1
|
Liu L, Zheng Z, Huang Y, Su H, Wu G, Deng Z, Li Y, Xie G, Li J, Zou F, Chen X. HSP90 N-terminal inhibition promotes mitochondria-derived vesicles related metastasis by reducing TFEB transcription via decreased HSP90AA1-HCFC1 interaction in liver cancer. Autophagy 2025; 21:639-663. [PMID: 39461872 PMCID: PMC11849932 DOI: 10.1080/15548627.2024.2421703] [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: 11/24/2023] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer cells compensate with increasing mitochondria-derived vesicles (MDVs) to maintain mitochondrial homeostasis, when canonical MAP1LC3B/LC3B (microtubule associated protein 1 light chain 3 beta)-mediated mitophagy is lacking. MDVs promote the transport of mitochondrial components into extracellular vesicles (EVs) and induce tumor metastasis. Although HSP90 (heat shock protein 90) chaperones hundreds of client proteins and its inhibitors suppress tumors, HSP90 inhibitors-related chemotherapy is associated with unexpected metastasis. Herein, we find that HSP90 inhibitor causes mitochondrial damage but stimulates the low LC3-induced MDVs and the release of MDVs-derived EVs. However, why LC3 decreases and what is the transcriptional regulatory mechanism of MDVs formation under HSP90 inhibition remain unknown. Because TFEB (transcription factor EB) is the most important mitophagy transcription factor, and the HSP90 client HCFC1 (host cell factor C1) regulates TFEB transcription, there should be a hidden connection between TFEB, HCFC1 and HSP90 in MDVs formation. Our results support the idea that HSP90 N-terminal inhibition reduces TFEB transcription via decreased HSP90AA1-HCFC1 interaction, which prevents HCFC1 from binding to the TFEB proximal promoter region. Decreased TFEB transcription and consequently reduced LC3, ultimately promoted MDVs formation. Blocking MDVs formation with the microtubule inhibitor nocodazole (NOC) activates the HCFC1-TFEB-LC3 axis, weakens HSP90 inhibitors-induced MDVs and the release of MDVs-derived EVs, inhibits the growth of tumor cell spheres and primary liver tumors, and reduces the extravasation of cancer cells to secondary metastatic sites. Taken together, these data suggest that combination therapy should be used to reduce the metastatic risk of low TFEB-triggered-MDVs formation caused by HSP90 inhibitors.Abbreviation: ACIs: ATP-competitive inhibitors; BaFA1: bafilomycin A1; CCCP: carbonyl cyanide 3-chlorophenylhydrazone; ChIP: chromatin immunoprecipitation; CHX: cycloheximide; CTD: C-terminal domain; EVs: extracellular vesicles; HCFC1: host cell factor C1; HSP90: heat shock protein 90; ILVs: intralumenal vesicles; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MD: middle domain; MDVs: mitochondria-derived vesicles; MQC: mitochondrial quality control; ΔΨm: mitochondrial membrane potential; MVBs: multivesicular bodies; NB: novobiocin; TEM: transmission electron microscopy; TFEB: transcription factor EB; TFs: transcription factors. NOC: nocodazole; NTD: N-terminal nucleotide binding domain; OCR: oxygen consumption rate; RFP: red fluorescent protein; ROS: reactive oxygen species; STA9090: Ganetespib; VPS35: VPS35 retromer complex component.
Collapse
Affiliation(s)
- Lixia Liu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhenming Zheng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yaling Huang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hairou Su
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guibing Wu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zihao Deng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yan Li
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guantai Xie
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jieyou Li
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xuemei Chen
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Zou J, Geng R, Zhang Z, Ji X, Yin Z, Wang D, Guo R, Chen L, Liu J. Novel role of the SOX4/CSNK2A1 axis in regulating TOP2A phosphorylation in breast cancer progression. FASEB J 2025; 39:e70315. [PMID: 39931818 DOI: 10.1096/fj.202401907rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 05/08/2025]
Abstract
This study examines the critical role of DNA topoisomerase II alpha (TOP2A) phosphorylation in breast cancer progression, regulated by the SRY-box transcription factor 4 (SOX4)/Casein kinase II subunit alpha 1 (CSNK2A1) axis. Using integrated transcriptomic and proteomic analyses, data were sourced from the Clinical Proteomic Tumor Analysis Consortium (CPTAC) and The Cancer Genome Atlas (TCGA) databases. To explore the dataset, differential analysis, kinase-substrate enrichment analysis (KSEA), and weighted gene co-expression network analysis (WGCNA) were performed. Immune profiling, combined with survival analysis, revealed the prognosis linked to different immune profiles in breast cancer patients. In vitro experiments assessed the effect of SOX4 on CSNK2A1 promoter activity through real-time quantitative polymerase chain reaction (RT-qPCR), Western blot, dual-luciferase reporter assays, and chromatin immunoprecipitation (ChIP). The phosphorylation level of TOP2A was also measured. Cell proliferation, migration, and invasion were evaluated using cell counting kit-8 (CCK-8), colony formation, and Transwell assays. In vivo studies extended to mouse models, where the effect of SOX4 on CSNK2A1-TOP2A phosphorylation was analyzed about tumor growth and metastasis. The results showed that upregulation of SOX4 increases CSNK2A1 transcription, which in turn promotes TOP2A phosphorylation and accelerates breast cancer progression. The clinical analysis identified three immune profiles, with the intermediate profile associated with a poorer prognosis, possibly due to enhanced TOP2A phosphorylation mediated by SOX4/CSNK2A1. Silencing SOX4 significantly reduced cell proliferation, migration, invasion, and tumor growth in vivo by lowering CSNK2A1-TOP2A phosphorylation. These findings highlight the therapeutic potential of targeting the SOX4/CSNK2A1 axis in breast cancer and provide insight into its mechanism through TOP2A phosphorylation.
Collapse
Affiliation(s)
- Jiaqiong Zou
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu, China
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ruiman Geng
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhengkun Zhang
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Xuxu Ji
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhaoru Yin
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Dingxue Wang
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Rong Guo
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Lihong Chen
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Ji Liu
- Department of Biochemistry & Molecular Biology, West China School of Basic Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Wu Y, Li N, Shang J, Jiang J, Liu X. Identification of cancer-associated fibroblast subtypes and prognostic model development in breast cancer: role of the RUNX1/SDC1 axis in promoting invasion and metastasis. Cell Biol Toxicol 2025; 41:21. [PMID: 39753834 PMCID: PMC11698906 DOI: 10.1007/s10565-024-09950-w] [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: 07/07/2024] [Accepted: 11/20/2024] [Indexed: 01/06/2025]
Abstract
In this study, we identified cancer-associated fibroblast (CAF) molecular subtypes and developed a CAF-based prognostic model for breast cancer (BRCA). The heterogeneity of cancer-associated fibroblasts (CAFs) and their significant involvement in the advancement of BRCA were discovered employing single-cell RNA sequencing. Notably, we discovered that the RUNX1/SDC1 axis enhances BRCA cell invasion and metastasis. RUNX1 transcriptionally upregulates SDC1, which facilitates extracellular matrix remodeling and promotes tumor cell migration. This finding highlights the vital contribution of CAFs to the tumor microenvironment and provides new potential targets for therapeutic intervention. The predictive model showcased remarkable precision in anticipating patient outcomes and could guide personalized treatment strategies.
Collapse
Affiliation(s)
- Yunhao Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Pancreatic and Thyroid Ward, Shenyang, 110004, P. R. China
| | - Nu Li
- Department of Breast surgery, The First Hospital of China Medical University, Shenyang, 110004, P.R. China
| | - Jin Shang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, P. R. China
| | - Jiazi Jiang
- Department of Emergency, The First Hospital of China Medical University, No.155 Nanjing Road, Heping District, Shenyang, 110001, Liaoning Province, P. R. China.
| | - Xiaoliang Liu
- Department of Emergency, The First Hospital of China Medical University, No.155 Nanjing Road, Heping District, Shenyang, 110001, Liaoning Province, P. R. China.
| |
Collapse
|
4
|
Qi H, Ma X, Ma Y, Jia L, Liu K, Wang H. Mechanisms of HIF1A-mediated immune evasion in gastric cancer and the impact on therapy resistance. Cell Biol Toxicol 2024; 40:87. [PMID: 39384651 PMCID: PMC11464584 DOI: 10.1007/s10565-024-09917-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 09/04/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND The high prevalence and detrimental effects on patient outcomes make gastric cancer (GC) a significant health issue that persists internationally. Existing treatment modalities exhibit limited efficacy, prompting the exploration of immune checkpoint inhibitors as a novel therapeutic approach. However, resistance to immunotherapy poses a significant challenge in GC management, necessitating a profound grasp of the intrinsic molecular pathways. METHODS This study focuses on investigating the immunosuppressive mechanisms of quiescent cancer cells (QCCs) in GC, particularly their resistance to T-cell-mediated immune responses. Utilizing mouse models, gene editing techniques, and transcriptome sequencing, we aim to elucidate the interactions between QCCs, immune cells, and key regulatory factors like HIF1A. Functional enrichment analysis will further underscore the role of glycolysis-related genes in mediating immunosuppression by QCCs. RESULTS The cancer cells that survived GC treated with T-cell therapy lost their proliferative ability. QCCs, as the main resistance force to immunotherapy, exhibit stronger resistance to CD8+ T-cell attack and possess higher cancer-initiating potential. Single-cell sequencing analysis revealed that the microenvironment in the QCCs region harbors more M2-type tumor-associated macrophages and fewer T cells. This microenvironment in the QCCs region leads to the downregulation of T-cell immune activation and alters macrophage metabolic function. Transcriptome sequencing of QCCs identified upregulated genes related to chemo-resistance, hypoxia, and glycolysis. In vitro cell experiments illustrated that HIF1A promotes the transcription of glycolysis-related genes, and silencing HIF1A in QCCs enhances T-cell proliferation and activation in co-culture systems, induces apoptosis in QCCs, and increases QCCs' sensitivity to immune checkpoint inhibitors. In vivo, animal experiments showed that silencing HIF1A in QCCs can inhibit GC growth and metastasis. CONCLUSION Unraveling the molecular mechanisms by which QCCs resist T-cell-mediated immune responses through immunosuppression holds promising implications for refining treatment strategies and enhancing patient outcomes in GC. By delineating these intricate interactions, this study contributes crucial insights into precision medicine and improved therapeutic outcomes in GC management.
Collapse
Affiliation(s)
- Hao Qi
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Xiaoyu Ma
- Departments of Gastrointestinal Endoscopy, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yu Ma
- Department of Nuclear Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Liuyu Jia
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Kuncong Liu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Honghu Wang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
5
|
Jiang P, Zhu X, Jiang Y, Li H, Luo Q. Targeting JUNB to modulate M2 macrophage polarization in preeclampsia. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167194. [PMID: 38663490 DOI: 10.1016/j.bbadis.2024.167194] [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: 11/11/2023] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 06/17/2024]
Abstract
Preeclampsia (PE) is a complex disorder affecting pregnant women, leading to significant maternal and fetal morbidity and mortality. Understanding the cellular dynamics and molecular mechanisms underlying PE is crucial for developing effective therapeutic strategies. This study utilized single-cell RNA sequencing (scRNA-seq) to delineate the cellular landscape of the placenta in PE, identifying 11 distinct cell subpopulations, with macrophages playing a pivotal role in mediating cell-cell communication. Specifically, the transcription factor JUNB was found to be a key gene in macrophages from PE samples, influencing the interaction between macrophages and both epithelial and endothelial cells. Functional experiments indicated that interference with JUNB expression promoted macrophage polarization towards an M2 phenotype, which facilitated trophoblast invasion, migration, and angiogenesis. Mechanistically, JUNB regulated the MIIP/PI3K/AKT pathway, as evidenced by gene expression analysis following JUNB knockdown. The study further demonstrated that targeting JUNB could activate the PI3K/AKT pathway by transcriptionally activating MIIP, thus promoting M2 polarization and potentially delaying the onset of PE. These findings present new insights into the pathogenesis of PE and suggest a novel therapeutic approach by modulating macrophage polarization.
Collapse
Affiliation(s)
- Peiyue Jiang
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, PR China
| | - Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, PR China
| | - Ying Jiang
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, PR China
| | - Hetong Li
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, PR China
| | - Qiong Luo
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, PR China.
| |
Collapse
|
6
|
Song C, Wang G, Liu M, Han S, Dong M, Peng M, Wang W, Wang Y, Xu Y, Liu L. Deciphering the SOX4/MAPK1 regulatory axis: a phosphoproteomic insight into IQGAP1 phosphorylation and pancreatic Cancer progression. J Transl Med 2024; 22:602. [PMID: 38943117 PMCID: PMC11212360 DOI: 10.1186/s12967-024-05377-3] [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: 01/14/2024] [Accepted: 06/06/2024] [Indexed: 07/01/2024] Open
Abstract
OBJECTIVE This study aims to elucidate the functional role of IQGAP1 phosphorylation modification mediated by the SOX4/MAPK1 regulatory axis in developing pancreatic cancer through phosphoproteomics analysis. METHODS Proteomics and phosphoproteomics data of pancreatic cancer were obtained from the Clinical Proteomic Tumor Analysis Consortium (CPTAC) database. Differential analysis, kinase-substrate enrichment analysis (KSEA), and independent prognosis analysis were performed on these datasets. Subtype analysis of pancreatic cancer patients was conducted based on the expression of prognostic-related proteins, and the prognosis of different subtypes was evaluated through prognosis analysis. Differential analysis of proteins in different subtypes was performed to identify differential proteins in the high-risk subtype. Clinical correlation analysis was conducted based on the expression of prognostic-related proteins, pancreatic cancer typing results, and clinical characteristics in the pancreatic cancer proteomics dataset. Functional pathway enrichment analysis was performed using GSEA/GO/KEGG, and most module proteins correlated with pancreatic cancer were selected using WGCNA analysis. In cell experiments, pancreatic cancer cells were grouped, and the expression levels of SOX4, MAPK1, and the phosphorylation level of IQGAP1 were detected by RT-qPCR and Western blot experiments. The effect of SOX4 on MAPK1 promoter transcriptional activity was assessed using a dual-luciferase assay, and the enrichment of SOX4 on the MAPK1 promoter was examined using a ChIP assay. The proliferation, migration, and invasion functions of grouped pancreatic cancer cells were assessed using CCK-8, colony formation, and Transwell assays. In animal experiments, the impact of SOX4 on tumor growth and metastasis through the regulation of MAPK1-IQGAP1 phosphorylation modification was studied by constructing subcutaneous and orthotopic pancreatic cancer xenograft models, as well as a liver metastasis model in nude mice. RESULTS Phosphoproteomics and proteomics data analysis revealed that the kinase MAPK1 may play an important role in pancreatic cancer progression by promoting IQGAP1 phosphorylation modification. Proteomics analysis classified pancreatic cancer patients into two subtypes, C1 and C2, where the high-risk C2 subtype was associated with poor prognosis, malignant tumor typing, and enriched tumor-related pathways. SOX4 may promote the occurrence of the high-risk C2 subtype of pancreatic cancer by regulating MAPK1-IQGAP1 phosphorylation modification. In vitro cell experiments confirmed that SOX4 promoted IQGAP1 phosphorylation modification by activating MAPK1 transcription while silencing SOX4 inhibited the proliferation, migration, and invasion of pancreatic cancer cells by reducing the phosphorylation level of MAPK1-IQGAP1. In vivo, animal experiments further confirmed that silencing SOX4 suppressed the growth and metastasis of pancreatic cancer by reducing the phosphorylation level of MAPK1-IQGAP1. CONCLUSION The findings of this study suggest that SOX4 promotes the phosphorylation modification of IQGAP1 by activating MAPK1 transcription, thereby facilitating the growth and metastasis of pancreatic cancer.
