1
|
Wang L, Ren Z, Wu L, Zhang X, Wang M, Niu H, He X, Wang H, Chen Y, Shi G, Qian X. HRD1 reduction promotes cholesterol-induced vascular smooth muscle cell phenotypic change via endoplasmic reticulum stress. Mol Cell Biochem 2024; 479:3021-3036. [PMID: 38145449 DOI: 10.1007/s11010-023-04902-0] [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: 07/14/2023] [Accepted: 11/07/2023] [Indexed: 12/26/2023]
Abstract
Phenotypic change of vascular smooth muscle cells (VSMCs) is the main contributor of vascular pathological remodeling in atherosclerosis. The endoplasmic reticulum (ER) is critical for maintaining VSMC function through elimination of misfolded proteins that impair VSMC cellular function. ER-associated degradation (ERAD) is an ER-mediated process that controls protein quality by clearing misfolded proteins. One of the critical regulators of ERAD is HRD1, which also plays a vital role in lipid metabolism. However, the function of HRD1 in VSMCs of atherosclerotic vessels remains poorly understood. The level of HRD1 expression was analyzed in aortic tissues of mice fed with a high-fat diet (HFD). The H&E and EVG (VERHOEFF'S VAN GIESON) staining were used to demonstrate pathological vascular changes. IF (immunofluorescence) and WB (western blot) were used to explore the signaling pathways in vivo and in vitro. The wound closure and transwell assays were also used to test the migration rate of VSMCs. CRISPR gene editing and transcriptomic analysis were applied in vitro to explore the cellular mechanism. Our data showed significant reduction of HRD1 in aortic tissues of mice under HFD feeding. VSMC phenotypic change and HRD1 downregulation were detected by cholesterol supplement. Transcriptomic and further analysis of HRD1-KO VSMCs showed that HRD1 deficiency induced the expression of genes related to ER stress response, proliferation and migration, but reduced the contractile-related genes in VSMCs. HRD1 deficiency also exacerbated the proliferation, migration and ROS production of VSMCs induced by cholesterol, which promoted the VSMC dedifferentiation. Our results showed that HRD1 played an essential role in the contractile homeostasis of VSMCs by negatively regulating ER stress response. Thus, HRD1 in VSMCs could serve as a potential therapeutic target in metabolic disorder-induced vascular remodeling.
Collapse
Affiliation(s)
- Linli Wang
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Zhitao Ren
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Lin Wu
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Ximei Zhang
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Min Wang
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Haiming Niu
- Department of Critical Care Medicine, Zhongshan People's Hospital, Zhongshan, 528400, China
| | - Xuemin He
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Heting Wang
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yanming Chen
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Guojun Shi
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Xiaoxian Qian
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
2
|
Cheng T, Gu ML, Xu WQ, Ye DW, Zha ZY, Fang WG, Mao LK, Ning J, Hu XB, Ding YH. Mechanism of lncRNA SNHG16 on kidney clear cell carcinoma cells by targeting miR-506-3p/ETS1/RAS/ERK molecular axis. Heliyon 2024; 10:e30388. [PMID: 38756581 PMCID: PMC11096951 DOI: 10.1016/j.heliyon.2024.e30388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Objective This study aimed to investigate the mechanism of long noncoding ribonucleic acid (lncRNA) SNHG16 on kidney clear cell carcinoma (KIRC) cells by targeting miR-506-3p/ETS proto-oncogene 1, transcription factor (ETS1)/RAS/Extracellular regulated protein kinases (ERK) molecular axis, thus to provide reference for clinical diagnosis and treatment of KIRC in the future. Methods Thirty-six patients with KIRC were enrolled in this study, and their carcinoma tissues and adjacent tissues were obtained for the detection of SNHG16/miR-506-3p/ETS1/RAS/ERK expression. Then, over-expressed SNHG16 plasmid and silenced plasmid were transfected into KIRC cells to observe the changes of their biological behavior. Results SNHG16 and ETS1 were highly expressed while miR-506- 3p was low expressed in KIRC tissues; the RAS/ERK signaling pathway was significantly activated in KIRC tissues (P < 0.05). After SNHG16 silence, KIRC cells showed decreased proliferation, invasion and migration capabilities and increased apoptosis rate; correspondingly, increase in SNHG16 expression achieved opposite results (P < 0.05). Finally, in the rescue experiment, the effects of elevated SNHG16 on KIRC cells were reversed by simultaneous increase in miR-506-3p, and the effects of miR-506-3p were reversed by ETS1. Activation of the RAS/ERK pathway had the same effect as increase in ETS1, which further worsened the malignancy of KIRC. After miR-506-3p increase and ETS1 silence, the RAS/ERK signaling pathway was inhibited (P < 0.05). At last, the rescue experiment (co-transfection) confirmed that the effect of SNHG16 on KIRC cells is achieved via the miR-506-3p/ETS1/RAS/ERK molecular axis. Conclusion SNHG16 regulates the biological behavior of KIRC cells by targeting the miR-506-3p/ETS1/RAS/ERK molecular axis.
