1
|
Wu F, Yu H. The role of the NOTCH1 signaling pathway in the maintenance of mesenchymal stem cell stemness and chondrocyte differentiation and its potential in the treatment of osteoarthritis. J Orthop Surg Res 2025; 19:772. [PMID: 39754211 PMCID: PMC11697486 DOI: 10.1186/s13018-024-05236-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/04/2024] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVE This study aims to explore the potential role of mesenchymal stem cells (MSCs) in the treatment of osteoarthritis (OA), particularly the function of the NOTCH1 signaling pathway in maintaining the stemness of MSCs and in chondrocyte differentiation. METHODS Utilizing diverse analytical techniques on an osteoarthritis dataset, we unveil distinct gene expression patterns and regulatory relationships, shedding light on potential mechanisms underlying the disease. Techniques used include the culture of MSCs, induction of differentiation into chondrocytes, establishment of stable cell lines, Western Blot, and immunofluorescence. Through the construction of lentiviruses overexpressing and knocking out NOTCH1, the effects of NOTCH1 on the stemness of MSCs and chondrocyte differentiation were investigated. Additionally, the effects of NOTCH1 on chondrocyte homeostasis and apoptosis were evaluated by adding the EZH2 inhibitor GSK126 and the endoplasmic reticulum stress inducer tunicamycin. RESULTS Experimental results demonstrated that NOTCH1 expression can influence the maintenance of MSC stemness and chondrocyte differentiation by regulating EZH2. Knockout of NOTCH1 decreased the expression of chondrocyte markers, while overexpression increased their expression. Under conditions of endoplasmic reticulum stress, NOTCH1 expression helped reduce the expression of stress-related proteins, maintain chondrocyte homeostasis, and inhibit apoptosis. CONCLUSION The NOTCH1 signaling pathway plays a crucial role in maintaining the stemness of MSCs, differentiating into chondrocytes, and in the treatment of osteoarthritis. NOTCH1 influences the differentiation fate of MSCs and the homeostasis of chondrocytes by regulating EZH2 and other related genes, offering new targets and strategies for the treatment of diseases like osteoarthritis.
Collapse
Affiliation(s)
- Fuming Wu
- Department of Knee Surgery, The First Hospital of Hebei Medical University, Hebei, China
| | - Haiquan Yu
- Department of Knee Surgery, The First Hospital of Hebei Medical University, Hebei, China.
| |
Collapse
|
2
|
Gong Y, Wei N, Shi P, Zhu G. CIRCTMCO3 ALLEVIATES SEPSIS-INDUCED ACUTE KIDNEY INJURY VIA REGULATING MIR-218-5P/ZEB2 AXIS. Shock 2025; 63:168-175. [PMID: 39454632 DOI: 10.1097/shk.0000000000002499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
ABSTRACT Background: Growing evidence has found the critical role of circular RNAs (circRNAs) in sepsis-induced acute kidney injury (S-AKI). CircTMCO3 has been found to be involved in tumor microenvironment changes of ovarian cancer. This study aimed to explore whether circTMCO3 functions in S-AKI, and if so, to elucidate the molecular mechanism. Methods: CircTMCO3 expression was analyzed in lipopolysaccharide (LPS)-induced HK-2 cells and in the kidney tissues of mice treated with cecal ligation and puncture (CLP), respectively. Furthermore, the effects of circTMCO3 on S-AKI and the related mechanisms were evaluated in both models through gain- and/or loss-of-function strategies. Results: CircTMCO3 expression was suppressed in both S-AKI models. Upregulation of circTMCO3 mitigated LPS-induced apoptosis, oxidative stress, and inflammation in HK-2 cells. In contrast, circTMCO3 downregulation exacerbated LPS-induced injuries in HK-2 cells. Intravenous injection of circTMCO3 lentivirus to increase circTMCO3 expression improved renal function and attenuated kidney injury in S-AKI mice, as evidenced by the decrease in serum creatinine and blood urea nitrogen concentrations, amelioration of tubular pathological injury, reduction of renal cell apoptosis, and mitigation of oxidative stress and proinflammatory cytokines (TNF-α, IL-1β, and IL-6). Moreover, circTMCO3 directly targeted miR-218-5p, and the mimic of which abolished the protective effect of circTMCO3 in cell models. ZEB2 was identified to be a target of miR-218-5p; its downregulation not only reversed the impacts of miR-218-5p inhibitor on S-AKI, but also mitigated the effects mediated by circTMCO3 upregulation in vitro . Conclusions: CircTMCO3 protects against S-AKI by regulating miR-218-5p/ZEB2 axis, thereby mediating antiapoptotic, antioxidant, and anti-inflammatory activities. This indicates that increasing circTMCO3 expression might be a future therapeutic method for S-AKI.
Collapse
Affiliation(s)
- Yingfeng Gong
- Graduate School of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Na Wei
- Graduate School of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Peipei Shi
- Neurological Intensive Care Unit, Suzhou Municipal Hospital of Anhui Province, Suzhou City, Anhui Province, China
| | - Gang Zhu
- Neurological Intensive Care Unit, Suzhou Municipal Hospital of Anhui Province, Suzhou City, Anhui Province, China
| |
Collapse
|
3
|
Han H, Zhang Y, Huang E, Zhou S, Huang Z, Qin K, Du X. The role of TBC1D15 in sepsis-induced acute lung injury: Regulation of mitochondrial homeostasis and mitophagy. Int J Biol Macromol 2024; 293:139289. [PMID: 39740704 DOI: 10.1016/j.ijbiomac.2024.139289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
Mitochondrial quality control is crucial in sepsis-induced acute lung injury (SI-ALI). Our study investigates how the intracellular protein TBC1D15 regulates mitochondrial quality to improve SI-ALI. We found TBC1D15 levels significantly decreased in the whole blood of sepsis patients, monocytes, lung tissue from SI-ALI mice, and the MLE-12 cellular model (mouse lung epithelial cells). Overexpression of TBC1D15 using adeno-associated viral and lentiviral vectors alleviated lung injury and inflammation in both mouse models and MLE-12 cells, while silencing TBC1D15 exacerbated inflammatory responses. Mechanistically, TBC1D15 overexpression dissociated mitochondria-lysosome contact duration, promoted mitophagy, and restored mitochondrial function. The protective effects of TBC1D15 were reversed by the mitophagy inhibitor Bafilomycin A1. Additionally, TBC1D15 knockdown prolonged mitochondria-lysosome contact time, resulting in worsened mitochondrial dysfunction and increased oxidative stress. Our findings indicate that SI-ALI is characterized by prolonged mitochondria-lysosome contact and impaired mitophagy. Thus, TBC1D15 overexpression presents a promising therapeutic strategy to mitigate mitochondrial dysfunction and reduce lung injury in septic conditions, suggesting potential clinical applications for SI-ALI treatment.