Collapse
Affiliation(s)
- Chao Song
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China
- Department of General Surgery, Qingpu Branch, Affiliated Zhongshan Hospital of Fudan University, Qingpu Branch, No. 1158 Park Road East, Qingpu District, Shanghai, PR China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, PR China
| | - Ganggang Wang
- Department of Hepatobiliary Surgery, Pudong Hospital, Fudan University, Shanghai, China
| | - Mengmeng Liu
- Department of Gastroenterology, Qingpu Branch, Affiliated Zhongshan Hospital of Fudan University, Shanghai, PR China
| | - Siyang Han
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China
| | - Meiyuan Dong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, PR China
| | - Maozhen Peng
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China
| | - Yicun Wang
- Department of General Surgery, Qingpu Branch, Affiliated Zhongshan Hospital of Fudan University, Qingpu Branch, No. 1158 Park Road East, Qingpu District, Shanghai, PR China.
| | - Yaolin Xu
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China.
| | - Liang Liu
- Department of Pancreatic Surgery, Affiliated Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai, PR China.
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, PR China.
| |
Collapse
|
7
|
Chen X, Li M, Li H, Liu M, Su J, Ji Y. Implications of IFNγ SNP rs2069705 in primary Sjögren's syndrome: transcriptional activation and B cell infiltration. Am J Physiol Cell Physiol 2024; 326:C1494-C1504. [PMID: 38406824 PMCID: PMC11371360 DOI: 10.1152/ajpcell.00661.2023] [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: 12/04/2023] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
Primary Sjögren's syndrome (pSS) is characterized by its autoimmune nature. This study investigates the role of the IFNγ SNP rs2069705 in modulating the susceptibility to pSS. Differential expression of IFNγ and BAFF was analyzed using the GEO database's mRNA microarray GSE84844. Genotyping of the IFNγ SNP rs2069705 was conducted via the dbSNP website. The JASPAR tool was used for predicting transcription factor bindings. Techniques such as dual-luciferase reporter assays, Chromatin immunoprecipitation, and analysis of a pSS mouse model were applied to study gene and protein interactions. A notable increase in the mutation frequency of IFNγ SNP rs2069705 was observed in MNCs from the exocrine glands of pSS mouse models. Bioinformatics analysis revealed elevated levels of IFNγ and BAFF in pSS samples. The model exhibited an increase in both CD20+ B cells and cells expressing IFNγ and BAFF. Knocking down IFNγ resulted in lowered BAFF expression and less lymphocyte infiltration, with BAFF overexpression reversing this suppression. Activation of the Janus kinase (JAK)/STAT1 pathway was found to enhance transcription in the BAFF promoter region, highlighting IFNγ's involvement in pSS. In addition, rs2069705 was shown to boost IFNγ transcription by promoting interaction between its promoter and STAT4. SNP rs2069705 in the IFNγ gene emerges as a pivotal element in pSS susceptibility, primarily by augmenting IFNγ transcription, activating the JAK/STAT1 pathway, and leading to B-lymphocyte infiltration in the exocrine glands.NEW & NOTEWORTHY The research employed a combination of bioinformatics analysis, genotyping, and experimental models, providing a multifaceted approach to understanding the complex interactions in pSS. We have uncovered that the rs2069705 SNP significantly affects the transcription of IFNγ, leading to altered immune responses and B-lymphocyte activity in pSS.
Collapse
Affiliation(s)
- Xi Chen
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang, China
| | - Min Li
- Department of Immunology, Mianyang Central Hospital, Mianyang, China
| | - Honglin Li
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang, China
| | - Miao Liu
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang, China
| | - Jianrong Su
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang, China
| | - Yuzhu Ji
- Department of Pathology, Mianyang Central Hospital, Mianyang, China
| |
Collapse
|
8
|
Zhang D, Chen K, Shan LS. Meta-analysis and transcriptomic analysis reveal that NKRF and ZBTB17 regulate the NF-κB signaling pathway, contributing to the shared molecular mechanisms of Alzheimer's disease and atherosclerosis. CNS Neurosci Ther 2024; 30:e14683. [PMID: 38738952 PMCID: PMC11090078 DOI: 10.1111/cns.14683] [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: 09/29/2023] [Revised: 12/26/2023] [Accepted: 01/15/2024] [Indexed: 05/14/2024] Open
Abstract
INTRODUCTION Alzheimer's disease (AD) and atherosclerosis (AS) are widespread diseases predominantly observed in the elderly population. Despite their prevalence, the underlying molecular interconnections between these two conditions are not well understood. METHODS Utilizing meta-analysis, bioinformatics methodologies, and the GEO database, we systematically analyzed transcriptome data to pinpoint key genes concurrently differentially expressed in AD and AS. Our experimental validations in mouse models highlighted the prominence of two genes, NKRF (NF-κB-repressing factor) and ZBTB17 (MYC-interacting zinc-finger protein 1). RESULTS These genes appear to influence the progression of both AD and AS by modulating the NF-κB signaling pathway, as confirmed through subsequent in vitro and in vivo studies. CONCLUSIONS This research uncovers a novel shared molecular pathway between AD and AS, underscoring the significant roles of NKRF and ZBTB17 in the pathogenesis of these disorders.
Collapse
Affiliation(s)
- Di Zhang
- Department of CardiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Keyan Chen
- Laboratory Animal Science of China Medical UniversityShenyangLiaoningChina
| | - Li Shen Shan
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
9
|
Wang G, Xu YL, Zhang XH, Tang L, Li Y. LncRNA HOTTIP regulates TLR4 promoter methylation by recruiting H3K4 methyltransferase MLL1 to affect apoptosis and inflammatory response of fibroblast-like synoviocyte in rheumatoid arthritis. Kaohsiung J Med Sci 2024; 40:335-347. [PMID: 38363110 DOI: 10.1002/kjm2.12805] [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: 08/29/2023] [Revised: 12/05/2023] [Accepted: 12/24/2023] [Indexed: 02/17/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease, and the role of HOXA transcript at the distal tip (HOTTIP) in its pathogenesis remains underexplored. This study investigates the mechanism by which HOTTIP influences apoptosis and the inflammatory response of fibroblast-like synoviocytes (FLS). An RA mouse model was established, and clinical scores were analyzed. Pathological changes in synovial tissues, bone mineral density (BMD) of the paws, serum tartrate-resistant acid phosphatase (TRAP) activity, and TNF-α and IL-1β levels were assessed. FLS were transfected, and cell proliferation and apoptosis were examined. The RNA-pull-down assay determined HOTTIP's interaction with mixed-lineage leukemia 1 (MLL1), while RNA immunoprecipitation assay measured HOTTIP expression pulled down by MLL1. The levels of MLL1 and toll-like receptor 4 (TLR4) after MLL1 overexpression based on HOTTIP silencing were determined. Chromatin immunoprecipitation (ChIP) was performed with H3K4me3 as an antibody, followed by the evaluation of TLR4 expression. HOTTIP expression was elevated in RA mouse synovial tissues. Inhibition of HOTTIP led to reduced clinical scores, inflammatory infiltration, synovial hyperplasia, TRAP activity, and TNF-α and IL-1β levels, along with increased BMD. In vitro Interference with HOTTIP suppressed RA-FLS apoptosis and inflammation. HOTTIP upregulated TLR4 expression by recruiting MLL1 to facilitate TLR4 promoter methylation. MLL1 overexpression reversed HOTTIP silencing-mediated repression of RA-FLS apoptosis. Activation of H3K4 methylation counteracted HOTTIP knockout, ameliorating the inflammatory response. HOTTIP regulates TLR4 expression by recruiting MLL1, leading to TLR4 promoter methylation, thereby suppressing RA-FLS proliferation and inducing cell apoptosis and inflammatory response in RA.
Collapse
Affiliation(s)
- Guan Wang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Sichuan Provincial Laboratory of Orthopaedic Engineering Luzhou, Sichuan, China
| | - Yu-Lin Xu
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Sichuan Provincial Laboratory of Orthopaedic Engineering Luzhou, Sichuan, China
| | - Xi-Hai Zhang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Sichuan Provincial Laboratory of Orthopaedic Engineering Luzhou, Sichuan, China
| | - Lian Tang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Sichuan Provincial Laboratory of Orthopaedic Engineering Luzhou, Sichuan, China
| | - Yao Li
- Laboratory Animal Center, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
10
|
Fang S, He T, You M, Zhu H, Chen P. Glucocorticoids promote steroid-induced osteonecrosis of the femoral head by down-regulating serum alpha-2-macroglobulin to induce oxidative stress and facilitate SIRT2-mediated BMP2 deacetylation. Free Radic Biol Med 2024; 213:208-221. [PMID: 38142952 DOI: 10.1016/j.freeradbiomed.2023.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/07/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
Our study investigated the possible molecular mechanism of glucocorticoid in steroid-induced osteonecrosis of the femoral head (SINFH) through regulating serum alpha-2-macroglobulin and SIRT2-mediated BMP2 deacetylation. Essential genes involved in glucocorticoid-induced SINFH were screened by transcriptome sequencing and analyzed by bioinformatics, followed by identifying downstream regulatory targets. Rat bone marrow mesenchymal stem cells were isolated and treated with methylprednisolone (MP) for in vitro cell experiments. Besides, a glucocorticoid-induced rat ONFH was established using the treatment of MP and LPS. ChIP-PCR detected the enrichment of SIRT2 in the promoter region of BMP2, and the deacetylation modification of SIRT2 on BMP2 was determined. Bioinformatics analysis revealed that glucocorticoids may induce ONFH through the SIRT2/BMP2 axis. In vitro cell experiments showed that glucocorticoids up-regulated SIRT2 expression in BMSCs by inducing oxidative stress, thereby promoting cell apoptosis. The up-regulation of SIRT2 expression may be due to the decreased ability of α2 macroglobulin to inhibit oxidative stress, and the addition of NOX protein inhibitor DPI could significantly inhibit SIRT2 expression. SIRT2 could promote histone deacetylation of the BMP2 promoter and inhibit its expression. In vitro cell experiments further indicated that knocking down SIRT2 could protect BMSC from oxidative stress and cell apoptosis induced by glucocorticoids by promoting BMP2 expression. In addition, animal experiments conducted also demonstrated that the knockdown of SIRT2 could improve glucocorticoid-induced ONFH through up-regulating BMP2 expression. Glucocorticoids could induce oxidative stress by down-regulating serum α2M to promote SIRT2-mediated BMP2 deacetylation, leading to ONFH.
Collapse
Affiliation(s)
- Shanhong Fang
- Department of Sports Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, PR China; Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China; Fujian Orthopaedics Research Institute, Fuzhou, 350000, PR China; Fujian Orthopedic Bone and Joint Disease and Sports Rehabilitation Clinical Medical Research Center, Fuzhou, 350000, PR China
| | - Tianmin He
- Department of Vascular Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China
| | - Mengqiang You
- Department of Sports Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, PR China; Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China
| | - Huixin Zhu
- Nursing Department, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China; Nursing Department, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China
| | - Peng Chen
- Department of Sports Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, PR China; Department of Orthopedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, PR China; Fujian Orthopaedics Research Institute, Fuzhou, 350000, PR China; Fujian Orthopedic Bone and Joint Disease and Sports Rehabilitation Clinical Medical Research Center, Fuzhou, 350000, PR China.
| |
Collapse
|
11
|
Li S, Zhang N, Yang Y, Liu T. Transcriptionally activates CCL28 expression to inhibit M2 polarization of macrophages and prevent immune escape in colorectal cancer cells. Transl Oncol 2024; 40:101842. [PMID: 38035446 PMCID: PMC10698578 DOI: 10.1016/j.tranon.2023.101842] [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: 07/10/2023] [Revised: 11/10/2023] [Accepted: 11/18/2023] [Indexed: 12/02/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the potential molecular mechanism of SPDEF in immune evasion of colorectal cancer (CRC) and examine its impact on macrophage M2 polarization using the TCGA and GEO databases. METHODS By combining TCGA and GEO databases, differential gene expression between CRC samples and standard tissue samples was analyzed to screen for immune-related genes (IRGs) associated with the prognosis of CRC patients. A predictive risk model was constructed based on 18 key IRGs, which were then validated using the GEO dataset. The relationship between transcription factors and IRGs was further explored to investigate their regulatory network in CRC. In vivo and in vitro experiments were carried out to validate these regulatory relationships and explore the function of SPDEF and CCL28 in CRC. RESULTS Twelve key IRGs associated with clinical and pathological characteristics of CRC patients were identified. Among them, CCL28 significantly impacted macrophage infiltration in CRC cells and may be a critical factor in immune evasion. In both in vitro and in vivo experiments, overexpression of SPDEF upregulated CCL28 expression, thereby suppressing M2 polarization of macrophages and inhibiting CRC cell proliferation and tumor growth. Notably, interference with CCL28 could reverse the effect of SPDEF overexpression. CONCLUSION SPDEF can suppress immune evasion of CRC cells by activating CCL28, which is achieved through the modulation of M2 polarization of macrophages. This provides a new research direction and potential therapeutic target for immunotherapy in CRC.
