1
|
Zheng Y, Zhang TN, Hao PH, Yang N, Du Y. Histone deacetylases and their inhibitors in kidney diseases. Mol Ther 2025:S1525-0016(25)00300-4. [PMID: 40263937 DOI: 10.1016/j.ymthe.2025.04.026] [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: 01/27/2025] [Revised: 03/18/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
Histone deacetylases (HDACs) have emerged as key regulators in the pathogenesis of various kidney diseases. This review explores recent advancements in HDAC research, focusing on their role in kidney development and their critical involvement in the progression of chronic kidney disease (CKD), acute kidney injury (AKI), autosomal dominant polycystic kidney disease (ADPKD), and diabetic kidney disease (DKD). It also discusses the therapeutic potential of HDAC inhibitors in treating these conditions. Various HDAC inhibitors have shown promise by targeting specific HDAC isoforms and modulating a range of biological pathways. Their protective effects include modulation of apoptosis, autophagy, inflammation, and fibrosis, underscoring their broad therapeutic potential for kidney diseases. However, further research is essential to improve the selectivity of HDAC inhibitors, minimize toxicity, overcome drug resistance, and enhance their pharmacokinetic properties. This review offers insights to guide future research and prevention strategies for kidney disease management.
Collapse
Affiliation(s)
- Yue Zheng
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Peng-Hui Hao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Yue Du
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Zhang L, Yang C, Liu X, He D, Lin T, Zhang Y, Wei G, Zhang D. Renal dysplasia development and chronic kidney disease. Pediatr Res 2025:10.1038/s41390-025-03950-0. [PMID: 40000855 DOI: 10.1038/s41390-025-03950-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 12/13/2024] [Accepted: 02/02/2025] [Indexed: 02/27/2025]
Abstract
Renal dysplasia is a common congenital birth defect in childhood, caused by fetal genetic defects, epigenetic modification disorders, or environmental factors. Maternal malnutrition, placental insufficiency, and exposure to harmful substances such as alcohol, angiotensin-converting enzyme inhibitors, and cocaine during pregnancy increase the risk of fetal renal dysplasia. The pathogenesis of this disease involves abnormal formation of renal units, leading to structural and functional abnormalities of the kidney. If left untreated, renal dysplasia can progress to chronic kidney disease (CKD) in children. This review explores the etiology and pathogenesis of renal dysplasia, emphasizing the intrinsic link between renal dysplasia and CKD through various pathological pathways. Additionally, we propose potential therapeutic agents targeting these mechanisms. We also highlight future research directions to further understand and address this issue. We hope this review will deepen clinicians' understanding of renal dysplasia and promote further laboratory research in this area. IMPACT: 1. This review comprehensively summarizes and elucidates the complex relationship between renal dysplasia and chronic kidney disease (CKD) based on previous research, offering new directions for related studies. 2. It expands upon conservative treatment approaches for renal dysplasia, providing more clinical options for therapeutic intervention.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pediatric Surgery, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Chunjiang Yang
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Xing Liu
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
- Department of Urology Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Dawei He
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
- Department of Urology Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Lin
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
- Department of Urology Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Guanghui Wei
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
- Department of Urology Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Deying Zhang
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China.
- Department of Urology Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Zeng X, Yuan X, Liao H, Wei Y, Wu Q, Zhu X, Li Q, Chen S, Hu M. The miR-665/SOST Axis Regulates the Phenotypes of Bone Marrow Mesenchymal Stem Cells and Osteoporotic Symptoms in Female Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2059-2075. [PMID: 39461772 DOI: 10.1016/j.ajpath.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/18/2024] [Accepted: 07/26/2024] [Indexed: 10/29/2024]
Abstract
Osteoporosis is a common degenerative skeletal disease among older people, especially postmenopausal women. Bone marrow mesenchymal stem cells (BMSCs), the progenitors of osteoblasts, are essential to the pathophysiology of osteoporosis. Herein, targeting miRNAs with differential expression in dysfunctional BMSCs was accomplished by bioinformatics analysis based on public databases. Target mRNAs were predicted and applied for signaling pathway and function enrichment annotations. In vitro and in vivo effects of selected miRNA on BMSC proliferation and osteogenesis were investigated, the putative binding between selected miRNA and predicted target mRNA was verified, and the co-effects of the miRNA/mRNA axis on BMSCs were determined. miRNA 665 (miR-665) was down-regulated in osteoporotic BMSCs compared with normal BMSCs and elevated in BMSCs experiencing osteogenic differentiation. In BMSCs, miR-665 overexpression promoted cell proliferation and osteogenic differentiation. miR-665 targeted the Wnt signaling inhibitor sclerostin (SOST) and inhibited SOST mRNA and protein expression. SOST overexpression inhibited BMSC cell proliferation and osteogenic differentiation. When co-transduced to BMSCs, SOST knockdown significantly reversed the effects of miR-665 on BMSCs. In ovariectomy (OVX)-induced osteoporosis model mice, OVX remarkably decreased bone mass, whereas miR-665 overexpression partially improved OVX-induced bone mass loss. miR-665 was down-regulated in osteoporotic BMSCs and up-regulated in osteogenically differentiated BMSCs. In conclusion, the miR-665/SOST axis modulates BMSC proliferation, osteogenic differentiation, and OVX-induced osteoporosis in mice, possibly through Wnt signaling.
Collapse
Affiliation(s)
- Xingxing Zeng
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Xianyu Yuan
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Hongchun Liao
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Yongfang Wei
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Qinxuan Wu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Xi Zhu
- Health Management, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Qingqing Li
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Shijie Chen
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Minghua Hu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China.
| |
Collapse
|
4
|
Liang L, Huang Y, Chen L, Shi Z, Wang H, Zhang T, Li Z, Mi J, Fan T, Lu Y, Chen F, Huang W, Hu K. Radioprotective efficacy of Astilbin in mitigating radiation-induced lung injury through inhibition of p53 acetylation. ENVIRONMENTAL TOXICOLOGY 2023; 38:2967-2980. [PMID: 37598414 DOI: 10.1002/tox.23931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/19/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023]
Abstract
Radiation-induced lung injury (RILI) is a common side effect in thoracic tumor patients undergoing radiotherapy. At present, there is no ideal radio-protective agent which is widely used in RILI treatment. Astilbin (AST), a bioactive flavonoid, exhibits various biological effects, including anti-inflammatory, antioxidant, and anti-fibrotic activities, which partly result from reducing oxidative stress and inflammation in various pathogenic conditions. However, the protective efficacy of AST to ameliorate RILI has not been reported. In this study, we employed network pharmacology, RNA sequencing, and experimental evaluation to reveal the effects and pharmacological mechanism of AST to treat RILI in vivo and in vitro. We observed that AST reduced radiation-induced apoptosis, DNA damage, inflammatory reactions, and the reactive oxygen species (ROS) level in human normal lung epithelial cells BEAS-2B. Further study showed that AST treatment significantly ameliorated RILI by reducing the radiation-induced pathology changes and inflammatory reaction of lung tissue in C57BL/6J mice. Mechanistically, the expression of epithelial-mesenchymal transition (EMT) markers and radiation-triggered acetylation of the p53 protein were alleviated by AST treatment. Furthermore, AST alleviated the acetylation of p53 after intervention of Trichostatin A (TSA). Our data indicate that AST can alleviate RILI by inhibiting inflammatory reactions and the EMT process through decreasing the expression of p53 acetylation. In conclusion, our study suggests that AST has great potential to be a new protective and therapeutic compound for RILI.
Collapse
Affiliation(s)
- Lixing Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Yaqin Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Liuyin Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Zhiling Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Housheng Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Tingting Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Zhixun Li
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Jinglin Mi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Ting Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Yushuang Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Fuli Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Weimei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Kai Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| |
Collapse
|
5
|
Sibony-Benyamini H, Aamar E, Enshell-Seijffers D. Hdac1 and Hdac2 regulate the quiescent state and survival of hair-follicle mesenchymal niche. Nat Commun 2023; 14:4820. [PMID: 37563109 PMCID: PMC10415406 DOI: 10.1038/s41467-023-40573-7] [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] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/02/2023] [Indexed: 08/12/2023] Open
Abstract
While cell division is essential for self-renewal and differentiation of stem cells and progenitors, dormancy is required to maintain the structure and function of the stem-cell niche. Here we use the hair follicle to show that during growth, the mesenchymal niche of the hair follicle, the dermal papilla (DP), is maintained quiescent by the activity of Hdac1 and Hdac2 in the DP that suppresses the expression of cell-cycle genes. Furthermore, Hdac1 and Hdac2 in the DP promote the survival of DP cells throughout the hair cycle. While during growth and regression this includes downregulation of p53 activity and the control of p53-independent programs, during quiescence, this predominantly involves p53-independent mechanisms. Remarkably, Hdac1 and Hdac2 in the DP during the growth phase also participate in orchestrating the hair cycle clock by maintaining physiological levels of Wnt signaling in the vicinity of the DP. Our findings not only provide insight into the molecular mechanism that sustains the function of the stem-cell niche in a persistently changing microenvironment, but also unveil that the same mechanism provides a molecular toolbox allowing the DP to affect and fine tune the microenvironment.
Collapse
Affiliation(s)
- Hadas Sibony-Benyamini
- The Laboratory of Developmental Biology, The Azrieli Faculty of Medicine, Bar Ilan University, 8 Henrietta Szold, Safed, Israel
| | - Emil Aamar
- The Laboratory of Developmental Biology, The Azrieli Faculty of Medicine, Bar Ilan University, 8 Henrietta Szold, Safed, Israel
| | - David Enshell-Seijffers
- The Laboratory of Developmental Biology, The Azrieli Faculty of Medicine, Bar Ilan University, 8 Henrietta Szold, Safed, Israel.
| |
Collapse
|
6
|
Nuclear SPHK2/S1P induces oxidative stress and NLRP3 inflammasome activation via promoting p53 acetylation in lipopolysaccharide-induced acute lung injury. Cell Death Dis 2023; 9:12. [PMID: 36653338 PMCID: PMC9847446 DOI: 10.1038/s41420-023-01320-5] [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: 11/11/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023]
Abstract
A bulk of evidence identified that macrophages, including resident alveolar macrophages and recruited macrophages from the blood, played an important role in the pathogenesis of acute respiratory distress syndrome (ARDS). However, the molecular mechanisms of macrophages-induced acute lung injury (ALI) by facilitating oxidative stress and inflammatory responses remain unclear. Herein, we noticed that the levels of mitochondrial reactive oxygen species (mtROS), SPHK2 and activated NLRP3 inflammasome were higher in peripheral blood mononuclear cells (PBMCs) of ARDS patients than that in healthy volunteers. Similar observations were recapitulated in LPS-treated RAW264.7 and THP-1 cells. After exposure to LPS, the SPHK2 enzymatic activity, NLRP3 inflammasome activation and mtROS were significantly upregulated in macrophages. Moreover, knockdown SPHK2 via shRNA or inhibition SPHK2 could prominently decrease LPS-induced M1 macrophage polarization, oxidative stress and NLRP3 inflammasome activation. Further study indicated that upregulated SPHK2 could increase nuclear sphingosine-1-phosphate (S1P) levels and then restrict the enzyme activity of HDACs to facilitate p53 acetylation. Acetylation of p53 reinforced its binding to the specific region of the NLRP3 promoter and drove expression of NLRP3. In the in vivo experiments, it was also observed that treating with Opaganib (ABC294640), a specific SPHK2 inhibitor, could observably alleviate LPS-induced ALI, evidencing by lowered infiltration of inflammatory cells, increased M2 macrophages polarization and reduced oxidative damage in lung tissues. Besides, SPHK2 inhibition can also decrease the accumulation of acetylated p53 protein and the activation of NLRP3 inflammasome. Taken together, our results demonstrated for the first time that nuclear S1P can regulate the acetylation levels of non-histone protein through affecting HDACs enzyme activities, linking them to oxidative stress and inflammation in response to environmental signals. These data provide a theoretical basis that SPHK2 may be an effective therapeutic target of ARDS.
