1
|
Lin C, Hsu JL, Hsu YT, Fan KC, Wu SS, Lin MH, Guh JH, Yu CW. Design and synthesis of novel HDAC6 inhibitor dimer as HDAC6 degrader for cancer treatment by palladium catalysed dimerisation. J Enzyme Inhib Med Chem 2025; 40:2468355. [PMID: 40013582 PMCID: PMC11869342 DOI: 10.1080/14756366.2025.2468355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The enigmatic histone deacetylase 6 (HDAC6) is one of a kind among its family. Recent reports revealed that HDAC6 CD1 exhibits E3 ligase activity. Inspired by these researches, we attempted to develop drugs targeting HDAC6 via novel mechanism. Herein, we report a palladium catalysed transformation and purification method for hydroxamic acid dimers, and series of HDAC6 inhibitor-based dimer showing outstanding biological activities and capability of inducing auto-degradation. Our proof-of-concept was highlighted with 2-amino benzamide-based HDAC6 inhibitor dimers that exhibit great HDAC6 inhibition activity (3.9-15.4 nM), good HDAC1/6 selectivity (95-577), and excellent cytotoxicity against human hormone-resistant prostate cancer (HRPC) PC-3 and non-small cell lung cancer (NSCLC) A549 cell lines (5.9-11.3 and 6.6-17.9 μM, respectively) while simultaneously inducing HDAC6 degradation. These dimers not only induce apoptosis and autophagy but also interfere with kinetochore attachment by the detection of BUBR1 phosphorylation at S670.
Collapse
Affiliation(s)
- Ching Lin
- School of Pharmacy, National Taiwan University, Taipei, ROC
| | - Jui-Ling Hsu
- School of Pharmacy, National Taiwan University, Taipei, ROC
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan City, ROC
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, ROC
| | - Yu-Tung Hsu
- School of Pharmacy, National Taiwan University, Taipei, ROC
| | - Kuo-Chen Fan
- School of Pharmacy, National Taiwan University, Taipei, ROC
| | - Sian-Siou Wu
- School of Pharmacy, National Taiwan University, Taipei, ROC
| | - Miao-Hsia Lin
- Department and Graduate Institute of Medical Microbiology, College of Medicine, National Taiwan University, Taipei, ROC
| | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, Taipei, ROC
| | - Chao-Wu Yu
- School of Pharmacy, National Taiwan University, Taipei, ROC
| |
Collapse
|
2
|
Li Y, Sun J, Li X, Yu W, Ren J, Wang B, Han X, Ma L, Sun X, Teng W, Gu X, Ding Q, Li B. Donepezil-induced degradation of hERG potassium channel via lysosomal pathway is exacerbated by hypoxia. Eur J Pharmacol 2025; 996:177549. [PMID: 40157707 DOI: 10.1016/j.ejphar.2025.177549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Donepezil (DPZ), an acetylcholinesterase inhibitor for Alzheimer's disease, has drawn attention for causing prolonged QT interval and torsade de pointes (TdP). Acquired long QT syndrome (acLQTS) is usually caused by blockage of the cardiac potassium current IKr/hERG, which is essential for cardiac repolarization. This study aimed to investigate DPZ's effect on hERG channel and its cardiotoxic mechanism, particularly focusing on whether hypoxia increases the risk of DPZ-induced acLQTS. To explore these, we employed western blotting to analyze protein levels, the patch clamp technique to measure hERG current and the action potentials of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Additionally, immunoprecipitation was utilized to detect protein-protein interactions. Finally, optical mapping monitored guinea pig ECGs and APD, providing in vivo insights. Our results indicate that 24-h incubation with DPZ inhibits hERG protein levels and current in the plasma membrane. Mechanistically, DPZ induces an imbalance in hERG protein acetylation/ubiquitination and decreases the stability of hERG by promoting HDAC6 expression, and the ubiquitinated hERG protein was degraded at lysosomes via K63-polyubiquitin chains. DPZ affects hERG membrane protein via two pathways: it accelerates endocytosis and directs degradation via CHMP3 (a sorting protein of ESCRT-III), while inhibiting recycling through Rab11. Hypoxia exacerbates DPZ-induced hERG degradation and APD prolongation in guinea pigs and hiPSC-CMs. Collectively, DPZ reduces hERG protein stability in the membrane, promoting its degradation in lysosomes. Hypoxia further exacerbates the risk of arrhythmia caused by DPZ. These findings remind us to pay attention to acLQTS induced by DPZ inhibition of hERG in clinical applications.