Collapse
Affiliation(s)
- Shiquan Li
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun 130000, China
| | - Nan Zhang
- Department of Burn Surgery, The First Hospital of Jilin University, Changchun 130000, China
| | - Yongping Yang
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun 130000, China
| | - Tongjun Liu
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
12
|
Zhang YJ, Yi DH. CDK1-SRC Interaction-Dependent Transcriptional Activation of HSP90AB1 Promotes Antitumor Immunity in Hepatocellular Carcinoma. J Proteome Res 2023; 22:3714-3729. [PMID: 37949475 DOI: 10.1021/acs.jproteome.3c00379] [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] [Indexed: 11/12/2023]
Abstract
This study aimed to analyze multiomics data and construct a regulatory network involving kinases, transcription factors, and immune genes in hepatocellular carcinoma (HCC) prognosis. The researchers used transcriptomic, proteomic, and clinical data from TCGA and GEO databases to identify immune genes associated with HCC. Statistical analysis, meta-analysis, and protein-protein interaction analyses were performed to identify key immune genes and their relationships. In vitro and in vivo experiments validated the CDK1-SRC-HSP90AB1 network's effects on HCC progression and antitumor immunity. A prognostic risk model was developed using clinicopathological features and immune infiltration. The immune genes LPA, BIRC5, HSP90AB1, ROBO1, and CCL20 were identified as the key prognostic factors. The CDK1-SRC-HSP90AB1 network promoted HCC cell proliferation and migration, with HSP90AB1 being transcriptionally activated by the CDK1-SRC interaction. Manipulating SRC or HSP90AB1 reversed the effects of CDK1 and SRC on HCC. The CDK1-SRC-HSP90AB1 network also influenced HCC tumor formation and antitumor immunity. Overall, this study highlights the importance of the CDK1-SRC-HSP90AB1 network as a crucial immune-regulatory network in the HCC prognosis.
Collapse
Affiliation(s)
- Yi-Jie Zhang
- Department of Hepatobiliary and Organ Transplantation, The First Affiliated Hospital of China Medical University, Shenyang 110001, P. R. China
- The Key Laboratory of Organ Transplantation of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang 110001, P. R. China
| | - De-Hui Yi
- Department of Hepatobiliary and Organ Transplantation, The First Affiliated Hospital of China Medical University, Shenyang 110001, P. R. China
- The Key Laboratory of Organ Transplantation of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang 110001, P. R. China
| |
Collapse
|
13
|
Chen X, Li W, Chang C. NR3C2 mediates oxidised low-density lipoprotein-induced human coronary endothelial cells dysfunction via modulation of NLRP3 inflammasome activation. Autoimmunity 2023; 56:2189135. [PMID: 36919662 DOI: 10.1080/08916934.2023.2189135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Nuclear receptor subfamily 3 group C member 2 (NR3C2) has been revealed to affect the progression of multiple inflammatory diseases, while NR3C2's efficacy in coronary artery disease (CAD) remains largely unsolved. The study intended to elucidate the possible mechanisms of NR3C2 in oxidised low density lipoprotein (ox-LDL)-induced inflammation in human coronary endothelial cells (HCAECs) via regulating NACHT, LRR, and PYD domains-containing protein 3 (NLRP3). Patients who underwent CT angiography or coronary angiography for suspected CAD in our hospital were collected. The patients were divided into the CAD and the non-CAD (NCAD) groups. The expression of NR3C2 and NLRP3 in the peripheral blood of patients in both groups was examined by RT-qPCR. HCAECs were treated with ox-LDL to establish the model. The expression of NR3C2 and NLRP3 in ox-LDL-induced HCAECs was tested by RT-qPCR. The proliferation of HCAECs was measured using CCK-8 assay, the apoptosis of HCAECs was assessed by flow cytometry, and the levels of inflammation-related factors IL-1β and IL-18 in the cell supernatant were evaluated by ELISA. The molecular mechanisms of these factors in the proliferation and apoptosis of HCAECs and in the inflammatory response were further determined by knockdown and overexpression systems. The relationship between NR3C2 and NLRP3 was determined by ChIP and luciferase activity assays and bioinformatics analysis. NR3C2 and NLRP3 levels were elevated in the serum of CAD patients. The ox-LDL treatment elevated NR3C2 levels, evoked apoptosis and inflammation, and impeded cell viability in HCAECs, whereas downregulation of NR3C2 increased cell viability and reduced apoptosis and inflammatory response in ox-LDL-induced inflammation in HCAECs. NR3C2 levels were positively correlated with NLRP3, and NR3C2 elevated NLRP3 expression through transcription. Overexpression of NLRP3 counteracted the impacts of silencing NR3C2 on cell viability, cell apoptosis, and inflammatory response in ox-LDL-induced HCAECs. Our research stresses that NR3C2 transcription promotes NLRP3 to induce inflammatory responses in ox-LDL-induced HCAECs.
Collapse
Affiliation(s)
- Xiaofan Chen
- Department of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weidong Li
- Department of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chengdong Chang
- Department of Pathology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Okuma N, Ito MA, Shimizu T, Hasegawa A, Ohmori S, Yoshida K, Matsuoka I. Amplification of poly(I:C)-induced interleukin-6 production in human bronchial epithelial cells by priming with interferon-γ. Sci Rep 2023; 13:21067. [PMID: 38030681 PMCID: PMC10687102 DOI: 10.1038/s41598-023-48422-9] [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: 08/04/2023] [Accepted: 11/27/2023] [Indexed: 12/01/2023] Open
Abstract
Proinflammatory cytokine interleukin (IL)-6 was associated with disease severity in patients with COVID-19. The mechanism underlying the excessive IL-6 production by SARS-Cov-2 infection remains unclear. Respiratory viruses initially infect nasal or bronchial epithelial cells that produce various inflammatory mediators. Here, we show that pretreatment of human bronchial epithelial cells (NCl-H292) with interferon (IFN)-γ (10 ng/mL) markedly increased IL-6 production induced by the toll-like receptor (TLR) 3 agonist poly(I:C) (1 µg/mL) from 0.4 ± 0.1 to 4.1 ± 0.4 ng/mL (n = 3, P < 0.01). A similar effect was observed in human alveolar A549 and primary bronchial epithelial cells. TLR3 knockdown using siRNA in NCl-H292 cells diminished the priming effects of IFN-γ on poly(I:C)-induced IL-6 production. Furthermore, the Janus kinase (JAK) inhibitor tofacitinib (1 µM) inhibited IFN-γ-induced upregulation of TLR3, and suppressed poly(I:C)-induced IL-6 production. Quantitative chromatin immunoprecipitation revealed that IFN-γ stimulated histone modifications at the IL-6 gene locus. Finally, IFN-γ priming significantly increased lung IL-6 mRNA and protein levels in poly(I:C)-administrated mice. Thus, priming bronchial epithelial cells with IFN-γ increases poly(I:C)-induced IL-6 production via JAK-dependent TLR3 upregulation and chromatin remodeling at the IL-6 gene locus. These mechanisms may be involved in severe respiratory inflammation following infection with RNA viruses.
Collapse
Affiliation(s)
- Norikazu Okuma
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, 370-0033, Japan
- Department of Pharmacy, Japan Community Health Care Organization Gunma Chuo Hospital, Maebashi-shi, Gunma, 371-0025, Japan
| | - Masa-Aki Ito
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, 370-0033, Japan.
| | - Tomoyoshi Shimizu
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, 370-0033, Japan
| | - Atsuya Hasegawa
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, 370-0033, Japan
| | - Shin'ya Ohmori
- Laboratory of Allergy, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, 370-0033, Japan
| | - Kazuki Yoshida
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, 370-0033, Japan
| | - Isao Matsuoka
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, 370-0033, Japan
| |
Collapse
|
15
|
Dong L, Gao L. JMJD3 and SNAI2 synergistically protect against Parkinson's disease by mediating the YAP/HIF1α signaling pathway in a mouse model. Hum Mol Genet 2023; 32:3040-3052. [PMID: 37453035 DOI: 10.1093/hmg/ddad115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
This study aimed to characterize the functional relevance and mechanistic basis of the histone demethylase Jumonji domain-containing protein-3 (JMJD3) in preserving dopaminergic neuron survival in Parkinson's disease (PD). Mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced lesions and MN9D dopaminergic neuronal cell lines exposed to 6-OHDA, respectively, were used to simulate in vivo and in vitro PD-like environments. PD-related genes with differential expressions were identified using RNA sequencing of hippocampal tissues collected from MPTP-lesioned mice. A specific lentiviral shRNA vector was used to investigate the effects of JMJD3 on neuron activities in vitro and PD-like phenotypes in vivo. JMJD3 was found to up-regulate the expression of Snail family transcriptional repressor 2 (SNAI2) through the inhibition of H3 on lysine 27 (H3K27me3) enrichment in the SNAI2 promoter region. As a result, the viability of 6-OHDA-exposed MN9D cells was stimulated, and cell apoptosis was diminished. Knockdown of SNAI2 decreased the expression of yes-associated protein (YAP) and HIF1α while also reducing the viability of 6-OHDA-exposed MN9D cells and increasing cell apoptosis. The in vivo experiments demonstrated that JMJD3 activated the SNAI2/YAP/HIF1α signaling pathway, inhibiting PD-like phenotypes in MPTP-lesioned mice. Thus, the findings provide evidence that JMJD3 inhibits the enrichment of H3K27me3 at the SNAI2 promoter, leading to the upregulation of SNAI2 expression and activation of the YAP/HIF1α signaling pathway, ultimately exerting a protective effect on PD mice. This finding suggests that targeting the JMJD3-SNAI2 pathway could be a promising therapeutic strategy for PD. Further in-depth studies are needed to elucidate the underlying mechanisms and identify potential downstream targets of this pathway.
Collapse
Affiliation(s)
- Li Dong
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Lianbo Gao
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| |
Collapse
|
16
|
Ding L, Hao K, Sang L, Shen X, Zhang C, Fu D, Qi X. ATF2-driven osteogenic activity of enoxaparin sodium-loaded polymethylmethacrylate bone cement in femoral defect regeneration. J Orthop Surg Res 2023; 18:646. [PMID: 37653390 PMCID: PMC10470168 DOI: 10.1186/s13018-023-04017-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/14/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Polymethylmethacrylate (PMMA) bone cement loaded with enoxaparin sodium (PMMA@ES) has been increasingly highlighted to affect the bone repair of bone defects, but the molecular mechanisms remain unclear. We addressed this issue by identifying possible molecular mechanisms of PMMA@ES involved in femoral defect regeneration based on bioinformatics analysis and network pharmacology analysis. METHODS The upregulated genes affecting the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) were selected through bioinformatics analysis, followed by intersection with the genes of ES-induced differentiation of BMSCs identified by network pharmacology analysis. PMMA@ES was constructed. Rat primary BMSCs were isolated and cultured in vitro in the proliferation medium (PM) and osteogenic medium (OM) to measure alkaline phosphatase (ALP) activity, mineralization of the extracellular matrix, and the expression of RUNX2 and OCN using gain- or loss-of-function experiments. A rat femoral bone defect model was constructed to detect the new bone formation in rats. RESULTS ATF2 may be a key gene in differentiating BMSCs into osteoblasts. In vitro cell assays showed that PMMA@ES promoted the osteogenic differentiation of BMSCs by increasing ALP activity, extracellular matrix mineralization, and RUNX2 and OCN expression in PM and OM. In addition, ATF2 activated the transcription of miR-335-5p to target ERK1/2 and downregulate the expression of ERK1/2. PMMA@ES induced femoral defect regeneration and the repair of femoral defects in rats by regulating the ATF2/miR-335-5p/ERK1/2 axis. CONCLUSION The evidence provided by our study highlighted the ATF2-mediated mechanism of PMMA@ES in the facilitation of the osteogenic differentiation of BMSCs and femoral defect regeneration.
Collapse
Affiliation(s)
- Luobin Ding
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, Shijiazhuang, 050000, People's Republic of China
| | - Kangning Hao
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Linchao Sang
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, Shijiazhuang, 050000, People's Republic of China
| | - Xiaoyu Shen
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Ce Zhang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Dehao Fu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, People's Republic of China.
| | - Xiangbei Qi
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China.
| |
Collapse
|
17
|
Wu F, Zhang P, Zhou G. The involvement of EGR1 in neuron apoptosis in the in vitro model of spinal cord injury via BTG2 up-regulation. Neurol Res 2023; 45:646-654. [PMID: 36759943 DOI: 10.1080/01616412.2023.2176633] [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: 02/11/2023]
Abstract
OBJECTIVE EGR1 has been implicated in the progression of spinal cord injury (SCI). Nevertheless, its specific mechanism in SCI remains to be investigated. Hence, this study explored the potential mechanism of EGR1 in SCI by focusing on neuron apoptosis. METHODS H2O2 was utilized to treat rat neurons-dorsal spinal cord (RN-dsc) for the construction of an in vitro model of SCI. Afterwards, cell survival, apoptosis, and LDH leakage were detected to evaluate the injury degree of H2O2-treated RN-dsc. The expression of apoptosis-related proteins was also measured. Additionally, EGR1 was silenced and/or BTG2 was overexpressed in RN-dsc before H2O2 treatment to assess the impacts of EGR1 and BTG2 on H2O2-induced RN-dsc. Jasper online website was utilized to predict binding sites of EGR1 on BTG2, and dual-luciferase reporter gene and chromatin immunoprecipitation (ChIP) assays were utilized to verify the binding between EGR1 and BTG2. RESULTS H2O2 treatment suppressed survival and promoted apoptosis in RN-dsc, accompanied by upregulated LDH, Bax, and cleaved-caspase-3 and down-regulated Bcl-2. Moreover, EGR1 and BTG2 were up-regulated in H2O2-induced RN-dsc. Mechanistically, EGR1 was bound to the promoter of BTG2 to transcriptionally activate BTG2. EGR1 knockdown diminished apoptosis and LDH, Bax, and cleaved-caspase-3 levels while elevating survival and Bcl-2 levels in H2O2-induced RN-dsc. These effects of EGR1 knockdown were abrogated by further BTG2 overexpression. DISCUSSION Conclusively, EGR1 promotes H2O2-induced apoptosis in RN-dsc by activating BTG2 transcription.