Collapse
|
7
|
Xu L, Xie H, Hu S, Zhao X, Han M, Liu Q, Feng P, Wang W, Li C. HDAC3 inhibition improves urinary-concentrating defect in hypokalaemia by promoting AQP2 transcription. Acta Physiol (Oxf) 2022; 234:e13802. [PMID: 35178888 DOI: 10.1111/apha.13802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 11/27/2022]
Abstract
AIM This study investigated whether enhanced histone acetylation, achieved by inhibiting histone deacetylases (HDACs), could prevent decreased aquaporin-2 (AQP2) expression during hypokalaemia. METHODS Male Wistar rats were fed a potassium-free diet with or without 4-phenylbutyric acid (4-PBA) or the selective HDAC3 inhibitor RGFP966 for 4 days. Primary renal inner medullary collecting duct (IMCD) cells and immortalized mouse cortical collecting duct (mpkCCD) cells were cultured in potassium-deprivation medium with or without HDAC inhibitors. RESULTS 4-PBA increased the levels of AQP2 mRNA and protein in the kidney inner medullae in hypokalaemic (HK) rats, which was associated with decreased urine output and increased urinary osmolality. The level of acetylated H3K27 (H3K27ac) protein was decreased in the inner medullae of HK rat kidneys; this decrease was mitigated by 4-PBA. The H3K27ac levels were decreased in IMCD and mpkCCD cells cultured in potassium-deprivation medium. Decreased H3K27ac in the Aqp2 promoter region was associated with reduced Aqp2 mRNA levels. HDAC3 protein expression was upregulated in mpkCCD and IMCD cells in response to potassium deprivation, and the binding of HDAC3 to the Aqp2 promoter was also increased. RGFP966 increased the levels of H3K27ac and AQP2 proteins and enhanced binding between H3K27ac and AQP2 in mpkCCD cells. Furthermore, RGFP966 reversed the hypokalaemia-induced downregulation of AQP2 and H3K27ac and alleviated polyuria in rats. RGFP966 increased interstitial osmolality in the kidney inner medullae of HK rats but did not affect urinary cAMP levels. CONCLUSION HDAC inhibitors prevented the downregulation of AQP2 induced by potassium deprivation, probably by enhancing H3K27 acetylation.
Collapse
Affiliation(s)
- Long Xu
- Institute of Hypertension Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
- Department of Physiology Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
| | - Haixia Xie
- Institute of Hypertension Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
- Department of Physiology Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
| | - Shan Hu
- Institute of Hypertension Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
- The School of Basic Medicine Guangzhou University of Chinese Medicine Guangzhou China
| | - Xiaoduo Zhao
- Institute of Hypertension Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
- Department of Pathophysiology Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
| | - Mengke Han
- Institute of Hypertension Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
- Department of Physiology Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
| | - Qiaojuan Liu
- Institute of Hypertension Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
- Department of Physiology Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
| | - Pinning Feng
- Department of Clinical Laboratory The First Affiliated Hospital Sun Yat‐sen University Guangzhou China
| | - Weidong Wang
- Institute of Hypertension Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
- Department of Pathophysiology Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
- Department of Nephrology The Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen China
| | - Chunling Li
- Institute of Hypertension Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
- Department of Physiology Zhongshan School of Medicine Sun Yat‐sen University Guangzhou China
| |
Collapse
|
8
|
MicroRNA-495 suppresses pre-eclampsia via activation of p53/PUMA axis. Cell Death Dis 2022; 8:132. [PMID: 35338123 PMCID: PMC8956677 DOI: 10.1038/s41420-022-00874-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 01/20/2022] [Accepted: 02/08/2022] [Indexed: 11/29/2022]
Abstract
Linkage between microRNAs (miRNAs) and pre-eclampsia (PE) has been documented. Here, we focused on miR-495 in PE and its underlying mechanism in regulation of trophoblast cells. Expression of miR-495, HDAC2, p53 and PUMA was determined in collected placental tissue samples. Loss- and gain-function was performed to determine the roles of miR-495, HDAC2, p53, and PUMA in biological processes of HTR8/SVneo cells and primary trophoblast cells. The relationships among miR-495, HDAC2, and p53 were pinpointed. PE patients presented with higher expression of miR-495, p53, and PUMA in placental tissues, but lower HDAC2. miR-495 negatively targeted HDAC2 expression. HDAC2 suppressed p53 expression via deacetylation. Overexpression of miR-495, p53, or PUMA inhibited biological properties of HTR8/SVneo cells and primary trophoblast cells, while opposite trends were observed in response to oe-HDAC2. In conclusion, miR-495 knockdown can suppress p53/PUMA axis by targeting HDAC2 to enhance biological behaviors of trophoblast cells, which may prevent occurrence of PE.
Collapse
|
9
|
Hyndman KA, Crossman DK. Kidney cell type-specific changes in the chromatin and transcriptome landscapes following epithelial Hdac1 and Hdac2 knockdown. Physiol Genomics 2022; 54:45-57. [PMID: 34890513 PMCID: PMC8791845 DOI: 10.1152/physiolgenomics.00102.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/21/2021] [Accepted: 12/08/2021] [Indexed: 02/03/2023] Open
Abstract
Recent studies have identified at least 20 different kidney cell types based upon chromatin structure and gene expression. Histone deacetylases (HDACs) are epigenetic transcriptional repressors via deacetylation of histone lysines resulting in inaccessible chromatin. We reported that kidney epithelial HDAC1 and HDAC2 activity is critical for maintaining a healthy kidney and preventing fluid-electrolyte abnormalities. However, to what extent does Hdac1/Hdac2 knockdown affect chromatin structure and subsequent transcript expression in the kidney? To answer this question, we used single nucleus assay for transposase-accessible chromatin-sequencing (snATAC-seq) and snRNA-seq to profile kidney nuclei from male and female, control, and littermate kidney epithelial Hdac1/Hdac2 knockdown mice. Hdac1/Hdac2 knockdown resulted in significant changes in the chromatin structure predominantly within the promoter region of gene loci involved in fluid-electrolyte balance such as the aquaporins, with both increased and decreased accessibility captured. Moreover, Hdac1/Hdac2 knockdown resulted different gene loci being accessible with a corresponding increased transcript number in the kidney, but among all mice only 24%-30% of chromatin accessibility agreed with transcript expression (e.g., open chromatin and increased transcript). To conclude, although chromatin structure does affect transcription, ∼70% of the differentially expressed genes cannot be explained by changes in chromatin accessibility and HDAC1/HDAC2 had a minimal effect on these global patterns. Yet, the genes that are targets of HDAC1 and HDAC2 are critically important for maintaining kidney function.
Collapse
Affiliation(s)
- Kelly A Hyndman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David K Crossman
- The UAB Genomics Core Facility, Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
10
|
Liu J, Li B, Li W, Pan T, Diao Y, Wang F. 6-Shogaol Inhibits Oxidative Stress-Induced Rat Vascular Smooth Muscle Cell Apoptosis by Regulating OXR1-p53 Axis. Front Mol Biosci 2022; 9:808162. [PMID: 35174215 PMCID: PMC8841977 DOI: 10.3389/fmolb.2022.808162] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/04/2022] [Indexed: 11/22/2022] Open
Abstract
Apoptosis of vascular smooth muscle cells (VSMCs) is closely related to the pathogenesis of cardiovascular diseases, and oxidative stress is an important cause of VSMCs' death. Inhibiting VSMCs apoptosis is an effective preventive strategy in slowing down the development of cardiovascular disease, especially for atherosclerosis. In this study, we found that oxidation resistance protein 1 (OXR1), a crucial participator for responding to oxidative stress, could modulate the expression of p53, the key regulator of cell apoptosis. Our results revealed that oxidative stress promoted VSMCs apoptosis by overexpression of the OXR1-p53 axis, and 6-shogaol (6S), a major biologically active compound in ginger, could effectively attenuate cell death by preventing the upregulated expression of the OXR1-p53 axis. Quantitative proteomics analysis revealed that the degradation of p53 mediated by OXR1 might be related to the enhanced assembly of SCF ubiquitin ligase complexes, which is reported to closely relate to the modification of ubiquitination or neddylation and subsequent degradation of p53.
Collapse
Affiliation(s)
- Jing Liu
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China
- Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian, China
| | - Bin Li
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China
- Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian, China
| | - Wenlian Li
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Taowen Pan
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China
- Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian, China
| | - Yunpeng Diao
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China
- Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian, China
| | - Fangjun Wang
- Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| |
Collapse
|
11
|
Li H, Kurtzeborn K, Kupari J, Gui Y, Siefker E, Lu B, Mätlik K, Olfat S, Montaño-Rodríguez AR, Huh SH, Costantini F, Andressoo JO, Kuure S. Postnatal prolongation of mammalian nephrogenesis by excess fetal GDNF. Development 2021; 148:268366. [PMID: 34032268 PMCID: PMC8180252 DOI: 10.1242/dev.197475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/26/2021] [Indexed: 01/21/2023]
Abstract
Nephron endowment, defined during the fetal period, dictates renal and related cardiovascular health throughout life. We show here that, despite its negative effects on kidney growth, genetic increase of GDNF prolongs the nephrogenic program beyond its normal cessation. Multi-stage mechanistic analysis revealed that excess GDNF maintains nephron progenitors and nephrogenesis through increased expression of its secreted targets and augmented WNT signaling, leading to a two-part effect on nephron progenitor maintenance. Abnormally high GDNF in embryonic kidneys upregulates its known targets but also Wnt9b and Axin2, with concomitant deceleration of nephron progenitor proliferation. Decline of GDNF levels in postnatal kidneys normalizes the ureteric bud and creates a permissive environment for continuation of the nephrogenic program, as demonstrated by morphologically and molecularly normal postnatal nephron progenitor self-renewal and differentiation. These results establish that excess GDNF has a bi-phasic effect on nephron progenitors in mice, which can faithfully respond to GDNF dosage manipulation during the fetal and postnatal period. Our results suggest that sensing the signaling activity level is an important mechanism through which GDNF and other molecules contribute to nephron progenitor lifespan specification. Summary: Dosage of neurotropic factor GDNF regulates nephron progenitors and in utero growth factor augmentation can extend postnatal lifespan and differentiation of nephron progenitors.