Collapse
Affiliation(s)
- Yuexin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jinyang Sun
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiaoxu Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wenting Yu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jiacheng Ren
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Baoqiang Wang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiaoxia Han
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lu Ma
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiang Sun
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wei Teng
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiwei Gu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Qirui Ding
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Baoxin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
3
|
Yao J, Sun X, Chen Y, Xu X, Feng J, Zhang M, Liu X, Shi X. Histone deacetylase 6 inhibition attenuates pathological cardiac hypertrophy by promoting autophagy through MAP1LC3B ubiquitination. J Pathol 2025; 266:217-229. [PMID: 40212005 DOI: 10.1002/path.6419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/17/2025] [Accepted: 02/24/2025] [Indexed: 05/08/2025]
Abstract
Cardiac hypertrophy is an adaptive response of the heart to pathological stimuli that may lead to cardiac dysfunction and heart failure. Histone deacetylase 6 (HDAC6) participates in the progression of multiple cardiovascular diseases, including chronic hypertension, ischemic stroke, and acute cardiac injury. A delicate balance of autophagy regulates heart homeostasis, whereas dysregulated autophagy is involved in myocardial hypertrophy. However, whether HDAC6 participates in pathological cardiac hypertrophy by regulating autophagy remains unclear. In this paper, we report for the first time that HDAC6 is involved in isoproterenol (ISO)-induced pathological cardiac hypertrophy by interacting with and ubiquitinating MAP1LC3B. First, the expression level of HDAC6 was found to be increased in cardiac hypertrophy models induced by ISO. HDAC6 overexpression promoted the expression of hypertrophic genes and enhanced cell surface area. Conversely, HDAC6 inhibition attenuated ISO-induced hypertrophic responses. Mechanistically, HDAC6 promoted hypertrophic responses by negatively regulating autophagy. Furthermore, HDAC6 interacted with MAP1LC3B and mediated its monoubiquitination, thereby contributing to reduced MAP1LC3B levels and impaired autophagy. Inhibition of HDAC6 activity in mice abrogated the hypertrophic effects of ISO by restoring MAP1LC3B expression. In summary, our data demonstrate that HDAC6 participates in ISO-induced cardiac hypertrophy by limiting the availability of MAP1LC3B and suppressing autophagy. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, PR China
| | - Yousheng Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Xuan Xu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Junxiao Feng
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Mingming Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Xiangdong Liu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, PR China
| |
Collapse
|
4
|
Chen Y, Ding K, Zheng S, Gao S, Xu X, Wu H, Zhou F, Wang Y, Xu J, Wang C, Ling C, Xu J, Wang L, Wu Q, Giamas G, Chen G, Zhang J, Yi C, Ji J. Post-translational modifications in DNA damage repair: mechanisms underlying temozolomide resistance in glioblastoma. Oncogene 2025:10.1038/s41388-025-03454-5. [PMID: 40419791 DOI: 10.1038/s41388-025-03454-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/04/2025] [Accepted: 05/15/2025] [Indexed: 05/28/2025]
Abstract
Temozolomide (TMZ) resistance is one of the critical factors contributing to the poor prognosis of glioblastoma (GBM). As a first-line chemotherapeutic agent for GBM, TMZ exerts its cytotoxic effects through DNA alkylation. However, its therapeutic efficacy is significantly compromised by enhanced DNA damage repair (DDR) mechanisms in GBM cells. Although several DDR-targeting drugs have been developed, their clinical outcomes remain suboptimal. Post-translational modifications (PTMs) in GBM cells play a pivotal role in maintaining the genomic stability of DDR mechanisms, including methylguanine-DNA methyltransferase-mediated repair, DNA mismatch repair dysfunction, base excision repair, and double-strand break repair. This review focuses on elucidating the regulatory roles of PTMs in the intrinsic mechanisms underlying TMZ resistance in GBM. Furthermore, we explore the feasibility of enhancing TMZ-induced cytotoxicity by targeting PTM-related enzymatic to disrupt key steps in PTM-mediated DDR pathways. By integrating current preclinical insights and clinical challenges, this work highlights the potential of modulating PTM-driven networks as a novel therapeutic strategy to overcome TMZ resistance and improve treatment outcomes for GBM patients.
