1
|
Shi R, Zhao W, Zhu L, Wang R, Wang D. Identification of basement membrane markers in diabetic kidney disease and immune infiltration by using bioinformatics analysis and experimental verification. IET Syst Biol 2023; 17:316-326. [PMID: 37776100 PMCID: PMC10725710 DOI: 10.1049/syb2.12078] [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: 05/22/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of chronic kidney disease worldwide. Basement membranes (BMs) are ubiquitous extracellular matrices which are affected in many diseases including DKD. Here, the authors aimed to identify BM-related markers in DKD and explored the immune cell infiltration in this process. The expression profiles of three datasets were downloaded from the Gene Expression Omnibus database. BM-related differentially expression genes (DEGs) were identified and Kyoto encyclopaedia of genes and genomes pathway enrichment analysis were applied to biological functions. Immune cell infiltration and immune function in the kidneys of patients with DKD and healthy controls were evaluated and compared using the ssGSEA algorithm. The association of hub genes and immune cells and immune function were explored. A total of 30 BM-related DEGs were identified. The functional analysis showed that BM-related DEGs were notably associated with basement membrane alterations. Crucially, BM-related hub genes in DKD were finally identified, which were able to distinguish patients with DKD from controls. Moreover, the authors observed that laminin subunit gamma 1(LAMC1) expression was significantly high in HK2 cells treated with high glucose. Immunohistochemistry results showed that, compared with those in db/m mouse kidneys, the levels of LAMC1 in db/db mouse kidneys were significantly increased. The biomarkers genes may prove crucial for DKD treatment as they could be targeted in future DKD treatment protocols.
Collapse
Affiliation(s)
- Rui Shi
- Department of NephrologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Wen‐Man Zhao
- Department of NephrologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Li Zhu
- Department of NephrologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Rui‐Feng Wang
- Department of NephrologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - De‐Guang Wang
- Department of NephrologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
2
|
Liu F, Wu Q, Dong Z, Liu K. Integrins in cancer: Emerging mechanisms and therapeutic opportunities. Pharmacol Ther 2023:108458. [PMID: 37245545 DOI: 10.1016/j.pharmthera.2023.108458] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Integrins are vital surface adhesion receptors that mediate the interactions between the extracellular matrix (ECM) and cells and are essential for cell migration and the maintenance of tissue homeostasis. Aberrant integrin activation promotes initial tumor formation, growth, and metastasis. Recently, many lines of evidence have indicated that integrins are highly expressed in numerous cancer types and have documented many functions of integrins in tumorigenesis. Thus, integrins have emerged as attractive targets for the development of cancer therapeutics. In this review, we discuss the underlying molecular mechanisms by which integrins contribute to most of the hallmarks of cancer. We focus on recent progress on integrin regulators, binding proteins, and downstream effectors. We highlight the role of integrins in the regulation of tumor metastasis, immune evasion, metabolic reprogramming, and other hallmarks of cancer. In addition, integrin-targeted immunotherapy and other integrin inhibitors that have been used in preclinical and clinical studies are summarized.
Collapse
Affiliation(s)
- Fangfang Liu
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| | - Qiong Wu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zigang Dong
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Kangdong Liu
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou, Henan 450001, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan 450000, China.
| |
Collapse
|
3
|
The potential role of FNDC5/irisin in various liver diseases: awakening the sleeping beauties. Expert Rev Mol Med 2022; 24:e23. [PMID: 35695040 DOI: 10.1017/erm.2022.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fibronectin type III domain-containing protein 5 (FNDC5) is a transmembrane protein and the precursor of irisin, which serves as a systemic exerkine/myokine with multiple origins. Since its discovery in 2012, this hormone-like polypeptide has rapidly evolved to a component significantly involved in a gamut of metabolic dysregulations and various liver diseases. After a decade of extensive investigation on FNDC5/irisin, we are still surrounded by lots of open questions regarding its diagnostic and therapeutic values. In this review, we first concentrated on the structure-function relationship of FNDC5/irisin. Next, we comprehensively summarised the current knowledge and research findings regarding pathogenic roles/therapeutic applications of FNDC5/irisin in the context of non-alcoholic fatty liver disease, fibrosis, liver injury due to multiple detrimental insults, hepatic malignancy and intrahepatic cholestasis of pregnancy. Moreover, the prominent molecules involved in the underlying mechanisms and signalling pathways were highlighted. As a result, emerging evidence reveals FNDC5/irisin may act as a proxy for diagnosing liver disease pathology, a sensitive biomarker for assessing damage severity, a predisposing factor for surveilling illness progression and a treatment option with protective/preventive impact, all of which are highly dependent on disease grading and contextually pathological features.