Collapse
Affiliation(s)
- Hao Li
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Kristen Kurtzeborn
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Jussi Kupari
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Yujuan Gui
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Edward Siefker
- Department of Developmental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Benson Lu
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Kärt Mätlik
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Soophie Olfat
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Ana R Montaño-Rodríguez
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Sung-Ho Huh
- Department of Developmental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Franklin Costantini
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Jaan-Olle Andressoo
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,GM-unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
12
|
Abstract
Histone deacetylases (HDACs) are part of the epigenetic machinery that regulates transcriptional processes. The current paradigm is that HDACs silence gene expression via regulation of histone protein lysine deacetylation, or by forming corepressor complexes with transcription factors. However, HDACs are more than just nuclear proteins, and they can interact and deacetylate a growing number of nonhistone proteins to regulate cellular function. Cancer-field studies have shown that deranged HDAC activity results in uncontrolled proliferation, inflammation, and fibrosis; all pathologies that also may occur in kidney disease. Over the past decade, studies have emerged suggesting that HDAC inhibitors may prevent and potentially treat various models of acute kidney injury. This review focuses on the physiology of kidney HDACs and highlights the recent advances using HDAC inhibitors to potentially treat kidney disease patients.
Collapse
Affiliation(s)
- Kelly A Hyndman
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
13
|
Effects of Environmental Conditions on Nephron Number: Modeling Maternal Disease and Epigenetic Regulation in Renal Development. Int J Mol Sci 2021; 22:ijms22084157. [PMID: 33923831 PMCID: PMC8073167 DOI: 10.3390/ijms22084157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/08/2021] [Accepted: 04/15/2021] [Indexed: 12/14/2022] Open
Abstract
A growing body of evidence suggests that low nephron numbers at birth can increase the risk of chronic kidney disease or hypertension later in life. Environmental stressors, such as maternal malnutrition, medication and smoking, can influence renal size at birth. Using metanephric organ cultures to model single-variable environmental conditions, models of maternal disease were evaluated for patterns of developmental impairment. While hyperthermia had limited effects on renal development, fetal iron deficiency was associated with severe impairment of renal growth and nephrogenesis with an all-proximal phenotype. Culturing kidney explants under high glucose conditions led to cellular and transcriptomic changes resembling human diabetic nephropathy. Short-term high glucose culture conditions were sufficient for long-term alterations in DNA methylation-associated epigenetic memory. Finally, the role of epigenetic modifiers in renal development was tested using a small compound library. Among the selected epigenetic inhibitors, various compounds elicited an effect on renal growth, such as HDAC (entinostat, TH39), histone demethylase (deferasirox, deferoxamine) and histone methyltransferase (cyproheptadine) inhibitors. Thus, metanephric organ cultures provide a valuable system for studying metabolic conditions and a tool for screening for epigenetic modifiers in renal development.
Collapse
|
14
|
Wei Y, Wang Z, Wei L, Li S, Qiu X, Liu C. MicroRNA-874-3p promotes testosterone-induced granulosa cell apoptosis by suppressing HDAC1-mediated p53 deacetylation. Exp Ther Med 2021; 21:359. [PMID: 33732332 PMCID: PMC7903439 DOI: 10.3892/etm.2021.9790] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
MicroRNA (miR)-874-3p is a newly identified miRNA that is involved in several pathological processes, including cancer, myocardial infarction, bone formation and erectile dysfunction. However, the role of miR-874-3p in polycystic ovary syndrome (PCOS) and granulosa cell (GC) apoptosis is not completely understood. The present study investigated the expression profile of miR-874-3p in PCOS by reverse transcription- quantitative PCR and the GC apoptosis by flow cytometry analysis. miR-874-3p expression was significantly upregulated in GCs isolated from patients with PCOS compared with patients without PCOS. In addition, miR-874-3p expression was positively correlated with GC apoptosis and testosterone levels in both patients with PCOS and patients without PCOS. Therefore, the present study also aimed to investigate the effects of miR-874-3p on testosterone-induced GC apoptosis. Compared with vehicle-treated GCs, miR-874-3p expression levels were significantly increased in testosterone-treated GCs, which was inhibited by the androgen receptor antagonist flutamide. GCs were transfected with either the miR-874-3p mimic or a miR-874-3p inhibitor. Compared with the control group, miR-874-3p mimic significantly enhanced GC apoptosis, whereas miR-874-3p inhibitor significantly decreased GC apoptosis. Moreover, histone deacetylase (HDAC) activity and HDAC1 expression levels were decreased in testosterone-treated GCs compared with vehicle-treated GCs. HDAC1 overexpression significantly attenuated the proapoptotic effects of testosterone. Additionally, miR-874-3p mimic and inhibitor significantly decreased and increased HDAC1 expression levels, respectively, compared with the control group. miR-874-3p inhibitor failed to attenuate HDAC1 overexpression-induced GC apoptosis. Furthermore, compared with the control group, testosterone treatment notably increased p53 expression and acetylation. Compared with the control group, western blotting analysis showed that miR-874-3p mimic notably increased p53 expression and acetylation, whereas miR-874-3p inhibitor markedly decreased p53 expression and acetylation. However, miR-874-3p inhibitor did not further decrease p53 acetylation and expression in cell overexpressing HDAC1. Collectively, the results of the present study indicated that miR-874-3p was upregulated in PCOS and promoted testosterone-induced GC apoptosis by suppressing HDAC1-mediated p53 deacetylation. Therefore, the present study improved the current understanding of the pathogenesis of PCOS and GC apoptosis.
Collapse
Affiliation(s)
- Youhua Wei
- Department of Medical Heredity and Prenatal Screening, Zaozhuang Maternal and Child Health Care Hospital, Zaozhuang, Shandong 277100, P.R. China
| | - Zhijun Wang
- Department of Medical Heredity and Prenatal Screening, Zaozhuang Maternal and Child Health Care Hospital, Zaozhuang, Shandong 277100, P.R. China
| | - Li Wei
- Department of Medical Heredity and Prenatal Screening, Zaozhuang Maternal and Child Health Care Hospital, Zaozhuang, Shandong 277100, P.R. China
| | - Shen Li
- Department of Obstetrics and Gynecology, Zaozhuang Maternal and Child Health Care Hospital, Zaozhuang, Shandong 277100, P.R. China
| | - Xuemei Qiu
- Department of Reproductive Center, Zaozhuang Maternal and Child Health Care Hospital, Zaozhuang, Shandong 277100, P.R. China
| | - Chengwen Liu
- Department of Obstetrics and Gynecology, Zaozhuang Maternal and Child Health Care Hospital, Zaozhuang, Shandong 277100, P.R. China
| |
Collapse
|
15
|
Bai L, Sun H, Jiang W, Yang L, Liu G, Zhao X, Hu H, Wang J, Gao S. DNA methylation and histone acetylation are involved in Wnt10b expression during the secondary hair follicle cycle in Angora rabbits. J Anim Physiol Anim Nutr (Berl) 2021; 105:599-609. [PMID: 33404138 DOI: 10.1111/jpn.13481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/03/2020] [Accepted: 11/02/2020] [Indexed: 12/25/2022]
Abstract
Secondary hair follicles (SHFs) in the Angora rabbit exhibit classic cyclic hair development, but the multiple molecular signals involved in hair cycling are yet to be explored in detail. In the present study, we investigated the expression pattern, methylation and histone H3 acetylation status of Wnt10b, as a molecular signal participating in hair cycling, during the SHF cycle in the Angora rabbit. Expression of Wnt10b at the anagen phase was significantly higher than that at both the telogen and catagen phases, suggesting that Wnt10b might serve as a critical activator during cyclic transition of SHFs. Methylation frequency of the fifth CpG site (CpG5-175 bp) in CpG islands at the anagen phase was lower than that at both the catagen and telogen phases. The methylation status of the CpG5 site was negatively correlated with Wnt10b expression. This indicated that the methylation of CpG5 might participate in Wnt10b transcriptional suppression in SHFs. Furthermore, histone H3 acetylation status in the regions-256~-11 bp and 98 ~ 361 bp were significantly lower at both the catagen and telogen phases than at the anagen phase. The histone H3 acetylation level was significantly positively correlated with Wnt10b expression. This confirmed that histone acetylation was likely involved in upregulating Wnt10b transcription in SHFs. Additionally, potential binding to the transcription factors ZF57 and HDBP was predicted within the CpG5 site. In conclusion, our findings reveal the epigenetic mechanism of Wnt10b transcription and provide a new insight into epigenetic regulation during the SHF cycle in the Angora rabbit.
Collapse
Affiliation(s)
- Liya Bai
- Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Haitao Sun
- Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Wenxue Jiang
- Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Liping Yang
- Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Gongyan Liu
- Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Xueyan Zhao
- Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Hongmei Hu
- Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jianying Wang
- Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Shuxia Gao
- Shandong Provincial Key Laboratory of Animal Disease Control & Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| |
Collapse
|
16
|
Liu H. The roles of histone deacetylases in kidney development and disease. Clin Exp Nephrol 2021; 25:215-223. [PMID: 33398599 PMCID: PMC7925501 DOI: 10.1007/s10157-020-01995-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 11/10/2020] [Indexed: 02/06/2023]
Abstract
Histone deacetylases (HDACs) are important epigenetic regulators that mediate deacetylation of both histone and non-histone proteins. HDACs, especially class I HDACs, are highly expressed in developing kidney and subject to developmental control. HDACs play an important role in kidney formation, especial nephron progenitor maintenance and differentiation. Several lines of evidence support the critical role of HDACs in the development and progression of various kidney diseases. HDAC inhibitors (HDACis) are very effective in the prevention and treatment of kidney diseases (including kidney cancer). A better understanting of the molecular mechanisms underlying the role(s) of HDACs in the pathogenesis and progression of renal disease are likely to be of great help in developing more effective and less toxic selective HDAC inhibitors and combinatorial therapeutics.