Collapse
Affiliation(s)
- Yike Chen
- Department of Plastic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kaikai Ding
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuyu Zheng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Songting Gao
- Guali Branch of the First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Xiaohui Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Haijian Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Fengqi Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Yongjie Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Jinfang Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Chun Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Chenhan Ling
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Jing Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Lin Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Qun Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, UK
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University, Oncology Department of the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- MOE Frontier Science Center for Brain Science & Brain-Machine Integration Zhejiang University, Hangzhou, Zhejiang, China.
| | - Chenggang Yi
- Department of Plastic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Jianxiong Ji
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Gu J, Yang M, Zhang L, Liu Y, Yan R, Pan D, Qian X, Hu H, Chu D, Hu C, Liu F, Cui H. Rhythmic TDP-43 affects RNA splicing of USP13, resulting in alteration of BMAL1 ubiquitination. J Cell Biol 2025; 224:e202405142. [PMID: 40202498 PMCID: PMC11980682 DOI: 10.1083/jcb.202405142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 11/20/2024] [Accepted: 02/11/2025] [Indexed: 04/10/2025] Open
Abstract
Circadian rhythm disorders are common characteristics of neurodegenerative diseases. The pathological aggregation of transactive response DNA-binding protein 43 (TDP-43) is associated with multiple neurodegenerative diseases, such as amyotrophic lateral sclerosis. However, the relationship between TDP-43 and circadian rhythm remains unknown. Here, we found that TDP-43 is rhythmically expressed both in vivo and in vitro. TDP-43 knockdown affected the expression of circadian genes, including BMAL1, CLOCK, CRY1, and PER2, and impaired autonomous circadian wheel behavior, cognitive functions, and balance abilities in mice. Furthermore, TDP-43 knockdown induced aberrant splicing of ubiquitin-specific peptidase 13 (USP13) and blocked USP13 rhythmic expression, enhancing the ubiquitination of BMAL1. Meanwhile, TDP-43 knockdown altered the rhythmic expression of phospho-AMPKα (Thr172) and platelet-type phosphofructokinase (PFKP), which may change cellular glucose uptake and ATP production. Our findings further the understanding of the role of TDP-43 dysfunction in circadian rhythm disruption in neurodegenerative diseases and provide new mechanistic evidence supporting the interaction between circadian rhythm disruption and neurodegeneration.
Collapse
Affiliation(s)
- Jianlan Gu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Mingming Yang
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Liti Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Yuxiao Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Ruolan Yan
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Danmin Pan
- Department of Cell Biology, School of Life Sciences, Nantong University, Nantong, China
| | - Xiaowei Qian
- Department of Cell Biology, School of Life Sciences, Nantong University, Nantong, China
| | - Hanjing Hu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Dandan Chu
- Department of Biochemistry and Molecular Biology, School of Medicine, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Chen Hu
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Hengxiang Cui
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Chi Z, Do VQ, Kausar R, Kim HK, Nguyen NTT, Le TPH, Lee J, Baek IJ, Lee SW, Kim JH, Lee SY. HDAC6 inhibition upregulates endothelial SOD3 expression via Sp1 acetylation and attenuates angiotensin II-induced hypertension. FEBS J 2025; 292:2624-2644. [PMID: 39957045 DOI: 10.1111/febs.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/16/2024] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
Extracellular superoxide dismutase (SOD3) plays an important role in maintaining vascular redox homeostasis by eliminating superoxides. The angiotensin II (AngII) peptide mediates vasoconstriction in part via reactive oxygen species (ROS) but has pathologic effects when elevated in adults. Histone deacetylase 6 (HDAC6) modulates the acetylation of non-histone substrates and is associated with hypertensive disorders. Here, we investigated the potential regulation of SOD3 by HDAC6 in human aortic endothelial cells (HAECs) and its implications for AngII-induced oxidative stress and hypertension. HDAC6 inhibition (via the specific inhibitor tubastatin A (TubA), gene knockdown, or a deacetylase activity-deficient mutant) significantly increased SOD3 protein and mRNA expression but did not affect SOD1 or SOD2 protein levels. Conversely, AngII downregulated SOD3 levels and increased ROS and superoxide levels; these effects were antagonized by TubA. We confirmed that the transcription factor Sp1 mediates TubA-induced as well as basal SOD3 expression. Notably, TubA strongly augmented Sp1 acetylation at lysine 703, which activated Sp1 binding to the proximal SOD3 promoter region and, consequently, SOD3 expression. Alternatively, AngII decreased Sp1 acetylation, and TubA-mediated SOD3 induction was reduced upon overexpression of an acetylation-resistant Sp1 mutant (K703R) compared to that by the wild-type protein. Consistent with these findings, aortic SOD3 expression was significantly higher in HDAC6-deficient mice than in wild-type mice. Moreover, AngII infusion-mediated blood pressure elevation was reduced in HDAC6-deficient mice compared with that in wild-type mice. Collectively, our results suggest that HDAC6 inhibition leads to SOD3 upregulation by enhancing Sp1 acetylation in HAECs, thereby mitigating AngII-induced oxidative stress and hypertension.