Collapse
Affiliation(s)
- Tao Cheng
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Ming-Li Gu
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Wei-Qiang Xu
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Da-Wen Ye
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Ze-Yu Zha
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Wen-Ge Fang
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Li-Kai Mao
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Jing Ning
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Xing-Bang Hu
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Yong-Hui Ding
- Department of Urology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| |
Collapse
|
3
|
Hu D, Ge Y, Ye L, Xi Y, Chen J, Zhu W, Wang Z, Sun Z, Su Y, Wang D, Xiao S, Qiu J. d-Galactose induces the senescence and phenotype switch of corpus cavernosum smooth muscle cells. J Cell Physiol 2024; 239:124-134. [PMID: 37942832 DOI: 10.1002/jcp.31150] [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: 08/17/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023]
Abstract
Studies regarding age-related erectile dysfunction (ED) based on naturally aging models are limited by their high costs, especially for the acquisition of primary cells from the corpus cavernosum. Herein, d-galactose ( d-gal) was employed to accelerate cell senescence, and the underlying mechanism was explored. As predominant functional cells involved in the erectile response, corpus cavernosum smooth muscle cells (CCSMCs) were isolated from 2-month-old rats. Following this, d-gal was introduced to induce cell senescence, which was verified via β-galactosidase staining. The effects of d-gal on CCSMCs were evaluated by terminal deoxynucleoitidyl transferase dUTP nick-end labeling (TUNEL), immunofluorescence staining, flow cytometry, western blot, and quantitative real-time polymerase chain reaction (qRT-PCR). Furthermore, RNA interference (RNAi) was carried out for rescue experiments. Subsequently, the influence of senescence on the corpus cavernosum was determined via scanning electron microscopy, qRT-PCR, immunohistochemistry, TUNEL, and Masson stainings. The results revealed that the accelerated senescence of CCSMCs was promoted by d-gal. Simultaneously, smooth muscle alpha-actin (alpha-SMA) expression was inhibited, while that of osteopontin (OPN) and Krüppel-like factor 4 (KLF4), as well as fibrotic and apoptotic levels, were elevated. After knocking down KLF4 expression in d-gal-induced CCSMCs by RNAi, the expression level of cellular alpha-SMA increased. Contrastingly, the OPN expression, apoptotic and fibrotic levels declined. In addition, cellular senescence acquired partial remission. Accordingly, in the aged corpus cavernosum, the fibrotic and apoptotic rates were increased, followed by downregulation in the expression of alpha-SMA and the concurrent upregulation in the expression of OPN and KLF4. Overall, our results signaled that d-gal-induced accelerated senescence of CCSMCs could trigger fibrosis, apoptosis and phenotypic switch to the synthetic state, potentially attributed to the upregulation of KLF4 expression, which may be a multipotential therapeutic target of age-related ED.