Collapse
Affiliation(s)
- Hongbing Liu
- Department of Pediatrics and The Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, SL-37, 1430 Tulane Avenue, New Orleans, LA, 70112, USA.
| |
Collapse
|
17
|
Wang F, Ngo J, Li Y, Liu H, Chen CH, Saifudeen Z, Sequeira-Lopez MLS, El-Dahr SS. Targeted disruption of the histone lysine 79 methyltransferase Dot1L in nephron progenitors causes congenital renal dysplasia. Epigenetics 2020; 16:1235-1250. [PMID: 33315499 DOI: 10.1080/15592294.2020.1861168] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The epigenetic regulator Dot1, the only known histone H3K79 methyltransferase, has a conserved role in organismal development and homoeostasis. In yeast, Dot1 is required for telomeric silencing and genomic integrity. In Drosophila, Dot1 (Grappa) regulates homoeotic gene expression. Dysregulation of DOT1L (human homologue of Dot1) causes leukaemia and is implicated in dilated cardiomyopathy. In mice, germline disruption of Dot1L and loss of H3K79me2 disrupt vascular and haematopoietic development. Targeted inactivation of Dot1L in principal cells of the mature collecting duct affects terminal differentiation and cell type patterning. However, the role of H3K79 methylation in mammalian tissue development has been questioned, as it is dispensable in the intestinal epithelium, a rapidly proliferating tissue. Here, we used lineage-specific Cre recombinase to delineate the role of Dot1L methyltransferase activity in the mouse metanephric kidney, an organ that develops via interactions between ureteric epithelial (Hoxb7) and mesenchymal (Six2) cell lineages. The results demonstrate that Dot1LHoxb7 is dispensable for ureteric bud branching morphogenesis. In contrast, Dot1LSix2 is critical for the maintenance and differentiation of Six2+ progenitors into epithelial nephrons. Dot1LSix2 mutant kidneys exhibit congenital nephron deficit and cystic dysplastic kidney disease. Molecular analysis implicates defects in key renal developmental regulators, such as Lhx1, Pax2 and Notch. We conclude that the developmental functions of Dot1L-H3K79 methylation in the kidney are lineage-restricted. The link between H3K79me and renal developmental pathways reaffirms the importance of chromatin-based mechanisms in organogenesis.
Collapse
Affiliation(s)
- Fenglin Wang
- Divisions of Pediatric Nephrology and Human Genetics, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jenny Ngo
- Divisions of Pediatric Nephrology and Human Genetics, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Yuwen Li
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Hongbing Liu
- Divisions of Pediatric Nephrology and Human Genetics, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Chao-Hui Chen
- Divisions of Pediatric Nephrology and Human Genetics, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Zubaida Saifudeen
- Divisions of Pediatric Nephrology and Human Genetics, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Maria Luisa S Sequeira-Lopez
- Division of Pediatric Nephrology, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Samir S El-Dahr
- Divisions of Pediatric Nephrology and Human Genetics, Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
18
|
Li Z, Li M, Wang D, Hou P, Chen X, Chu S, Chai D, Zheng J, Bai J. Post-translational modifications of EZH2 in cancer. Cell Biosci 2020; 10:143. [PMID: 33308321 PMCID: PMC7731458 DOI: 10.1186/s13578-020-00505-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Enhancer of zeste homolog 2 (EZH2), as a main component of Polycomb Repressive Complex 2, catalyzes histone H3K27me3 to silence its target gene expression. EZH2 upregulation results in cancer development and poor prognosis of cancer patients. Post-translational modifications (PTMs) are important biological events in cancer progression. PTMs regulate protein conformation and diversity functions. Recently, mounting studies have demonstrated that EZH2 stability, histone methyltransferase activity, localization, and binding partners can be regulated by PTMs, including phosphorylation, O-GlcNAcylation, acetylation, methylation and ubiquitination. However, the detailed molecular mechanisms of the EZH2-PTMs and whether other types of PTMs occur in EZH2 remain largely unclear. This review presents an overview of different roles of EZH2 modification and EZH2-PTMs crosstalk during tumorigenesis and cancer metastasis. We also discussed the therapeutic potential of targeting EZH2 modifications for cancer therapy.
Collapse
Affiliation(s)
- Zhongwei Li
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China
| | - Minle Li
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China
| | - Diandian Wang
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China
| | - Pingfu Hou
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China
| | - Xintian Chen
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China
| | - Sufang Chu
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China.
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu Province, China.
| |
Collapse
|
19
|
Ilimaquinone inhibits neovascular age-related macular degeneration through modulation of Wnt/β-catenin and p53 pathways. Pharmacol Res 2020; 161:105146. [PMID: 32814173 DOI: 10.1016/j.phrs.2020.105146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 11/22/2022]
Abstract
Neovascular age-related macular degeneration (nAMD) is a common cause of irreversible vision loss in the elderly. Anti-vascular endothelial growth factor has been effective in treating pathological ocular neovascularization, but it has limitations including the need for repeated intraocular injections for the maintenance of therapeutic effects in most patients and poor or non-response to this agent in some patients. in vitro cellular studies were conducted using retinal pigment epithelial cell lines (ARPE-19 and hTERT-RPE1), human umbilical vein endothelial cells (HUVECs), and human umbilical vein smooth muscle cells (HUVSMCs). in vivo efficacy of ilimaquinone (IQ) was tested in laser-induced choroidal neovascularization mouse and rabbit models. Tissue distribution study was performed in male C57BL6/J mice. IQ, 4,9-friedodrimane-type sesquiterpenoid isolated from the marine sponge, repressed the expression of angiogenic/inflammatory factors and restored the expression of E-cadherin in retinal pigment epithelial cells by inhibiting the Wnt/β-catenin pathway. In addition, it selectively inhibited proliferation and tube formation of HUVECs by activating the p53 pathway. Topical and intraperitoneal administration of IQ significantly reduced choroidal neovascularization in rabbits and mice with laser-induced choroidal neovascularization. Notably, IQ by the oral route of exposure was highly permeable to the eyes and suppressed abnormal vascular leakage by downregulation of β-catenin and stabilization of p53 in vivo. Our findings demonstrate that IQ functions through regulation of p53 and Wnt/β-catenin pathways with conceivable advantages over existing cytokine-targeted anti-angiogenic therapies.
Collapse
|
20
|
Hyndman KA, Speed JS, Mendoza LD, Allan JM, Colson J, Sedaka R, Jin C, Jung HJ, El-Dahr S, Pollock DM, Pollock JS. Fluid-electrolyte homeostasis requires histone deacetylase function. JCI Insight 2020; 5:137792. [PMID: 32673289 PMCID: PMC7455138 DOI: 10.1172/jci.insight.137792] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/09/2020] [Indexed: 12/27/2022] Open
Abstract
Histone deacetylase (HDAC) enzymes regulate transcription through epigenetic modification of chromatin structure, but their specific functions in the kidney remain elusive. We discovered that the human kidney expresses class I HDACs. Kidney medulla-specific inhibition of class I HDACs in the rat during high-salt feeding results in hypertension, polyuria, hypokalemia, and nitric oxide deficiency. Three new inducible murine models were used to determine that HDAC1 and HDAC2 in the kidney epithelium are necessary for maintaining epithelial integrity and maintaining fluid-electrolyte balance during increased dietary sodium intake. Moreover, single-nucleus RNA-sequencing determined that epithelial HDAC1 and HDAC2 are necessary for expression of many sodium or water transporters and channels. In performing a systematic review and meta-analysis of serious adverse events associated with clinical HDAC inhibitor use, we found that HDAC inhibitors increased the odds ratio of experiencing fluid-electrolyte disorders, such as hypokalemia. This study provides insight on the mechanisms of potential serious adverse events with HDAC inhibitors, which may be fatal to critically ill patients. In conclusion, kidney tubular HDACs provide a link between the environment, such as consumption of high-salt diets, and regulation of homeostatic mechanisms to remain in fluid-electrolyte balance.
Collapse
Affiliation(s)
- Kelly A Hyndman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Joshua S Speed
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Luciano D Mendoza
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - John M Allan
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jackson Colson
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Randee Sedaka
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chunhua Jin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hyun Jun Jung
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Samir El-Dahr
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jennifer S Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
21
|
Zhou J, Zhou H, Liu C, Huang L, Lu D, Gao C. HDAC1-mediated deacetylation of LSD1 regulates vascular calcification by promoting autophagy in chronic renal failure. J Cell Mol Med 2020; 24:8636-8649. [PMID: 32596952 PMCID: PMC7412400 DOI: 10.1111/jcmm.15494] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 05/19/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic renal failure (CRF) is commonly associated with various adverse consequences including pathological vascular calcification (VC), which represents a significant clinical concern. Existing literature has suggested the involvement of histone deacetylases (HDACs) in the progression of CRF‐induced VC. However, the underlying molecular mechanisms associated with HDACs remain largely unknown. Therefore, we established the adenine‐induced CRF rat model and in vitro VC models based on vascular smooth muscle cells (VSMCs) to examine HDAC1/lysine demethylase 1A (LSD1)/SESN2 as a novel molecular pathway in CRF‐induced VC. Our initial results demonstrated that HDAC1 reduced the formation of VC in vivo and in vitro. HDAC1 was found to deacetylate LSD1, which subsequently led to impaired transcriptional activity in CRF‐induced VC. Moreover, our results illustrated that LSD1 diminished the enrichment of H3K4me2 at the SESN2 promoter. Autophagy was identified as a vasculo‐protective element against calcification in VC. Finally, we found that the inhibitory effects of HDAC1 overexpression on VC were partially abolished via over‐expressed LSD1 in adenine‐induced CRF model rats and in high phosphate‐induced VSMCs. Taken together, these results highlight the crucial role of HDAC1 as an antagonistic factor in the progression of VC in CRF, and also revealed a novel regulatory mechanism by which HDAC1 operates. These findings provide significant insight and a fresh perspective into promising novel treatment strategies by up‐regulating HDAC1 in CRF.
Collapse
Affiliation(s)
- Jiajun Zhou
- Kidney Department, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Han Zhou
- Queen Mary College of Nanchang University, Nanchang, China
| | - Caixin Liu
- Clinical Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Lin Huang
- Kidney Department, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Dongmei Lu
- Kidney Department, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Chaoqing Gao
- Kidney Department, Yijishan Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
22
|
Huang B, Liu Z, Vonk A, Zeng Z, Li Z. Epigenetic regulation of kidney progenitor cells. Stem Cells Transl Med 2020; 9:655-660. [PMID: 32163228 PMCID: PMC7214665 DOI: 10.1002/sctm.19-0289] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/26/2020] [Accepted: 02/17/2020] [Indexed: 12/15/2022] Open
Abstract
The reciprocal interactions among the different embryonic kidney progenitor populations lay the basis for proper kidney organogenesis. During kidney development, three types of progenitor cells, including nephron progenitor cells, ureteric bud progenitor cells, and interstitial progenitor cells, generate the three major kidney structures—the nephrons, the collecting duct network, and the stroma, respectively. Epigenetic mechanisms are well recognized for playing important roles in organism development, in fine‐tuned control of physiological activities, and in responses to environment stimuli. Recently, evidence supporting the importance of epigenetic mechanisms underlying kidney organogenesis has emerged. In this perspective, we summarize the research progress and discuss the potential contribution of novel stem cell, organoid, and next‐generation sequencing tools in advancing this field in the future.