Collapse
Affiliation(s)
- Zhexi Chi
- Department of Anesthesiology and Pain Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Van Quan Do
- Department of Anesthesiology and Pain Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Rukhsana Kausar
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Korea
| | - Hyun Kyung Kim
- Department of Anesthesiology and Pain Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Nga Thi Thanh Nguyen
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea
| | - Truc Phan Hoang Le
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea
| | - Jungwoo Lee
- Department of Anesthesiology and Pain Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - In-Jeoung Baek
- Department of Cell and Genetic Engineering, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-Wook Lee
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Hyung Kim
- Department of Anesthesiology and Pain Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Sang Yoon Lee
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea
| |
Collapse
|
7
|
Iegiani G, Pallavicini G, Pezzotta A, Brix A, Ferraro A, Gai M, Boda E, Bielas SL, Pistocchi A, Di Cunto F. CITK modulates BRCA1 recruitment at DNA double strand breaks sites through HDAC6. Cell Death Dis 2025; 16:320. [PMID: 40254670 PMCID: PMC12009987 DOI: 10.1038/s41419-025-07655-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025]
Abstract
Citron Kinase (CITK) is a protein encoded by the CIT gene, whose pathogenic variants underlie microcephalic phenotypes that characterize MCPH17 syndrome. In neural progenitors, CITK loss leads to microtubule instability, resulting in mitotic spindle positioning defects, cytokinesis failure, and accumulation of DNA double strand breaks (DSBs), ultimately resulting in TP53-dependent senescence and apoptosis. Although DNA damage accumulation has been associated with impaired homologous recombination (HR), the role of CITK in this process and whether microtubule dynamics are involved is still unknown. In this report we show that CITK is required for proper BRCA1 localization at sites of DNA DSBs. We found that CITK's scaffolding, rather than its catalytic activity, is necessary for maintaining BRCA1 interphase levels in progenitor cells during neurodevelopment. CITK regulates the nuclear levels of HDAC6, a modulator of both microtubule stability and DNA damage repair. Targeting HDAC6 in CITK-deficient cells increases microtubule stability and recovers BRCA1 localization defects and DNA damage levels to that detected in controls. In addition, the CIT-HDAC6 axis is functionally relevant in a MCPH17 zebrafish model, as HDAC6 targeting recovers the head size phenotype produced by interfering with the CIT orthologue gene. These data provide novel insights into the functional interplay between HR and microtubule dynamics and into the pathogenesis of CITK based MCPH17, which may be relevant for development of therapeutic strategies.
Collapse
Affiliation(s)
- Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Alex Pezzotta
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Alessia Brix
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Alessia Ferraro
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Marta Gai
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Enrica Boda
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy
| | - Stephanie L Bielas
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anna Pistocchi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy.
- Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Torino, Italy.
| |
Collapse
|
8
|
Duan J, Chen T, Li Q, Zhang Y, Lu T, Xue J, Sun Y, Gao L, Zhang Y. Protein arginine methyltransferase 6 enhances immune checkpoint blockade efficacy via the STING pathway in MMR-proficient colorectal cancer. J Immunother Cancer 2025; 13:e010639. [PMID: 40086819 PMCID: PMC11907083 DOI: 10.1136/jitc-2024-010639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The emergence of immunotherapy has revolutionized the paradigm of cancer treatment with immune checkpoint blockades (ICB) in solid cancers, including colorectal cancer (CRC). However, only a small subset of CRC patients harboring deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) benefits from ICB therapy. A very limited response to ICB therapy has been achieved in MMR-proficient CRC, representing a significant challenge limiting the clinical application of immunotherapy. MMR is the critical DNA repair pathway that maintains genomic integrity by correcting DNA mismatches, which is mediated by the MutSα or MutSβ complex consisting of MSH2 with MSH6 and MSH3, respectively. Given that MMR status directs effective immune response, we sought to determine whether targeting MMR capacity boosts ICB efficacy. METHODS Azoxymethane/dextran sodium sulfate (AOM/DSS)-induced CRC and xenograft model were used to evaluate the function of PRMT6 and response to PRMT6 inhibitor EPZ020411 and combination therapy of PD1 and EPZ020411. Biochemical assays were performed to elucidate the underlying mechanism of PRMT6-mediated MSH2 methylation and immune evasion. RESULTS We have identified PRMT6 as a crucial regulator of MMR capacity via MSH2 dimethylation at R171 and R219. Such a modification abrogates its MMR capacity and prevents the recruitment of MSH3 and MSH6. PRMT6 loss or inhibition triggers cytosolic DNA accumulation and cGAS-STING signaling activation, leading to enhanced immune response in PRMT6-deficient colon tumors or xenografts. Pharmacological inhibition of PRMT6 using EPZ020411 promotes mutagenesis and destabilizes MutSα or MutSβ assembly, and prolonged EPZ020411 exposure maintains an MSI-like phenotype in microsatellite stability (MSS) cells. EPZ020411 treatment sensitizes ICB efficacy of MSS cells, but not MSI cells in vivo. Similar effects have been observed in MSS colon tumors induced by AOM/DSS. CONCLUSIONS Our study provides a preclinical proof of concept to overcome resistance to immunotherapy by targeting PRMT6 in CRC with MSS.
Collapse
Affiliation(s)
- Jinlin Duan
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Pathology, Tongren Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tao Chen
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University, Shanghai, China
| | - Qiwei Li
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Zhang
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Shanghai, China
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Ting Lu
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Junyan Xue
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yang Sun
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Ling Gao
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yonglong Zhang
- Laboratory of Targeted Therapy and Precision Medicine, Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
- Department of General Surgery, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Sun L, Fan G, Zhang Z, Chang D, Zhang X, Zhang T, Geng J, Zhang X, Lin M, Hu C, Zhou J, Wang M, Cao L, Zhang M, He B, Zhang S, Wang C. Phosphorylation of SIRT7 by ATM causes DNA mismatch repair downregulation and adaptive mutability during chemotherapy. Cell Rep 2025; 44:115269. [PMID: 39908142 DOI: 10.1016/j.celrep.2025.115269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/03/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Drug resistance significantly limits the efficacy of chemotherapy. The DNA mismatch repair (MMR) system maintains genomic stability by correcting DNA errors. During DNA-damaging treatments, cancer cells transiently increase their adaptive mutability, also known as microsatellite instability (MSI), to evade therapeutic pressure through MMR downregulation, conferring drug resistance. However, an understanding of the underlying mechanisms of MMR protein downregulation under DNA-damaging drugs remains limited. Our study reveals a negative correlation between SIRT7 protein levels and MMR core protein MSH2 levels in cervical and lung cancer tissues. SIRT7 destabilizes MSH2, promoting MSI and mutagenesis. Molecularly, DNA damage triggers ATM kinase-dependent phosphorylation and subcellular redistribution of SIRT7. Phosphorylated SIRT7 interacts with and deacetylates MSH2, impairing MMR, and inducing MSI and drug resistance. Our findings suggest that SIRT7 drives MMR downregulation under therapeutic stress and that ATM-dependent phosphorylation of SIRT7 may serve as a predictive biomarker for chemotherapeutic efficacy and a target for cancer treatment.