Collapse
Affiliation(s)
- Daoyuan Hu
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunlong Ge
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lei Ye
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuhang Xi
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jialiang Chen
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wenliang Zhu
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhenqing Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhuolun Sun
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, München, Germany
| | - Ying Su
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dejuan Wang
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shiwei Xiao
- Department of Urology, Guizhou Province People's Hospital, Guiyang, China
| | - Jianguang Qiu
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
4
|
Zhang H, Liu G, Mao X, Yang L, Wang B, Yuan X. LncRNA MEG3 induces endothelial differentiation of mouse derived adipose-derived stem cells by targeting MiR-145-5p/KLF4. Mol Biol Rep 2022; 49:8495-8505. [PMID: 35802277 DOI: 10.1007/s11033-022-07671-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/01/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The present study aimed to investigate the mechanisms through which long non-coding RNA (lncRNA) maternally expressed 3 (MEG3) affected the endothelial differentiation of mouse derived adipose-derived stem cells (ADSCs). MATERIALS AND METHODS ADSCs were isolated and identified by specific surface marker detection. The effects of lncRNA MEG3 on endothelial differentiation of ADSCs were also detected via quantitative PCR, western blotting, immunofluorescence and Matrigel angiogenesis assays. In addition, using target gene prediction tools and luciferase reporter assays, the downstream target gene was demonstrated. RESULTS LncRNA MEG3 targeted and reduced the expression levels of microRNA-145-5p (miR-145-5p), which upregulated the expression levels of Krüppel like factor 4 (KLF4), promoting endothelial differentiation of ADSCs. CONCLUSION LncRNA MEG3 induced endothelial differentiation of ADSCs by targeting miR-145-5p/KLF4, which may provide novel insights to illustrate the mechanism of endothelial differentiation of ADSCs.
Collapse
Affiliation(s)
- Hailong Zhang
- Department of Dermatology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150006, Heilongjiang, People's Republic of China
| | - Gang Liu
- Department of Medicine, Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150006, Heilongjiang, People's Republic of China
| | - Xu Mao
- Department of Health Center, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang, People's Republic of China
| | - Lei Yang
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, No. 33 of West Dazhi Street, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Bingyu Wang
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, No. 33 of West Dazhi Street, Harbin, 150001, Heilongjiang, People's Republic of China.
| | - Xingxing Yuan
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, No. 33 of West Dazhi Street, Harbin, 150001, Heilongjiang, People's Republic of China.
| |
Collapse
|
5
|
Yang M, Huang X, Shen F, Yi J, Meng Y, Chen Y. Lef1 is transcriptionally activated by Klf4 and suppresses hyperoxia-induced alveolar epithelial cell injury. Exp Lung Res 2022; 48:213-223. [PMID: 35950640 DOI: 10.1080/01902148.2022.2108945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
PURPOSE Bronchopulmonary dysplasia (BPD) is a long-term respiratory condition. More than a quarter of extremely premature newborns are harmed by BPD. At present, there are no apparent effective drugs or treatments for the condition. In this study, we aimed to investigate the functional role and mechanism of lymphoid enhancer-binding factor 1 (Lef1) in BPD in vitro. MATERIALS AND METHODS Blood samples from BPD patients and healthy volunteers were gathered, and an in vitro model of BPD was developed in alveolar epithelial cells (AECs) MLE-12 induced by hyperoxia. Then expression of krüppel-like factor 4 (KLF4/Klf4) and LEF1/Lef1 were evaluated. After Lef1 overexpressing plasmid and the vector were transfected into hyperoxia-induced MLE-12 cells, cell proliferation assays were carried out. Cell apoptosis was investigated by a flow cytometry assay, and apoptosis related proteins Bcl-2, cleaved-caspase 3 and 9 were analyzed by a western blot assay. The binding between Klf4 and Lef1 promoter predicted on the JASPAR website was verified using luciferase and ChIP assays. For further study of the mechanism of Klf4 and Lef1 in BPD, gain-of-function experiments were performed. RESULTS The mRNA levels of KLF4/Klf4 and LEF1/Lef1 were diminished in clinical BPD serum samples and hyperoxia-induced MLE-12 cells. Overexpression of Lef1 stimulated AEC proliferation and suppressed AEC apoptosis induced by hyperoxia. Mechanically, Klf4 bound to Lef1's promoter region and aids transcription. Moreover, the results of gain-of-function experiments supported that Klf4 could impede AEC damage induced by hyperoxia via stimulating Lef1. CONCLUSION Klf4 and Lef1 expression levels were declined in hyperoxia-induced AECs, and Lef1 could be transcriptionally activated by Klf4 and protect against hyperoxia-induced AEC injury in BPD. As a result, Lef1 might become a prospective therapeutic target for BPD.