Collapse
Affiliation(s)
- Biao Huang
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zhenqing Liu
- Division of Oral Biology and Medicine, School of Dentistry, UCLA, Los Angeles, California, USA
| | - Ariel Vonk
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zipeng Zeng
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zhongwei Li
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
23
|
Chatterjee B, Ghosh K, Kanade SR. Resveratrol modulates epigenetic regulators of promoter histone methylation and acetylation that restores BRCA1, p53, p21 CIP1 in human breast cancer cell lines. Biofactors 2019; 45:818-829. [PMID: 31317586 DOI: 10.1002/biof.1544] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/27/2019] [Indexed: 01/09/2023]
Abstract
The epigenetic enzymes catalyze posttranslational modifications (PTMs) of histones, which functionally determine gene expression at the chromatin level. Resveratrol (RVT) a much studied anti-cancer natural molecule is known for restoration of BRCA1, p53, and p21 in cancer cells. We aimed to investigate the role of histone methylation and acetylation on upregulation of these tumor suppressor genes. Our results suggest RVT significantly increase expression of BRCA1, p53, and p21, while decreased expression of protein arginine methyltransferase 5 (PRMT5) and enhancer of Zeste homolog 2 (EZH2) at a 20 μM concentration by 48 hr in both MCF-7 and MDA-MB-231 breast cancer cells. Also, there was an overall loss of H4R3me2s (catalytic product of PRMT5) and H3K27me3 (catalytic product of PRMT5). In contrast, RVT exposure caused a significant decrease in lysine deacetylase (KDAC) activity and expression of KDAC1-3, whereas the expression of lysine acetyltransferase KAT2A/3B was increased compared to the unexposed cells. As an outcome, RVT increased global level of H3K9ac and H3K27ac marks. The chromatin immunoprecipitation showed 20 μM RVT exposure significantly reduced the enrichment of repressive histone marks (H4R3me2s and H3K27me3) while the abundance of activating histone marks (H3K9/27ac) within the proximal promoter region of BRCA1, p53, and p21 was increased. We hypothesize RVT by affecting the expression and function of methylation and acetylation enzymes altered the epigenetic modifications on promoter histones that restored expression of these critically important tumor suppressor genes.
Collapse
Affiliation(s)
- Biji Chatterjee
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Kasargod, Kerala, India
| | - Krishna Ghosh
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Kasargod, Kerala, India
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Central University P.O., Hyderabad, Telangana, India
| | - Santosh R Kanade
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Kasargod, Kerala, India
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Central University P.O., Hyderabad, Telangana, India
| |
Collapse
|
24
|
Aboukhatwa SM, Hanigan TW, Taha TY, Neerasa J, Ranjan R, El-Bastawissy EE, Elkersh MA, El-Moselhy TF, Frasor J, Mahmud N, McLachlan A, Petukhov PA. Structurally Diverse Histone Deacetylase Photoreactive Probes: Design, Synthesis, and Photolabeling Studies in Live Cells and Tissue. ChemMedChem 2019; 14:1096-1107. [PMID: 30921497 PMCID: PMC6548601 DOI: 10.1002/cmdc.201900114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/28/2019] [Indexed: 01/27/2023]
Abstract
Histone deacetylase (HDAC) activity is modulated in vivo by post-translational modifications and formation of multiprotein complexes. Novel chemical tools to study how these factors affect engagement of HDAC isoforms by HDAC inhibitors (HDACi) in cells and tissues are needed. In this study, a synthetic strategy to access chemically diverse photoreactive probes (PRPs) was developed and used to prepare seven novel HDAC PRPs 9-15. The class I HDAC isoform engagement by PRPs was determined in biochemical assays and photolabeling experiments in live SET-2, HepG2, HuH7, and HEK293T cell lines and in mouse liver tissue. Unlike the HDAC protein abundance and biochemical activity against recombinant HDACs, the chemotype of the PRPs and the type of cells were key in defining the engagement of HDAC isoforms in live cells. Our findings suggest that engagement of HDAC isoforms by HDACi in vivo may be substantially modulated in a cell- and tissue-type-dependent manner.
Collapse
Affiliation(s)
- Shaimaa M Aboukhatwa
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Thomas W Hanigan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| | - Taha Y Taha
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| | - Jayaprakash Neerasa
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| | - Rajeev Ranjan
- Section of Hematology/Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Eman E El-Bastawissy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Mohamed A Elkersh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University, Alexandria, 21311, Egypt
| | - Tarek F El-Moselhy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Jonna Frasor
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Nadim Mahmud
- Section of Hematology/Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Alan McLachlan
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Pavel A Petukhov
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| |
Collapse
|
25
|
Wang MH, Wu CH, Huang TY, Sung HW, Chiou LL, Lin SP, Lee HS. Nerve-mediated expression of histone deacetylases regulates limb regeneration in axolotls. Dev Biol 2019; 449:122-131. [PMID: 30826398 DOI: 10.1016/j.ydbio.2019.02.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 02/13/2019] [Accepted: 02/22/2019] [Indexed: 02/07/2023]
Abstract
Axolotls have amazing abilities to regenerate their lost limbs. Nerve and wound epidermis have great impacts on this regeneration. Histone deacetylases (HDACs) have been shown to play roles in the regeneration of amphibian tails and limbs. In this study, a bi-phasic up-regulation of HDAC1 was noted before early differentiation stage of axolotl limb regeneration. Limb regeneration was delayed in larvae incubated with an HDAC inhibitor MS-275. Local injection of MS-275 or TSA, another HDAC inhibitor, into amputation sites of the juveniles did not interfere with wound healing but more profoundly inhibited local HDAC activities and blastema formation/limb regeneration. Elevation of HDAC1 expression was more apparent in wound epidermis than in mesenchyme. Prior denervation prohibited this elevation and limb regeneration. Supplementation of nerve factors BMP7, FGF2, and FGF8 in the stump ends after amputation on denervated limbs not only enabled HDAC1 up-regulation but also led to more extent of limb regeneration. In conclusion, nerve-mediated HDAC1 expression is required for blastema formation and limb regeneration.
Collapse
Affiliation(s)
- Mu-Hui Wang
- Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ting-Yu Huang
- Liver Disease Prevention and Treatment Research Foundation, Taipei, Taiwan
| | - Hung-Wei Sung
- Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Ling-Ling Chiou
- Liver Disease Prevention and Treatment Research Foundation, Taipei, Taiwan
| | - Shau-Ping Lin
- Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan; Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan; Center of Systems Biology, National Taiwan University, Taipei, Taiwan; The Research Center of Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| | - Hsuan-Shu Lee
- Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
26
|
Sun D, Yu M, Li Y, Xing H, Gao Y, Huang Z, Hao W, Lu K, Kong C, Shimozato O, Ozaki T, Zhu Y. Histone deacetylase 2 is involved in DNA damage-mediated cell death of human osteosarcoma cells through stimulation of the ATM/p53 pathway. FEBS Open Bio 2019; 9:478-489. [PMID: 30868056 PMCID: PMC6396148 DOI: 10.1002/2211-5463.12585] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 11/30/2018] [Accepted: 12/20/2018] [Indexed: 11/24/2022] Open
Abstract
Tumor suppressor p53 is a short‐lived nuclear transcription factor, which becomes stabilized and activated in response to a wide variety of cellular stresses. Around 50% of human cancer tissues carry p53 mutations, and certain p53 mutations contribute to chemoresistance. In the present study, we found that histone deacetylase 2 (HDAC2) acts as a co‐activator of tumor suppressor p53 and participates in the early molecular events following DNA damage. Anti‐cancer drug adriamycin (ADR) treatment induced cell death in p53‐wild‐type human osteosarcoma U2OS cells, and this was accompanied by a remarkable accumulation of p53 and γH2AX. HDAC2 gene silencing significantly decreased the sensitivity of U2OS cells to ADR and attenuated p53‐dependent DNA damage responses, such as ADR‐mediated phosphorylation of ataxia telangiectasia mutated (ATM) and p53, as well as accumulation of γH2AX and cleaved poly (ADP‐ribose) polymerase. However, HDAC2 knockdown had a marginal effect on p53‐null human lung cancer H1299 cells following ADR exposure. In contrast, forced expression of HA‐HDAC2 promoted cell death and stimulated the transcriptional activity of p53. Moreover, p53 and HDAC2 were found to co‐precipitate with ATM. Together, our present results strongly suggest that the p53–HDAC2 axis plays a vital role in the regulation of the DNA damage response and also contributes to chemosensitivity of cancer cells.
Collapse
Affiliation(s)
- Dan Sun
- Department of Urology The First Hospital of China Medical University Shenyang China
| | - Meng Yu
- Department of Reproductive Biology and Transgenic Animal China Medical University Shenyang China
| | - Yuanyuan Li
- Department of Molecular Medicine Life Science Institute Saga Medical Center KOSEIKAN Saga Japan
| | - Haotian Xing
- Department of Urology The First Hospital of China Medical University Shenyang China
| | - Ying Gao
- Department of Urology The First Hospital of China Medical University Shenyang China
| | - Zhihong Huang
- Department of Urology The First Hospital of China Medical University Shenyang China
| | - Wenjun Hao
- Department of Urology The First Hospital of China Medical University Shenyang China
| | - Kaining Lu
- Department of Urology The First Hospital of China Medical University Shenyang China
| | - Chuize Kong
- Department of Urology The First Hospital of China Medical University Shenyang China
| | - Osamu Shimozato
- Laboratory of DNA Damage Signaling Chiba Cancer Center Research Institute Chiba Japan
| | - Toshinori Ozaki
- Laboratory of DNA Damage Signaling Chiba Cancer Center Research Institute Chiba Japan
| | - Yuyan Zhu
- Department of Urology The First Hospital of China Medical University Shenyang China
| |
Collapse
|
27
|
Tham MS, Smyth IM. Cellular and molecular determinants of normal and abnormal kidney development. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 8:e338. [DOI: 10.1002/wdev.338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/07/2018] [Accepted: 11/14/2018] [Indexed: 01/21/2023]
Affiliation(s)
- Ming S. Tham
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute, Monash University Melbourne Victoria Australia
| | - Ian M. Smyth
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute, Monash University Melbourne Victoria Australia
- Department of Biochemistry and Molecular Biology Monash Biomedicine Discovery Institute, Monash University Melbourne Victoria Australia
| |
Collapse
|
28
|
Nishikawa M, Yuri S, Kimura H, Yanagawa N, Hamon M, Hauser P, Zhao L, Jo OD, Yanagawa N. Comprehensive analysis of chromatin signature and transcriptome uncovers functional lncRNAs expressed in nephron progenitor cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:58-70. [PMID: 30416088 DOI: 10.1016/j.bbagrm.2018.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/25/2018] [Accepted: 09/12/2018] [Indexed: 01/21/2023]
Abstract
Emerging evidence from recent studies has unraveled the roles of long noncoding RNAs (lncRNAs) in the function of various tissues. However, little is known about the roles of lncRNAs in kidney development. In our present study, we aimed to identify functional lncRNAs in one of the three lineages of kidney progenitor cells, i.e., metanephric mesenchymal (MM) cells. We conducted comprehensive analyses of the chromatin signature and transcriptome by RNA-seq and ChIP-seq. We found seventeen lncRNAs that were expressed specifically in MM cells with an active chromatin signature, while remaining silenced in a bivalent chromatin state in non-MM cells. Out of these MM specific lncRNAs, we identified a lncRNA, Gm29418, in a distal enhancer region of Six2, a key regulatory gene of MM cells. We further identified three transcript variants of Gm29418 by Rapid Amplification of cDNA Ends (RACE), and confirmed that the transcription-start-sites (TSSs) of these variants were consistent with the result of Cap Analysis Gene Expression (CAGE). In support of the enhancer-like function of Gm29418 on Six2 expression, we found that knock-down of Gm29418 by two independent anti-sense locked nucleic acid (LNA) phosphorothioate gapmers suppressed Six2 mRNA expression levels in MM cells. We also found that over-expression of Gm29418 led to an increase in Six2 mRNA expression levels in a mouse MM cell line. In conclusion, we identified a lncRNA, Gm29418, in nephron progenitor cells that has an enhancer-like function on a key regulatory gene, Six2.
