strategies under development.
Collapse
Affiliation(s)
- Angelo Giuseppe Condorelli
- Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant' Onofrio 4, 00165, Rome, Italy.
| | - May El Hachem
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant' Onofrio 4, 00165, Rome, Italy
| | - Giovanna Zambruno
- Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant' Onofrio 4, 00165, Rome, Italy
| | - Alexander Nystrom
- Department of Dermatology, Medical Faculty, Medical Center, University of Freiburg, Freiburg, Germany
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy.,IDI-IRCCS, via Monti di Creta 104, 00167, Rome, Italy
| | - Daniele Castiglia
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, via Monti di Creta 104, 00167, Rome, Italy
| |
Collapse
|
20
|
Chen L, Zhang X, Yu Z, Song Y, Wang T, Yang K, Li S, Wang J, Su Y, Song B. Inhibition of Notch Intracellular Domain Suppresses Cell Activation and Fibrotic Factors Production in Hypertrophic Scar Fibroblasts Versus Normal Skin Fibroblasts. Ann Plast Surg 2021; 86:400-405. [PMID: 32881749 DOI: 10.1097/sap.0000000000002540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Hypertrophic scar (HS) is the most common complication after skin injury with unknown etiopathogenesis. There is increasing evidence to suggest that aberrant Notch signaling contributes directly to skin pathogenesis and altered expression of the Notch intracellular domain (NICD) identified in HS. Therefore, the aim of this study was to investigate the effects of Notch signaling pathway in HS pathogenesis. METHODS Hypertrophic scar and normal skin samples were collected. Notch intracellular domain expression was detected by immunohistochemistry staining and fibroblasts were separated from the samples. We compared fibrotic factors production, cell viability, migration and apoptosis of HS fibroblasts (HFB) versus normal skin fibroblasts (NFB) by real time quantitative polymerase chain reaction, MTS, cell scratch assay and flow cytometry respectively under the impact of inhibition of Notch signaling by NICD-small-interfering RNA (SiRNA). RESULTS The results showed that NICD was overexpressed in the dermis of HS tissues. Inhibition of Notch signaling by NICD-SiRNA suppressed the production of the fibrotic factors including collagen 1, collagen 3, α-SMA, and TGF-β1 by HFB and NFB. Cell viability and migration were reduced in NICD-SiRNA-treated NFB and HFB, whereas cell apoptosis was enhanced by NICD-SiRNA. CONCLUSIONS Conclusively, the study demonstrates a potential role for Notch signaling in HS progression, and targeting this pathway may provide a novel strategy for treatment of HS.
Collapse
Affiliation(s)
- Lin Chen
- From the Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Molecular Mechanisms of Renal Progenitor Regulation: How Many Pieces in the Puzzle? Cells 2021; 10:cells10010059. [PMID: 33401654 PMCID: PMC7823786 DOI: 10.3390/cells10010059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/26/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
Kidneys of mice, rats and humans possess progenitors that maintain daily homeostasis and take part in endogenous regenerative processes following injury, owing to their capacity to proliferate and differentiate. In the glomerular and tubular compartments of the nephron, consistent studies demonstrated that well-characterized, distinct populations of progenitor cells, localized in the parietal epithelium of Bowman capsule and scattered in the proximal and distal tubules, could generate segment-specific cells in physiological conditions and following tissue injury. However, defective or abnormal regenerative responses of these progenitors can contribute to pathologic conditions. The molecular characteristics of renal progenitors have been extensively studied, revealing that numerous classical and evolutionarily conserved pathways, such as Notch or Wnt/β-catenin, play a major role in cell regulation. Others, such as retinoic acid, renin-angiotensin-aldosterone system, TLR2 (Toll-like receptor 2) and leptin, are also important in this process. In this review, we summarize the plethora of molecular mechanisms directing renal progenitor responses during homeostasis and following kidney injury. Finally, we will explore how single-cell RNA sequencing could bring the characterization of renal progenitors to the next level, while knowing their molecular signature is gaining relevance in the clinic.
Collapse
|
22
|
Abd El-Rhman RH, El-Naga RN, Gad AM, Tadros MG, Hassaneen SK. Dibenzazepine Attenuates Against Cisplatin-Induced Nephrotoxicity in Rats: Involvement of NOTCH Pathway. Front Pharmacol 2020; 11:567852. [PMID: 33381027 PMCID: PMC7768080 DOI: 10.3389/fphar.2020.567852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 11/06/2020] [Indexed: 02/05/2023] Open
Abstract
Cisplatin is one of the standard anti-cancer agents that are used to treat variety of solid tumors. Nevertheless, due to the accumulation of cisplatin in the renal epithelial cells, nephrotoxicity was found to be the main side effect that limits its clinical use. The current study was conducted to assess the potential nephroprotective effect of dibenzazepine, a Notch inhibitor, against cisplatin-induced nephrotoxicity in rats as well as the possible mechanisms underlying this nephroprotection. The rats were pre-treated with 2 mg/kg dibenzazepine for 7 days before giving a single nephrotoxic dose of cisplatin (7 mg/kg). Cisplatin induced acute nephrotoxicity, where blood urea nitrogen and serum creatinine levels were significantly increased. Besides, lipid peroxidation was markedly elevated and the levels of reduced glutathione and catalase were significantly reduced. Also, the tissue levels of the pro-inflammatory mediators; IL-1β, TNF-α, and NF-kB, were significantly increased in the cisplatin group. The pre-treatment with dibenzazepine significantly mitigated the nephrotoxic effects of cisplatin, the oxidative stress and inflammatory status as well as decreased caspase-3 expression, as compared to the cisplatin group. Furthermore, the up-regulation of Notch-1 and Hes-1 was found to be involved in cisplatin-induced nephrotoxicity and their expression was significantly reduced by dibenzazepine. The nephroprotective effect of dibenzazepine was further confirmed by the histopathological assessment. Moreover, dibenzazepine pre-treatment of hela and PC3 cells in vitro did not antagonize the cisplatin anti-cancer activity. In conclusion, these findings show that dibenzazepine provides protection against cisplatin-induced nephrotoxicity. Moreover, the up-regulation of the Notch pathway was shown to play a role in the pathogenesis of cisplatin-induced renal injury.
Collapse
Affiliation(s)
| | - Reem N. El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amany M. Gad
- Department of Pharmacology, Egyptian Drug Authority (ED), Giza, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University, East Kantara Branch, New City, El Ismailia, Egypt
| | - Mariane G. Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | | |
Collapse
|
23
|
Tang H, Guo Y, Zhang Z, Li Z, Zhang Y, Li Y, Kang X, Han R. Integrative analysis of long non-coding RNA and mRNA in broilers with valgus-varus deformity. PLoS One 2020; 15:e0239450. [PMID: 32970714 PMCID: PMC7514040 DOI: 10.1371/journal.pone.0239450] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 09/07/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Bone abnormality and leg disease in commercial broiler flocks are increasingly prominent, causing serious economic losses to the broiler breeding industry. Valgus-varus deformity (VVD) is a common deformity of the long bone in broilers that manifests as an outward or inward deviation of the tibiotarsus or tarsometatarsus. There is a paucity of studies on the molecular mechanisms of VVD. RESULTS In this study, 6 cDNA libraries were constructed from spleen samples from VVD birds and normal birds. A total of 1951 annotated lncRNAs, 7943 novel lncRNAs and 30252 mRNAs were identified by RNA-sequencing. In addition, 420 differentially expressed (DE) mRNAs and 124 differentially expressed lncRNAs (adjusted P-value < 0.05) were obtained. A total of 16 dysregulated genes were confirmed by qPCR to be consistent with the results of the RNA-Seq. The functional lncRNA-mRNA co-expression network was constructed using differentially expressed mRNAs and target genes of the differentially expressed lncRNAs. 11 DE genes were obtained from the analysis. In order to gain insight into the interactions of genes, lncRNAs and pathways associated with VVD, we focused on the following pathways, which are involved in immunity and bone development: the Jak-stat signaling pathway, Toll-like receptor signaling pathway, Wnt-signaling pathway, mTOR signaling pathway, VEGF signaling pathway, Notch signaling pathway, TGF-beta signaling pathway and Fanconi anemia pathway. All together, 30 candidate DE genes were obtained from these pathways. We then analyzed the interaction between the DE genes and their corresponding lncRNAs. From these interaction network analyses we found that GARS, NFIC, PIK3R1, BMP6, NOTCH1, ACTB and CREBBP were the key core nodes of these networks. CONCLUSION This study showed that differentially expressed genes and signaling pathways were related to immunity or bone development. These results increase the understanding of the molecular mechanisms of VVD and provide some reference for the etiology and pathogenesis of VVD.