Collapse
Affiliation(s)
- Hanghang Han
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yingying Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Enhao Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Siyu Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zijin Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China
| | - Ke Qin
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China.
| | - Xueke Du
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
4
|
Yao X, Yang S, Chen L, Lin F, Ruan Y, Rao T, Cheng F. The bach1/G9a/Slc7a11 axis epigenetically promotes renal fibrosis by mediated ferroptosis. Int Immunopharmacol 2024; 143:113363. [PMID: 39393269 DOI: 10.1016/j.intimp.2024.113363] [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: 06/07/2024] [Revised: 09/21/2024] [Accepted: 10/05/2024] [Indexed: 10/13/2024]
Abstract
A high percentage of individuals with renal fibrosis are susceptible to developing chronic kidney disease (CKD), and conventional therapy fails to halt the progression of renal fibrosis and CKD. Here, we assessed the potential functions of G9a in a unilateral ureteral obstruction (UUO)-induced renal fibrosis mouse model. The expression of G9a was significantly increased in the fibrotic kidneys of patients and mice. G9a knockout inhibited inflammatory cytokine production and collagen deposition in mice, whereas its overexpression aggravated renal fibrosis in mice. In vitro, the knockdown of G9a alleviated the production of inflammatory cytokines and renal fibrosis. G9a, a histone methyltransferase, interacts with transcription factor Bach1 and activates ferroptosis by suppressing the transcription of Slc7a11 via dimethylation of histone 3 lysine 9 (H3K9me2) both in vivo and in vitro. Collectively, our findings indicate that G9a could be an attractive therapeutic target for renal fibrosis.
Collapse
Affiliation(s)
- Xiaobing Yao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Songyuan Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lijia Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuan Ruan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
5
|
Mimura I, Chen Z, Natarajan R. Epigenetic alterations and memory: key players in the development/progression of chronic kidney disease promoted by acute kidney injury and diabetes. Kidney Int 2024:S0085-2538(24)00918-9. [PMID: 39725223 DOI: 10.1016/j.kint.2024.10.031] [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: 02/25/2024] [Revised: 09/20/2024] [Accepted: 10/08/2024] [Indexed: 12/28/2024]
Abstract
Chronic kidney disease (CKD) is a highly prevalent global public health issue and can progress to renal failure. Survivors of acute kidney injury (AKI) have an increased risk of progressing to CKD by 8.8-fold and kidney failure by 3.1-fold. Further, 20% to 40% of individuals with diabetes will develop CKD, also known as diabetic kidney disease (DKD). Thus, preventing these kidney diseases can positively impact quality-of-life and life-expectancy outcomes for affected individuals. Frequent episodes of hyperglycemia and renal hypoxia are implicated in the pathophysiology of CKD. Prior periods of hyperglycemia/uncontrolled diabetes can result in development/progression of DKD even after achieving normoglycemia, a phenomenon known as metabolic memory or legacy effect. Similarly, in AKI, hypoxic memory is stored in renal cells even after recovery from the initial AKI episode and can transition to CKD. Epigenetic mechanisms involving DNA methylation, chromatin histone post-translational modifications, and noncoding RNAs are implicated in both metabolic and hypoxic memory, collectively known as "epigenetic memory." This epigenetic memory is generally reversible and provides a therapeutic avenue to ameliorate persistent disease progression due to hyperglycemia and hypoxia and prevent/ameliorate CKD progression. Indeed, therapeutic strategies targeting epigenetic memory are effective at preventing CKD development/progression in experimental models of AKI and DKD. Here, we review the latest in-depth evidence for epigenetic features in DKD and AKI, and in epigenetic memories of AKI-to-CKD transition or DKD development and progression, followed by translational and clinical implications of these epigenetic changes for the treatment of these widespread kidney disorders.
Collapse
Affiliation(s)
- Imari Mimura
- Division of Nephrology and Endocrinology, the University of Tokyo School of Medicine, Tokyo Japan.
| | - Zhuo Chen
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA.
| |
Collapse
|
6
|
Li Z, Mao C, Zhao Y, Zhao Y, Yi H, Liu J, Liang J. The STING antagonist SN-011 ameliorates cisplatin induced acute kidney injury via suppression of STING/NF-κB-mediated inflammation. Int Immunopharmacol 2024; 146:113876. [PMID: 39709905 DOI: 10.1016/j.intimp.2024.113876] [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: 09/17/2024] [Revised: 11/17/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
Acute kidney injury (AKI) is a critical clinical syndrome associated with both innate and adaptive immune responses and thus increases mortality. Nevertheless, specific therapeutics for AKI are scarce so far. Recent studies have revealed that knockout of STING alleviate AKI, suggesting that STING could be an attractive target for AKI therapy. SN-011, a promising STING inhibitor, has not been reported in studies of its anti-AKI activity. In this study, we sought to examine the effects of SN-011 on AKI and explore its underlying mechanism. Our findings indicate that SN-011 could modulate the NF-κB and MAPK pathways, suppress the expression of inflammatory factors, and decrease ROS release in the cisplatin-induced cell model. In addition, SN-011 blocked the nuclear translocation of NF-κB p65, further mitigating the inflammatory response. In vivo, SN-011 enhanced survival rates and alleviated renal dysfunction. According to gene set enrichment analysis of sequencing data from mouse kidneys, we further confirm that SN-011 modulates the NF-κB and MAPK pathways. Our study suggests that SN-011 could be an attractive anti-inflammatory agent for further anti-AKI research.