Collapse
Affiliation(s)
- Lianhui Sun
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Guangjian Fan
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Zhuqing Zhang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Dong Chang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Xiaoyu Zhang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Tongqing Zhang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Jichuan Geng
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Xiaoxia Zhang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Menghan Lin
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Chen Hu
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Jiaqi Zhou
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Mengxue Wang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Liu Cao
- Health Sciences Institute, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| | - Mary Zhang
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R., Detroit, MI 48201, USA
| | - Baokun He
- Institute of Chinese Materia Medica, The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, China
| | - Shengping Zhang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China.
| | - Chuangui Wang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China.
| |
Collapse
|
10
|
Zheng H, Yang X, Zhong H, Song C, Wu Z, Yang H. HDAC6 Facilitates PRV and VSV Infection by Inhibiting Type I Interferon Production. Viruses 2025; 17:90. [PMID: 39861880 PMCID: PMC11768819 DOI: 10.3390/v17010090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
HDAC6 modulates viral infection through diverse mechanisms. Here, we investigated the role of HDAC6 in influencing viral infection in pig cells with the aim of exploiting the potential antiviral gene targets in pigs. Using gene knockout and overexpression strategies, we found that HDAC6 knockout greatly reduced PRV and VSV infectivity, whereas HDAC6 overexpression increased their infectivity in PK15 cells. Mechanistic studies identified HDAC6 as a DNA damage inhibitor in PK15 cells. HDAC6 overexpression attenuated DNA damage levels, which can further reduce type I IFN production to promote viral infection. Conversely, HDAC6 deficiency can limit viral infection by increasing DNA damage-mediated type I IFN production. This work demonstrates that HDAC6 affects the infection process of multiple viruses by modulating type I IFN production, highlighting a regulatory role of HDAC6 linking host immune response and viral infection levels in pig cells.
Collapse
Affiliation(s)
- Hu Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Xiaohui Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Haiwen Zhong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Changxu Song
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu 527400, China
| | - Huaqiang Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu 527400, China
| |
Collapse
|
11
|
Lohner H, Han X, Ren J, Liang S, Liang R, Wang H. HDAC6-Mediated FoxO1 Acetylation And Phosphorylation Control Periodontal Inflammatory Responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627820. [PMID: 39713362 PMCID: PMC11661216 DOI: 10.1101/2024.12.10.627820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Post-translational modifications (PTMs) are critical regulators of protein function and cellular signaling. While histone deacetylation by histone deacetylases (HDACs) is well established, the role of specific HDACs in modulating non-histone protein PTMs, particularly in an infectious context, is poorly understood. Here, we reveal a pivotal role for HDAC6 in orchestrating periodontal inflammation through its dual regulatory effects on FoxO1 acetylation and phosphorylation. Using Porphyromonas gingivalis , a key periodontal pathogen, as a model pathogen, we observed that infection induces HDAC6 activation, driving inflammatory responses via modulating FoxO1 activity. HDAC6 depletion increased FoxO1 acetylation and phosphorylation, leading to its cytoplasmic sequestration and subsequent suppression of FoxO1- mediated pro-inflammatory cytokine production in macrophages. Mechanistically, HDAC6 deficiency not only directly enhances the acetylation of FoxO1 but also upregulates the expression of Rictor, a critical component of the mTORC2 complex, thereby promoting Akt phosphorylation and subsequently FoxO1 phosphorylation. This results in its cytoplasmic retention and attenuated inflammatory transcriptional activity. Functional studies demonstrated that HDAC6 depletion suppressed the production of key inflammatory mediators, including TNFα, IL-6, IL-12p40, and MIP-2, while promoting macrophage polarization toward anti-inflammatory M2 phenotypes. In vivo , using oral gavage infection and ligature-induced mouse periodontitis models, HDAC6 deficiency significantly reduced inflammatory cell infiltration in gingival tissues and protected against alveolar bone loss. These findings establish HDAC6 as a central regulator of periodontal inflammation, acting through the coordinated modulation of FoxO1 acetylation and phosphorylation. Beyond its role in oral pathology, HDAC6 may serve as a promising therapeutic target for managing inflammatory diseases linked to immune dysregulation.