Collapse
Affiliation(s)
- Hehe Tang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yaping Guo
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Zhenzhen Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Zhuanjian Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou, China
| | - Yanhua Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yuanfang Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xiangtao Kang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou, China
| | - Ruili Han
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Zhengzhou, China
| |
Collapse
|
24
|
Wu J, Dong X, Li W, Zhao L, Zhou L, Sun S, Li H. Dibenzazepine promotes cochlear supporting cell proliferation and hair cell regeneration in neonatal mice. Cell Prolif 2020; 53:e12872. [PMID: 32677724 PMCID: PMC7507434 DOI: 10.1111/cpr.12872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/04/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Objectives To investigate the role of dibenzazepine (DBZ) in promoting supporting cell (SC) proliferation and hair cell (HC) regeneration in the inner ear. Materials and Methods Postnatal day 1 wild‐type or neomycin‐damaged mouse cochleae were cultured with DBZ. Immunohistochemistry and scanning electron microscopy were used to examine the morphology of cochlear cells, and high‐throughput RNA‐sequencing was used to measure gene expression levels. Results We found that DBZ promoted SC proliferation and HC regeneration in a dose‐dependent manner in both normal and damaged cochleae. In addition, most of the newly regenerated HCs induced by DBZ had visible and relatively mature stereocilia bundle structures. Finally, RNA sequencing detected the differentially expressed genes between DBZ treatment and controls, and interaction networks were constructed for the most highly differentially expressed genes. Conclusions Our study demonstrates that DBZ can significantly promote SC proliferation and increase the number of mitotically regenerated HCs with relatively mature stereocilia bundles in the neonatal mouse cochlea by inhibiting Notch signalling and activating Wnt signalling, suggesting the DBZ might be a new therapeutic target for stimulating HC regeneration.
Collapse
Affiliation(s)
- Jingfang Wu
- Otorhinolaryngology Department of Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Biomedical Sciences, Fudan University School of Basic Medical Sciences, Shanghai, China
| | - Xinran Dong
- Molecular Medical Center, Children's Hospital of Fudan University, Shanghai, China
| | - Wen Li
- Otorhinolaryngology Department of Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Biomedical Sciences, Fudan University School of Basic Medical Sciences, Shanghai, China
| | - Liping Zhao
- Otorhinolaryngology Department of Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Biomedical Sciences, Fudan University School of Basic Medical Sciences, Shanghai, China
| | - Li Zhou
- Shanghai High School, Shanghai, China
| | - Shan Sun
- Otorhinolaryngology Department of Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Biomedical Sciences, Fudan University School of Basic Medical Sciences, Shanghai, China
| | - Huawei Li
- Otorhinolaryngology Department of Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Biomedical Sciences, Fudan University School of Basic Medical Sciences, Shanghai, China.,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Fan J, Shen W, Lee SR, Mathai AE, Zhang R, Xu G, Gillies MC. Targeting the Notch and TGF-β signaling pathways to prevent retinal fibrosis in vitro and in vivo. Am J Cancer Res 2020; 10:7956-7973. [PMID: 32724452 PMCID: PMC7381727 DOI: 10.7150/thno.45192] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022] Open
Abstract
Rationale: The Notch and transforming growth factor-β (TGFβ) signaling pathways are two intracellular mechanisms that control fibrosis in general but whether they play a major role in retinal fibrosis is less clear. Here we study how these two signaling pathways regulate Müller cell-dominated retinal fibrosis in vitro and in vivo. Methods: Human MIO-M1 Müller cells were treated with Notch ligands and TGFβ1, either alone or in combination. Western blots were performed to study changes in γ-secretase proteases, Notch downstream effectors, endogenous TGFβ1, phosphorylated Smad3 (p-Smad3) and extracellular matrix (ECM) proteins. We also studied the effects of RO4929097, a selective γ-secretase inhibitor, on expression of ECM proteins after ligand stimulation. Müller cell viability was studied by AlamarBlue and cytotoxicity by lactate cytotoxicity assays. Finally, we studied changes in Notch and TGFβ signaling and tested the effect of intravitreal injections of the Notch pathway inhibitor RO4929097 on retinal fibrosis resulted from Sodium iodate (NaIO3)-induced retinal injury in mice. We also studied the safety of intravitreal injections of RO4929097 in normal mice. Results: Treatment of Müller cells with Notch ligands upregulated γ-secretase proteases and Notch downstream effectors, with increased expression of endogenous TGFβ1, TGFβ receptors and p-Smad3. TGFβ1 upregulated the expression of proteins associated with both signaling pathways in a similar manner. Notch ligands and TGFβ1 had additive effects on overexpression of ECM proteins in Müller cells which were inhibited by RO4929097. Notch and TGFβ ligands stimulated Müller cell proliferation which was inhibited by RO4929097 without damaging the cells. NaIO3-induced retinal injury activated both Notch and TGFβ signaling pathways in vivo. Intravitreal injection of RO4929097 prevented Müller cell gliosis and inhibited overexpression of ECM proteins in this murine model. We found no safety concerns for up to 17 days after an intravitreal injection of RO4929097. Conclusions: Inhibiting Notch signaling might be an effective way to prevent retinal fibrosis. This study is of clinical significance in developing a treatment for preventing fibrosis in proliferative vitreoretinopathy, proliferative diabetic retinopathy and wet age-related macular degeneration.
Collapse
|
26
|
Abudukeyoumu A, Li MQ, Xie F. Transforming growth factor-β1 in intrauterine adhesion. Am J Reprod Immunol 2020; 84:e13262. [PMID: 32379911 DOI: 10.1111/aji.13262] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023] Open
Abstract
Intrauterine adhesion (IUA), led by trauma to the basal layer, can prevent the endometrium from growing, resulting in complications in females, such as infertility and amenorrhea. Transforming growth factor-β1 (TGF-β1) plays a crucial role in inducing and promoting the differentiation and proliferation of mesenchymal cells, in the secretion of extracellular matrix-associated components, and is a major cytokine in initiating and terminating tissue repair downstream of the TGF-β/Smad signaling pathway. Some evidence supports that TGF-β1 is closely associated with the occurrence and development of IUA, and is regarded as an early risk factor of disease recurrence. Furthermore, the role of TGF-β1 has been demonstrated to be potentially regulated by a variety of cytokines, hormones, enzymes, and microRNAs. This review provides an overview of the expression, function, and regulation of TGF-β1 in IUA, with a brief discussion and perspectives on its future clinical implications on the diagnosis and treatment of IUA.
Collapse
Affiliation(s)
- Ayitila Abudukeyoumu
- Laboratory for Reproductive Immunology, Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Medical Center of Diagnosis and Treatment for Cervical Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Feng Xie
- Laboratory for Reproductive Immunology, Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Medical Center of Diagnosis and Treatment for Cervical Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Zhu QQ, Yang XY, Zhang XJ, Yu CJ, Pang QQ, Huang YW, Wang XJ, Sheng J. EGCG targeting Notch to attenuate renal fibrosisviainhibition of TGFβ/Smad3 signaling pathway activation in streptozotocin-induced diabetic mice. Food Funct 2020; 11:9686-9695. [DOI: 10.1039/d0fo01542c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
EGCG may improve renal fibrosis by targeting Notchviainhibition of the TGFβ/Smad3 pathway in diabetic mice.