Collapse
Affiliation(s)
- Ziyang Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Can Mao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yixin Zhao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yanbin Zhao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Hanyu Yi
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Jin Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China.
| | - Jinqiang Liang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China.
| |
Collapse
|
7
|
Zhou J, Xiong KL, Wang HX, Sun WW, Ke H, Zhang SJ, Dong ZW, Fan L. BATF2/SINHCAF regulates the quantity and function of macrophages infected with Mycobacterium Tuberculosis via regulation of TTC23 through Wnt/β-catenin pathway. Int J Biol Macromol 2024; 288:138639. [PMID: 39672395 DOI: 10.1016/j.ijbiomac.2024.138639] [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: 10/02/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
Elucidating the pathogenic mechanism of Tuberculosis (TB) can contribute to control TB. Basic leucine zipper transcription factor ATF-like 2 (BATF2) belonging to a large family of leucine zipper transcription factors (TFs) termed bZip proteins, had been verified to have important value in the diagnosis of TB. However, its role and mechanism in TB had not been elucidated. The study aimed to explore its function and molecular mechanism in macrophages infected with Mycobacterium tuberculosis (Mtb). The results indicated that BATF2 inhibited cell proliferation, promoted inflammatory response and impaired the antibacterial and antigen-presenting capacity in macrophages for T cells through regulating its downstream gene TTC23 by interacting with SINHCAF. Above roles and regulations were dependent on β-catenin functions in macrophages infected with Mtb. Clinical samples verified that the expressions of BATF2 and TTC23 were significantly higher in the blood of patients with pulmonary TB compared with health controls. Altogether, BATF2 interacted with SINHCAF to regulate the quantity and function of macrophages during Mtb infection by targeting TTC23 through Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China
| | - Kun-Long Xiong
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China
| | - Hong-Xiu Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Wen Sun
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China
| | - Hui Ke
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China
| | - Shao-Jun Zhang
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China
| | - Zheng-Wei Dong
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lin Fan
- Department of Tuberculosis, Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai Key Lab of Tuberculosis, Shanghai, China.
| |
Collapse
|
8
|
Huang G, Xu J, Li Y, Song L, Wen C, Ruan Q, Wen Z, Qi J, Deng J, Liu Y. Corynoxine exerts the anti-tumor effect on esophageal squamous cell carcinoma principally via the EZH2-DUSP5-ERK1/2-mediated cell growth inhibition. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156103. [PMID: 39383633 DOI: 10.1016/j.phymed.2024.156103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Esophageal cancer is one of the most prevalent malignant tumors and the sixth largest cause of tumor-associated death worldwide. Squamous cell carcinoma (ESCC) accounts for 85 % of all esophageal cancer cases. ESCC treatment remains to be significantly difficult. Corynoxine (Cory) is a tetracyclic hydroxyindole alkaloid isolated from Uncaria macrophylla. It is unclear whether Cory has an anti-tumor effect on ESCC. PURPOSE To determine the anti-tumor activity of Cory and the associated mechanisms in ESCC. STUDY DESIGN Cory's effects on proliferation, apoptosis, migration, and invasion, as well as the underlying molecular causes were assessed using two ESCC cell lines, KYSE150 and TE-1. A xenograft mouse model was then applied to evaluate the anti-tumor activity of Cory in vivo. METHODS Western blot, assays including CCK-8, colony formation, EdU staining, TUNEL staining, cell scratch and Transwell, and a xenograft mouse model were used in this study. RESULTS Cory suppressed cell growth, provoked cell apoptosis, and hindered cell migration and invasion of ESCC cells. DUSP5 knockdown reduced the Cory-induced cell death and restored cell migration and invasion through ERK1/2 activation. Further analyses showed that Cory promoted DUSP5 expression via inhibiting EZH2 expression, leading to inactivation of ERK1/2 signaling and the subsequent cell growth inhibition of ESCC. In vivo experiments disclosed that Cory suppressed tumor growth of ESCC through upregulating DUSP5 expression. CONCLUSIONS Cory plays an anti-tumor role in ESCC by regulating EZH2-DUSP5-ERK1/2 signaling pathway. Cory may be promising to be a novel therapy for treating ESCC.
Collapse
Affiliation(s)
- Gang Huang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; The People's Hospital of Beilun District, Ningbo 315000, China
| | - Jiale Xu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yingchao Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Liangtao Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Chunmei Wen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Qingqing Ruan
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Zhikai Wen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035 China
| | - Jinxia Qi
- Biobank, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jie Deng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Yu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
9
|
Deng R, Yang H, Zhong W, Zhou J, Huang G, Zeng K. CITED2 Mediates Metabolic Reprogramming in Renal Tubular Epithelial Cells via the AKT Signaling Pathway to Induce Sepsis-Associated Acute Kidney Injury. J Inflamm Res 2024; 17:9485-9505. [PMID: 39600684 PMCID: PMC11590677 DOI: 10.2147/jir.s486596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Background Sepsis-associated acute kidney injury (S-AKI) is a prevalent and severe clinical complication in intensive care units (ICUs) and is associated with high mortality and poor prognosis. The dysfunction of renal tubular epithelial cells (TECs), particularly through their metabolic reprogramming, plays a critical role in the onset and progression of S-AKI. CITED2 is shown to regulate a variety of cellular processes, but its specific impact on TECs metabolism and S-AKI pathogenesis remains unclear. The aim of this study was to investigate the role of CITED2 in the metabolic reprogramming of TECs and its effects on inflammation and kidney injury in S-AKI. Material and Methods The C57BL/6 mouse model of S-AKI was established using cecal ligation and puncture (CLP). We assessed the inflammatory responses, glucose metabolism and CITED2 expression in the kidneys of septic mice. Additionally, the effect of CITED2 on TECs metabolism and inflammation was evaluated using in vivo and in vitro models. CITED2 silencing and overexpression were employed to elucidate its regulatory role, focusing on the AKT signaling pathway. Results S-AKI causes structural and functional kidney damage, aggravated inflammatory responses, and dysregulated glucose metabolism, accompanied by increased expression of CITED2. CITED2 silencing attenuated TECs metabolic dysfunction and reduced inflammation, thereby protecting the kidney from injury. Conversely, CITED2 overexpression exacerbated TECs metabolic dysfunction, promoted inflammatory responses, and worsened kidney injury. Mechanistically, CITED2 regulates TEC metabolism through the AKT signaling pathway, promoting S-AKI-related inflammation and contributing to kidney injury. Conclusion CITED2 drives the metabolic reprogramming of TECs through the AKT signaling pathway, thereby aggravating the inflammatory response and leading to kidney injury, highlighting its critical role in S-AKI. Targeting CITED2 inhibition may represent a novel therapeutic approach for managing S-AKI.