Collapse
Affiliation(s)
- Min Yang
- Department of Respiratory, Hunan Children's Hospital, Changsha, China
| | | | - Fang Shen
- Research Institute of Children, Hunan Children's Hospital, Changsha, China
| | - Juanjuan Yi
- Department of Neonate, Hunan Children's Hospital, Changsha, China
| | - Yanni Meng
- Department of Respiratory, Hunan Children's Hospital, Changsha, China
| | - Yanping Chen
- Department of Respiratory, Hunan Children's Hospital, Changsha, China
| |
Collapse
|
6
|
Liu Q, Cui Y, Ding N, Zhou C. Knockdown of circ_0003928 ameliorates high glucose-induced dysfunction of human tubular epithelial cells through the miR-506-3p/HDAC4 pathway in diabetic nephropathy. Eur J Med Res 2022; 27:55. [PMID: 35392987 PMCID: PMC8991937 DOI: 10.1186/s40001-022-00679-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/20/2022] [Indexed: 11/10/2022] Open
Abstract
Background Previous data have indicated the importance of circular RNA (circRNA) in the pathogenesis of diabetic nephropathy (DN). The study is designed to investigate the effects of circ_0003928 on oxidative stress and apoptosis of high glucose (HG)-treated human tubular epithelial cells (HK-2) and the underlying mechanism. Methods The DN cell model was established by inducing HK-2 cells using 30 mmol/L D-glucose. RNA expression of circ_0003928, miR-506-3p and histone deacetylase 4 (HDAC4) was detected by quantitative real-time polymerase chain reaction. Cell viability and proliferation were investigated by cell counting kit-8 and 5-Ethynyl-29-deoxyuridine (EdU) assays, respectively. Oxidative stress was evaluated by commercial kits. Caspase 3 activity and cell apoptotic rate were assessed by a caspase 3 activity assay and flow cytometry analysis, respectively. Protein expression was detected by Western blotting analysis. The interactions among circ_0003928, miR-506-3p and HDAC4 were identified by dual-luciferase reporter and RNA pull-down assays. Results Circ_0003928 and HDAC4 expression were significantly upregulated, while miR-506-3p was downregulated in the serum of DN patients and HG-induced HK-2 cells. HG treatment inhibited HK-2 cell proliferation, but induced oxidative stress and cell apoptosis; however, these effects were reversed after circ_0003928 depletion. Circ_0003928 acted as a miR-506-3p sponge, and HDAC4 was identified as a target gene of miR-506-3p. Moreover, the circ_0003928/miR-506-3p/HDAC4 axis regulated HG-induced HK-2 cell dysfunction. Conclusion Circ_0003928 acted as a sponge for miR-506-3p to regulate HG-induced oxidative stress and apoptosis of HK-2 cells through HDAC4, which suggested that circ_0003928 might be helpful in the therapy of DN. Supplementary Information The online version contains supplementary material available at 10.1186/s40001-022-00679-y.
Collapse
Affiliation(s)
- Qiong Liu
- Department of Nephrology, Hebei General Hospital, Shijiazhuang, China
| | - Yuanyuan Cui
- Department of Endocrine Rheumatology and Immunology, People's Hospital of Gaotang County, Gaotang, China
| | - Nan Ding
- Department of Clinical Laboratory, Hebei General Hospital, Shijiazhuang, China
| | - Changxue Zhou
- Department of Kidney Internal Medicine, Zaozhuang Municipal Hospital, No. 41 Longtou Road, Central District, Zaozhuang, 277100, China.
| |
Collapse
|