Collapse
Affiliation(s)
- Qiang-Qiang Zhu
- Key Laboratory of Pu-er Tea Science
- Ministry of Education
- Yunnan Agricultural University
- Kunming
- China
| | - Xiao-Ying Yang
- Key Laboratory of Pu-er Tea Science
- Ministry of Education
- Yunnan Agricultural University
- Kunming
- China
| | - Xiao-Juan Zhang
- Key Laboratory of Pu-er Tea Science
- Ministry of Education
- Yunnan Agricultural University
- Kunming
- China
| | - Cai-Jun Yu
- Key Laboratory of Pu-er Tea Science
- Ministry of Education
- Yunnan Agricultural University
- Kunming
- China
| | - Qian-Qian Pang
- Key Laboratory of Pu-er Tea Science
- Ministry of Education
- Yunnan Agricultural University
- Kunming
- China
| | - Ye-wei Huang
- Key Laboratory of Pu-er Tea Science
- Ministry of Education
- Yunnan Agricultural University
- Kunming
- China
| | - Xuan-jun Wang
- Key Laboratory of Pu-er Tea Science
- Ministry of Education
- Yunnan Agricultural University
- Kunming
- China
| | - Jun Sheng
- Key Laboratory of Pu-er Tea Science
- Ministry of Education
- Yunnan Agricultural University
- Kunming
- China
| |
Collapse
|
28
|
Epithelial and interstitial Notch1 activity contributes to the myofibroblastic phenotype and fibrosis. Cell Commun Signal 2019; 17:145. [PMID: 31718671 PMCID: PMC6849313 DOI: 10.1186/s12964-019-0455-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022] Open
Abstract
Background Notch1 signalling is a stem-cell-related pathway that is essential for embryonic development, tissue regeneration and organogenesis. However, the role of Notch1 in the formation of myofibroblasts and fibrosis in kidneys following injury remains unknown. Methods The activity of Notch1 signalling was evaluated in fibrotic kidneys in CKD patients and in ureteral obstructive models in vivo and in cultured fibroblasts and TECs in vitro. In addition, the crosstalk of Notch1 with TGF-β1/Smad2/3 signalling was also investigated. Results Notch1 activity was elevated in fibrotic kidneys of rat models and patients with chronic kidney disease (CKD). Further study revealed that epithelial and interstitial Notch1 activity correlated with an α-SMA-positive myofibroblastic phenotype. In vitro, injury stimulated epithelial Notch1 activation and epithelial-mesenchymal transition (EMT), resulting in matrix deposition in tubular epithelial cells (TECs). Additionally, interstitial Notch1 activation in association with fibroblast-myofibroblast differentiation (FMD) in fibroblasts mediated a myofibroblastic phenotype. These TGF-β1/Smad2/3-dependent phenotypic transitions were abolished by Notch1 knockdown or a specific antagonist, DAPT, and were exacerbated by Notch1 overexpression or an activator Jagged-1-Fc chimaera protein. Interestingly, as a major driving force behind the EMT and FMD, TGF-β1, also induced epithelial and interstitial Notch1 activity, indicating that TGF-β1 may engage in crosstalk with Notch1 signalling to trigger fibrogenesis. Conclusion These findings suggest that epithelial and interstitial Notch1 activation in kidneys following injury contributes to the myofibroblastic phenotype and fibrosis through the EMT in TECs and to the FMD in fibroblasts by targeting downstream TGF-β1/Smad2/3 signalling.
Collapse
|
29
|
Chen JX, Yi XJ, Gu PL, Gao SX. The role of KDR in intrauterine adhesions may involve the TGF-β1/Smads signaling pathway. ACTA ACUST UNITED AC 2019; 52:e8324. [PMID: 31596310 PMCID: PMC6787948 DOI: 10.1590/1414-431x20198324] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/30/2019] [Indexed: 01/10/2023]
Abstract
The aim of this study was to investigate the role of kinase-insert domain-containing receptor (KDR) in intrauterine adhesions (IUA) and its mechanism. The Case group consisted of 92 patients diagnosed with IUA, and the Control group included 86 patients with uterine septum who had normal endometrium verified with an uteroscope. In addition, 50 rats were randomly assigned into Control, Sham, Model, NC-siRNA, and KDR-siRNA groups. Rats in the Model, NC-siRNA, and KDR-siRNA groups were induced by uterine curettage and lipopolysaccharide (LPS) treatment to establish the IUA model. Then, immunohistochemistry was applied for detection of VEGF and KDR expression, HE staining was used for observation of the endometrial morphology and gland counting, Masson staining for measurement of the degree of endometrial fibrosis, and qRT-PCR and western blot for the expression of KDR, VEGF, MMP-9, as well as TGF-β1/Smads pathway-related proteins. Compared with the Control group, the mRNA and protein expressions of KDR were significantly higher in IUA endometrial tissues, and the expression of KDR was positively correlated to the severity of IUA. In addition, the injection of si-KDR increased the number of endometrial glands, reduced the area of fibrosis, inhibited mRNA and protein expression of KDR and VEGF, up-regulated the expression of MMP-9 and Smad7, and decreased the expression level of TGF-β1, p-Smad2, p-Smad3, and Smad4 in rats with IUA. Highly-expressed KDR was related to patients' severity of IUA, and silencing KDR may prevent the occurrence and development of IUA via TGF-β1/Smads signaling pathway and up-regulating the expression of MMP-9.
Collapse
Affiliation(s)
- Jian Xia Chen
- Department of Reproductive Medicine, Linyi Central Hospital, Linyi City, Shandong, China
| | - Xi Juan Yi
- Department of Reproductive Medicine, Linyi Central Hospital, Linyi City, Shandong, China
| | - Pei Ling Gu
- Department of Reproductive Medicine, Linyi Central Hospital, Linyi City, Shandong, China
| | - Shan Xia Gao
- Department of Reproductive Medicine, Linyi Central Hospital, Linyi City, Shandong, China
| |
Collapse
|
30
|
Notch Signaling Inhibition by LY411575 Attenuates Osteoblast Differentiation and Decreased Ectopic Bone Formation Capacity of Human Skeletal (Mesenchymal) Stem Cells. Stem Cells Int 2019; 2019:3041262. [PMID: 31534459 PMCID: PMC6724428 DOI: 10.1155/2019/3041262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 06/23/2019] [Indexed: 11/19/2022] Open
Abstract
Background Chemical biology approaches using small molecule inhibitors targeting specific signaling pathways are useful tools to dissect the molecular mechanisms governing stem cell differentiation and for their possible use in therapeutic interventions. Methods Stem cell signaling small molecule library functional screen was performed employing human bone marrow skeletal (mesenchymal) stem cells (hBMSCs). Alkaline phosphatase (ALP) activity and formation of mineralized matrix visualized by Alizarin red staining were employed as markers for osteoblastic differentiation. Global gene expression profiling was conducted using the Agilent microarray platform, and data normalization and bioinformatics were performed using GeneSpring software. Pathway analyses were conducted using the Ingenuity Pathway Analysis (IPA) tool. In vivo ectopic bone formation was performed using hBMSC mixed with hydroxyapatite–tricalcium phosphate granules that were implanted subcutaneously in 8-week-old female nude mice. Hematoxylin and eosin staining and Sirius red staining were performed to identify bone formation in vivo. Results Among the tested molecules, LY411575, a potent γ-secretase and Notch signaling inhibitor, exhibited significant inhibitory effects on osteoblastic differentiation of hBMSCs manifested by reduced ALP activity, mineralized matrix formation, and decreased osteoblast-specific gene expression as well as in vivo ectopic bone formation. Global gene expression profiling of LY411575-treated cells revealed changes in multiple signaling pathways, including focal adhesion, insulin, TGFβ, IL6, and Notch signaling, and decreased the expression of genes associated with functional categories of tissue development. Among the affected signaling networks were TGFβ1, SPP1, and ERK regulatory networks. Conclusions We identified γ-secretase inhibitor (LY411575) as a potent regulator of osteoblastic differentiation of hBMSC that may be useful as a therapeutic option for treating conditions associated with ectopic bone formation.
Collapse
|
31
|
Wang XF, Zhang BH, Lu XQ, Wang RQ. DLX5 gene regulates the Notch signaling pathway to promote glomerulosclerosis and interstitial fibrosis in uremic rats. J Cell Physiol 2019; 234:21825-21837. [PMID: 31297803 DOI: 10.1002/jcp.28032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 11/30/2018] [Indexed: 01/31/2023]
Abstract
Uremia largely results from the accumulation of organic waste products normally cleared by the kidneys, which commonly accompanies kidney failure and chronic kidney disease. However, genetic investigations in a uremia remain largely unclear. This study aimed to determine the expression patterns of distal-less homeobox 5 (DLX5) in uremia rat model and further to study its effects on glomerulosclerosis and interstitial fibrosis. Uremic expression chip was applied to screen differentially expressed genes in uremia. Next, we used small interfering RNA-mediated RNA interference to specifically silence DLX5 in experimental uremic rats to understand the regulatory mechanism of DLX5. To understand effect of Notch1 signaling pathway in uremia, we also treated experimental uremic rats with γ-secretase inhibitor (GSI), an inhibitor of Notch1 signaling pathway. The expression of fibronectin (FN), laminin (LN), transforming growth factor-β1 (TGF-β1), Hes1, Hes5, and Jagged2 was determined. The semiquantitative assessment was applied to verify the effects of DLX5 on glomerulosclerosis. In the uremic expression chip, we found that DLX5 was upregulated in uremia samples, and considered to regulate the Notch signaling pathway. We found that small interfering RNA-mediated DLX5 inhibition or Notch1 signaling pathway inhibitory treatment relieved and delayed the kidney injury and glomerulosclerosis in uremia. Meanwhile, inhibition of DLX5 or Nothch1 signaling pathway reduced expression of FN, LN, Nothch1, TGF-β1, Hes1, Hes5, and Jagged2. Intriguingly, we discovered that Notch1 signaling pathway was inhibited after silencing DLX5. In conclusion, these findings highlight that DLX5 regulates Notch signaling, which may, in turn, promote complications of uremia such as kidney fibrosis, providing a novel therapeutic target for treating uremia.