Collapse
Affiliation(s)
- Ruiming Deng
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, 350004, People’s Republic of China
- Department of Anesthesiology, Ganzhou People’s Hospital, Ganzhou City, Jiangxi Province, 341000, People’s Republic of China
| | - Hang Yang
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, 350004, People’s Republic of China
| | - Weibo Zhong
- Department of Anesthesiology, Ganzhou People’s Hospital, Ganzhou City, Jiangxi Province, 341000, People’s Republic of China
| | - Juan Zhou
- Department of Thyroid and Breast Surgery, Ganzhou People’s Hospital, Ganzhou City, Jiangxi Province, 341000, People’s Republic of China
| | - Guiming Huang
- Department of Anesthesiology, Ganzhou People’s Hospital, Ganzhou City, Jiangxi Province, 341000, People’s Republic of China
| | - Kai Zeng
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, 350004, People’s Republic of China
| |
Collapse
|
10
|
Wang Y, Chen Y, Yang J, Sun W, Zhang X. Histone demethylase JMJD1C advances macrophage foam cell formation and atherosclerosis progression by promoting the transcription of PCSK9. J Physiol Biochem 2024:10.1007/s13105-024-01058-3. [PMID: 39511107 DOI: 10.1007/s13105-024-01058-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
Macrophage is considered as a critical driving factor in the progression of atherosclerosis (AS), and epigenetic heterogeneity contributes important mechanisms in this process. Here, we identified that a histone demethylase jumonji domain-containing protein 1 C (JMJD1C) is a promising biomarker for atherosclerotic cerebral infarction through clinical analysis. Then, AOPE-/- mice fed with a high fat diet and RAW264.7 cells induced by oxidized low-density lipoprotein (ox-LDL) were used as AS models to verify the function of JMJD1C in AS development in vivo and in vitro. JMJD1C knockdown significantly reduced plaque area, inflammation and endothelial damage in AS model mice, and also alleviated foam cell formation, inflammatory cytokines production and cell apoptosis in ox-LDL-treated RAW264.7 cells. Mechanistically, JMJD1C promoted the transcription of proprotein convertase subtilisin/kexin type 9 (PCSK9) through mediating H3 Lysine 9 demethylation. The effects of JMJD1C knockdown on ox-LDL-induced macrophages were blocked by PCSK9 overexpression. Altogether, our study proves that JMJD1C advances macrophage foam cell formation, inflammation and apoptosis to accelerate AS progression through H3 demethylation of PCSK9. The findings underscore the important role of JMJD1C-mediated histone modification in macrophage regulation and AS progression, which brings a new insight into the pathobiology of AS.
Collapse
Affiliation(s)
- Yiming Wang
- Second Department of Neurology, Xinjiang Uygur Autonomous Region Chinese Medicine Hospital, 116 Huanghe Road, Shayibak District, Urumqi, Xinjiang, 830000, China
| | - Yifei Chen
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Jianbo Yang
- Second Department of Neurology, Xinjiang Uygur Autonomous Region Chinese Medicine Hospital, 116 Huanghe Road, Shayibak District, Urumqi, Xinjiang, 830000, China
| | - Wei Sun
- Second Department of Neurology, Xinjiang Uygur Autonomous Region Chinese Medicine Hospital, 116 Huanghe Road, Shayibak District, Urumqi, Xinjiang, 830000, China
| | - Xiaoning Zhang
- Second Department of Neurology, Xinjiang Uygur Autonomous Region Chinese Medicine Hospital, 116 Huanghe Road, Shayibak District, Urumqi, Xinjiang, 830000, China.
| |
Collapse
|
11
|
Xu T, Shen Y, Guo R, Luo C, Niu Y, Luo Z, Zhu Z, Wu Z, Zhao X, Luo H, Gao Y. Mutual regulation between histone methyltransferase Suv39h1 and the Wnt/β-catenin signaling pathway promoted cell proliferation and inhibited apoptosis in bone marrow mesenchymal stem cells exposed to hydroquinone. Toxicology 2024; 508:153932. [PMID: 39179171 DOI: 10.1016/j.tox.2024.153932] [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: 06/21/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Hydroquinone (HQ), a metabolite of benzene, is frequently utilized as a surrogate for benzene in in vitro studies and is associated with the development of acute myeloid leukemia (AML). In the hemotoxicity caused by benzene and HQ, cell apoptosis plays a key role. However, the molecular mechanisms underlying HQ are unknown. Studies have indicated that Suv39h1 is involved in regulating cell division and proliferation by regulating histone H3K9me3. Meanwhile, the Wnt/β-catenin signaling pathway also plays a significant role in cell proliferation and apoptosis. Therefore, this study was aimed at exploring the regulatory role of Suv39h1 and the Wnt/β-catenin signaling pathway in the effects of HQ on bone marrow mesenchymal stem cells (BMSCs), as well as its influence on cell proliferation and apoptosis. The results demonstrated that HQ elevated the levels of Suv39h1 and H3K9me3 and activated the Wnt/β-catenin signaling pathway by upregulating β-catenin, Wnt2b, C-myc, and Cyclin D1 and downregulating Wnt5a, resulting in an increase in cell growth and a decrease in apoptosis. Suv39h1 knockdown inhibited the Wnt/β-catenin signaling pathway. Meanwhile, inhibition of the Wnt/β-catenin signaling pathway resulted in the down-regulation of Suv39h1 and H3K9me3 in BMSCs. They both promoted cell proliferation and inhibited apoptosis in the effects of HQ on BMSCs by downregulating the expression of Cyt-C, Bax, Caspase 3, and Caspase 9 and upregulating the expression of Bcl-xl. Therefore, we concluded that Suv39h1 and the Wnt/β-catenin signaling pathway may mutually regulate each other in the effects of HQ on BMSCs in order to ameliorate the altered function of BMSCs.