Collapse
|
4
|
Antioxidant Effects of Irisin in Liver Diseases: Mechanistic Insights. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3563518. [PMID: 35035659 PMCID: PMC8759828 DOI: 10.1155/2022/3563518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/19/2021] [Accepted: 12/10/2021] [Indexed: 02/08/2023]
Abstract
Oxidative stress is a crucial factor in the development of various liver diseases. Irisin, a metabolic hormone discovered in 2012, is mainly produced by proteolytic cleavage of fibronectin type III domain containing 5 (FNDC5) in skeletal muscles. Irisin is induced by physical exercise, and a rapidly growing body of literature suggests that irisin is, at least partially, responsible for the beneficial effects of regular exercise. The major biological function of irisin is believed to be involved in the maintenance of metabolic homeostasis. However, recent studies have suggested the therapeutic potential of irisin against a variety of liver diseases involving its antioxidative function. In this review, we aim to summarize the accumulating evidence demonstrating the antioxidative effects of irisin in liver diseases, with an emphasis on the current understanding of the potential molecular mechanisms.
Collapse
|
5
|
Wu Q, Jiao B, Gui W, Zhang Q, Wang F, Han L. Long non-coding RNA SNHG1 promotes fibroblast-to-myofibroblast transition during the development of pulmonary fibrosis induced by silica particles exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:112938. [PMID: 34741930 DOI: 10.1016/j.ecoenv.2021.112938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 06/13/2023]
Abstract
Inhaling silica dust in the environment can cause progressive pulmonary fibrosis, then silicosis. Silicosis is the most harmful occupational disease in the world, so the study of the mechanism is of great significance for the prevention and treatment of silicosis. Long non-coding RNAs (lncRNAs) are important players in the pathological process of fibrotic diseases. However, the function of specific lncRNA in regulating pulmonary fibrosis remains elusive. In this study, a mouse model of pulmonary fibrosis via intratracheal instillation of silica particles was established, and the differential expression of lnc-SNHG1 and miR-326 in lung tissues and TGF-β1-treated fibroblasts was detected by the qRT-PCR method. Short interfering RNA (siRNA) and plasmid were designed for knockdown or overexpression of lnc-SNHG1 in fibroblasts. MiRNA simulant was designed for overexpression of miR-326 in vivo and in vitro. Dual-luciferase reporter system, immunofluorescence, western blot, wound healing and transwell assay were performed to investigate the function and the underlying mechanisms of lnc-SNHG1. As a result, we found that lnc-SNHG1 was highly expressed in fibrotic lung tissues of mice and TGF-β1-treated fibroblasts. Moreover, the high expression of lnc-SNHG1 facilitated the migration and invasion of fibroblasts and the secretion of fibrotic molecules, while the low expression of lnc-SNHG1 exerted the opposite effects. Further mechanism studies showed that miR-326 was the potential target of lnc-SNHG1, and there is a negative correlation between the expression levels of lnc-SNHG1 and miR-326. Combined with mitigating fibrotic effects of miR-326 in a mouse model of silica particles exposure, we revealed that lnc-SNHG1 significantly sponged miR-326 and facilitated the expression of SP1, thus accelerating fibroblast-to-myofibroblast transition and synergistically promoting the development of pulmonary fibrosis. Our study uncovered a key mechanism by which lnc-SNHG1 regulated pulmonary fibrosis through miR-326/SP1 axis, and lnc-SNHG1 is a potential target for the prevention and treatment of silicosis.