Collapse
Affiliation(s)
- Xin-Fang Wang
- Department of Blood Purification, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Bei-Hao Zhang
- Department of Blood Purification, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Xiao-Qing Lu
- Department of Blood Purification, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Rui-Qiang Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
32
|
Yang L, Liu X, Zhang N, Chen L, Xu J, Tang W. Investigation of circular RNAs and related genes in pulmonary fibrosis based on bioinformatics analysis. J Cell Biochem 2019; 120:11022-11032. [PMID: 30767300 PMCID: PMC6593700 DOI: 10.1002/jcb.28380] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 12/06/2018] [Indexed: 01/24/2023]
Abstract
Pulmonary fibrosis is a lethal inflammatory disease. In this study, we aimed to explore the potential-related circular RNAs (circRNAs) and genes that are associated with pulmonary fibrosis. Pulmonary fibrosis rat models were constructed and the fibrosis deposition was detected using hematoxylin and eosin and Masson staining. The differentially expressed circRNAs were obtained through RNA sequencing. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were further performed to uncover the key function and pathways in pulmonary fibrosis. The interaction networks between circRNAs and their downstream micro RNAs (miRNAs) and genes were constructed by Cytoscape Software. The quantitative polymerase chain reaction was performed to validate the expression of 10 candidate circRNAs and five of them were performed ringwise sequencing in pulmonary fibrosis rats. We further selected five candidate circRNAs target miRNAs and messenger RNAs and validated by real-time polymerase chain reaction. The pulmonary fibrosis models were successfully constructed according to the pathological examination. circRNAs were differentially expressed between the pulmonary fibrosis and normal pulmonary tissues. GO analysis verified that the differentially expressed circRNAs were significantly clustered in the cellular component, molecular function, and biological process. In the KEGG analysis, circRNAs were enriched in the following pathways: antigen processing and presentation, phagosome, PI3K-AKt signaling pathway, HTLV-I infection, and Herpes simplex infection. After validation in pulmonary fibrosis rat models, it was found that five of those circRNAs (chr9:113534327|113546234 [down], chr1:200648164|200672411 [down], chr5:150850432|150865550 [up], chr20:14319170|14326640 [down], and chr10:57634023|57634588 [down]) showed a relatively consistent trend with predictions. Validation of these circRNAs target miRNAs and genes showed that chr9:113534327|113546234, chr20:14319170|14326640, and chr10:57634023|57634588 were implicated in Notch1 activated transforming growth factor-β (TGF-β) signaling pathway. The study demonstrated that a series of circRNAs are differentially expressed in pulmonary fibrosis rats. These circRNAs, especially TGF-β- and Notch1-related circRNAs might play an important role in regulating pulmonary fibrogenesis.
Collapse
Affiliation(s)
- Liteng Yang
- Department of Respiratory MedicineShenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen UniversityGuangdongShenzhenChina
| | - Xin Liu
- Department of Traditional Chinese Medicine, Zunyi Medical and Pharmaceutical CollegeGuizhouZunyiChina
| | - Ning Zhang
- Department of Respiratory MedicineShenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen UniversityGuangdongShenzhenChina
| | - Lifang Chen
- Department of Respiratory MedicineShenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen UniversityGuangdongShenzhenChina
| | - Jingyi Xu
- Department of Respiratory MedicineShenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen UniversityGuangdongShenzhenChina
| | - Wencheng Tang
- Department of Respiratory MedicineShenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen UniversityGuangdongShenzhenChina
| |
Collapse
|
33
|
Zmorzyński S, Styk W, Filip AA, Krasowska D. The Significance of NOTCH Pathway in the Development of Fibrosis in Systemic Sclerosis. Ann Dermatol 2019; 31:365-371. [PMID: 33911613 PMCID: PMC7992759 DOI: 10.5021/ad.2019.31.4.365] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/11/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disorder characterized by the fibrosis of skin, heart, lung, and kidney as well. Excessive activation of fibroblasts is associated with higher expression of Notch1 and/or Notch3 genes. The constitutive expression of NOTCH genes was described in epithelial cells: epidermal keratinocytes, hair follicle cells and sebaceous glands. The NOTCH signalling pathway may be involved in the development of fibrosis, myofibroblast formation and the process of epithelial-mesenchymal transition. Activation of the NOTCH pathway leads to morphological, phenotypic and functional changes in epithelial cells. Furthermore, inhibition of Notch signalling prevent the development of fibrosis in different models, among them, bleomycin-induced fibrosis and in the Task-1 mause model. Molecular mechanisms, including the role of NOTCH signaling pathway, associated with fibrosis in SSc have not been completely recognized.
Collapse
Affiliation(s)
- Szymon Zmorzyński
- Department of Cancer Genetics with Cytogenetic Laboratory, Medical University of Lublin, Poland
| | - Wojciech Styk
- Department of Cancer Genetics with Cytogenetic Laboratory, Medical University of Lublin, Poland
| | - Agata Anna Filip
- Department of Cancer Genetics with Cytogenetic Laboratory, Medical University of Lublin, Poland
| | - Dorota Krasowska
- Chair and Department of Dermatology, Venerology and Paediatric Dermatology, Medical University of Lublin, Poland
| |
Collapse
|
34
|
Genz B, Coleman MA, Irvine KM, Kutasovic JR, Miranda M, Gratte FD, Tirnitz-Parker JEE, Olynyk JK, Calvopina DA, Weis A, Cloonan N, Robinson H, Hill MM, Al-Ejeh F, Ramm GA. Overexpression of miRNA-25-3p inhibits Notch1 signaling and TGF-β-induced collagen expression in hepatic stellate cells. Sci Rep 2019; 9:8541. [PMID: 31189969 PMCID: PMC6561916 DOI: 10.1038/s41598-019-44865-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 05/23/2019] [Indexed: 02/06/2023] Open
Abstract
During chronic liver injury hepatic stellate cells (HSCs), the principal source of extracellular matrix in the fibrotic liver, transdifferentiate into pro-fibrotic myofibroblast-like cells - a process potentially regulated by microRNAs (miRNAs). Recently, we found serum miRNA-25-3p (miR-25) levels were upregulated in children with Cystic Fibrosis (CF) without liver disease, compared to children with CF-associated liver disease and healthy individuals. Here we examine the role of miR-25 in HSC biology. MiR-25 was detected in the human HSC cell line LX-2 and in primary murine HSCs, and increased with culture-induced activation. Transient overexpression of miR-25 inhibited TGF-β and its type 1 receptor (TGFBR1) mRNA expression, TGF-β-induced Smad2 phosphorylation and subsequent collagen1α1 induction in LX-2 cells. Pull-down experiments with biotinylated miR-25 revealed Notch signaling (co-)activators ADAM-17 and FKBP14 as miR-25 targets in HSCs. NanoString analysis confirmed miR-25 regulation of Notch- and Wnt-signaling pathways. Expression of Notch signaling pathway components and endogenous Notch1 signaling was downregulated in miR-25 overexpressing LX-2 cells, as were components of Wnt signaling such as Wnt5a. We propose that miR-25 acts as a negative feedback anti-fibrotic control during HSC activation by reducing the reactivity of HSCs to TGF-β-induced collagen expression and modulating the cross-talk between Notch, Wnt and TGF-β signaling.