Collapse
Affiliation(s)
- Tao Xu
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| | - Yilin Shen
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| | - Runmin Guo
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| | - Chiheng Luo
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| | - Yibo Niu
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| | - Zhilong Luo
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| | - Zhongxin Zhu
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| | - Zehui Wu
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| | - Xinyu Zhao
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| | - Hao Luo
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| | - Yuting Gao
- Shunde Women and Children's Hospital of Guangdong Medical University, School of Public Health, Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong, China.
| |
Collapse
|
12
|
Xu T, Shen Y, Guo R, Luo C, Niu Y, Luo Z, Zhu Z, Wu Z, Zhao X, Luo H, Gao Y. Mutual regulation between histone methyltransferase Suv39h1 and the Wnt/β-catenin signaling pathway promoted cell proliferation and inhibited apoptosis in bone marrow mesenchymal stem cells exposed to hydroquinone. Toxicology 2024; 508:153932. [DOI: https:/doi.org/10.1016/j.tox.2024.153932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
|
13
|
Marquez VE. 3-Deazaneplanocin A (DZNep): A Drug That Deserves a Second Look. J Med Chem 2024; 67:17964-17979. [PMID: 39392180 DOI: 10.1021/acs.jmedchem.4c01566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The emerging data compiled during the past five years on 3-deazaneplanocin (DZNep) provide compelling evidence to reevaluate this drug as a better alternative over the specific catalytic inhibitors of histone methyl transferases (HTMs). The indirect mechanism of DZNep via inhibition of AdoHcy-ase, once considered a liability due to possible side effects, has now shown to be rather beneficial as additional pathways targeted by DZNep are important contributors to its superior anticancer properties. Furthermore, DZNep has demonstrated the ability to induce proteasomal degradation of its target and reduce toxicity in combination with well-established antitumor therapies in animal models. In addition, DZNep has shown important effects in suppressing fibrosis and inflammation in liver, kidney, peritoneum, and airways. Finally, inhibition of mRNA m6A methylation by DZNep suppresses the synthesis of the viral genome in SARS-Cov-2 infection and promises to have important therapeutic value when combined with its potent antiviral efficacy and anti-inflammatory effects.
Collapse
Affiliation(s)
- Victor E Marquez
- Chemical Biology Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, United States
| |
Collapse
|
14
|
Pan S, Yuan T, Xia Y, Yu W, Li H, Rao T, Ye Z, Li L, Zhou X, Cheng F. SMYD2 Promotes Calcium Oxalate-Induced Glycolysis in Renal Tubular Epithelial Cells via PTEN Methylation. Biomedicines 2024; 12:2279. [PMID: 39457592 PMCID: PMC11504487 DOI: 10.3390/biomedicines12102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Damage to renal tubular cells (RTCs) represents a critical pathological manifestation in calcium oxalate (CaOx) stone disease, but the underlying mechanism remains elusive. Energy metabolism reprogramming is a vital influencer of RTC survival, and SMYD2 is a histone methylation transferase that has been extensively implicated in various metabolic disorders. Hence, this research aimed to identify whether SMYD2 induces the reprogramming of energy metabolism in RTCs exposed to CaOx nephrolithiasis. Methods: Kidney samples were obtained from patients who underwent laparoscopic nephrectomy for non-functioning kidneys caused by nephrolithiasis. The glyoxylate-induced CaOx stone mice model was established and treated with AZ505. The SMYD2-knockout HK-2 cell line was constructed. Histological changes were evaluated by HE, VK, Tunel, Masson stainings. The molecular mechanism was explored through co-immunoprecipitation and western blotting. Results: The results found that SMYD2 upregulation led to energy reprogramming to glycolysis in human kidney tissue samples and in mice with CaOx nephrolithiasis. We also identified the substantial involvement of glycolysis in the induction of apoptosis, inflammation, and epithelial-mesenchymal transition (EMT) in HK-2 cells caused by calcium oxalate monohydrate (COM). In vivo and in vitro results demonstrated that SMYD2 inhibition reduces glycolysis, kidney injury, and fibrosis. Mechanistically, SMYD2 was found to promote metabolic reprogramming of RTCs toward glycolysis by activating the AKT/mTOR pathway via methylated PTEN, which mediates CaOx-induced renal injury and fibrosis. Conclusions: Our findings reveal an epigenetic regulatory role of SMYD2 in metabolic reprogramming in CaOx nephrolithiasis and associated kidney injury, suggesting that targeting SMYD2 and glycolysis may represent a potential therapeutic strategy for CaOx-induced kidney injury and fibrosis.