Collapse
Affiliation(s)
- Masaki Nishikawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Shunsuke Yuri
- Nara Institute of Science & Technology, Nara 630-0192, Japan
| | | | - Naomi Yanagawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Morgan Hamon
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Peter Hauser
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Lifu Zhao
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA
| | - Oak D Jo
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Norimoto Yanagawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
29
|
Liu H, Chen S, Yao X, Li Y, Chen CH, Liu J, Saifudeen Z, El-Dahr SS. Histone deacetylases 1 and 2 regulate the transcriptional programs of nephron progenitors and renal vesicles. Development 2018; 145:dev.153619. [PMID: 29712641 DOI: 10.1242/dev.153619] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 04/20/2018] [Indexed: 12/17/2022]
Abstract
Nephron progenitor cells (NPCs) are Six2-positive metanephric mesenchyme cells, which undergo self-renewal and differentiation to give rise to nephrons until the end of nephrogenesis. Histone deacetylases (HDACs) are a group of epigenetic regulators that control cell fate, but their role in balancing NPC renewal and differentiation is unknown. Here, we report that NPC-specific deletion of Hdac1 and Hdac2 genes in mice results in early postnatal lethality owing to renal hypodysplasia and loss of NPCs. HDAC1/2 interact with the NPC renewal regulators Six2, Osr1 and Sall1, and are co-bound along with Six2 on the Six2 enhancer. Although the mutant NPCs differentiate into renal vesicles (RVs), Hdac1/2 mutant kidneys lack nascent nephrons or mature glomeruli, a phenocopy of Lhx1 mutants. Transcriptional profiling and network analysis identified disrupted expression of Lhx1 and its downstream genes, Dll1 and Hnf1a/4a, as key mediators of the renal phenotype. Finally, although HDAC1/2-deficient NPCs and RVs overexpress hyperacetylated p53, Trp53 deletion failed to rescue the renal dysgenesis. We conclude that the epigenetic regulators HDAC1 and HDAC2 control nephrogenesis via interactions with the transcriptional programs of nephron progenitors and renal vesicles.
Collapse
Affiliation(s)
- Hongbing Liu
- Department of Pediatrics and The Tulane Hypertension & Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Shaowei Chen
- Department of Pediatrics and The Tulane Hypertension & Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Xiao Yao
- Department of Pediatrics and The Tulane Hypertension & Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Yuwen Li
- Department of Pediatrics and The Tulane Hypertension & Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Chao-Hui Chen
- Department of Pediatrics and The Tulane Hypertension & Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jiao Liu
- Department of Pediatrics and The Tulane Hypertension & Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Zubaida Saifudeen
- Department of Pediatrics and The Tulane Hypertension & Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Samir S El-Dahr
- Department of Pediatrics and The Tulane Hypertension & Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
30
|
Kanda K, Sakamoto J, Matsumoto Y, Ikuta K, Goto N, Morita Y, Ohno M, Nishi K, Eto K, Kimura Y, Nakanishi Y, Ikegami K, Yoshikawa T, Fukuda A, Kawada K, Sakai Y, Ito A, Yoshida M, Kimura T, Chiba T, Nishi E, Seno H. Nardilysin controls intestinal tumorigenesis through HDAC1/p53-dependent transcriptional regulation. JCI Insight 2018; 3:91316. [PMID: 29669932 DOI: 10.1172/jci.insight.91316] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 03/20/2018] [Indexed: 02/06/2023] Open
Abstract
Colon cancer is a complex disease affected by a combination of genetic and epigenetic factors. Here we demonstrate that nardilysin (N-arginine dibasic convertase; NRDC), a metalloendopeptidase of the M16 family, regulates intestinal tumorigenesis via its nuclear functions. NRDC is highly expressed in human colorectal cancers. Deletion of the Nrdc gene in ApcMin mice crucially suppressed intestinal tumor development. In ApcMin mice, epithelial cell-specific deletion of Nrdc recapitulated the tumor suppression observed in Nrdc-null mice. Moreover, epithelial cell-specific overexpression of Nrdc significantly enhanced tumor formation in ApcMin mice. Notably, epithelial NRDC controlled cell apoptosis in a gene dosage-dependent manner. In human colon cancer cells, nuclear NRDC directly associated with HDAC1, and controlled both acetylation and stabilization of p53, with alterations of p53 target apoptotic factors. These findings demonstrate that NRDC is critically involved in intestinal tumorigenesis through its epigenetic regulatory function, and targeting NRDC may lead to a novel prevention or therapeutic strategy against colon cancer.
Collapse
Affiliation(s)
| | - Jiro Sakamoto
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Kozo Ikuta
- Department of Gastroenterology and Hepatology, and
| | | | - Yusuke Morita
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mikiko Ohno
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kiyoto Nishi
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto, Japan
| | - Yuto Kimura
- Department of Gastroenterology and Hepatology, and
| | | | | | | | | | - Kenji Kawada
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshiharu Sakai
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihiro Ito
- Chemical Genetics Laboratory, RIKEN, Wako, Saitama, Japan.,Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Minoru Yoshida
- Chemical Genetics Laboratory, RIKEN, Wako, Saitama, Japan.,Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Eiichiro Nishi
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, and
| |
Collapse
|
31
|
Gregath A, Lu QR. Epigenetic modifications-insight into oligodendrocyte lineage progression, regeneration, and disease. FEBS Lett 2018; 592:1063-1078. [PMID: 29427507 DOI: 10.1002/1873-3468.12999] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/28/2018] [Accepted: 02/02/2018] [Indexed: 12/11/2022]
Abstract
Myelination by oligodendrocytes in the central nervous system permits high-fidelity saltatory conduction from neuronal cell bodies to axon terminals. Dysmyelinating and demyelinating disorders impair normal nervous system functions. Consequently, an understanding of oligodendrocyte differentiation that moves beyond the genetic code into the field of epigenetics is essential. Chromatin reprogramming is critical for steering stage-specific differentiation processes during oligodendrocyte development. Fine temporal control of chromatin remodeling through ATP-dependent chromatin remodelers and sequential histone modifiers shapes a chromatin regulatory landscape conducive to oligodendrocyte fate specification, lineage differentiation, and maintenance of cell identity. In this Review, we will focus on the biological functions of ATP-dependent chromatin remodelers and histone deacetylases in myelinating oligodendrocyte development and implications for myelin regeneration in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alexander Gregath
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Qing Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, OH, USA
| |
Collapse
|
32
|
Jing H, Su X, Gao B, Shuai Y, Chen J, Deng Z, Liao L, Jin Y. Epigenetic inhibition of Wnt pathway suppresses osteogenic differentiation of BMSCs during osteoporosis. Cell Death Dis 2018; 9:176. [PMID: 29416009 PMCID: PMC5833865 DOI: 10.1038/s41419-017-0231-0] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/27/2017] [Accepted: 12/13/2017] [Indexed: 12/12/2022]
Abstract
Disrupted Wnt signaling in osteoblastic-lineage cells leads to bone formation defect in osteoporosis. However, the factors repressing Wnt signaling are unclear. In our study, we found that Wnt signaling was suppressed persistently in bone marrow-derived mesenchymal stem cells (BMSCs) during osteoporosis. Accordingly, histone acetylation levels on Wnt genes (Wnt1, Wnt6, Wnt10a, and Wnt10b) were declined in BMSCs from OVX mice. By screening the family of histone acetyltransferase, we identified that GCN5 expression increased during osteogenic differentiation of BMSCs, whereas decreased after osteoporosis. Further analysis revealed that GCN5 promoted osteogenic differentiation of BMSCs by increasing acetylation on histone 3 lysine 9 loci on the promoters of Wnt genes. Reduced GCN5 expression suppressed Wnt signaling, resulting in osteogenic defect of BMSCs from OVX mice. Moreover, restoring GCN5 levels recovered BMSC osteogenic differentiation, and attenuated bone loss in OVX mice. Taken together, our study demonstrated that disrupted histone acetylation modification in BMSCs lead to bone formation defect during osteoporosis. The findings also introduced a novel therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Huan Jing
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China
| | - Xiaoxia Su
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Bo Gao
- Department of Orthopaedic Surgery. Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yi Shuai
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China
| | - Ji Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China.,Department of Oral Implantology, School of Stomatology, State Key Laboratory of Military Stomatology, The Fourth Military Medical University, Xi'an, Shanxi, 710032, China
| | - Zhihong Deng
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Li Liao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China. .,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China. .,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
33
|
Hornig E, Heppt MV, Graf SA, Ruzicka T, Berking C. Inhibition of histone deacetylases in melanoma-a perspective from bench to bedside. Exp Dermatol 2018; 25:831-838. [PMID: 27792246 DOI: 10.1111/exd.13089] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2016] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDACs) are critically involved in epigenetic gene regulation through alterations of the chromatin status of DNA. Aberrant expression, dysregulation of their enzymatic activity or imbalances between HDACs and histone acetyltransferases are likely involved in the development and progression of cancer. Pharmacologic inhibition of HDACs shows potent antitumor activity in a panel of malignancies such as colon or gastric cancer and multiple myeloma. In this review, we summarize the current knowledge of HDACs in melanoma and evaluate the application of HDAC inhibition from an experimental and clinical perspective. The molecular functions of HDACs can be classified into histone and non-histone effects with diverse implications in proliferation, cell cycle progression and apoptosis. HDAC inhibition results in G1 cell cycle arrest, induces apoptosis and increases the immunogenicity of melanoma cells. Some studies proposed that HDAC inhibition may overcome the resistance of melanoma cells to BRAF inhibition. Several inhibitors such as vorinostat, entinostat and valproic acid have recently been tested in phase I and early phase II trials, yet most agents show limited efficacy and tolerability as single agents. The most frequent adverse events of HDAC inhibition comprise haematological toxicity, fatigue, nausea and laboratory abnormalities. Existing evidence supports the hypothesis that HDAC inhibitors (HDACi) may sensitize melanoma cells to immunotherapy and targeted therapy and hence bear therapeutic potential concurrent with immune checkpoint blockade or BRAF and MEK inhibition.