Collapse
Affiliation(s)
- Berit Genz
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Mater Research, Translational Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Miranda A Coleman
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Katharine M Irvine
- Mater Research, Translational Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Jamie R Kutasovic
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Personalised Medicine Team, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mariska Miranda
- Personalised Medicine Team, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Francis D Gratte
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia.,School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, Australia
| | - Janina E E Tirnitz-Parker
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - John K Olynyk
- Department of Gastroenterology & Hepatology, Fiona Stanley Fremantle Hospital Group, Murdoch, Western Australia, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Diego A Calvopina
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anna Weis
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Nicole Cloonan
- Genomic Biology Lab, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Harley Robinson
- Precision & Systems Biomedicine, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Michelle M Hill
- Precision & Systems Biomedicine, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Fares Al-Ejeh
- Personalised Medicine Team, QIMR-Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Grant A Ramm
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia. .,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
35
|
Myofibroblast in Kidney Fibrosis: Origin, Activation, and Regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:253-283. [DOI: 10.1007/978-981-13-8871-2_12] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
36
|
Su X, Wei Y, Cao J, Wu X, Mou D, Luo J, Li A, Zuo GW, Tang M. CCN3 and DLL1 co-regulate osteogenic differentiation of mouse embryonic fibroblasts in a Hey1-dependent manner. Cell Death Dis 2018; 9:1188. [PMID: 30538222 PMCID: PMC6289993 DOI: 10.1038/s41419-018-1234-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023]
Abstract
Notch signaling pathway is one of the most important pathways to regulate intercellular signal transduction and is crucial in the regulation of bone regeneration. Nephroblastoma overexpressed (NOV or CCN3) serves as a non-canonical secreted ligand of Notch signaling pathway and its role in the process of osteogenic differentiation of mesenchymal stem cells (MSCs) was undefined. Here we conducted a comprehensive study on this issue. In vivo and in vitro studies have shown that CCN3 significantly inhibited the early and late osteogenic differentiation of mouse embryonic fibroblasts (MEFs), the expression of osteogenesis-related factors, and the subcutaneous ectopic osteogenesis of MEFs in nude mice. In mechanism studies, we found that CCN3 significantly inhibited the expression of BMP9 and the activation of BMP/Smad and BMP/MAPK signaling pathways. There was also a mutual inhibition between CCN3 and DLL1, one of the classic membrane protein ligands of Notch signaling pathway. Additionally, we further found that Hey1, the target gene shared by BMP and Notch signaling pathways, partially reversed the inhibitory effect of CCN3 on osteoblastic differentiation of MEFs. In summary, our findings suggested that CCN3 significantly inhibited the osteogenic differentiation of MEFs. The inhibitory effect of CCN3 was mainly through the inhibition of BMP signaling and the mutual inhibition with DLL1, so as to inhibit the expression of Hey1, the target gene shared by BMP and Notch signaling pathways.
Collapse
Affiliation(s)
- Xin Su
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, 400016, Chongqing, China
| | - Yalin Wei
- Xi'an No.4 Hospital, 710004, Xi'an, China
| | - Junjie Cao
- Department of Clinical Laboratory, Xi'an No.5 Hospital, 710082, Xi'an, China
| | - Xiulin Wu
- Department of Geriatrics, Army Military Medical University, 400038, Chongqing, China
| | - Daiyong Mou
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, 400016, Chongqing, China
| | - Jinyong Luo
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, 400016, Chongqing, China
| | - Aifang Li
- Department of Clinical Laboratory, Xi'an Chest Hospital, 710100, Xi'an, China
| | - Guo-Wei Zuo
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, 400016, Chongqing, China.
| | - Min Tang
- Key Laboratory of Diagnostic Medicine designated by the Chinese Ministry of Education, Chongqing Medical University, 400016, Chongqing, China.
| |
Collapse
|
37
|
Zhou X, Fang Y, Wan L, Xu Q, Huang H, Zhu R, Wu Q, Liu J. Notch signaling inhibits cardiac fibroblast to myofibroblast transformation by antagonizing TGF‐β1/Smad3 signaling. J Cell Physiol 2018; 234:8834-8845. [PMID: 30317638 DOI: 10.1002/jcp.27543] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 09/13/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Xue‐liang Zhou
- Department of Cardiac Surgery The First Affiliated Hospital, Nanchang University Nanchang China
| | - Yi‐hu Fang
- Department of Cardiac Surgery The First Affiliated Hospital, Nanchang University Nanchang China
| | - Li Wan
- Department of Cardiac Surgery The First Affiliated Hospital, Nanchang University Nanchang China
| | - Qi‐rong Xu
- Department of Cardiac Surgery The First Affiliated Hospital, Nanchang University Nanchang China
| | - Huang Huang
- Department of Cardiac Surgery The First Affiliated Hospital, Nanchang University Nanchang China
| | - Rong‐rong Zhu
- Department of Obstetrics and Gynecology Jiangxi Province Hospital of Integrated Traditional Chinese and Western Medicine China
| | - Qi‐cai Wu
- Department of Cardiac Surgery The First Affiliated Hospital, Nanchang University Nanchang China
| | - Ji‐chun Liu
- Department of Cardiac Surgery The First Affiliated Hospital, Nanchang University Nanchang China
| |
Collapse
|
38
|
Healy L, Nicholls K, Gibson R, Stella D, Bogwitz M, Taylor J, Walsh M, Donaldson L, Winship I. Absence of renal phenotype in hereditary haemorrhagic telangiectasia. Intern Med J 2018; 48:1255-1257. [PMID: 30288899 DOI: 10.1111/imj.14059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/21/2018] [Accepted: 04/22/2018] [Indexed: 11/27/2022]
Abstract
Hereditary haemorrhagic telangiectasia is characterised by abnormal blood vessel formation, producing telangiectasia and arteriovenous malformations in multiple organs. Information regarding possible renal involvement in hereditary haemorrhagic telangiectasia is limited. This study assessed renal structure and function in 11 patients with genetically confirmed diagnosis and known arteriovenous malformations in lung, liver, gastrointestinal tract or brain. All had significant current or past epistaxis. Despite the vascularity of the kidneys, we found no evidence of renal involvement. This observation warrants further consideration.
Collapse
Affiliation(s)
- Lachlan Healy
- Department of Genomic Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kathleen Nicholls
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Nephrology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Robert Gibson
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Radiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Damien Stella
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Radiology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Michael Bogwitz
- Department of Genomic Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jessica Taylor
- Department of Genomic Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Maie Walsh
- Department of Genomic Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
| | - Liz Donaldson
- Department of Genomic Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Ingrid Winship
- Department of Genomic Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
39
|
PAX2 may induce ADAM10 expression in renal tubular epithelial cells and contribute to epithelial-to-mesenchymal transition. Int Urol Nephrol 2018; 50:1729-1741. [PMID: 30117015 PMCID: PMC6133107 DOI: 10.1007/s11255-018-1956-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 08/07/2018] [Indexed: 11/30/2022]
Abstract
Purpose We sought to investigate the role of PAX2 in renal epithelial-to-mesenchymal transition (EMT), examining the influence of PAX2 on ADAM10 expression during renal EMT and ADAM10 expression in fibrotic kidneys. Methods A rat renal tubular epithelial cell line, NRK52E, was transfected with lentivirus carrying PAX2, and E-cadherin and α-SMA expressions were measured. The influence of PAX2 on ADAM10 promoter activity was evaluated using chromatin immunoprecipitation (CHIP) and dual-luciferase reporter assay. We also treated NRK52E with ADAM10-specific over-expression vector and inhibitors and measured E-cadherin and α-SMA expression. In vivo, Wistar rats (n = 36) were subjected to unilateral ureteral obstruction (UUO) (n = 18) or sham surgery (n = 18), with tissues from post-operative day 3, 7, and 14 days examined, and PAX2/ADAM10 activity measured. ADAM10 expression was also assessed in kidneys from patients with chronic kidney disease (CKD). Results In NRK52E overexpressing PAX2, ADAM10 and α-SMA levels were increased, while E-cadherin levels were decreased. CHIP and dual-luciferase reporter assay showed that PAX2 directly bound to a specific site within the ADAM10 promoter, and over-expression of PAX2 significantly activated ADAM10 transcription. NRK52E with ADAM10 over-expression also significantly decreased E-cadherin and increased α-SMA levels. In the fibrotic kidneys of rats with UUO, E-cadherin levels were increased and α-SMA levels were decreased, and expression of PAX2 and ADAM10 increased. ADAM10 expression also elevated in the renal tissues of CKD patients. Conclusions PAX2 directly increased expression of ADAM10, the presence of which contributed to EMT in renal tubular epithelia and hence plays an important role in renal fibrosis.