Collapse
|
15
|
Zhao Q, Zhang R, Wang Y, Li T, Xue J, Chen Z. FOXQ1, deubiquitinated by USP10, alleviates sepsis-induced acute kidney injury by targeting the CREB5/NF-κB signaling axis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167331. [PMID: 38960057 DOI: 10.1016/j.bbadis.2024.167331] [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: 11/22/2023] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Sepsis-induced acute kidney injury (S-AKI) is a severe and frequent complication that occurs during sepsis. This study aimed to understand the role of FOXQ1 in S-AKI and its potential upstream and downstream regulatory mechanisms. A cecal ligation and puncture induced S-AKI mouse model in vivo and an LPS-induced HK-2 cell model in vitro were used. FOXQ1 was significantly upregulated in CLP mice and downregulated in the LPS-induced HK-2 cells. Upregulation of FOXQ1 improved kidney injury and dysfunction in CLP mice. Overexpression of FOXQ1 remarkably suppressed the apoptosis and inflammatory response via down-regulating oxidative stress indicators and pro-inflammatory factors (IL-1β, IL-6, and TNF-α), both in vivo and in vitro. From online analysis, the CREB5/NF-κB axis was identified as the downstream target of FOXQ1. FOXQ1 transcriptionally activated CREB5, upregulating its expression. Overexpression of FOXQ1 suppressed the phosphorylation level and nucleus transport of p65. Rescue experiments showed that CREB5 mediates the protective role of FOXQ1 on S-AKI. Furthermore, FOXQ1 was identified as a substrate of USP10, a deubiquitinating enzyme. Ectopic expression of USP10 reduced the ubiquitination of FOXQ1, promoting its protein stability. USP10 upregulation alleviated LPS-induced cell apoptosis and inflammatory response, while suppression of FOXQ1 augmented these trends. Collectively, our results suggest that FOXQ1, deubiquitinated by USP10, plays a protective role in S-AKI induced inflammation and apoptosis by targeting CREB5/NF-κB axis.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ran Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yu Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Zhiguang Chen
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
16
|
Zhang Y, Zhang Y. Knockdown of USP7 alleviates atherosclerosis in ApoE-deficient mice by regulating EZH2 expression. Open Life Sci 2024; 19:20220929. [PMID: 39310812 PMCID: PMC11416069 DOI: 10.1515/biol-2022-0929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 09/25/2024] Open
Abstract
Atherosclerosis (AS) is a chronic vascular disease associated with lipid accumulation. Understanding the molecular mechanisms of AS is essential. Ubiquitin-specific protease 7 (USP7) is a deubiquitination enzyme involved in various cellular processes, including lipid metabolism. In this study, we aimed to elucidate the role of USP7 in AS progression and its underlying mechanism using ApoE-deficient mice. We found that USP7 ablation improved the morphological characteristics of AS in these mice. USP7 knockdown reduced inflammation, evidenced by decreases in inflammatory markers IL-6, TNF-α, and IL-1β by 35, 40, and 38%, respectively (p < 0.01). Additionally, USP7 depletion reduced oxidative stress, indicated by a 30% reduction in malondialdehyde levels and increases in superoxide dismutase and glutathione peroxidase levels by 25 and 28%, respectively (p < 0.01). Moreover, USP7 knockdown blocked lipid accumulation in aortic tissue cells. Mechanistically, USP7 knockdown inhibited enhancer of Zeste Homolog 2 (EZH2) expression, thereby suppressing AS progression. In conclusion, USP7 depletion alleviated AS progression in ApoE-deficient mice by targeting EZH2 expression. USP7 may serve as a therapeutic target for AS.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Yanchun Zhang
- Department of Cardiology, Shuyang Hospital of Traditional Chinese Medicine, No. 28 Shanghai Road, Shuyang, Jiangsu, 223600, China
| |
Collapse
|
17
|
Yuan N, Chen Y, Yan Y, Wang F, Xu X, Wang M, Diao J, Xiao W. Myricetin alleviates renal tubular epithelial-mesenchymal transition via NOX4/NF- κB/snail axis in diabetic nephropathy based on network pharmacology analysis. Heliyon 2024; 10:e35234. [PMID: 39224244 PMCID: PMC11367043 DOI: 10.1016/j.heliyon.2024.e35234] [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: 04/02/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Diabetic nephropathy (DN), a leading cause of end-stage renal disease, remains a formidable challenge in diabetes management due to the complex nature of its pathogenesis, particularly the epithelial-mesenchymal transition (EMT) process. Our innovative study leverages network pharmacology to explore the therapeutic potentials of Myricetin, a natural flavonoid, focusing on its effects against NOX4, a critical mediator in DN progression. This investigation marks a pioneering approach by integrating network pharmacology to predict and elucidate the inhibitory relationship between Myricetin and NOX4. Utilizing a high-fat diet/streptozotocin (HFD/STZ) induced DN mouse model, we delved into the effects of Myricetin on renal EMT processes. Through network pharmacology analyses coupled with molecular docking studies, we identified and confirmed Myricetin's binding efficacy to NOX4. Extensive in vitro and in vivo experiments further established Myricetin's significant impact on mitigating EMT by modulating the NOX4-NF-κB-Snail signaling pathway. Results from our research demonstrated notable improvements in renal function and reductions in tissue fibrosis among treated HFD/STZ mice. By curtailing NOX4 expression, Myricetin effectively reduced reactive oxygen species (ROS) production, thereby inhibiting NF-κB activation and subsequent Snail expression, crucial steps in the EMT pathway. Supported by both theoretical predictions and empirical validations, this study unveils the mechanism underlying Myricetin's modulation of EMT in DN through disrupting the NOX4-NF-κB-Snail axis. These findings not only contribute a new therapeutic avenue for DN treatment but also underscore the utility of network pharmacology in advancing drug discovery processes.
Collapse
Affiliation(s)
- Ningning Yuan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yuchi Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yangtian Yan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Fujing Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xinyao Xu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Mingqing Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jianxin Diao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wei Xiao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| |
Collapse
|
18
|
Lu D, Tu L, Hu Y, Cai X. Prognostic value of systemic inflammatory response index for acute kidney injury and the prognosis of pediatric patients in critical care units. PLoS One 2024; 19:e0306884. [PMID: 39208322 PMCID: PMC11361669 DOI: 10.1371/journal.pone.0306884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/25/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND We proposed a link between the first systemic inflammatory response index (SIRI) and acute kidney injury (AKI), as well as the prognosis of pediatric patients in intensive care units (PICU). METHODS This study comprised 5114 children from the pediatric-specific intensive care (PIC) database. SIRI was estimated as a neutrophil monocyte lymphocyte ratio. All patients were arbitrarily allocated to the training set (n = 3593) and the validation cohort (n = 1521) and divided into two groups depending on their SIRI levels. The diagnostic value of SIRI for pediatric ICU patients was subsequently determined using LASSO regression models. RESULTS After controlling for additional confounding variables in the training set, the higher SIRI value (≥ 0.59) had a greater risk of AKI (adjusted odds ratio, OR, 3.95, 95% confidence interval, 95%CI, 2.91-5.36, P<0.001) and in-hospital mortality (hazard ratio, HR, 5.01, 95%CI 2.09-12.03, P<0.001). Similar findings were discovered in the validation set. Furthermore, the suggested nomogram derived from SIRI and other clinical metrics showed outstanding calibration capability as well as therapeutic usefulness in both groups. CONCLUSIONS SIRI is a reliable and useful factor for AKI and fatality in pediatric ICU patients, and the proposed nomogram based on SIRI yields an appropriate prediction value for critically sick pediatric patients.