Collapse
Affiliation(s)
- Fangqian Wu
- Department of Orthopedics (Spine Surgery), Jiangxi Fuzhou First People's Hospital, Fuzhou, Jiangxi, P.R. China
| | - Ping Zhang
- Department of Neurology, Jiangxi Provincial People's Hospital (The First Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, P.R. China
| | - Guohui Zhou
- Department of Orthopedics (Spine Surgery), Jiangxi Fuzhou First People's Hospital, Fuzhou, Jiangxi, P.R. China
| |
Collapse
|
18
|
Wang Z, Tao T, Tao Y. ChIP and ChIRP Assays in Ferroptosis. Methods Mol Biol 2023; 2712:19-27. [PMID: 37578693 DOI: 10.1007/978-1-0716-3433-2_3] [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] [Indexed: 08/15/2023]
Abstract
Ferroptosis is characterized by the accumulation of lipid peroxidation driven by iron. As a regulated cell death, ferroptosis plays a critical role in various diseases and exhibits great therapeutic potentials. However, the mechanisms underlying ferroptosis, including its occurrence, execution, and regulation, remain poorly understood, which is necessary for developing effective therapeutic strategies. In this chapter, we summarize chromatin immunoprecipitation (ChIP) assay for the research of proteins-chromatin interactions. Moreover, Chromatin Isolation by RNA Purification (ChIRP) trial is introduced to investigate the interactions between lncRNA and chromatin. The application of ChIP and ChIRP is expected to explore the transcription and epigenetic regulation of ferroptosis deeply for therapeutic benefits.
Collapse
Affiliation(s)
- Zuli Wang
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou, China
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tania Tao
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Yongguang Tao
- NHC Key Laboratory of Carcinogenesis, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
19
|
Wu F, Li X, Looso M, Liu H, Ding D, Günther S, Kuenne C, Liu S, Weissmann N, Boettger T, Atzberger A, Kolahian S, Renz H, Offermanns S, Gärtner U, Potente M, Zhou Y, Yuan X, Braun T. Spurious transcription causing innate immune responses is prevented by 5-hydroxymethylcytosine. Nat Genet 2023; 55:100-111. [PMID: 36539616 PMCID: PMC9839451 DOI: 10.1038/s41588-022-01252-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
Generation of functional transcripts requires transcriptional initiation at regular start sites, avoiding production of aberrant and potentially hazardous aberrant RNAs. The mechanisms maintaining transcriptional fidelity and the impact of spurious transcripts on cellular physiology and organ function have not been fully elucidated. Here we show that TET3, which successively oxidizes 5-methylcytosine to 5-hydroxymethylcytosine (5hmC) and other derivatives, prevents aberrant intragenic entry of RNA polymerase II pSer5 into highly expressed genes of airway smooth muscle cells, assuring faithful transcriptional initiation at canonical start sites. Loss of TET3-dependent 5hmC production in SMCs results in accumulation of spurious transcripts, which stimulate the endosomal nucleic-acid-sensing TLR7/8 signaling pathway, thereby provoking massive inflammation and airway remodeling resembling human bronchial asthma. Furthermore, we found that 5hmC levels are substantially lower in human asthma airways compared with control samples. Suppression of spurious transcription might be important to prevent chronic inflammation in asthma.
Collapse
Affiliation(s)
- Fan Wu
- grid.418032.c0000 0004 0491 220XDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Xiang Li
- grid.418032.c0000 0004 0491 220XDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mario Looso
- grid.418032.c0000 0004 0491 220XDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hang Liu
- grid.418032.c0000 0004 0491 220XDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Dong Ding
- grid.418032.c0000 0004 0491 220XDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Günther
- grid.418032.c0000 0004 0491 220XDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Carsten Kuenne
- grid.418032.c0000 0004 0491 220XDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Shuya Liu
- grid.418032.c0000 0004 0491 220XDepartment of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany ,grid.13648.380000 0001 2180 3484Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Norbert Weissmann
- grid.440517.3Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany ,grid.8664.c0000 0001 2165 8627Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Thomas Boettger
- grid.418032.c0000 0004 0491 220XDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ann Atzberger
- grid.418032.c0000 0004 0491 220XDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Saeed Kolahian
- grid.10253.350000 0004 1936 9756Philipps University of Marburg - Medical Faculty, Center for Tumor- and Immunobiology (ZTI), Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Marburg, Germany
| | - Harald Renz
- grid.10253.350000 0004 1936 9756Philipps University of Marburg - Medical Faculty, Center for Tumor- and Immunobiology (ZTI), Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Marburg, Germany
| | - Stefan Offermanns
- grid.418032.c0000 0004 0491 220XDepartment of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ulrich Gärtner
- Institute for Anatomy und Cell Biology, Giessen, Germany
| | - Michael Potente
- grid.418032.c0000 0004 0491 220XAngiogenesis and Metabolism Laboratory, Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Yonggang Zhou
- grid.418032.c0000 0004 0491 220XDepartment of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Xuejun Yuan
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany. .,Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany.
| |
Collapse
|
20
|
Tan W, Zhang J, Liu L, Liang M, Li J, Deng Z, Zheng Z, Deng Y, Liu C, Li Y, Xie G, Zhang J, Zou F, Chen X. Hsp90 Inhibitor STA9090 induced VPS35 related extracellular vesicle release and metastasis in hepatocellular carcinoma. Transl Oncol 2022; 26:101502. [PMID: 36137350 PMCID: PMC9493061 DOI: 10.1016/j.tranon.2022.101502] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/28/2022] [Indexed: 11/05/2022] Open
Abstract
Heat shock protein 90 (Hsp90) has been an important therapeutic target for cancer therapy for decades. Unexpectedly, the monotherapy of N-terminal Hsp90 inhibitor STA9090 related clinical trials halted in phase III, and metastases were reported in animal models with the treatment of N-terminal Hsp90 inhibitors. Vacuolar protein sorting-associated protein 35 (VPS35) plays a vital role in endosome-derived EV (extracellular vesicle) traffic in neurodegeneration diseases, but no vps35 related EV were reported in tumors till now. Since tumor derived EVs contributes to metastasis and VPS35 is recently found to be involved in the invasion and metastasis of hepatocellular carcinoma (HCC), whether N-terminal Hsp90 inhibitor STA9090 induced EVs generation and the role of VPS35 in it were explored in this study. We found that N-terminal Hsp90 inhibitor STA9090 upregulated Bclaf1 and VPS35 levels, increased the secretion of EVs, and STA9090-induced-EVs promoted the invasion of HepG2 cells. As the clinical data suggested that the increased Bclaf1 and VPS35 levels correlated with increased metastasis and poorer prognosis in HCC, we focused on the Bclaf1-VPS35-EVs axis to further explore the mechanism of VPS35-related metastasis. The results demonstrated that Bclaf1 facilitated the transcription of VPS35 via bZIP domain, and knockdown of Bclaf1 or VPS35 alleviated pro-metastatic capability of STA9090-induced-EVs. All the results revealed the role of Bclaf1-VPS35-EVs axis on metastasis of HCC, and VPS35 knockdown decreased Hsp90 Inhibitor STA9090 induced extracellular vesicle release and metastasis, which provided a new combination therapeutic strategy to inhibit the metastasis of HCC caused by N-terminal Hsp90 inhibitor induced extracellular vesicles.
Collapse
Affiliation(s)
- Wenchong Tan
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jinxin Zhang
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lixia Liu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Manfeng Liang
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jieyou Li
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zihao Deng
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhenming Zheng
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yaotang Deng
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Chenyang Liu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yan Li
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guantai Xie
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiajie Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Fei Zou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China.
| | - Xuemei Chen
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China.
| |
Collapse
|
21
|
Ghosh S, Nelson JF, Cobb GMC, Etchells JP, de Lucas M. Light regulates xylem cell differentiation via PIF in Arabidopsis. Cell Rep 2022; 40:111075. [PMID: 35858547 PMCID: PMC9638722 DOI: 10.1016/j.celrep.2022.111075] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 01/15/2022] [Accepted: 06/18/2022] [Indexed: 11/23/2022] Open
Abstract
The balance between cell proliferation and differentiation in the cambium defines the formation of plant vascular tissues. As cambium cells proliferate, subsets of daughter cells differentiate into xylem or phloem. TDIF-PXY/TDR signaling is central to this process. TDIF, encoded by CLE41 and CLE44, activates PXY/TDR receptors to maintain proliferative cambium. Light and water are necessary for photosynthesis; thus, vascular differentiation must occur upon light perception to facilitate the transport of water and minerals to the photosynthetic tissues. However, the molecular mechanism controlling vascular differentiation in response to light remains elusive. In this study we show that the accumulation of PIF transcription factors in the dark promotes TDIF signaling and inhibits vascular cell differentiation. On the contrary, PIF inactivation by light leads to a decay in TDIF activity, which induces vascular cell differentiation. Our study connects light to vascular differentiation and highlights the importance of this crosstalk to fine-tune water transport. Active CLE peptide TDIF inhibits xylem differentiation in etiolated seedlings The expression of the TDIF precursor CLE44 is rapidly inhibited by light PIF transcription factors are necessary for TDIF expression in the dark Blue light signaling prevents TDIF expression, which promotes xylem differentiation
Collapse
Affiliation(s)
- Shraboni Ghosh
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Joseph F Nelson
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | | | - J Peter Etchells
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Miguel de Lucas
- Department of Biosciences, Durham University, Durham DH1 3LE, UK.
| |
Collapse
|
22
|
Zhang Q, Zhu Z, Guan J, Hu Y, Zhou W, Ye W, Lin B, Weng S, Chen Y, Zheng C. Hes1 Controls Proliferation and Apoptosis in Chronic Lymphoblastic Leukemia Cells by Modulating PTEN Expression. Mol Biotechnol 2022; 64:1419-1430. [PMID: 35704163 DOI: 10.1007/s12033-022-00476-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 03/02/2022] [Indexed: 12/26/2022]
Abstract
Hairy and enhancer of split homolog-1 (HES1), regulated by the Notch, has been reported to play important roles in the immune response and cancers, such as leukemia. In this study, we aim to explore the effect of HES1-mediated Notch1 signaling pathway in chronic lymphocytic leukemia (CLL). Reverse transcription quantitative polymerase chain reaction and Western blot assay were conducted to determine the expression of HES1, Notch1, and PTEN in B lymphocytes of peripheral blood samples of 60 CLL patients. We used lentivirus-mediated overexpression or silencing of HES1 and the Notch1 signaling pathway inhibitor, MW167, to detect the interaction among HES1, Notch1, and PTEN in CLL MEC1 and HG3 cells. MTT assay and flow cytometry were employed for detection of biological behaviors of CLL cells. HES1 and Notch1 showed high expression, but PTEN displayed low expression in B lymphocytes of peripheral blood samples of patients with CLL in association with poor prognosis. HES1 bound to the promoter region of PTEN and reduced PTEN expression. Overexpression of HES1 activated the Notch1 signaling pathway, thus promoting the proliferation of CLL cells, increasing the proportion of cells arrested at the S phase and limiting the apoptosis of CLL cells. Collectively, HES1 can promote activation of the Notch1 signaling pathway to cause PTEN transcription inhibition and the subsequent expression reduction, thereby promoting the proliferation and inhibiting the apoptosis of CLL cells.
Collapse
Affiliation(s)
- Qikai Zhang
- Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Zongsi Zhu
- Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Jiaqiang Guan
- Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Yingying Hu
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Wenjin Zhou
- Department of Chemotherapy, Cancer Hospital of The University of Chinese Academy of Science, Wenzhou Campus, Wenzhou, 325000, People's Republic of China
| | - Wanchun Ye
- Department of Chemotherapy, Cancer Hospital of The University of Chinese Academy of Science, Wenzhou Campus, Wenzhou, 325000, People's Republic of China
| | - Bijing Lin
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Shanshan Weng
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Yuemiao Chen
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Cuiping Zheng
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China.
| |
Collapse
|
23
|
Xing G, Liu Z, Huang L, Zhao D, Wang T, Yuan H, Wu Y, Li L, Long Q, Zhou Y, Hao Z, Liu Y, Lu J, Li S, Zhu J, Wang B, Wang J, Liu J, Chen J, Pei D, Liu X, Chen K. MAP2K6 remodels chromatin and facilitates reprogramming by activating Gatad2b-phosphorylation dependent heterochromatin loosening. Cell Death Differ 2022; 29:1042-1054. [PMID: 34815549 PMCID: PMC9090911 DOI: 10.1038/s41418-021-00902-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 11/03/2021] [Accepted: 11/10/2021] [Indexed: 11/09/2022] Open
Abstract
Somatic cell reprogramming is an ideal model for studying epigenetic regulation as it undergoes dramatic chromatin remodeling. However, a role for phosphorylation signaling in chromatin protein modifications for reprogramming remains unclear. Here, we identified mitogen-activated protein kinase kinase 6 (Mkk6) as a chromatin relaxer and found that it could significantly enhance reprogramming. The function of Mkk6 in heterochromatin loosening and reprogramming requires its kinase activity but does not depend on its best-known target, P38. We identified Gatad2b as a novel target of Mkk6 phosphorylation that acts downstream to elevate histone acetylation levels and loosen heterochromatin. As a result, Mkk6 over-expression facilitates binding of Sox2 and Klf4 to their targets and promotes pluripotency gene expression during reprogramming. Our studies not only reveal an Mkk phosphorylation mediated modulation of chromatin status in reprogramming, but also provide new rationales to further investigate and improve the cell fate determination processes.
Collapse
Affiliation(s)
- Guangsuo Xing
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Zichao Liu
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Luyuan Huang
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Danyun Zhao
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Tao Wang
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Hao Yuan
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Yi Wu
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Linpeng Li
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Qi Long
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Yanshuang Zhou
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Zhihong Hao
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Yang Liu
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Jianghuan Lu
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Shiting Li
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Jieying Zhu
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Bo Wang
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Junwei Wang
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Jing Liu
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Jiekai Chen
- grid.410737.60000 0000 8653 1072CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530 Guangzhou, China ,grid.428926.30000 0004 1798 2725Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, China
| | - Duanqing Pei
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China. .,Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China. .,Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
| | - Keshi Chen
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou Medical University, 510530, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China.
| |
Collapse
|
24
|
Cao Y, Tu Y, Xiong J, Tan S, Luo L, Wu A, Shu X, Jie Z, Li Z. microRNA-15b-5p encapsulated by M2 macrophage-derived extracellular vesicles promotes gastric cancer metastasis by targeting BRMS1 and suppressing DAPK1 transcription. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:152. [PMID: 35449111 PMCID: PMC9027839 DOI: 10.1186/s13046-022-02356-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUND Extracellular vesicles (EVs) derived from tumor-associated macrophages are implicated in the progression and metastasis of gastric cancer (GC) via the transfer of molecular cargo RNAs. We aimed to decipher the impact of microRNA (miR)-15b-5p transferred by M2 macrophage-derived EVs in the metastasis of GC. METHODS Expression of miR-15b-5p was assessed and the downstream genes of miR-15b-5p were analyzed. GC cells were subjected to gain- and loss-of function experiments for miR-15b-5p, BRMS1, and DAPK1. M2 macrophage-derived EVs were extracted, identified, and subjected to co-culture with GC cells and their biological behaviors were analyzed. A lung metastasis model in nude mice was established to determine the effects of miR-15b-5p on tumor metastasis in vivo. RESULTS miR-15b-5p was upregulated in GC tissues and cells as well as in M2 macrophage-derived EVs. miR-15b-5p promoted the proliferative and invasive potentials, and epithelial-mesenchymal transition (EMT) of GC cells. M2 macrophage-derived EVs could transfer miR-15b-5p into GC cells where it targeted BRMS1 by binding to its 3'UTR. BRMS1 was enriched in the DAPK1 promoter region and promoted its transcription, thereby arresting the proliferative and invasive potentials, and EMT of GC cells. In vivo experiments demonstrated that orthotopic implantation of miR-15b-5p overexpressing GC cells in nude mice displayed led to enhanced tumor metastasis by inhibiting the BRMS1/DAPK1 axis. CONCLUSIONS Overall, miR-15b-5p delivered by M2 macrophage-derived EVs constitutes a molecular mechanism implicated in the metastasis of GC, and may thus be considered as a novel therapeutic target for its treatment.