Collapse
Affiliation(s)
- Qiuyun Wu
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou 221004, China.
| | - Biyang Jiao
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Wenwen Gui
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Qianyi Zhang
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Feng Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Lei Han
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| |
Collapse
|
6
|
Network Pharmacology/Metabolomics-Based Validation of AMPK and PI3K/AKT Signaling Pathway as a Central Role of Shengqi Fuzheng Injection Regulation of Mitochondrial Dysfunction in Cancer-Related Fatigue. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5556212. [PMID: 34326918 PMCID: PMC8302405 DOI: 10.1155/2021/5556212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/06/2021] [Accepted: 06/12/2021] [Indexed: 12/17/2022]
Abstract
Chinese herbal medicines have multiple targets and properties, and their use in multidisciplinary cancer therapies has consequently received increasing attention. Here, we have investigated the possible active ingredients associated with cancer-related fatigue (CRF) in the Shengqi Fuzheng Injection (SFI). In vitro cell models were used to measure the regulation effects of SFI on CRF. Metabolomic analysis was used to identify the potential genes and pathways in C2C12 mouse myoblasts treated with SFI, and the interaction of compounds and CRF targets was predicted using network pharmacology and molecular docking analyses. The putative pathways were further verified using immuno-blotting assays. The results showed that SFI significantly inhibited muscle cell apoptosis and increased the mitochondrial membrane potential of muscle cells. The network pharmacology analysis results identified 36 candidate compounds, and 244 potential targets were yielded by SFI, and they shared 10 key targets associated with cancer-related fatigue. According to the enrichment analysis and experimental validation, SFI might ameliorate muscle cell mitochondrial function by activating AMPK and inhibiting the PI3K/Akt signaling pathways, and the expression changes of mitochondrial metabolic enzymes MnSOD and apoptosis-associated proteins Bax and Bcl-2 were also triggered. The functions and mechanisms of SFI in anticancer-related fatigue were found here to be at least partly due to the targeting of the AMPK and PI3K/Akt signaling pathways, and this has highlighted new potential applications for network pharmacology when researching Chinese Medicines.
Collapse
|
7
|
LncRNA KCNQ1OT1 knockdown inhibits viability, migration and epithelial-mesenchymal transition in human lens epithelial cells via miR-26a-5p/ITGAV/TGF-beta/Smad3 axis. Exp Eye Res 2020; 200:108251. [PMID: 32950535 DOI: 10.1016/j.exer.2020.108251] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Long noncoding RNA potassium voltage-gated channel subfamily Q member 1 opposite strand/antisense transcript 1 (KCNQ1OT1) takes part in diabetic cataract progression. This research aims to analyze the function and mechanism of KCNQ1OT1 on viability, migration and epithelial-mesenchymal transition (EMT) in lens epithelial cells. METHODS 20 diabetic cataract posterior lens capsule tissues and normal samples were collected. Lens epithelial cells (SRA01/04) were stimulated via high glucose (HG). The levels of KCNQ1OT1, miR-26a-5p, integrin αV (ITGAV), TGF-β, Smad3 and phosphorylated (p)-Smad3 were measured via quantitative real-time polymerase chain reaction or Western blot. Cell viability, migration and EMT were analyzed via MTT, wound healing, transwell and Western blot assays. The target relationship between miR-26a-5p and KCNQ1OT1 or ITGAV was determined via luciferase reporter assay. RESULTS KCNQ1OT1 was up-regulated and miR-26a-5p level was reduced in diabetic cataract tissues and HG-treated SRA01/04 cells. Silence of KCNQ1OT1 or miR-26a-5p up-regulation repressed cell viability, migration and EMT in SRA01/04 cells stimulated via HG. KCNQ1OT1 could target miR-26a-5p and controlled cell viability, migration and EMT via regulating miR-26a-5p. ITGAV was targeted via miR-26a-5p and positively regulated via KCNQ1OT1. ITGAV overexpression promoted cell viability, migration and EMT in HG-treated SRA01/04 cells, which were mitigated by KCNQ1OT1 silence. KCNQ1OT1 knockdown mitigated HG-induced the activation of TGF-β/Smad3 signaling by regulating miR-26a-5p. CONCLUSION KCNQ1OT1 knockdown represses cell viability, migration and EMT through miR-26a-5p/ITGAV/TGF-β/Smad3 axis in SRA01/04 cells under HG condition, providing a new target for the treatment of diabetic cataract.
Collapse
|