Collapse
Affiliation(s)
- Eva Hornig
- Department of Dermatology and Allergy, Munich University Hospital (LMU), Munich, Germany
| | - Markus V Heppt
- Department of Dermatology and Allergy, Munich University Hospital (LMU), Munich, Germany
| | - Saskia A Graf
- Department of Dermatology and Allergy, Munich University Hospital (LMU), Munich, Germany
| | - Thomas Ruzicka
- Department of Dermatology and Allergy, Munich University Hospital (LMU), Munich, Germany
| | - Carola Berking
- Department of Dermatology and Allergy, Munich University Hospital (LMU), Munich, Germany.
| |
Collapse
|
34
|
Takenaka T, Inoue T, Miyazaki T, Kobori H, Nishiyama A, Ishii N, Hayashi M, Suzuki H. Klotho suppresses the renin-angiotensin system in adriamycin nephropathy. Nephrol Dial Transplant 2018; 32:791-800. [PMID: 27798196 DOI: 10.1093/ndt/gfw340] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/10/2016] [Indexed: 11/13/2022] Open
Abstract
Backgrounds Klotho protein interacts with the transforming growth factor β (TGF-β) receptor and Wnt, which contribute to the progression of renal disease, inhibiting their signals. Renal and circulating klotho levels are diminished in chronic kidney disease. Methods Experiments were performed to assess whether supplementation of klotho protein could have protective effects on the kidney. Rats were injected with adriamycin (5 mg/kg) and divided into three groups: those treated with vehicle, those treated with klotho protein and those treated with klotho plus 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD). Rats without adriamycin treatment were used as a control. Results Adriamycin reduced the serum klotho concentration and renal expression of klotho and E-cadherin. Adriamycin also increased the renal expression of Wnt, TGF-β, and angiotensinogen, as well as the renal abundance of β-catenin and angiotensin II. Klotho supplementation suppressed adriamycin-induced elevations of β-catenin and angiotensin II with sustained Wnt expression. Combined treatment with klotho and TDZD reversed the klotho-induced improvements in the renal abundance of β-catenin and angiotensin II as well as the expression of TGF-β and angiotensinogen without affecting E-cadherin. Conclusions Our data indicate that Wnt is involved in the pathogenesis of adriamycin nephropathy. Furthermore, klotho supplementation inhibited Wnt signaling, ameliorating renal angiotensin II. Finally, klotho protein appears to suppress epithelial-mesenchymal transition by inhibiting TGF-β and Wnt signaling.
Collapse
Affiliation(s)
- Tsuneo Takenaka
- Department of Medicine, International University of Health and Welfare, 8-10-16 Akasaka, Minato, Tokyo 107-0052, Japan
| | - Tsutomu Inoue
- Department of Nephrology, Saitama Medical University, Iruma, Saitama, Japan
| | - Takashi Miyazaki
- Department of Nephrology, Saitama Medical University, Iruma, Saitama, Japan
| | - Hiroyuki Kobori
- Department of Medicine, International University of Health and Welfare, 8-10-16 Akasaka, Minato, Tokyo 107-0052, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University, Kida, Kagawa, Japan
| | - Naohito Ishii
- Department of Clinical Chemistry, Kitasato University, Sagamihara, Kanagawa, Japan
| | | | - Hiromichi Suzuki
- Department of Nephrology, Saitama Medical University, Iruma, Saitama, Japan
| |
Collapse
|
35
|
Ma Y, Yuan S, Tian X, Lin S, Wei S, Hu T, Chen S, Li X, Chen S, Wu D, Wang M, Guo D. ABIN1 inhibits HDAC1 ubiquitination and protects it from both proteasome- and lysozyme-dependent degradation. J Cell Biochem 2017; 119:3030-3043. [PMID: 29058807 DOI: 10.1002/jcb.26428] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022]
Abstract
ABIN1, an important immune regulator, has been shown to be involved in various cellular functions, such as immunity, development, tissue homeostasis, and tumor progression. It inhibits TNF- and TLR-induced NF-κB signaling activation and the consequent gene expression. Despite its functional significance, the mechanism of ABIN1 in the regulation of various cellular functions remains unclear. In this study, we identified HDAC1, a key regulator of eukaryotic gene expression and many important cellular events, including cell proliferation, differentiation, cancer and immunity, as an interacting partner of ABIN1. The results showed that ABIN1 acted as a modulator to down-regulate HDAC1 ubiquitination via three different linkages, thereby stabilizing HDAC1 by inhibiting its lysosomal and proteasomal degradation. Interestingly, the inhibitory function of ABIN1 required direct binding with HDAC1. Moreover, the level of p53, which was a tumor suppressor and a well-studied substrate of HDAC1, was under the regulation of ABIN1 via the modulation of HDAC1 levels, suggesting that ABIN1 was physiologically significant in tumor progression. This study has revealed a new function of ABIN1 in mediating HDAC1 modification and stability.
Collapse
Affiliation(s)
- Yuhong Ma
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Sen Yuan
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Xuezhang Tian
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Shanchuan Lin
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Shangmou Wei
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Tongtong Hu
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Shiyou Chen
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, PR China
| | - Xueqing Li
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Shuliang Chen
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Dongcheng Wu
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Min Wang
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Deyin Guo
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, PR China.,School of Basic Medicine (Shenzhen), Sun Yat-Sen University, Guangzhou, PR China
| |
Collapse
|
36
|
El-Dahr SS, Li Y, Liu J, Gutierrez E, Hering-Smith KS, Signoretti S, Pignon JC, Sinha S, Saifudeen Z. p63+ ureteric bud tip cells are progenitors of intercalated cells. JCI Insight 2017; 2:89996. [PMID: 28469077 DOI: 10.1172/jci.insight.89996] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/23/2017] [Indexed: 01/13/2023] Open
Abstract
During renal branching morphogenesis, ureteric bud tip cells (UBTC) serve as the progenitor epithelium for all cell types of the collecting duct. While the transcriptional circuitry of ureteric bud (UB) branching has been intensively studied, the transcriptional control of UBTC differentiation has been difficult to ascertain. This is partly due to limited knowledge of UBTC-specific transcription factors that mark the progenitor state. Here, we identify the transcription factor p63 (also known as TP63), a master regulator of basal stem cells in stratified epithelia, as a specific marker of mouse and human UBTC. Nuclear p63 marks Ret+ UBTC transiently and is silenced by the end of nephrogenesis. Lineage tracing revealed that a subset of UBTC expressing the ΔNp63 isoform (N-terminus truncated p63) is dedicated to generating cortical intercalated cells. Germline targeting of ΔNp63 in mice caused a marked reduction in intercalated cells near the time of birth, indicating that p63 not only marks UBTC, but also is essential for their differentiation. We conclude that the choice of UBTC progenitors to differentiate is determined earlier than previously recognized and that UBTC progenitors are prepatterned and fate restricted. These findings prompt the rethinking of current paradigms of collecting duct differentiation and may have implications for regenerative renal medicine.
Collapse
Affiliation(s)
| | | | | | | | | | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard School of Medicine, Boston, Massachusetts, USA
| | - Jean-Christophe Pignon
- Department of Pathology, Brigham and Women's Hospital, Harvard School of Medicine, Boston, Massachusetts, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine, University of Buffalo, New York, USA
| | | |
Collapse
|
37
|
Wanner N, Bechtel-Walz W. Epigenetics of kidney disease. Cell Tissue Res 2017; 369:75-92. [PMID: 28286899 DOI: 10.1007/s00441-017-2588-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 02/15/2017] [Indexed: 02/06/2023]
Abstract
DNA methylation and histone modifications determine renal programming and the development and progression of renal disease. The identification of the way in which the renal cell epigenome is altered by environmental modifiers driving the onset and progression of renal diseases has extended our understanding of the pathophysiology of kidney disease progression. In this review, we focus on current knowledge concerning the implications of epigenetic modifications during renal disease from early development to chronic kidney disease progression including renal fibrosis, diabetic nephropathy and the translational potential of identifying new biomarkers and treatments for the prevention and therapy of chronic kidney disease and end-stage kidney disease.
Collapse
Affiliation(s)
- Nicola Wanner
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Center for Systems Biology (ZBSA), Albert-Ludwigs-University, Freiburg, Germany. .,Renal Division, University Hospital Freiburg, Breisacher Strasse 66, 79106, Freiburg, Germany.
| | - Wibke Bechtel-Walz
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Renal Division, University Hospital Freiburg, Breisacher Strasse 66, 79106, Freiburg, Germany.
| |
Collapse
|
38
|
Vriend J, Marzban H. The ubiquitin-proteasome system and chromosome 17 in cerebellar granule cells and medulloblastoma subgroups. Cell Mol Life Sci 2017; 74:449-467. [PMID: 27592301 PMCID: PMC11107675 DOI: 10.1007/s00018-016-2354-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/17/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022]
Abstract
Chromosome 17 abnormalities are often observed in medulloblastomas (MBs), particularly those classified in the consensus Groups 3 and 4. Herein we review MB signature genes associated with chromosome 17 and the relationship of these signature genes to the ubiquitin-proteasome system. While clinical investigators have not focused on the ubiquitin-proteasome system in relation to MB, a substantial amount of data on the topic has been hidden in the form of supplemental datasets of gene expression. A supplemental dataset associated with the Thompson classification of MBs shows that a subgroup of MB with 17p deletions is characterized by reduced expression of genes for several core particle subunits of the beta ring of the proteasome (β1, β4, β5, β7). One of these genes (PSMB6, the gene for the β1 subunit) is located on chromosome 17, near the telomeric end of 17p. By comparison, in the WNT group of MBs only one core proteasome subunit, β6, associated with loss of a gene (PSMB1) on chromosome 6, was down-regulated in this dataset. The MB subgroups with the worst prognosis have a significant association with chromosome 17 abnormalities and irregularities of APC/C cyclosome genes. We conclude that the expression of proteasome subunit genes and genes for ubiquitin ligases can contribute to prognostic classification of MBs. The therapeutic value of targeting proteasome subunits and ubiquitin ligases in the various subgroups of MB remains to be determined separately for each classification of MB.
Collapse
Affiliation(s)
- Jerry Vriend
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Rm134, BMSB, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| | - Hassan Marzban
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Rm134, BMSB, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
- Children's Hospital Research Institute of Manitoba (CHRIM), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
39
|
Dong F, Xie K, Chen Y, Yang Y, Mao Y. Polycistronic tRNA and CRISPR guide-RNA enables highly efficient multiplexed genome engineering in human cells. Biochem Biophys Res Commun 2017; 482:889-895. [PMID: 27890617 PMCID: PMC5284736 DOI: 10.1016/j.bbrc.2016.11.129] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 01/16/2023]
Abstract
CRISPR/Cas9 has been widely used for genomic editing in many organisms. Many human diseases are caused by multiple mutations. The CRISPR/Cas9 system provides a potential tool to introduce multiple mutations in a genome. To mimic complicated genomic variants in human diseases, such as multiple gene deletions or mutations, two or more small guide RNAs (sgRNAs) need to be introduced all together. This can be achieved by separate Pol III promoters in a construct. However, limited enzyme sites and increased insertion size lower the efficiency to make a construct. Here, we report a strategy to quickly assembly multiple sgRNAs in one construct using a polycistronic-tRNA-gRNA (PTG) strategy. Taking advantage of the endogenous tRNA processing system in mammalian cells, we efficiently express multiple sgRNAs driven using only one Pol III promoter. Using an all-in-one construct carrying PTG, we disrupt the deacetylase domain in multiple histone deacetylases (HDACs) in human cells simultaneously. We demonstrate that multiple HDAC deletions significantly affect the activation of the Wnt-signaling pathway. Thus, this method enables to efficiently target multiple genes and provide a useful tool to establish mutated cells mimicking human diseases.