Collapse
|
40
|
Schütte-Nütgen K, Edeling M, Mendl G, Krahn MP, Edemir B, Weide T, Kremerskothen J, Michgehl U, Pavenstädt H. Getting a Notch closer to renal dysfunction: activated Notch suppresses expression of the adaptor protein Disabled-2 in tubular epithelial cells. FASEB J 2018; 33:821-832. [PMID: 30052485 DOI: 10.1096/fj.201800392rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Reactivation of Notch signaling in kidneys of animal models and patients with chronic kidney disease (CKD) has been shown to contribute to epithelial injury and fibrosis development. Here, we investigated the mechanisms of Notch-induced injury in renal epithelial cells. We performed genome-wide transcriptome analysis to identify Notch target genes using an in vitro system of cultured tubular epithelial cells expressing the intracellular domain of Notch1. One of the top downregulated genes was Disabled-2 ( Dab2). With the use of Drosophila nephrocytes as a model system, we found that Dab (the Drosophila homolog of Dab2) knockdown resulted in a significant filtration defect, indicating that loss of Dab2 plays a functional role in kidney disease development. We showed that Dab2 expression in cultured tubular epithelial cells is involved in endocytic regulation and that it also protects cells from TGF-β-induced epithelial-to-mesenchymal transition. In vivo correlation studies indicated its additional role in renal ischemia-induced injury. Together, these data suggest that Dab2 plays a versatile role in the kidney and may impact on acute and CKDs.-Schütte-Nütgen, K., Edeling, M., Mendl, G., Krahn, M. P., Edemir, B., Weide, T., Kremerskothen, J., Michgehl, U., Pavenstädt, H. Getting a Notch closer to renal dysfunction: activated Notch suppresses expression of the adaptor protein Disabled-2 in tubular epithelial cells.
Collapse
Affiliation(s)
| | - Maria Edeling
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| | - Gudrun Mendl
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| | - Michael P Krahn
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| | - Bayram Edemir
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and.,Department of Hematology and Oncology, Internal Medicine IV, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Thomas Weide
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| | | | - Ulf Michgehl
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| | - Hermann Pavenstädt
- Internal Medicine D, University Hospital Muenster, Muenster, Germany; and
| |
Collapse
|
41
|
Chen Q, Zhao T, Xie X, Yu D, Wu L, Yu W, Sun W. MicroRNA-663 regulates the proliferation of fibroblasts in hypertrophic scars via transforming growth factor-β1. Exp Ther Med 2018; 16:1311-1317. [PMID: 30116380 PMCID: PMC6090240 DOI: 10.3892/etm.2018.6350] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 05/15/2017] [Indexed: 12/11/2022] Open
Abstract
The present study determined the expression of microRNA (miR)-663 in hypertrophic scar (HS) tissues and investigate the regulatory mechanisms of miR-663 in HS. A total of 51 patients diagnosed with HS between December 2013 and February 2016 were included in the present study. HS tissues (experimental group) and HS-adjacent tissues (control group) were collected. Primary fibroblasts were obtained from HS tissue and transfected with small-interfering RNA against transforming growth factor (TGF)-β1 or miR-663 mimics. Reverse-transcription quantitative PCR was used to determine the levels of TGF-β1 mRNA and miR-663. Western blot analysis was performed to determine TGF-β1 protein expression. An MTT assay was employed to detect the proliferation of fibroblasts, and a dual luciferase reporter assay was performed to identify the binding of miR-663 with TGF-β1 mRNA. TGF-β1 was found to have a regulatory role in HS at the transcriptional level. The expression of TGF-β1 was upregulated in HS tissues, and knockdown of TGF-β1 in cultured fibroblasts led to inhibition of proliferation. The expression of miR-663 was downregulated in HS. miR-663 was revealed to regulate the expression of TGF-β1 by binding with the 3′-untranslated region of TGF-β1 mRNA. Elevated expression of miR-663 inhibited the proliferation of fibroblasts by regulating TGF-β1 expression. The present study demonstrated that upregulation of TGF-β1 in HS tissues is associated with the downregulation of miR-663 expression. miR-663 may regulate the proliferation of fibroblasts in HS and the expression of associated proteins.
Collapse
Affiliation(s)
- Qi Chen
- Department of Plastic and Cosmetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Tianlan Zhao
- Department of Plastic and Cosmetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xiaoming Xie
- Department of Plastic and Cosmetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Daojiang Yu
- Department of Plastic and Cosmetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Lijun Wu
- Department of Plastic and Cosmetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Wenyuan Yu
- Department of Plastic and Cosmetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Wei Sun
- Department of Plastic and Cosmetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
42
|
Yu H, Ju J, Liu J, Li D. Aberrant expression of miR-663 and transforming growth factor-β1 in nasal polyposis in children. Exp Ther Med 2018; 15:4550-4556. [PMID: 29849780 DOI: 10.3892/etm.2018.5927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 04/11/2017] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to investigate the expression of microRNA (miR)-663 and its regulatory effects on the pathogenesis of nasal polyposis in children. Nasal polyp tissue, as well as serum and peripheral blood eosinophils were collected from 35 children diagnosed with nasal polypectomy between August 2013 and August 2015. As a control, the inferior nasal concha, serum and peripheral blood eosinophils were collected from 46 patients with nasal septal deviation complicated by inferior turbinate hypertrophy or patients with simple inferior turbinate hypertrophy who had undergone surgical removal of the inferior nasal concha. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to measure the expression of miR-663 and transforming growth factor-β1 (TGF-β1) in the nasal polyp tissue, serum and peripheral blood eosinophils of patients with nasal polyposis and controls. Western blotting was used to measure the expression of TGF-β1 protein in nasal tissue and eosinophils and an enzyme-linked immunosorbent assay was used to measure serum level of TGF-β1 protein. A dual luciferase reporter assay was used to determine whether TGF-β1 was a target gene of miR-663. Compared with the control group, levels of TGF-β1 mRNA and protein were significantly increased in all three types of specimens from pediatric patients with nasal polyposis (P<0.05). miR-663 expression was significantly decreased in nasal polyp tissue and peripheral blood eosinophils (P<0.05). The dual luciferase reporter assay confirmed that TGF-β1 was a target gene of miR-663. The current study suggests that the upregulation of TGF-β1 may be associated with the downregulation of miR-663 in nasal polyposis in children. miR-663 may have regulatory effects on the pathogenesis of nasal polyposis by regulating TGF-β1 and may be developed as a genetic marker of nasal polyposis in children.
Collapse
Affiliation(s)
- Hailing Yu
- Department of Otolaryngology, The Women and Children's Hospital of Qingdao, Qingdao, Shandong 266033, P.R. China
| | - Jianbao Ju
- Department of Otolaryngology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266033, P.R. China
| | - Jingdong Liu
- Department of Medicine, The Women and Children's Hospital of Qingdao, Qingdao, Shandong 266033, P.R. China
| | - Da Li
- Department of Otolaryngology, The Women and Children's Hospital of Qingdao, Qingdao, Shandong 266033, P.R. China
| |
Collapse
|
43
|
González-Foruria I, Santulli P, Chouzenoux S, Carmona F, Chapron C, Batteux F. Dysregulation of the ADAM17/Notch signalling pathways in endometriosis: from oxidative stress to fibrosis. Mol Hum Reprod 2018; 23:488-499. [PMID: 28486700 DOI: 10.1093/molehr/gax028] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/28/2017] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION Is oxidative stress associated with the A disintegrin and metalloproteases (ADAM) metallopeptidase domain 17 (ADAM17)/Notch signalling pathway and fibrosis in the development of endometriosis? SUMMARY ANSWER Oxidative stress is correlated with hyperactivation of the ADAM17/Notch signalling pathway and a consequent increase in fibrosis in patients with endometriosis. WHAT IS KNOWN ALREADY It is nowadays accepted that oxidative stress plays an important role in the onset and progression of endometriosis. Oxidative stress is able to induce the synthesis of some members of the 'ADAM' family, such as ADAM17. ADAM17/Notch signalling is dysregulated in other profibrotic and inflammatory diseases. STUDY DESIGN, SIZE, DURATION This was a prospective laboratory study conducted in a tertiary-care university hospital between January 2011 and April 2013. We investigated non-pregnant, younger than 42-year-old patients (n = 202) during surgery for a benign gynaecological condition. PARTICIPANTS/MATERIALS, SETTING, METHODS After complete surgical exploration of the abdominopelvic cavity, 121 women with histologically proven endometriosis and 81 endometriosis-free control women were enrolled. Peritoneal fluid (PF) samples were obtained from all the study participants during surgery in order to detect advanced oxidation protein products (AOPPs) and metalloproteinase activity of ADAM17. Stromal cells from endometrial specimens (n = 8) were obtained from endometrium of control patients (Cs), and from eutopic (Es) and ectopic (Ps) endometrium of patients with deep infiltrating endometriosis (DIE) (n = 8). ADAM17, Notch and the fibrosis markers α-smooth muscle actin (α-SMA) and type-I collagen were assessed using immunoblotting in all the endometrial samples obtained. Additionally, fibrosis was assessed after using Notch cleavage inhibitors (DAPT and FLI-06). Notch and fibrosis were also evaluated after stimulation of stromal endometrial cells with ADAM17 purified protein, increasing concentrations of H2O2 and primary cell culture supernatants. MAIN RESULTS AND THE ROLE OF CHANCE Patients with DIE presented higher PF AOPP and ADAM17 protein levels than controls (P < 0.01 and P < 0.05, respectively). In addition, these two markers were positively correlated (r = 0.614; P < 0.001). At the cellular level, ADAM17 activity was increased in Es and Ps compared to Cs (P < 0.001 and P < 0.01, respectively). Furthermore, Ps presented hyperactivation of Notch signalling (P < 0.05) and augmentation of fibrosis markers (P = 0.009 for α-SMA and P = 0.015 for type-I collagen) compared to controls. The use of DAPT and FLI-06 reduced both fibrosis markers in Ps but not in Cs. Stimulation with ADAM17, H2O2 and Ps supernatant culture significantly increased Notch and fibrosis in both Ps and Cs. LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION The control group consisted of women who underwent surgery for benign gynaecological conditions, which could lead to biases because some of these conditions may cause alterations in oxidative stress and the ADAM17/Notch pathways. The small sample size of endometrial biopsies used for each group of patients (n = 8) is a limitation of the study, and results should be interpreted with caution. WIDER IMPLICATIONS OF THE FINDINGS We propose a novel pathway in endometriosis pathogenesis that correlates oxidative stress, hyperactivation of ADAM17/Notch signalling and a consequent increase in fibrosis. This study suggests that Notch signalling plays a key role in the fibrotic processes that take place in ectopic lesions of patients with DIE, as already observed in other pro-fibrotic diseases. STUDY FUNDING AND COMPETING INTEREST(S) This work was supported by grants from University Paris Descartes, INSERM and Fundación Alfonso Martín Escudero. The authors have no competing interests to declare.