Collapse
Affiliation(s)
- Yi Cao
- Department of General Surgery, Jiangxi Province, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Road, Nanchang, 330006, People's Republic of China
| | - Yi Tu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Jianbo Xiong
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Shengxing Tan
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Lianghua Luo
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Ahao Wu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Xufeng Shu
- Department of General Surgery, Jiangxi Province, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Road, Nanchang, 330006, People's Republic of China
| | - Zhigang Jie
- Department of General Surgery, Jiangxi Province, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Road, Nanchang, 330006, People's Republic of China.
| | - Zhengrong Li
- Department of General Surgery, Jiangxi Province, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Road, Nanchang, 330006, People's Republic of China.
| |
Collapse
|
25
|
Sevoflurane Offers Neuroprotection in a Cerebral Ischemia/Reperfusion Injury Rat Model Through the E2F1/EZH2/TIMP2 Regulatory Axis. Mol Neurobiol 2022; 59:2219-2231. [PMID: 35064540 DOI: 10.1007/s12035-021-02602-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/15/2021] [Indexed: 10/19/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury contributes considerably to the poor prognosis in patients with ischemic stroke. This study is aimed to delineate the molecular mechanistic actions by which sevoflurane protects against cerebral I/R injury. A rat model of cerebral I/R injury was established and pre-treated with sevoflurane, in which hippocampal neuron apoptosis was found to be repressed and the level of E2F transcription factor 1 (E2F1) was observed to be down-regulated. Then, the up-regulated expression of E2F1 was validated in rats with cerebral I/R injury, responsible for stimulated neuron apoptosis. Further, the binding of E2F1 to enhancer of zeste homolog 2 (EZH2) and EZH2 to tissue inhibitor of metalloproteinases-2 (TIMP2) was identified. The stimulative effect of the E2F1/EZH2/TIMP2 regulatory axis on neuron apoptosis was subsequently demonstrated through functional assays. After that, it was substantiated in vivo that sevoflurane suppressed the apoptosis of hippocampal neurons in rats with cerebral I/R injury by down-regulating E2F1 to activate the EZH2/TIMP2 axis. Taken together, our data elucidated that sevoflurane reduced neuron apoptosis through mediating the E2F1/EZH2/TIMP2 regulatory axis, thus protecting rats against cerebral I/R injury.
Collapse
|
26
|
Mesejo C, Marzal A, Martínez-Fuentes A, Reig C, de Lucas M, Iglesias DJ, Primo-Millo E, Blázquez MA, Agustí M. Reversion of fruit-dependent inhibition of flowering in Citrus requires sprouting of buds with epigenetically silenced CcMADS19. THE NEW PHYTOLOGIST 2022; 233:526-533. [PMID: 34403516 DOI: 10.1111/nph.17681] [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: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 05/16/2023]
Abstract
In Citrus, the response to environmental floral inductive signals is inhibited by the presence of developing fruits. The mechanism involves epigenetic activation of the CcMADS19 locus (FLC orthologue), encoding a floral repressor. To understand how this epigenetic regulation is reverted to allow flowering in the following season, we have forced precocious sprouting of axillary buds in fruit-bearing shoots, and examined the competence to floral inductive signals of old and new leaves derived from them. We have found that CcMADS19 is enriched in repressive H3K27me3 marks in young, but not old leaves, revealing that axillary buds retain a silenced version of the floral repressor that is mitotically transmitted to the newly emerging leaves, which are able to induce flowering. Therefore, we propose that flowering in Citrus is necessarily preceded by vegetative sprouting, so that the competence to respond to floral inductive signals is reset in the new leaves.
Collapse
Affiliation(s)
- Carlos Mesejo
- Instituto Agroforestal Mediterráneo, Universitat Politècnica de València, València, 46022, Spain
| | - Andrés Marzal
- Instituto Agroforestal Mediterráneo, Universitat Politècnica de València, València, 46022, Spain
| | - Amparo Martínez-Fuentes
- Instituto Agroforestal Mediterráneo, Universitat Politècnica de València, València, 46022, Spain
| | - Carmina Reig
- Instituto Agroforestal Mediterráneo, Universitat Politècnica de València, València, 46022, Spain
| | - Miguel de Lucas
- Department of Biosciences, Durham University, Stockton Rd, Durham, DH1 3LE, UK
| | - Domingo J Iglesias
- Centro de Citricultura y Producción Vegetal, IVIA-GV, Moncada, València, 46113, Spain
| | - Eduardo Primo-Millo
- Centro de Citricultura y Producción Vegetal, IVIA-GV, Moncada, València, 46113, Spain
| | - Miguel A Blázquez
- Instituto de Biología Molecular y Celular de Plantas, CSIC-UPV, València, 46022, Spain
| | - Manuel Agustí
- Instituto Agroforestal Mediterráneo, Universitat Politècnica de València, València, 46022, Spain
| |
Collapse
|
27
|
Measuring the buffering capacity of gene silencing in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A 2021; 118:2111841118. [PMID: 34857629 PMCID: PMC8670432 DOI: 10.1073/pnas.2111841118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2021] [Indexed: 11/18/2022] Open
Abstract
Gene silencing, once established, is stably maintained for several generations. Despite the high fidelity of the inheritance of the silent state, individual components of silenced chromatin are in constant flux. Models suggest that silent loci can tolerate fluctuations in Sir proteins and histone acetylation levels, but the level of tolerance is unknown. To understand the quantitative relationships between H4K16 acetylation, Sir proteins, and silencing, we developed assays to quantitatively alter a H4K16 acetylation mimic allele and Sir protein levels and measure the effects of these changes on silencing. Our data suggest that a two- to threefold change in levels of histone marks and specific Sir proteins affects the stability of the silent state of a large chromatin domain. Gene silencing in budding yeast is mediated by Sir protein binding to unacetylated nucleosomes to form a chromatin structure that inhibits transcription. Transcriptional silencing is characterized by the high-fidelity transmission of the silent state. Despite its relative stability, the constituent parts of the silent state are in constant flux, giving rise to a model that silent loci can tolerate such fluctuations without functional consequences. However, the level of tolerance is unknown, and we developed methods to measure the threshold of histone acetylation that causes the silent chromatin state to switch to the active state as well as to measure the levels of the enzymes and structural proteins necessary for silencing. We show that loss of silencing required 50 to 75% acetyl-mimic histones, though the precise levels were influenced by silencer strength and upstream activating sequence (UAS) enhancer/promoter strength. Measurements of repressor protein levels necessary for silencing showed that reducing SIR4 gene dosage two- to threefold significantly weakened silencing, though reducing the gene copy numbers for Sir2 or Sir3 to the same extent did not significantly affect silencing suggesting that Sir4 was a limiting component in gene silencing. Calculations suggest that a mere twofold reduction in the ability of acetyltransferases to acetylate nucleosomes across a large array of nucleosomes may be sufficient to generate a transcriptionally silent domain.
Collapse
|
28
|
Halabian R, Valizadeh Arshad, Ahmadi A, Saeedi P, Azimzadeh Jamalkandi S, Alivand MR. Laboratory methods to decipher epigenetic signatures: a comparative review. Cell Mol Biol Lett 2021; 26:46. [PMID: 34763654 PMCID: PMC8582164 DOI: 10.1186/s11658-021-00290-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
Epigenetics refers to nucleotide sequence-independent events, and heritable changes, including DNA methylation and histone modification (as the two main processes), contributing to the phenotypic features of the cell. Both genetics and epigenetics contribute to determining the outcome of regulatory gene expression systems. Indeed, the flexibility of epigenetic effects and stability of genetic coding lead to gene regulation complexity in response signals. Since some epigenetic changes are significant in abnormalities such as cancers and neurodegenerative diseases, the initial changes, dynamic and reversible properties, and diagnostic potential of epigenomic phenomena are subject to epigenome-wide association studies (EWAS) for therapeutic aims. Based on recent studies, methodological developments are necessary to improve epigenetic research. As a result, several methods have been developed to explore epigenetic alterations at low, medium, and high scales, focusing on DNA methylation and histone modification detection. In this research field, bisulfite-, enzyme sensitivity- and antibody specificity-based techniques are used for DNA methylation, whereas histone modifications are gained based on antibody recognition. This review provides a mechanism-based understanding and comparative overview of the most common techniques for detecting the status of epigenetic effects, including DNA methylation and histone modifications, for applicable approaches from low- to high-throughput scales.
Collapse
Affiliation(s)
- Raheleh Halabian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Valizadeh Arshad
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute For Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Pardis Saeedi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Mollasadra Ave., 14359-16471, Tehran, Iran.
| | - Mohammad Reza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
29
|
Du Z, Shi X, Guan A. lncRNA H19 facilitates the proliferation and differentiation of human dental pulp stem cells via EZH2-dependent LATS1 methylation. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 25:116-126. [PMID: 34401209 PMCID: PMC8339349 DOI: 10.1016/j.omtn.2021.04.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 04/20/2021] [Indexed: 12/11/2022]
Abstract
Human dental pulp stem cells (hDPSCs) have been recognized as a candidate cell source for tissue engineering. Long non-coding RNAs (lncRNAs) are differentially expressed in inflamed human dental pulp tissues. The present study is aimed at investigating the role of lncRNA H19 in the differentiation potential of hDPSCs. hDPSCs were successfully isolated and cultured, followed by conducting gain and loss-of-function experiments on lncRNA H19 and large tumor suppressor 1 (LATS1) to elucidate their respective biological functions in hDPSCs. lncRNA H19 was able to promote, whereas LATS1 was found to inhibit the differentiation, proliferation, and migration capabilities of hDPSCs. LATS1 was found to activate the Hippo-Yes-associated protein (YAP) signaling pathway by decreasing levels of YAP and Tafazzin (TAZ). The effects of lncRNA H19 on hDPSCs were achieved by repressing LATS1 through enhancer of zeste homolog 2-induced trimethylation of histone 3 at lysine 27. Finally, hDPSCs overexpressing lncRNA H19 and/or LATS1 were transplanted into nude mice. It was shown that lncRNA H19 inhibited LATS1 to promote the production of odontoblasts in vivo. Taken together, lncRNA H19 serves as a contributor to the differentiation potential of hDPSCs via the inhibition of LATS1, therefore highlighting novel therapeutic targets for dental pulp repair.
Collapse
Affiliation(s)
- Zhen Du
- Department of Stomatology, Linyi People's Hospital, Linyi 276000, Shandong Province, P.R. China
| | - Xiaoming Shi
- Department of Stomatology, Linyi People's Hospital, Linyi 276000, Shandong Province, P.R. China
| | - Aizhong Guan
- Department of Stomatology, Linyi People's Hospital, Linyi 276000, Shandong Province, P.R. China
| |
Collapse
|
30
|
Hong S, Li S, Bi M, Yu H, Yan Z, Liu T, Wang H. lncRNA ILF3-AS1 promotes proliferation and metastasis of colorectal cancer cells by recruiting histone methylase EZH2. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:1012-1023. [PMID: 34141456 PMCID: PMC8167202 DOI: 10.1016/j.omtn.2021.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 04/07/2021] [Indexed: 12/11/2022]
Abstract
The role of long non-coding RNA (lncRNA) has been displayed in colorectal cancer (CRC). Here, we aimed to discuss the role of lncRNA interleukin enhancer-binding factor 3-antisense RNA 1 (ILF3-AS1)/enhancer of zeste homolog 2 (EZH2)/cyclin-dependent kinase inhibitor 2 (CDKN2A)/histone 3 (H3) lysine 27 trimethylation (H3K27me3) in cell proliferation and metastasis of CRC. ILF3-AS1, EZH2, and CDKN2A levels in CRC tissues and cells were detected. The relationship between ILF3-AS1/EZH2 expression and the clinicopathological features of CRC was analyzed. High/low expression of ILF3-AS1/EZH2 plasmids were composed to explore the function of ILF3-AS1/EZH2 in invasion, migration, proliferation, colony formation, and apoptosis of CRC cells. The growth status of nude mice was observed to verify the in vitro results from in vivo experiment. ILF3-AS1 and EZH2 increased, whereas CDKN2A reduced in CRC tissues and cells. ILF3-AS1 and EZH2 expression was linked to Dukes stage, distant metastasis, vascular invasion, and lymph node metastasis of CRC patients. Depleted ILF3-AS1 or reduced EZH2 suppressed proliferation, migration, colony-formation, and invasion ability, as well as facilitated apoptosis of CRC cells and attenuated the tumor growth in CRC mice. ILF3-AS1 accelerates the proliferation and metastasis of CRC cells by recruiting histone methylase EZH2 to induce trimethylation of H3K27 and downregulate CDKN2A.