Collapse
Affiliation(s)
- Fengping Dong
- Department of Biology, Pennsylvania State University, PA 16802, USA
| | - Kabin Xie
- Department of Plant Pathology and Environmental Microbiology, Pennsylvania State University, PA 16802, USA; College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yueying Chen
- Department of Plant Pathology and Environmental Microbiology, Pennsylvania State University, PA 16802, USA
| | - Yinong Yang
- Department of Plant Pathology and Environmental Microbiology, Pennsylvania State University, PA 16802, USA.
| | - Yingwei Mao
- Department of Biology, Pennsylvania State University, PA 16802, USA.
| |
Collapse
|
40
|
Epigenetics of Renal Development and Disease. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2016; 89:565-573. [PMID: 28018145 PMCID: PMC5168832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An understanding of epigenetics is indispensable to our understanding of gene regulation under normal and pathological states. This knowledge will help with designing better therapeutic approaches in regenerative tissue medicine. Epigenetics allows us to parse out the mechanisms by which transcriptional regulators gain access to specific gene loci thereby imprinting epigenetic information affecting chromatin function. This epigenetic memory forms the basis of cell lineage specification in multicellular organisms. Post-translational modifications to DNA and histones in the nucleosome core form characteristic epigenetic codes which are distinct for self-renewing and primed progenitor cell populations. Studies of chromatin modifiers and modifications in renal development and disease have been gaining momentum. Both congenital and adult renal diseases have a gene-environment component, which involves alterations to the epigenetic information imprinted during development. This epigenetic memory must be characterized to establish optimal treatment of both acute and chronic renal diseases.
Collapse
|
41
|
Lu H, Li G, Zhou C, Jin W, Qian X, Wang Z, Pan H, Jin H, Wang X. Regulation and role of post-translational modifications of enhancer of zeste homologue 2 in cancer development. Am J Cancer Res 2016; 6:2737-2754. [PMID: 28042497 PMCID: PMC5199751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 08/09/2016] [Indexed: 06/06/2023] Open
Abstract
Post-translational modifications (PTMs) are critical molecular events which alter protein conformation after their synthesis and diversity protein properties by modulating their stability, localization, interacting partners or the activity of their substrates, consequently exerting pivotal roles in regulating the functions of many important eukaryotic proteins. It has been well acknowledged that PTMs are of great importance in a broad range of biological processes such as gene regulation, cell proliferation, differentiation and apoptosis, tissue development, diseases, tumor progression and drug resistance. As the core and contributing catalytic subunit of Polycomb repressive complex 2(PRC2), Enhancer of zeste homolog 2 (EZH2) is a master epigenetic regulator, often serving as a highly conserved histone methyltransferase (HMTase) to induce histone H3 lysine 27 trimethylation (H3K27me3) and repress gene transcription and expression. Dysregulated EZH2 expression is frequently associated with cancer development and poor prognosis in a wide variety of cancers. Considered its essential role in carcinogenesis, EZH2 is a potential candidate for cancer targeted therapy. Remarkably, mounting evidence highlights that EZH2 expression, activity and stability can be regulated by PTMs including phosphorylation, acetylation, ubiquitination, sumoylation and GlcNAcylation aside from its well-validated modifications in transcriptional and post-transcriptional levels. However, the precise regulatory mechanisms underlying EZH2 PTMs and whether other types of PTMs orchestrate in EZH2 remain largely unclear. In this review, we summarize current advances in the understanding of EZH2 regulation by PTMs and their associated biological functions during tumorigenesis.
Collapse
Affiliation(s)
- Haiqi Lu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
- Laboratory of Cancer Biology, Provincial Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Guangliang Li
- Department of Medical Oncology, Zhejiang Cancer HospitalHangzhou, Zhejiang, China
| | - Chenyi Zhou
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Wei Jin
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Xiaoling Qian
- Laboratory of Cancer Biology, Provincial Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Zhuo Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
- Laboratory of Cancer Biology, Provincial Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Provincial Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| |
Collapse
|
42
|
Mateo Sánchez S, Freeman SD, Delacroix L, Malgrange B. The role of post-translational modifications in hearing and deafness. Cell Mol Life Sci 2016; 73:3521-33. [PMID: 27147466 PMCID: PMC11108544 DOI: 10.1007/s00018-016-2257-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/21/2016] [Accepted: 04/26/2016] [Indexed: 12/20/2022]
Abstract
Post-translational modifications (PTMs) are key molecular events that modify proteins after their synthesis and modulate their ultimate functional properties by affecting their stability, localisation, interaction potential or activity. These chemical changes expand the size of the proteome adding diversity to the molecular pathways governing the biological outcome of cells. PTMs are, thus, crucial in regulating a variety of cellular processes such as apoptosis, proliferation and differentiation and have been shown to be instrumental during embryonic development. In addition, alterations in protein PTMs have been implicated in the pathogenesis of many human diseases, including deafness. In this review, we summarize the recent progress made in understanding the roles of PTMs during cochlear development, with particular emphasis on the enzymes driving protein phosphorylation, acetylation, methylation, glycosylation, ubiquitination and SUMOylation. We also discuss how these enzymes may contribute to hearing impairment and deafness.
Collapse
Affiliation(s)
- Susana Mateo Sánchez
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium
| | - Stephen D Freeman
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium
| | - Laurence Delacroix
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium
| | - Brigitte Malgrange
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium.
| |
Collapse
|
43
|
Epigenetics mechanisms in renal development. Pediatr Nephrol 2016; 31:1055-60. [PMID: 26493068 PMCID: PMC4841758 DOI: 10.1007/s00467-015-3228-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 01/03/2023]
Abstract
Appreciation for the role of epigenetic modifications in the diagnosis and treatment of diseases is fast gaining attention. Treatment of chronic kidney disease stemming from diabetes or hypertension as well as Wilms tumor will all profit from knowledge of the changes in the epigenomic landscapes. To do so, it is essential to characterize the epigenomic modifiers and their modifications under normal physiological conditions. The transcription factor Pax2 was identified as a major epigenetic player in the early specification of the kidney. Notably, the progenitors of all nephrons that reside in the cap mesenchyme display a unique bivalent histone signature (expressing repressive epigenetic marks alongside activation marks) on lineage-specific genes. These cells are deemed poised for differentiation and commitment to the nephrogenic lineage. In response to the appropriate inducing signal, these genes lose their repressive histone marks, which allow for their expression in nascent nephron precursors. Such knowledge of the epigenetic landscape and the resultant cell fate or behavior in the developing kidney will greatly improve the overall success in designing regenerative strategies and tissue reprogramming methodologies from pluripotent cells.
Collapse
|
44
|
Vakilian H, Mirzaei M, Sharifi Tabar M, Pooyan P, Habibi Rezaee L, Parker L, Haynes PA, Gourabi H, Baharvand H, Salekdeh GH. DDX3Y, a Male-Specific Region of Y Chromosome Gene, May Modulate Neuronal Differentiation. J Proteome Res 2015; 14:3474-83. [PMID: 26144214 DOI: 10.1021/acs.jproteome.5b00512] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although it is apparent that chromosome complement mediates sexually dimorphic expression patterns of some proteins that lead to functional differences, there has been insufficient evidence following the manipulation of the male-specific region of the Y chromosome (MSY) gene expression during neural development. In this study, we profiled the expression of 23 MSY genes and 15 of their X-linked homologues during neural cell differentiation of NTERA-2 human embryonal carcinoma cell line (NT2) cells in three different developmental stages using qRT-PCR, Western blotting, and immunofluorescence. The expression level of 12 Y-linked genes significantly increased over neural differentiation, including RBMY1, EIF1AY, DDX3Y, HSFY1, BPY2, PCDH11Y, UTY, RPS4Y1, USP9Y, SRY, PRY, and ZFY. We showed that siRNA-mediated knockdown of DDX3Y, a DEAD box RNA helicase enzyme, in neural progenitor cells impaired cell cycle progression and increased apoptosis, consequently interrupting differentiation. Label-free quantitative shotgun proteomics based on a spectral counting approach was then used to characterize the proteomic profile of the cells after DDX3Y knockdown. Among 917 reproducibly identified proteins detected, 71 proteins were differentially expressed following DDX3Y siRNA treatment compared with mock treated cells. Functional grouping indicated that these proteins were involved in cell cycle, RNA splicing, and apoptosis, among other biological functions. Our results suggest that MSY genes may play an important role in neural differentiation and demonstrate that DDX3Y could play a multifunctional role in neural cell development, probably in a sexually dimorphic manner.
Collapse
Affiliation(s)
- Haghighat Vakilian
- Department of Stem Cells Biology & Technology, Royan Institute , Banihashem Sq., Banihashem St., Ressalat highway, Tehran, Iran
| | - Mehdi Mirzaei
- Department of Chemistry and Biomolecular Sciences, Macquarie University , Sydney, New South Wales 2109, Australia
| | - Mehdi Sharifi Tabar
- Department of Stem Cells Biology & Technology, Royan Institute , Banihashem Sq., Banihashem St., Ressalat highway, Tehran, Iran
| | - Paria Pooyan
- Department of Stem Cells Biology & Technology, Royan Institute , Banihashem Sq., Banihashem St., Ressalat highway, Tehran, Iran
| | - Lida Habibi Rezaee
- Department of Stem Cells Biology & Technology, Royan Institute , Banihashem Sq., Banihashem St., Ressalat highway, Tehran, Iran
| | - Lindsay Parker
- Department of Chemistry and Biomolecular Sciences, Macquarie University , Sydney, New South Wales 2109, Australia
| | - Paul A Haynes
- Department of Chemistry and Biomolecular Sciences, Macquarie University , Sydney, New South Wales 2109, Australia
| | - Hamid Gourabi
- Department of Genetics at Reproductive Biomedicine Research Center, Royan Institute , Banihashem Sq., Banihashem St., Ressalat highway, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells Biology & Technology, Royan Institute , Banihashem Sq., Banihashem St., Ressalat highway, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture , Sharif Esfahani Blvd, Park Street, Tehran, Iran
| | - Ghasem Hosseini Salekdeh
- Department of Stem Cells Biology & Technology, Royan Institute , Banihashem Sq., Banihashem St., Ressalat highway, Tehran, Iran.,Seed and Plant Improvement Institute's Campus, Agricultural Biotechnology Research Institute of Iran , Mahdasht Road, Karaj, Iran
| |
Collapse
|