Collapse
Affiliation(s)
- Iñaki González-Foruria
- Département Développement, Reproduction et Cancer, Institut Cochin, INSERM U1016, Equipe Pr. Batteux, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre, Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynaecology Obstetrics II and Reproductive Medicine, 75679 Paris Cedex 14, France.,Institut Clínic of Gynecology, Obstetrics and Neonatology, Hospital Clínic. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine - University of Barcelona, Barcelona, Spain
| | - Pietro Santulli
- Département Développement, Reproduction et Cancer, Institut Cochin, INSERM U1016, Equipe Pr. Batteux, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre, Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynaecology Obstetrics II and Reproductive Medicine, 75679 Paris Cedex 14, France
| | - Sandrine Chouzenoux
- Département Développement, Reproduction et Cancer, Institut Cochin, INSERM U1016, Equipe Pr. Batteux, Paris, France
| | - Francisco Carmona
- Institut Clínic of Gynecology, Obstetrics and Neonatology, Hospital Clínic. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Faculty of Medicine-University of Barcelona, Barcelona, Spain
| | - Charles Chapron
- Département Développement, Reproduction et Cancer, Institut Cochin, INSERM U1016, Equipe Pr. Batteux, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre, Centre Hospitalier Universitaire (CHU) Cochin, Department of Gynaecology Obstetrics II and Reproductive Medicine, 75679 Paris Cedex 14, France
| | - Frédéric Batteux
- Département Développement, Reproduction et Cancer, Institut Cochin, INSERM U1016, Equipe Pr. Batteux, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre, Centre Hospitalier Universitaire (CHU) Cochin, Service d'immunologie biologique, 75679 Paris Cedex 14, France
| |
Collapse
|
44
|
Wyss JC, Kumar R, Mikulic J, Schneider M, Aebi JD, Juillerat-Jeanneret L, Golshayan D. Targeted γ-secretase inhibition of Notch signaling activation in acute renal injury. Am J Physiol Renal Physiol 2018; 314:F736-F746. [DOI: 10.1152/ajprenal.00414.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The Notch pathway has been reported to control tissue damage in acute kidney diseases. To investigate potential beneficial nephroprotective effects of targeting Notch, we developed chemically functionalized γ-secretase inhibitors (GSIs) targeting γ-glutamyltranspeptidase (γ-GT) and/or γ-glutamylcyclotransferase (γ-GCT), two enzymes overexpressed in the injured kidney, and evaluated them in in vivo murine models of acute tubular and glomerular damage. Exposure of the animals to disease-inducing drugs together with the functionalized GSIs improved proteinuria and, to some extent, kidney dysfunction. The expression of genes involved in the Notch pathway, acute inflammatory stress responses, and the renin-angiotensin system was enhanced in injured kidneys, which could be downregulated upon administration of functionalized GSIs. Immunohistochemistry staining and Western blots demonstrated enhanced activation of Notch1 as detected by its cleaved active intracellular domain during acute kidney injury, and this was downregulated by concomitant treatment with the functionalized GSIs. Thus targeted γ-secretase-based prodrugs developed as substrates for γ-GT/γ-GCT have the potential to selectively control Notch activation in kidney diseases with subsequent regulation of the inflammatory stress response and the renin-angiotensin pathways.
Collapse
Affiliation(s)
- Jean-Christophe Wyss
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Rajesh Kumar
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Josip Mikulic
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Manfred Schneider
- Medicinal Chemistry, Roche Pharma Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche, Basel, Switzerland
| | - Johannes D. Aebi
- Medicinal Chemistry, Roche Pharma Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche, Basel, Switzerland
| | - Lucienne Juillerat-Jeanneret
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- University Institute of Pathology, CHUV and UNIL, Lausanne, Switzerland
| | - Dela Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
45
|
Ni MM, Wang YR, Wu WW, Xia CC, Zhang YH, Xu J, Xu T, Li J. Novel Insights on Notch signaling pathways in liver fibrosis. Eur J Pharmacol 2018; 826:66-74. [PMID: 29501868 DOI: 10.1016/j.ejphar.2018.02.051] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 02/27/2018] [Accepted: 02/28/2018] [Indexed: 12/11/2022]
Abstract
Liver fibrosis is characterized by an increased and altered deposition of extracellular matrix (ECM) proteins that make up excessive tissue scarring and promote chronic liver injury. Activation of hepatic stellate cells (HSCs) is a pivotal cellular event in the progression of liver fibrosis. However, the mechanisms involved in the development of liver fibrosis are only now beginning to be unveiled. The Notch pathway is a fundamental and highly conserved pathway able to control cell-fate, including cell proliferation, differentiation, apoptosis, regeneration and other cellular activities. Recently, the deregulation of Notch cascade has been found involved in many pathological processes, including liver fibrosis. These data give evidence for a role for Notch signaling in liver fibrosis. In addition,more and more date are available on the role of Notch pathways in the process. Therefore, this review focuses on the current knowledge about the Notch signaling pathway, which dramatically takes part in HSC activation and liver fibrosis, and look ahead on new perspectives of Notch signaling pathway research. Furthermore, we will summarize this new evidence on the different interactions in Notch signaling pathway-regulated liver fibrosis, and support the potentiality of putative biomarkers and unique therapeutic targets.
Collapse
Affiliation(s)
- Ming-Ming Ni
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, No.72 Guangzhou Road, Nanjing 210001,China
| | - Ya-Rui Wang
- TCM Research Institution, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001,China
| | - Wen-Wen Wu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, No.72 Guangzhou Road, Nanjing 210001,China
| | - Chong-Cai Xia
- TCM Research Institution, Nanjing Municipal Hospital of T.C.M, The Third Affiliated Hospital of Nanjing University of T.C.M, Nanjing 210001,China
| | - Yi-He Zhang
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, No.72 Guangzhou Road, Nanjing 210001,China
| | - Jing Xu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, No.72 Guangzhou Road, Nanjing 210001,China.
| | - Tao Xu
- Institute for Liver Diseases of Anhui Medical University(AMU), Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Jun Li
- Institute for Liver Diseases of Anhui Medical University(AMU), Anhui Medical University, Hefei 230032, Anhui Province, China
| |
Collapse
|
46
|
Tschumperlin DJ, Ligresti G, Hilscher MB, Shah VH. Mechanosensing and fibrosis. J Clin Invest 2018; 128:74-84. [PMID: 29293092 DOI: 10.1172/jci93561] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tissue injury disrupts the mechanical homeostasis that underlies normal tissue architecture and function. The failure to resolve injury and restore homeostasis gives rise to progressive fibrosis that is accompanied by persistent alterations in the mechanical environment as a consequence of pathological matrix deposition and stiffening. This Review focuses on our rapidly growing understanding of the molecular mechanisms linking the altered mechanical environment in injury, repair, and fibrosis to cellular activation. In particular, our focus is on the mechanisms by which cells transduce mechanical signals, leading to transcriptional and epigenetic responses that underlie both transient and persistent alterations in cell state that contribute to fibrosis. Translation of these mechanobiological insights may enable new approaches to promote tissue repair and arrest or reverse fibrotic tissue remodeling.