Collapse
Affiliation(s)
- Sen Hong
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun 130021, Jilin, People’s Republic of China
| | - Shiquan Li
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun 130021, Jilin, People’s Republic of China
| | - Miaomiao Bi
- Department of Ophthalmology, The China-Japan Union Hospital of Jilin University, Jilin University, Changchun 130022, Jilin, People’s Republic of China
| | - Haiyao Yu
- Chief Pharmacist, Changchun Food and Drug Inspection Center, Changchun, Jilin, People’s Republic of China
| | - Zhenkun Yan
- Endoscopy Center, The China-Japan Union Hospital of Jilin University, Changchun 130022, Jilin, People’s Republic of China
| | - Tao Liu
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun 130021, Jilin, People’s Republic of China
| | - Helei Wang
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130021, Jilin, People’s Republic of China
| |
Collapse
|
31
|
Silencing of long non-coding RNA FOXD2-AS1 inhibits the progression of gallbladder cancer by mediating methylation of MLH1. Gene Ther 2021; 28:306-318. [PMID: 32917950 DOI: 10.1038/s41434-020-00187-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 07/13/2020] [Accepted: 08/05/2020] [Indexed: 01/29/2023]
Abstract
Evidence has documented the tumor-promoting properties of long non-coding RNA (lncRNA) FOXD2 adjacent opposite strand RNA 1 (FOXD2-AS1) in many cancers. However, little is known about its role in gallbladder cancer. Here, we aimed to characterize the functional relevance of lncRNA FOXD2-AS1 in gallbladder cancer and the possible mechanisms associated with methylation of MutL homolog-1 (MLH1). Initially, microarray-based gene expression profiling of gallbladder cancer was employed to identify differentially expressed lncRNAs. Next, the expression of lncRNA FOXD2-AS1 was examined, and the cell line presenting with the highest lncRNA FOXD2-AS1 expression was selected for subsequent experimentation. Then, the interaction between lncRNA FOXD2-AS1 and MLH1 was identified. The effect of lncRNA FOXD2-AS1 on proliferation, migration, invasion, and apoptosis as well as tumorigenicity of transfected GBC-SD cells was examined with gain- and loss-of-function experiments. We found that lncRNA FOXD2-AS1 was highly expressed, while MLH1 was poorly expressed in gallbladder cancer cells. Besides, lncRNA FOXD2-AS1 could promote MLH1 methylation by recruiting DNMT1 to the MLH1 promoter, and consequently inhibit MLH1 transcription. Silencing of lncRNA FOXD2-AS1 or overexpression of MLH1 inhibited gallbladder cancer cell proliferation, invasion, and migration, while facilitating cell apoptosis in vitro as well as retarding tumor growth in vivo. Thus, silencing of lncRNA FOXD2-AS1 suppressed the progression of gallbladder cancer via upregulation of MLH1 by inhibiting MLH1 promoter methylation. These findings present lncRNA FOXD2-AS1 knockdown as a potential candidate for the treatment of gallbladder cancer.
Collapse
|
32
|
Pan W, Song X, Hu Q, Zhang Y. miR-485 inhibits histone deacetylase HDAC5, HIF1α and PFKFB3 expression to alleviate epilepsy in cellular and rodent models. Aging (Albany NY) 2021; 13:14416-14432. [PMID: 34021541 PMCID: PMC8202868 DOI: 10.18632/aging.203058] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/05/2020] [Indexed: 01/15/2023]
Abstract
We investigated the role of microRNA (miR)-485 and its downstream signaling molecules on mediating epilepsy in cellular and rat models. We established a cellular epilepsy model by exposing hippocampal neurons to magnesium and a rat model by treating ICR mice with lithium chloride (127 mg/kg) and pilocarpine (30 mg/kg). We confirmed that miR-485 could bind and inhibit histone deacetylase 5 (HDAC5) and then measured expression of miR-485 and in mice and cells. Cells were transfected with overexpression or knockdown of miR-485, HDAC5, hypoxia-inducible factor-1alpha (HIF1α), or 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 enzyme (PFKFB3) to verify their roles in apoptosis, oxidative stress, and inflammation in epileptic hippocampal neurons. Binding relationship between miR-485, HDAC5, HIF1α, and PFKFB3 was verified. Oxidative stress and inflammation marker levels in epilepsy model mice were assessed. miR-485 was downregulated and HDAC5 was upregulated in cell and animal model of epilepsy. Seizure, neuronal apoptosis, oxidative stress (increased SOD and GSH-Px expression and decreased MDA and 8-OHdG expression) and inflammation (reduced IL-1β, TNF-α, and IL-6 expression) were reduced by miR-485 in epileptic cells. HIF1α and PFKFB3 expression was reduced by HDAC5 knockdown in cells, which was recapitulated in vivo. Thus, miR-485 alleviates neuronal damage and epilepsy by inhibiting HDAC5, HIF1α, and PFKFB3.
Collapse
Affiliation(s)
- Wei Pan
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Xingyu Song
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Qibo Hu
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yunfeng Zhang
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| |
Collapse
|
33
|
Chen P, Duan Y, Lu X, Chen L, Zhang W, Wang H, Hu R, Liu S. RB1CC1 functions as a tumor-suppressing gene in renal cell carcinoma via suppression of PYK2 activity and disruption of TAZ-mediated PDL1 transcription activation. Cancer Immunol Immunother 2021; 70:3261-3275. [PMID: 33837850 DOI: 10.1007/s00262-021-02913-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 03/08/2021] [Indexed: 10/21/2022]
Abstract
Rb1-inducible coiled-coil 1 (RB1CC1) has been demonstrated to function as an inhibitor of proline-rich/Ca-activated tyrosine kinase 2 (PYK2) by binding to the kinase domain of PYK2, which promotes the proliferation, invasion, and migration of renal cell carcinoma (RCC) cells. Additionally, in breast cancer, PYK2 positively regulates the expression of transcriptional co-activator with PDZ-binding motif (TAZ) which in turn can enhance PDL1 levels in breast and lung cancer cells. The current study was performed to decipher the impact of RB1CC1 in the progression of RCC via regulation of the PYK2/TAZ/PDL1 signaling axis. Expression of RB1CC1 and PYK2 was quantified in clinical tissue samples from RCC patients. The relationship between TAZ and PYK2, TAZ and PDL1 was then validated. The cellular processes of doxorubicin (DOX)-induced human RCC cell lines including the abilities of proliferation, colony formation, sphere formation and apoptosis, as well as the tumorigenicity of transfected cells, were evaluated after the alteration of RB1CC1 expression. RB1CC1 exhibited decreased expression in RCC tissues and was positively correlated with patient survival. RB1CC1 could inhibit the activity of PYK2, which in turn stimulated the stability of TAZ protein by phosphorylating TAZ. Meanwhile, TAZ protein activated PDL1 transcription by binding to the promoter region of PDL1. RB1CC1 overexpression or PYK2 knockdown could help everolimus (EVE) to inhibit tumor proliferation and activate immune response. Taken together, RB1CC1 can potentially augment the response of RCC cells to immunotherapy by suppressing the PYK2/TAZ/PDL1 signaling axis.
Collapse
Affiliation(s)
- Pingfeng Chen
- Department of Urology, First Affiliated Hospital, University of South China, No. 69, Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Youjun Duan
- Department of Urology, First Affiliated Hospital, University of South China, No. 69, Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Xinsheng Lu
- Department of Urology, First Affiliated Hospital, University of South China, No. 69, Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Libo Chen
- Department of Urology, First Affiliated Hospital, University of South China, No. 69, Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Wang Zhang
- Department of Urology, First Affiliated Hospital, University of South China, No. 69, Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Hao Wang
- Department of Urology, First Affiliated Hospital, University of South China, No. 69, Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Rong Hu
- Department of Radiology, First Affiliated Hospital, University of South China, No. 69, Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China.
| | - Shimin Liu
- Department of Urology, First Affiliated Hospital, University of South China, No. 69, Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China.
| |
Collapse
|
34
|
Ötvös K, Miskolczi P, Marhavý P, Cruz-Ramírez A, Benková E, Robert S, Bakó L. Pickle Recruits Retinoblastoma Related 1 to Control Lateral Root Formation in Arabidopsis. Int J Mol Sci 2021; 22:ijms22083862. [PMID: 33917959 PMCID: PMC8068362 DOI: 10.3390/ijms22083862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/04/2021] [Accepted: 04/06/2021] [Indexed: 12/31/2022] Open
Abstract
Lateral root (LR) formation is an example of a plant post-embryonic organogenesis event. LRs are issued from non-dividing cells entering consecutive steps of formative divisions, proliferation and elongation. The chromatin remodeling protein PICKLE (PKL) negatively regulates auxin-mediated LR formation through a mechanism that is not yet known. Here we show that PKL interacts with RETINOBLASTOMA-RELATED 1 (RBR1) to repress the LATERAL ORGAN BOUNDARIES-DOMAIN 16 (LBD16) promoter activity. Since LBD16 function is required for the formative division of LR founder cells, repression mediated by the PKL–RBR1 complex negatively regulates formative division and LR formation. Inhibition of LR formation by PKL–RBR1 is counteracted by auxin, indicating that, in addition to auxin-mediated transcriptional responses, the fine-tuned process of LR formation is also controlled at the chromatin level in an auxin-signaling dependent manner.
Collapse
Affiliation(s)
- Krisztina Ötvös
- Department of Plant Physiology, Umeå Plant Science Center, Umeå University, S-901 87 Umeå, Sweden
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria; (P.M.); (E.B.)
- Bioresources Unit, AIT Austrian Institute of Technology, 3430 Tulln, Austria
- Correspondence: (K.Ö.); (L.B.); Tel.: +46-907867970 (K.Ö.); Fax: +46-907866676 (K.Ö.)
| | - Pál Miskolczi
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Center, Swedish University of Agricultural Sciences, S-901 87 Umeå, Sweden; (P.M.); (S.R.)
| | - Peter Marhavý
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria; (P.M.); (E.B.)
| | - Alfredo Cruz-Ramírez
- Laboratory of Molecular and Developmental Complexity at Laboratorio Nacional de Genómica para la Biodiversidad, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, (CINVESTAV-IPN), 36590 Irapuato, Mexico;
| | - Eva Benková
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria; (P.M.); (E.B.)
| | - Stéphanie Robert
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Center, Swedish University of Agricultural Sciences, S-901 87 Umeå, Sweden; (P.M.); (S.R.)
| | - László Bakó
- Department of Plant Physiology, Umeå Plant Science Center, Umeå University, S-901 87 Umeå, Sweden
- Correspondence: (K.Ö.); (L.B.); Tel.: +46-907867970 (K.Ö.); Fax: +46-907866676 (K.Ö.)
| |
Collapse
|
35
|
Hu A, Hong F, Li D, Jin Y, Kon L, Xu Z, He H, Xie Q. Long non-coding RNA ROR recruits histone transmethylase MLL1 to up-regulate TIMP3 expression and promote breast cancer progression. J Transl Med 2021; 19:95. [PMID: 33653378 PMCID: PMC7927245 DOI: 10.1186/s12967-020-02682-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/15/2020] [Indexed: 01/11/2023] Open
Abstract
Background As a significant cause of cancer deaths worldwide, breast cancer continues to be a troublesome malignancy. Long non-coding RNAs (lncRNAs) have been implicated in the development of breast cancer. Abnormal methylation has been associated with unfavorable breast cancer prognosis. Herein, the current study aimed to elucidate the role of lncRNA ROR in breast cancer. Methods RT-qPCR was performed to determine whether lncRNA ROR was highly expressed in breast cancer tissues, while lncRNA ROR expression was detected in both the nuclear and cytoplasm of breast cancer cells. MCF-7 cells were subsequently introduced with oe-lncRNA ROR, sh-lncRNA ROR to explore the effects of lncRNA ROR on cell proliferation, invasion and apoptosis. Results RIP, RNA pull-down and ChIP assays provided evidence suggesting that lncRNA ROR recruited transmethylase MLL1 to promote H3K4 trimethylation that enhanced TIMP3 transcription. The rescue experiments demonstrated that lncRNA ROR knockdown could inhibit the progression of breast cancer via the downregulation of TIMP3. Finally, the in vivo experiment findings consistently highlighted the suppressive effects of lncRNA ROR silencing on tumor growth. Conclusion Taken together, our study demonstrates that silencing of lncRNA ROR inhibits breast cancer progression via repression of transmethylase MLL1 and TIMP3, emphasizing the potential of lncRNA ROR as a novel target against breast cancer.
Collapse
Affiliation(s)
- Aixia Hu
- Department of Pathology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou, 450003, Henan, People's Republic of China. .,Henan University People's Hospital, Zhengzhou, 450003, People's Republic of China.
| | - Fan Hong
- Henan University People's Hospital, Zhengzhou, 450003, People's Republic of China
| | - Daohong Li
- Department of Pathology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou, 450003, Henan, People's Republic of China
| | - Yuwei Jin
- Department of Pathology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou, 450003, Henan, People's Republic of China
| | - Lingfei Kon
- Department of Pathology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou, 450003, Henan, People's Republic of China.,Henan University People's Hospital, Zhengzhou, 450003, People's Republic of China
| | - Ziguang Xu
- Department of Pathology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou, 450003, Henan, People's Republic of China
| | - Hui He
- Department of Pathology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou, 450003, Henan, People's Republic of China
| | - Qi Xie
- Department of Pathology, Henan Provincial People's Hospital, No. 7, Weiwu Road, Zhengzhou, 450003, Henan, People's Republic of China
| |
Collapse
|
36
|
Mann SN, Hadad N, Nelson Holte M, Rothman AR, Sathiaseelan R, Ali Mondal S, Agbaga MP, Unnikrishnan A, Subramaniam M, Hawse J, Huffman DM, Freeman WM, Stout MB. Health benefits attributed to 17α-estradiol, a lifespan-extending compound, are mediated through estrogen receptor α. eLife 2020; 9:59616. [PMID: 33289482 PMCID: PMC7744101 DOI: 10.7554/elife.59616] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic dysfunction underlies several chronic diseases, many of which are exacerbated by obesity. Dietary interventions can reverse metabolic declines and slow aging, although compliance issues remain paramount. 17α-estradiol treatment improves metabolic parameters and slows aging in male mice. The mechanisms by which 17α-estradiol elicits these benefits remain unresolved. Herein, we show that 17α-estradiol elicits similar genomic binding and transcriptional activation through estrogen receptor α (ERα) to that of 17β-estradiol. In addition, we show that the ablation of ERα completely attenuates the beneficial metabolic effects of 17α-E2 in male mice. Our findings suggest that 17α-E2 may act through the liver and hypothalamus to improve metabolic parameters in male mice. Lastly, we also determined that 17α-E2 improves metabolic parameters in male rats, thereby proving that the beneficial effects of 17α-E2 are not limited to mice. Collectively, these studies suggest ERα may be a drug target for mitigating chronic diseases in male mammals.