Collapse
Affiliation(s)
- Danchi Lu
- Department of Emergency, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Lijuan Tu
- Department of Emergency, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yugang Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaofang Cai
- Department of Emergency, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
19
|
Chen L, Wang R, Lv X, Kan M, Zhang H, Qiu W, Chen S, Zhao J, Wen X, Meng X, Wang H, Zang H. Hepatic-derived BMP9 is involved in hepatic fibrosis-induced kidney injury through inhibition of renal VEGFA. Biochem Pharmacol 2024; 226:116371. [PMID: 38885771 DOI: 10.1016/j.bcp.2024.116371] [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: 12/20/2023] [Revised: 05/01/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Clinical observations suggest that acute kidney injury (AKI) occurs in approximately 20-50% of hospitalized cirrhotic patients, suggesting a link between the liver and kidney. Bone morphogenetic protein 9 (BMP9) is a protein produced primarily by the liver and can act on other tissues at circulating systemic levels. Previous studies have demonstrated that controlling abnormally elevated BMP9 in acute liver injury attenuates liver injury; however, reports on whether BMP9 plays a role in liver injury-induced AKI are lacking. By testing we found that liver injury in mice after bile duct ligation (BDL) was accompanied by a significant upregulation of the kidney injury marker kidney injury molecule (KIM-1). Interestingly, all these impairments were alleviated in the kidneys of hepatic BMP9 knockout (BMP9-KO) mice. Peritubular capillary injury is a key process leading to the progression of AKI, and previous studies have demonstrated that vascular endothelial growth factor A (VEGFA) plays a key role in maintaining the renal microvascular system. In animal experiments, we found that high levels of circulating BMP9 had an inhibitory effect on VEGFA expression, while renal tubular epithelial cell injury was effectively attenuated by VEGFA supplementation in the hypoxia-enriched-oxygen (H/R) constructs of the AKI cell model in both humans and mice. Overall, we found that elevated BMP9 in hepatic fibrosis can affect renal homeostasis by regulating VEGFA expression. Therefore, we believe that targeting BMP9 therapy may be a potential means to address the problem of clinical liver fibrosis combined with AKI.
Collapse
Affiliation(s)
- Le Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ruonan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaodong Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Kan
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongtao Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Qiu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shao Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiuling Zhao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hua Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongmei Zang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
20
|
Beamish JA, Watts JA, Dressler GR. Gene regulation in regeneration after acute kidney injury. J Biol Chem 2024; 300:107520. [PMID: 38950862 PMCID: PMC11325799 DOI: 10.1016/j.jbc.2024.107520] [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: 06/03/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/03/2024] Open
Abstract
Acute kidney injury (AKI) is a common condition associated with significant morbidity, mortality, and cost. Injured kidney tissue can regenerate after many forms of AKI. However, there are no treatments in routine clinical practice to encourage recovery. In part, this shortcoming is due to an incomplete understanding of the genetic mechanisms that orchestrate kidney recovery. The advent of high-throughput sequencing technologies and genetic mouse models has opened an unprecedented window into the transcriptional dynamics that accompany both successful and maladaptive repair. AKI recovery shares similar cell-state transformations with kidney development, which can suggest common mechanisms of gene regulation. Several powerful bioinformatic strategies have been developed to infer the activity of gene regulatory networks by combining multiple forms of sequencing data at single-cell resolution. These studies highlight not only shared stress responses but also key changes in gene regulatory networks controlling metabolism. Furthermore, chromatin immunoprecipitation studies in injured kidneys have revealed dynamic epigenetic modifications at enhancer elements near target genes. This review will highlight how these studies have enhanced our understanding of gene regulation in injury response and regeneration.
Collapse
Affiliation(s)
- Jeffrey A Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason A Watts
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Gregory R Dressler
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
21
|
Pan H, Xu R, Zhang Y. Role of SPRY4 in health and disease. Front Oncol 2024; 14:1376873. [PMID: 38686189 PMCID: PMC11056578 DOI: 10.3389/fonc.2024.1376873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
SPRY4 is a protein encoding gene that belongs to the Spry family. It inhibits the mitogen-activated protein kinase (MAPK) signaling pathway and plays a role in various biological functions under normal and pathological conditions. The SPRY4 protein has a specific structure and interacts with other molecules to regulate cellular behavior. It serves as a negative feedback inhibitor of the receptor protein tyrosine kinases (RTK) signaling pathway and interferes with cell proliferation and migration. SPRY4 also influences inflammation, oxidative stress, and cell apoptosis. In different types of tumors, SPRY4 can act as a tumor suppressor or an oncogene. Its dysregulation is associated with the development and progression of various cancers, including colorectal cancer, glioblastoma, hepatocellular carcinoma, perihilar cholangiocarcinoma, gastric cancer, breast cancer, and lung cancer. SPRY4 is also involved in organ development and is associated with ischemic diseases. Further research is ongoing to understand the expression and function of SPRY4 in specific tumor microenvironments and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Hao Pan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renjie Xu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Jin J, Liu XM, Shao W, Meng XM. Nucleic acid and protein methylation modification in renal diseases. Acta Pharmacol Sin 2024; 45:661-673. [PMID: 38102221 PMCID: PMC10943093 DOI: 10.1038/s41401-023-01203-6] [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: 06/27/2023] [Accepted: 11/18/2023] [Indexed: 12/17/2023] Open
Abstract
Although great efforts have been made to elucidate the pathological mechanisms of renal diseases and potential prevention and treatment targets that would allow us to retard kidney disease progression, we still lack specific and effective management methods. Epigenetic mechanisms are able to alter gene expression without requiring DNA mutations. Accumulating evidence suggests the critical roles of epigenetic events and processes in a variety of renal diseases, involving functionally relevant alterations in DNA methylation, histone methylation, RNA methylation, and expression of various non-coding RNAs. In this review, we highlight recent advances in the impact of methylation events (especially RNA m6A methylation, DNA methylation, and histone methylation) on renal disease progression, and their impact on treatments of renal diseases. We believe that a better understanding of methylation modification changes in kidneys may contribute to the development of novel strategies for the prevention and management of renal diseases.
Collapse
Affiliation(s)
- Juan Jin
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
- Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Xue-Mei Liu
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Wei Shao
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, China.