Collapse
Affiliation(s)
| | | | - Moira B Hilscher
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
47
|
Amini-Nik S, Yousuf Y, Jeschke MG. Scar management in burn injuries using drug delivery and molecular signaling: Current treatments and future directions. Adv Drug Deliv Rev 2018; 123:135-154. [PMID: 28757325 PMCID: PMC5742037 DOI: 10.1016/j.addr.2017.07.017] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
In recent decades, there have been tremendous improvements in burn care that have allowed patients to survive severe burn injuries that were once fatal. However, a major limitation of burn care currently is the development of hypertrophic scars in approximately 70% of patients. This significantly decreases the quality of life for patients due to the physical and psychosocial symptoms associated with scarring. Current approaches to manage scarring include surgical techniques and non-surgical methods such as laser therapy, steroid injections, and compression therapy. These treatments are limited in their effectiveness and regularly fail to manage symptoms. As a result, the development of novel treatments that aim to improve outcomes and quality of life is imperative. Drug delivery that targets the molecular cascades of wound healing to attenuate or prevent hypertrophic scarring is a promising approach that has therapeutic potential. In this review, we discuss current treatments for scar management after burn injury, and how drug delivery targeting molecular signaling can lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Saeid Amini-Nik
- Sunnybrook Research Institute, Toronto, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada.
| | - Yusef Yousuf
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada
| | - Marc G Jeschke
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada; Department of Immunology, University of Toronto, Toronto, Canada; Ross-Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada.
| |
Collapse
|
48
|
Notch-mediated Sox9 + cell activation contributes to kidney repair after partial nephrectomy. Life Sci 2017; 193:104-109. [PMID: 29198839 DOI: 10.1016/j.lfs.2017.11.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/16/2017] [Accepted: 11/26/2017] [Indexed: 12/25/2022]
Abstract
AIMS Partial nephrectomy is a surgical technique as an alternative for traditional radical nephrectomy. The advantage of partial nephrectomy technique is nephron-sparing, however, whether the remaining kidney tissue could regenerate the lost nephron is still unknown. The current work is to investigate the kidney tissue repair process and the related cellular and molecular mechanism. MAIN METHODS We used a novel unilateral partial nephrectomy mouse model to study kidney repair, and focused on a population of Sox9+ progenitor cells to study their pivotal role in the regenerative process. Kidney function after nephrectomy was measured using creatinine and urea nitrogen assay kit. Wound healing was assessed by Masson Trichrome Staining. Tissue regeneration was tested by Sox9+ cells immunofluorescence staining. The differentiation potential of Sox9+ cells were assessed by immunoanalysis with various tubular cell markers. Notch activation was determined by qPCR and Western blotting. KEY FINDINGS After partial nephrectomy, we found that massive Sox9+ cells emerged one day after the surgery and lasted for up to 20days. The Sox9+ cells had proliferative capacity and could give rise to epithelial cells of proximal tubule, Henle's loop, distal tubule, collecting duct, and the parietal layer of glomerulus. We also found that the activation of Sox9+ cells was mediated by Notch signaling pathway. SIGNIFICANCE The current study reveals that Notch-mediated Sox9+ cell activation can contribute to kidney tubule regeneration after unilateral partial nephrectomy in mice.
Collapse
|
49
|
Cannito S, Novo E, Parola M. Therapeutic pro-fibrogenic signaling pathways in fibroblasts. Adv Drug Deliv Rev 2017; 121:57-84. [PMID: 28578015 DOI: 10.1016/j.addr.2017.05.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/28/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023]
Abstract
Myofibroblasts (MFs) play a critical role in the progression of chronic inflammatory and fibroproliferative diseases in different tissues/organs, whatever the etiology. Fibrosis is preceded and sustained by persistent injury and inflammatory response in a profibrogenic scenario involving mutual interactions, operated by several mediators and pathways, of MFs and related precursor cells with innate immunity cells and virtually any cell type in a defined tissue. These interactions, mediators and related signaling pathways are critical in initiating and perpetuating the differentiation of precursor cells into MFs that in different tissues share peculiar traits and phenotypic responses, including the ability to proliferate, produce ECM components, migrate and contribute to the modulation of inflammatory response and tissue angiogenesis. Literature studies related to liver, lung and kidney fibrosis have outlined a number of MF-related core regulatory fibrogenic signaling pathways conserved across these different organs and potentially targetable in order to develop effective antifibrotic therapeutic strategies.
Collapse
|
50
|
Xu L, Zhang Y, Guo R, Shen W, Qi Y, Wang Q, Guo Z, Qi C, Yin H, Wang J. HES1 promotes extracellular matrix protein expression and inhibits proliferation and migration in human trabecular meshwork cells under oxidative stress. Oncotarget 2017; 8:21818-21833. [PMID: 28423527 PMCID: PMC5400626 DOI: 10.18632/oncotarget.15631] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/07/2017] [Indexed: 01/06/2023] Open
Abstract
Glaucoma is the leading cause of irreversible blindness. The most prevalent form of glaucoma is primary open-angle glaucoma (POAG). Oxidative stress is one of the major pathogenic factors of the POAG, and can elicit molecular and functional changes in trabecular meshwork cells, causing increased aqueous humor outflow resistance and elevated intraocular pressure. However, the regulatory mechanisms underlying oxidative stress-induced cell phenotypic changes remain elusive. Herein, we exposed primary human trabecular meshwork cells to the oxidative stress induced by 300 μM H2O2 for 2 h, and found significantly up-regulated expression of extracellular matrix proteins and a transcription factor, hairy and enhancer of split-1 (HES1). The cell functions, including migration and proliferation, were impaired by the oxidative stress. Furthermore, HES1 shRNA abrogated the extracellular matrix protein up-regulation and rescued the functional defects caused by the oxidative stress; conversely, HES1 overexpression resulted in the molecular and functional changes similar to those induced by H2O2. These results suggest that HES1 promotes extracellular matrix protein expression and inhibits proliferative and migratory functions in the trabecular meshwork cells under oxidative stress, thereby providing a novel pathogenic mechanism underlying and a potential therapeutic target to the POAG.
Collapse
Affiliation(s)
- Linqi Xu
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, College of Optometry and Ophthalmology, Tianjin Medical University, Tianjin 300384, China.,Tianjin Research Center of Basic Medical Sciences and Department of Cell Biology, Tianjin Medical University, Tianjin 300070, China
| | - Yan Zhang
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, College of Optometry and Ophthalmology, Tianjin Medical University, Tianjin 300384, China
| | - Ruru Guo
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, College of Optometry and Ophthalmology, Tianjin Medical University, Tianjin 300384, China
| | - Wencui Shen
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, College of Optometry and Ophthalmology, Tianjin Medical University, Tianjin 300384, China.,Tianjin Research Center of Basic Medical Sciences and Department of Cell Biology, Tianjin Medical University, Tianjin 300070, China
| | - Yan Qi
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, College of Optometry and Ophthalmology, Tianjin Medical University, Tianjin 300384, China.,Tianjin Research Center of Basic Medical Sciences and Department of Cell Biology, Tianjin Medical University, Tianjin 300070, China
| | - Qingsong Wang
- Tianjin Research Center of Basic Medical Sciences and Department of Cell Biology, Tianjin Medical University, Tianjin 300070, China
| | - Zhenglong Guo
- Tianjin Research Center of Basic Medical Sciences and Department of Cell Biology, Tianjin Medical University, Tianjin 300070, China
| | - Chen Qi
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, College of Optometry and Ophthalmology, Tianjin Medical University, Tianjin 300384, China
| | - Haifang Yin
- Tianjin Research Center of Basic Medical Sciences and Department of Cell Biology, Tianjin Medical University, Tianjin 300070, China
| | - Jiantao Wang
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, College of Optometry and Ophthalmology, Tianjin Medical University, Tianjin 300384, China
| |
Collapse
|