Collapse
Affiliation(s)
- Shivani N Mann
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Niran Hadad
- The Jackson Laboratory, Bar Harbor, United States
| | - Molly Nelson Holte
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Alicia R Rothman
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Roshini Sathiaseelan
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Samim Ali Mondal
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Martin-Paul Agbaga
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Archana Unnikrishnan
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | | | - John Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, United States
| | - Willard M Freeman
- Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, United States.,Oklahoma City Veterans Affairs Medical Center, Oklahoma City, United States
| | - Michael B Stout
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| |
Collapse
|
37
|
Zhao H, Xu Q. Long non-coding RNA DLX6-AS1 mediates proliferation, invasion and apoptosis of endometrial cancer cells by recruiting p300/E2F1 in DLX6 promoter region. J Cell Mol Med 2020; 24:12572-12584. [PMID: 32951317 PMCID: PMC7686961 DOI: 10.1111/jcmm.15810] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 07/30/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
Endometrial cancer features abnormal growth of cells of the inner lining of the uterus with the potential to invade to other organs. Accumulating evidence suggests that aberrant expression of long non‐coding RNA (lncRNA) may facilitate cancer progression. The aim of the present study was to identify the molecular mechanisms of the lncRNA known as DLX6 antisense RNA 1 (DLX6‐AS1) in endometrial cancer. Microarray‐based analysis was utilized to predict expression profile and possible function pattern of DLX6‐AS1 and DLX6 in endometrial cancer, and their expression was quantified in 78 clinically obtained endometrial cancer tissues and also in cell lines. We next assessed the effects of DLX6‐AS1 and DLX6 on proliferation, invasion and apoptosis of endometrial cancer cells. A mouse xenograft model was established to confirm DLX6‐AS1 functions and explore its underlying regulatory mechanisms in vivo. DLX6‐AS1 and DLX6 were highly expressed in endometrial cancer tissues and cells, and their silencing weakened the proliferative and invasive abilities of endometrial cancer cells and tumours, while promoting apoptosis. Mechanistic investigations indicated that DLX6‐AS1 formed a triplex structure with DLX6 via interaction with p300/E2F1 acetyltransferase. Thus, we find that functional up‐regulation of DLX6‐AS1 can promote endometrial cancer progression via a novel triplex mechanism that may prove to be great clinical significance for future treatments of endometrial cancer.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Gynaecology and Obstetrics, Linyi People's Hospital, Linyi, China
| | - Qian Xu
- Department of Gynaecology and Obstetrics, Linyi People's Hospital, Linyi, China
| |
Collapse
|
38
|
Chen L, Fan X, Zhu J, Chen X, Liu Y, Zhou H. LncRNA MAGI2-AS3 inhibits the self-renewal of leukaemic stem cells by promoting TET2-dependent DNA demethylation of the LRIG1 promoter in acute myeloid leukaemia. RNA Biol 2020; 17:784-793. [PMID: 32174258 PMCID: PMC7549675 DOI: 10.1080/15476286.2020.1726637] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 12/19/2022] Open
Abstract
The presence or absence of cytogenetic mutations is proposed to be responsible for the pathogenesis of acute myeloid leukaemia (AML). However, the current classification system is inadequate to elucidate the molecular heterogeneity of the disease, and therapy failures frequently occur. Leukaemia stem cells (LSCs) initiate and maintain the clonal hierarchy of AML and exhibit properties of self-renewal remaining recalcitrant to conventional chemotherapy. In this study, we identified a novel long non-coding RNA (lncRNA) MAGI2 antisense RNA 3 (MAGI2-AS3) in AML and investigated its functional role in regulating LSCs self-renewal. LSCs were identified by immunoprofiling of CD34+ CD123+ in AML patients' marrow. MAGI2-AS3 exhibited a poor expression level in LSCs than the normal human haematopoietic stem cells. Lentivirus-mediated upregulation of MAGI2-AS3 or leucine-rich repeats and Ig-like domains 1 (LRIG1) impaired LSCs self-renewal. MAGI2-AS3-overexpressed LSCs acquired the ability of self-renewal following lentivirus-mediated knockdown of LRIG1. Methylation-dependent inhibition of LRIG1 was evident in LSCs. MAGI2-AS3 was found to induce occupancy of TET2 at the LRIG1 promoter. Lentivirus-mediated downregulation of TET2 could impair MAGI2-AS3-mediated elevation of LRIG1 and neutralize the inhibitory effect of MAGI2-AS3 on LSCs self-renewal. In vivo analysis indicated an elevated overall survival of NOD/SCID mice injected with LSCs in the presence of MAGI2-AS3. Altogether, the key findings support the potential of lncRNA MAGI2-AS3 to serve as a novel candidate for the improvement of AML treatment.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Cell Line, Tumor
- Cell Self Renewal/genetics
- DNA Demethylation
- DNA-Binding Proteins/metabolism
- Dioxygenases
- Gene Expression Regulation, Leukemic
- Guanylate Kinases/genetics
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Membrane Glycoproteins/genetics
- Neoplastic Stem Cells/metabolism
- Promoter Regions, Genetic
- Protein Binding
- Proto-Oncogene Proteins/metabolism
- RNA Interference
- RNA, Antisense
- RNA, Long Noncoding
Collapse
Affiliation(s)
- Lijuan Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Fan
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jianhua Zhu
- Laboratory of Clinical Immunology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuexin Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiling Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Zhou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
39
|
Alqahtani A, Skelton A, Eley L, Annavarapu S, Henderson DJ, Chaudhry B. Isolation and next generation sequencing of archival formalin-fixed DNA. J Anat 2020; 237:587-600. [PMID: 32426881 PMCID: PMC7476199 DOI: 10.1111/joa.13209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 11/29/2022] Open
Abstract
DNA from archived organs is presumed unsuitable for genomic studies because of excessive formalin‐fixation. As next generation sequencing (NGS) requires short DNA fragments, and Uracil‐N‐glycosylase (UNG) can be used to overcome deamination, there has been renewed interest in the possibility of genomic studies using these collections. We describe a novel method of DNA extraction capable of providing PCR amplicons of at least 400 bp length from such excessively formalin‐fixed human tissues. When compared with a leading commercial formalin‐fixed DNA extraction kit, our method produced greater yields of DNA and reduced sequence variations. Analysis of PCR products using bacterial sub‐cloning and Sanger sequencing from UNG‐treated DNA unexpectedly revealed increased sequence variations, compared with untreated samples. Finally, whole exome NGS was performed on a myocardial sample fixed in formalin for 2 years and compared with lymphocyte‐derived DNA (as a gold standard) from the same patient. Despite the reduction in the number and quality of reads in the formalin‐fixed DNA, we were able to show that bioinformatic processing by joint calling and variant quality score recalibration (VQSR) increased the sensitivity four‐fold to 56% and doubled specificity to 68% when compared with a standard hard‐filtering approach. Thus, high‐quality DNA can be extracted from excessively formalin‐fixed tissues and bioinformatic processing can optimise sensitivity and specificity of results. Sequencing of several sub‐cloned amplicons is an important methodological step in assessing DNA quality.
Collapse
Affiliation(s)
- Ahlam Alqahtani
- Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew Skelton
- Bioinformatic Support Unit, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Lorraine Eley
- Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Srinivas Annavarapu
- Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Department of Cellular Pathology, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Deborah J Henderson
- Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Bill Chaudhry
- Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
40
|
Differential activation of human pregnane X receptor PXR by isomeric mono-methylated indoles in intestinal and hepatic in vitro models. Toxicol Lett 2020; 324:104-110. [DOI: 10.1016/j.toxlet.2020.02.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/27/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
|
41
|
Chen K, Long Q, Xing G, Wang T, Wu Y, Li L, Qi J, Zhou Y, Ma B, Schöler HR, Nie J, Pei D, Liu X. Heterochromatin loosening by the Oct4 linker region facilitates Klf4 binding and iPSC reprogramming. EMBO J 2020; 39:e99165. [PMID: 31571238 PMCID: PMC6939195 DOI: 10.15252/embj.201899165] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 01/13/2023] Open
Abstract
The success of Yamanaka factor reprogramming of somatic cells into induced pluripotent stem cells suggests that some factor(s) must remodel the nuclei from a condensed state to a relaxed state. How factor-dependent chromatin opening occurs remains unclear. Using FRAP and ATAC-seq, we found that Oct4 acts as a pioneer factor that loosens heterochromatin and facilitates the binding of Klf4 and the expression of epithelial genes in early reprogramming, leading to enhanced mesenchymal-to-epithelial transition. A mutation in the Oct4 linker, L80A, which shows impaired interaction with the BAF complex component Brg1, is inactive in heterochromatin loosening. Oct4-L80A also blocks the binding of Klf4 and retards MET. Finally, vitamin C or Gadd45a could rescue the reprogramming deficiency of Oct4-L80A by enhancing chromatin opening and Klf4 binding. These studies reveal a cooperation between Oct4 and Klf4 at the chromatin level that facilitates MET at the cellular level and shed light into the research of multiple factors in cell fate determination.
Collapse
Affiliation(s)
- Keshi Chen
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| | - Qi Long
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| | - Guangsuo Xing
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
- Institute of Physical Science and Information TechnologyAnhui UniversityHefeiChina
| | - Tianyu Wang
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| | - Yi Wu
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| | - Linpeng Li
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| | - Juntao Qi
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| | - Yanshuang Zhou
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| | - Bochao Ma
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| | - Hans R Schöler
- Department for Cell and Developmental BiologyMax Planck Institute for Molecular BiomedicineMünsterGermany
| | - Jinfu Nie
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| | - Duanqing Pei
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative BiologyJoint School of Life SciencesHefei Institute of Stem Cell and Regenerative MedicineGuangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhou Medical UniversityGuangzhouChina
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangdong Provincial Key Laboratory of Stem Cell and Regenerative MedicineSouth China Institute for Stem Cell Biology and Regenerative MedicineInstitute for Stem Cell and RegenerationGuangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhouChina
| |
Collapse
|
42
|
Agustí M, Mesejo C, Muñoz-Fambuena N, Vera-Sirera F, de Lucas M, Martínez-Fuentes A, Reig C, Iglesias DJ, Primo-Millo E, Blázquez MA. Fruit-dependent epigenetic regulation of flowering in Citrus. THE NEW PHYTOLOGIST 2020; 225:376-384. [PMID: 31273802 DOI: 10.1111/nph.16044] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/26/2019] [Indexed: 06/09/2023]
Abstract
In many perennial plants, seasonal flowering is primarily controlled by environmental conditions, but in certain polycarpic plants, environmental signals are locally gated by the presence of developing fruits initiated in the previous season through an unknown mechanism. Polycarpy is defined as the ability of plants to undergo several rounds of reproduction during their lifetime, alternating vegetative and reproductive meristems in the same individual. To understand how fruits regulate flowering in polycarpic plants, we focused on alternate bearing in Citrus trees that had been experimentally established as fully flowering or nonflowering. We found that the presence of the fruit causes epigenetic changes correlating with the induction of the CcMADS19 floral repressor, which prevents the activation of the floral promoter CiFT2 even in the presence of the floral inductive signals. By contrast, newly emerging shoots display an opposite epigenetic scenario associated with CcMADS19 repression, thereby allowing the activation of CiFT2 the following cold season.
Collapse
Affiliation(s)
- Manuel Agustí
- Instituto Agroforestal Mediterráneo, Universitat Politècnica de València, 46022, Valencia, Spain
| | - Carlos Mesejo
- Instituto Agroforestal Mediterráneo, Universitat Politècnica de València, 46022, Valencia, Spain
| | - Natalia Muñoz-Fambuena
- Instituto Agroforestal Mediterráneo, Universitat Politècnica de València, 46022, Valencia, Spain
| | - Francisco Vera-Sirera
- Instituto de Biología Molecular y Celular de Plantas, CSIC-UPV, 46022, Valencia, Spain
| | - Miguel de Lucas
- Department of Biosciences, Durham University, Stockton Rd, Durham, DH1 3LE, UK
| | - Amparo Martínez-Fuentes
- Instituto Agroforestal Mediterráneo, Universitat Politècnica de València, 46022, Valencia, Spain
| | - Carmina Reig
- Instituto Agroforestal Mediterráneo, Universitat Politècnica de València, 46022, Valencia, Spain
| | - Domingo J Iglesias
- Centro de Citricultura y Producción Vegetal, IVIA-GV, 46113, Moncada, Valencia, Spain
| | - Eduardo Primo-Millo
- Centro de Citricultura y Producción Vegetal, IVIA-GV, 46113, Moncada, Valencia, Spain
| | - Miguel A Blázquez
- Instituto de Biología Molecular y Celular de Plantas, CSIC-UPV, 46022, Valencia, Spain
| |
Collapse
|
43
|
Chen H, Yang S, Shao R. Long non-coding XIST raises methylation of TIMP-3 promoter to regulate collagen degradation in osteoarthritic chondrocytes after tibial plateau fracture. Arthritis Res Ther 2019; 21:271. [PMID: 31815654 PMCID: PMC6902347 DOI: 10.1186/s13075-019-2033-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 10/17/2019] [Indexed: 02/07/2023] Open
Abstract
Background Hypermethylation of gene promoters has been regarded as an epigenetic regulator for gene inactivation in the development of several diseases. In the current study, we aimed to explore how long noncoding RNA X-inactive specific transcript (lncRNA XIST) function in collagen degradation in chondrocytes of osteoarthritis (OA) after tibial plateau fracture by regulating tissue inhibitor of metalloproteinase-3 (TIMP-3) promoter methylation. Methods In silico analysis was used to screen differentially expressed lncRNAs in cartilage tissues of OA. Chondrocytes were then successfully isolated from normal and OA cartilage tissues and identified, with the expressions of lncRNA XIST and TIMP-3 examined. The methylation levels of TIMP-3 promoter were determined by MS-PCR. The binding of lncRNA XIST to DNA methyltransferase and the binding of TIMP-3 promoter to DNA methyltransferase were determined by a series of experiments, including RIP, RNA pull-down, and ChIP assays. Results The differentially expressed lncRNA XIST was determined in OA. In addition, cartilage tissues of OA showed upregulation of lncRNA XIST and downregulation of TIMP-3. LncRNA XIST was primarily localized in the nucleus and was capable of binding to the promoter of TIMP-3. The silencing of lncRNA XIST decreased the methylation levels of TIMP-3 promoter and increased the expressions of TIMP-3, which consequently inhibited collagen degradation in OA chondrocytes. Furthermore, TIMP-3 over-expression reversed the effect of lncRNA XIST on collagen degradation in OA chondrocytes. Conclusion Collectively, lncRNA XIST raises collagen degradation in OA chondrocytes after tibial plateau fracture by accelerating the methylation of TIMP-3 promoter by recruiting DNA methyltransferase.