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-Inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
23
|
Liu R, Xu R, Yan S, Li P, Jia C, Sun H, Sheng K, Wang Y, Zhang Q, Guo J, Xin X, Li X, Guo D. Hi-C, a chromatin 3D structure technique advancing the functional genomics of immune cells. Front Genet 2024; 15:1377238. [PMID: 38586584 PMCID: PMC10995239 DOI: 10.3389/fgene.2024.1377238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/13/2024] [Indexed: 04/09/2024] Open
Abstract
The functional performance of immune cells relies on a complex transcriptional regulatory network. The three-dimensional structure of chromatin can affect chromatin status and gene expression patterns, and plays an important regulatory role in gene transcription. Currently available techniques for studying chromatin spatial structure include chromatin conformation capture techniques and their derivatives, chromatin accessibility sequencing techniques, and others. Additionally, the recently emerged deep learning technology can be utilized as a tool to enhance the analysis of data. In this review, we elucidate the definition and significance of the three-dimensional chromatin structure, summarize the technologies available for studying it, and describe the research progress on the chromatin spatial structure of dendritic cells, macrophages, T cells, B cells, and neutrophils.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Dianhao Guo
- School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
24
|
Yan X, Xia Y, Li B, Ye Z, Li L, Yuan T, Song B, Yu W, Rao T, Ning J, Lin F, Mei S, Mao Z, Zhou X, Li W, Cheng F. The SOX4/EZH2/SLC7A11 signaling axis mediates ferroptosis in calcium oxalate crystal deposition-induced kidney injury. J Transl Med 2024; 22:9. [PMID: 38169402 PMCID: PMC10763321 DOI: 10.1186/s12967-023-04793-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024] Open
Abstract
Epigenetic regulation is reported to play a significant role in the pathogenesis of various kidney diseases, including renal cell carcinoma, acute kidney injury, renal fibrosis, diabetic nephropathy, and lupus nephritis. However, the role of epigenetic regulation in calcium oxalate (CaOx) crystal deposition-induced kidney injury remains unclear. Our study demonstrated that the upregulation of enhancer of zeste homolog 2 (EZH2)-mediated ferroptosis facilitates CaOx-induced kidney injury. CaOx crystal deposition promoted ferroptosis in vivo and in vitro. Usage of liproxstatin-1 (Lip-1), a ferroptosis inhibitor, mitigated CaOx-induced kidney damage. Single-nucleus RNA-sequencing, RNA-sequencing, immunohistochemical and western blotting analyses revealed that EZH2 was upregulated in kidney stone patients, kidney stone mice, and oxalate-stimulated HK-2 cells. Experiments involving in vivo EZH2 knockout, in vitro EZH2 knockdown, and in vivo GSK-126 (an EZH2 inhibitor) treatment confirmed the protective effects of EZH2 inhibition on kidney injury and ferroptosis. Mechanistically, the results of RNA-sequencing and chromatin immunoprecipitation assays demonstrated that EZH2 regulates ferroptosis by suppressing solute carrier family 7, member 11 (SLC7A11) expression through trimethylation of histone H3 lysine 27 (H3K27me3) modification. Additionally, SOX4 regulated ferroptosis by directly modulating EZH2 expression. Thus, this study demonstrated that SOX4 facilitates ferroptosis in CaOx-induced kidney injury through EZH2/H3K27me3-mediated suppression of SLC7A11.
Collapse
Affiliation(s)
- Xinzhou Yan
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Yuqi Xia
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Bojun Li
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Zehua Ye
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Lei Li
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Tianhui Yuan
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Baofeng Song
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Weimin Yu
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Ting Rao
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Jinzhuo Ning
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Shuqin Mei
- Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China
| | - Zhiguo Mao
- Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China.
| | - Wei Li
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China.
| | - Fan Cheng
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, People's Republic of China.
| |
Collapse
|
25
|
Wang J, Wan X, Le Q. Cross-regulation between SOX9 and the canonical Wnt signalling pathway in stem cells. Front Mol Biosci 2023; 10:1250530. [PMID: 37664185 PMCID: PMC10469848 DOI: 10.3389/fmolb.2023.1250530] [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: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023] Open
Abstract
SOX9, a member of the SRY-related HMG-box transcription factors, has been reported to critically regulate fetal development and stem cell homeostasis. Wnt signalling is a highly conserved signalling pathway that controls stem cell fate decision and stemness maintenance throughout embryonic development and adult life. Many studies have shown that the interactions between SOX9 and the canonical Wnt signalling pathway are involved in many of the physiological and pathological processes of stem cells, including organ development, the proliferation, differentiation and stemness maintenance of stem cells, and tumorigenesis. In this review, we summarize the already-known molecular mechanism of cross-interactions between SOX9 and the canonical Wnt signalling pathway, outline its regulatory effects on the maintenance of homeostasis in different types of stem cells, and explore its potential in translational stem cell therapy.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Xichen Wan
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Qihua Le
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Research Center, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Myopia Key Laboratory of Ministry of Health, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| |
Collapse
|
26
|
Sun L, Li X, Luo H, Guo H, Zhang J, Chen Z, Lin F, Zhao G. EZH2 can be used as a therapeutic agent for inhibiting endothelial dysfunction. Biochem Pharmacol 2023; 213:115594. [PMID: 37207700 DOI: 10.1016/j.bcp.2023.115594] [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: 02/26/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023]
Abstract
Enhancer of zeste homolog 2 (EZH2) is a catalytic subunit of polycomb repressor complex 2 and plays important roles in endothelial cell homeostasis. EZH2 functionally methylates lysine 27 of histone H3 and represses gene expression through chromatin compaction. EZH2 mediates the effects of environmental stimuli by regulating endothelial functions, such as angiogenesis, endothelial barrier integrity, inflammatory signaling, and endothelial mesenchymal transition. Numerous studies have been conducted to determine the significance of EZH2 in endothelial function. The aim of this review is to provide a concise summary of the roles EZH2 plays in endothelial function and elucidate its therapeutic potential in cardiovascular diseases.
Collapse
Affiliation(s)
- Li Sun
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Xuefang Li
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Hui Luo
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Huige Guo
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Jie Zhang
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Zhigang Chen
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Fei Lin
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China.
| | - Guoan Zhao
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China.
| |
Collapse
|