Collapse
Affiliation(s)
- Hongwei Chen
- Department of Orthopedic Surgery, Yiwu Central Hospital, the Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu, 322000, People's Republic of China
| | - Shengdi Yang
- Department of Hand-Foot Microsurgery, Lanshi Hospital, Lanzhou, 730050, People's Republic of China
| | - Ruyi Shao
- Department of Orthopedics, Zhuji People's Hospital, No. 9, Jianmin Road, Zhuji, 311800, Zhejiang Province, People's Republic of China.
| |
Collapse
|
44
|
Silencing of long noncoding RNA HOXA11-AS inhibits the Wnt signaling pathway via the upregulation of HOXA11 and thereby inhibits the proliferation, invasion, and self-renewal of hepatocellular carcinoma stem cells. Exp Mol Med 2019; 51:1-20. [PMID: 31757938 PMCID: PMC6874533 DOI: 10.1038/s12276-019-0328-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/12/2019] [Accepted: 07/26/2019] [Indexed: 01/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related deaths, but its molecular mechanisms are not yet well characterized. Long noncoding RNAs (lncRNAs) play crucial roles in tumorigenesis, including that of HCC. However, the role of homeobox A11 antisense (HOXA11-AS) in determining HCC stem cell characteristics remains to be explained; hence, this study aimed to investigate the effects of HOXA11-AS on HCC stem cell characteristics. Initially, the expression patterns of HOXA11-AS and HOXA11 in HCC tissues, cells, and stem cells were determined. HCC stem cells, successfully sorted from Hep3B and Huh7 cells, were transfected with short hairpin or overexpression plasmids for HOXA11-AS or HOXA11 overexpression and depletion, with an aim to study the influences of these mediators on the self-renewal, proliferation, migration, and tumorigenicity of HCC stem cells in vivo. Additionally, the potential relationship and the regulatory mechanisms that link HOXA11-AS, HOXA11, and the Wnt signaling pathway were explored through treatment with Dickkopf-1 (a Wnt signaling pathway inhibitor). HCC stem cells showed high expression of HOXA11-AS and low expression of HOXA11. Both HOXA11-AS silencing and HOXA11 overexpression suppressed the self-renewal, proliferation, migration, and tumorigenicity of HCC stem cells in vivo, as evidenced by the decreased expression of cancer stem cell surface markers (CD133 and CD44) and stemness-related transcription factors (Nanog, Sox2, and Oct4). Moreover, silencing HOXA11-AS inactivated the Wnt signaling pathway by decreasing the methylation level of the HOXA11 promoter, thereby inhibiting HCC stem cell characteristics. Collectively, this study suggested that HOXA11-AS silencing exerts an antitumor effect, suppressing HCC development via Wnt signaling pathway inactivation by decreasing the methylation level of the HOXA11 promoter. A long RNA molecule promotes the growth of liver cancer cells through its inhibitory effects on gene regulation. The HOXA11 gene controls cell proliferation and tissue development, and several studies have suggested that HOXA11-AS, an RNA that regulates this gene, may play a role in certain cancers. Researchers led by Min Guo at Hainan General Hospital in Haikou, China, have now obtained evidence linking HOXA11-AS to the growth of hepatocellular carcinoma cells. After determining that this RNA is consistently highly expressed in such cells, the authors demonstrated that it can stimulate cellular proliferation and invasive behavior through its suppressive effects on HOXA11 and other genes. This inhibition results from HOXA11-AS-induced chemical modification of these DNA sequences. The authors hypothesize that this same mechanism could also contribute to growth of other tumor subtypes.
Collapse
|
45
|
Ying Y, Mao Y, Yao M. NLRP3 Inflammasome Activation by MicroRNA-495 Promoter Methylation May Contribute to the Progression of Acute Lung Injury. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:801-814. [PMID: 31734560 PMCID: PMC6861628 DOI: 10.1016/j.omtn.2019.08.028] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/01/2019] [Accepted: 08/17/2019] [Indexed: 12/18/2022]
Abstract
Acute lung injury (ALI) is a pulmonary disorder that causes acute respiratory failure, thus leading to relative high mortality worldwide. However, the molecular mechanisms of ALI remain largely unknown. MicroRNA (miRNA)-dependent control of gene expression at a post-transcriptional level has been recently reported. Herein, we identify a candidate miRNA, miR-495, that affects the progression of ALI. Alveolar macrophages (NR8383) were treated with 1 μg/mL lipopolysaccharide (LPS) to establish a cell-injury model. Combined with the data from western blot, methylation-specific PCR, methylated DNA immunoprecipitation, and chromatin immunoprecipitation assays, NLRP3 inflammasome activation and methylation-dependent repression of miR-495 were found in LPS-exposed NR8383 cells. Dual-luciferase reporter gene assay and miR-495 gain-of-function experiments confirmed that NLRP3 was a target of miR-495. Next, the expression of miR-495 and NLRP3 was overexpressed or silenced to assess their effects on NLRP3 inflammasome activation, alveolar macrophage inflammation, and pyroptosis in vitro. As demonstrated, overexpressed miR-495 alleviated alveolar macrophage inflammation and pyroptosis and inhibited NLRP3 inflammasome activation by negatively regulating the NLRP3 gene. Consistently, elevated miR-495 alleviated lung injury and reduced the neutrophil infiltration and inflammation in rat models of LPS-induced ALI. Taken together, the data in our study demonstrated that methylation of the miR-495 promoter could downregulate miR-495, whose elevation could attenuate the activation of the NLRP3 inflammasome to protect against ALI, which provides novel therapeutic targets for ALI treatment.
Collapse
Affiliation(s)
- Youguo Ying
- Department of Intensive Care Unit, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, P.R. China
| | - Yong Mao
- Department of Intensive Care Unit, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, P.R. China
| | - Min Yao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, P.R. China.
| |
Collapse
|
46
|
Ritter N, Ali T, Kopitchinski N, Schuster P, Beisaw A, Hendrix DA, Schulz MH, Müller-McNicoll M, Dimmeler S, Grote P. The lncRNA Locus Handsdown Regulates Cardiac Gene Programs and Is Essential for Early Mouse Development. Dev Cell 2019; 50:644-657.e8. [PMID: 31422919 DOI: 10.1016/j.devcel.2019.07.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 05/10/2019] [Accepted: 07/10/2019] [Indexed: 11/29/2022]
Abstract
Precisely controlled gene regulatory networks are required during embryonic development to give rise to various structures, including those of the cardiovascular system. Long non-coding RNA (lncRNA) loci are known to be important regulators of these genetic programs. We have identified a novel and essential lncRNA locus Handsdown (Hdn), active in early heart cells, and show by genetic inactivation that it is essential for murine development. Hdn displays haploinsufficiency for cardiac development as Hdn-heterozygous adult mice exhibit hyperplasia in the right ventricular wall. Transcriptional activity of the Hdn locus, independent of its RNA, suppresses its neighboring gene Hand2. We reveal a switch in a topologically associated domain in differentiation of the cardiac lineage, allowing the Hdn locus to directly interact with regulatory elements of the Hand2 locus.
Collapse
Affiliation(s)
- Nicole Ritter
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Tamer Ali
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Faculty of Science, Benha University, Benha 13518, Egypt
| | - Nina Kopitchinski
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Peggy Schuster
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Arica Beisaw
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - David A Hendrix
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA; School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, OR, USA
| | - Marcel H Schulz
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; German Center for Cardiovascular Regeneration (DZHK), Partner site Rhein-Main, 60590 Frankfurt am Main, Germany
| | - Michaela Müller-McNicoll
- RNA Regulation Group, Institute of Cell Biology and Neuroscience, Goethe University, Max-von-Laue-Strasse 13, 60438 Frankfurt am Main, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; German Center for Cardiovascular Regeneration (DZHK), Partner site Rhein-Main, 60590 Frankfurt am Main, Germany
| | - Phillip Grote
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
47
|
Denisenko O, Mar D, Trawczynski M, Bomsztyk K. Chromatin changes trigger laminin genes dysregulation in aging kidneys. Aging (Albany NY) 2019; 10:1133-1145. [PMID: 29846172 PMCID: PMC5990391 DOI: 10.18632/aging.101453] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
Abstract
Dysregulation of gene expression is a hallmark of aging. We examined epigenetic mechanisms that mediate aberrant expression of laminin genes in aging rat kidneys. In old animals, no alterations were found in the levels of abundant laminin mRNAs, whereas Lama3, b3, and c2 transcripts were increased compared to young animals. Lamc2 showed the strongest changes at the mRNA and protein levels. Lamc2 upregulation was transcriptional, as indicated by the elevated RNA polymerase II density at the gene. Furthermore, aging is associated with the loss of H3K27m3 and 5mC silencing modifications at the Lamc2 gene. Western blot analysis revealed no changes in cellular levels of H3K27m3 and cognate enzyme Ezh2 in old kidneys. Thus, the decrease in H3K27m3 at Lamc2 resulted from the re-distribution of this mark among genomic sites. Studies in kidney cells in vitro showed that reducing H3K27m3 density with Ezh2 inhibitor had no effect on Lamc2 expression, suggesting that this modification plays little role in gene upregulation in aging kidney. In contrast, treatment with DNA methylation inhibitor 2'-deoxy-5-azacytidine was sufficient to upregulate Lamc2 gene. We suggest that the loss of 5mC at silenced laminin genes drives their de-repression during aging, contributing to the age-related decline in renal function.
Collapse
Affiliation(s)
- Oleg Denisenko
- Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Daniel Mar
- Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | | | - Karol Bomsztyk
- Department of Medicine, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
48
|
Li W, Huang K, Wen F, Cui G, Guo H, He Z, Zhao S. LINC00184 silencing inhibits glycolysis and restores mitochondrial oxidative phosphorylation in esophageal cancer through demethylation of PTEN. EBioMedicine 2019; 44:298-310. [PMID: 31201145 PMCID: PMC6606559 DOI: 10.1016/j.ebiom.2019.05.055] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Total lesion glycolysis has been reported to be a satisfactory predictor of survival in patients with locally advanced esophageal cancer (EC). The aim of the present study is to investigate the function of long intergenic non-protein coding RNA 184 (LINC00184) on the EC cell glycolysis and mitochondrial oxidative phosphorylation (OXPHOS). METHODS The expression of LINC00184 was determined to be highly expressed and PTEN was poorly expressed in EC tissues and cells by RT-qPCR. In order to evaluate the effects of LINC00184 on cellular process in vitro and in vivo, gain- and loss-of-function approaches were performed to alter the expression of LINC00184 and PTEN in EC cells. RESULTS Silencing of LINC00184 was observed to inhibit the proliferation, migration, invasion, colony formation, and glycolysis of EC cells and tumour growth, while the mitochondrial OXPHOS was restored. By recruiting DNMT1, LINC00184 enhanced the promoter methylation of PTEN. Inhibition of PTEN promoter methylation suppressed EC glycolysis, whereas, improved mitochondrial OXPHOS. Mechanically, LINC00184 modulated glycolysis and mitochondrial OXPHOS in EC cells through induction of the Akt phosphorylation. After blockage of Akt signaling pathway by an Akt inhibitor, LY294002, the regulatory effects of LINC00184 on the glycolysis and mitochondrial OXPHOS of EC cells were reversed. CONCLUSION Taken together, the LINC00184/PTEN/Akt axis mediates glycolysis and mitochondrial OXPHOS in EC cells. This study highlighted a potential intervention target for treating EC.
Collapse
Affiliation(s)
- Weihao Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Kai Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Fengbiao Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Guanghui Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Haizhou Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zhanfeng He
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Song Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
49
|
Li H, Han S, Sun Q, Yao Y, Li S, Yuan C, Zhang B, Jing B, Wu J, Song Y, Wang H. Long non-coding RNA CDKN2B-AS1 reduces inflammatory response and promotes cholesterol efflux in atherosclerosis by inhibiting ADAM10 expression. Aging (Albany NY) 2019; 11:1695-1715. [PMID: 30926762 PMCID: PMC6461186 DOI: 10.18632/aging.101863] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/06/2019] [Indexed: 06/09/2023]
Abstract
INTRODUCTION Long non-coding RNAs (lncRNAs) play key roles in the development of atherosclerosis through the inflammatory pathway. This study aimed to investigate the role of lncRNA cyclin-dependent kinase inhibitor 2B antisense RNA 1 (CDKN2B-AS1) in atherosclerosis via its function in A disintegrin and metalloprotease 10 (ADAM10). METHODS Initially, the expression of CDKN2B-AS1 and ADAM10 in atherosclerotic plaque tissues and THP-1 macrophage-derived foam cells was determined, after which the cholesterol efflux rate of macrophages was calculated. Interaction between CDKN2B-AS1 and ADAM10 was analyzed, after which, expression of CDKN2B-AS1 and ADAM10 were altered to explore their effects on inflammatory response and cholesterol efflux. The aforementioned findings were further intended to be validated by the atherosclerosis mouse model experiments. RESULTS Atherosclerotic plaque tissue and THP-1 macrophage-derived foam cells exhibited downregulated CDKN2B-AS1 and upregulated ADAM10. Upon overexpressing CDKN2B-AS1 or silencing ADAM10, lipid accumulation was reduced and cholesterol efflux was increased. CDKN2B-AS1 located in the nucleus could bind to DNA methyltransferase 1 (DNMT1) to enhance methylation of ADAM10 promoter, leading to suppressed atherosclerotic inflammatory response and promoted cholesterol efflux. CONCLUSION Altogether, lncRNA CDKN2B-AS1 can inhibit the transcription of ADAM10 via DNMT1-mediated ADAM10 DNA methylation, consequently preventing inflammatory response of atherosclerosis and promoting cholesterol efflux.
Collapse
Affiliation(s)
- Haocheng Li
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
- Equal contribution
| | - Song Han
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
- Equal contribution
| | - Qingfeng Sun
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Ye Yao
- Department of Cardiac Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Shiyong Li
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Chao Yuan
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Bo Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Bao Jing
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Jia Wu
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Ye Song
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| | - Haiyang Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P. R. China
| |
Collapse
|
50
|
Abstract
Chromatin immunoprecipitation (ChIP) is a powerful and sensitive technique that is widely used to study DNA-protein interactions. It enables an unbiased genome-wide analysis of transcriptional changes during several biological processes including cellular differentiation. Here, we describe a step-by-step protocol to identify histone modifications, transcription factor, and co-factor binding to chromatin in skeletal myoblasts. We discuss critical steps during cell harvesting, sonication, and immunoprecipitation and provide notes to evade common pitfalls.
Collapse
Affiliation(s)
- Vinay Kumar Rao
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
| | - Shilpa Rani Shankar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reshma Taneja
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|