Collapse
|
12
|
Spallotta F, Illi B. The Role of HDAC6 in Glioblastoma Multiforme: A New Avenue to Therapeutic Interventions? Biomedicines 2024; 12:2631. [PMID: 39595195 PMCID: PMC11591585 DOI: 10.3390/biomedicines12112631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Despite the great advances in basic research results, glioblastoma multiforme (GBM) still remains an incurable tumour. To date, a GBM diagnosis is a death sentence within 15-18 months, due to the high recurrence rate and resistance to conventional radio- and chemotherapy approaches. The effort the scientific community is lavishing on the never-ending battle against GBM is reflected by the huge number of clinical trials launched, about 2003 on 10 September 2024. However, we are still far from both an in-depth comprehension of the biological and molecular processes leading to GBM onset and progression and, importantly, a cure. GBM is provided with high intratumoral heterogeneity, immunosuppressive capacity, and infiltrative ability due to neoangiogenesis. These features impact both tumour aggressiveness and therapeutic vulnerability, which is further limited by the presence in the tumour core of niches of glioblastoma stem cells (GSCs) that are responsible for the relapse of this brain neoplasm. Epigenetic alterations may both drive and develop along GBM progression and also rely on changes in the expression of the genes encoding histone-modifying enzymes, including histone deacetylases (HDACs). Among them, HDAC6-a cytoplasmic HDAC-has recently gained attention because of its role in modulating several biological aspects of GBM, including DNA repair ability, massive growth, radio- and chemoresistance, and de-differentiation through primary cilia disruption. In this review article, the available information related to HDAC6 function in GBM will be presented, with the aim of proposing its inhibition as a valuable therapeutic route for this deadly brain tumour.
Collapse
Affiliation(s)
- Francesco Spallotta
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy;
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Barbara Illi
- Institute of Molecular Biology and Pathology, National Research Council (IBPM-CNR), 00185 Rome, Italy
| |
Collapse
|
13
|
Hu H, Wang Q, Zhang Y, Yang S, Shen A, Yan J, Zhao D, Hu B. Effects of a novel HDAC6-selective inhibitor's radiosensitization on cancer cells. Mol Biol Rep 2024; 51:1151. [PMID: 39537948 DOI: 10.1007/s11033-024-10084-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The radiation sensitivity of tumor cells is a critical determinant of their therapeutic response to radiotherapy. Histone deacetylase 6 (HDAC6), beyond its known role in modulating tubulin acetylation and influencing cell motility, is also involved in the DNA damage response, potentially enhancing tumor cell radiosensitivity. Targeted HDAC6 inhibitors have shown substantial promise in preclinical studies aimed at increasing radiosensitivity and inhibiting cellular migration. METHODS A new HDAC inhibitor, named OXHA, was designed by substituting the phenyl cap of SAHA with an N,5-diphenyloxazole-2-carboxamide group. The inhibitory activity of OXHA was evaluated via in vitro enzymatic assays. Its effects on tumor cell migration and radiosensitization potential were assessed using scratch wound healing assays, micronucleus formation, and clonogenic survival assays. RESULT Enzymatic assays confirmed OXHA's selective inhibition of HDAC6. Compared to SAHA, OXHA significantly increased α-tubulin acetylation while minimally impacting histone H3 acetylation, indicating a high selectivity for HDAC6. In combination with X-ray irradiation, OXHA markedly impaired wound healing in A549 and HepG2 cells, enhanced micronucleus formation, and reduced clonogenic survival across multiple tumor lines. CONCLUSION OXHA exhibits potent and selective HDAC6 inhibition, effectively impeding tumor cell migration and enhancing radiosensitivity across multiple cell lines. These findings suggest that OXHA has strong potential as a therapeutic strategy to improve radiotherapy efficacy.
Collapse
Affiliation(s)
- Huixiao Hu
- School of Public Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Qi Wang
- School of Public Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yuni Zhang
- School of Public Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Shuhua Yang
- School of Public Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Aihua Shen
- School of Public Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Junfang Yan
- School of Public Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Denggao Zhao
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, China.
| | - Burong Hu
- School of Public Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
14
|
Peng J, Liu H, Liu Y, Liu J, Zhao Q, Liu W, Niu H, Xue H, Sun J, Wu J. HDAC6 mediates tumorigenesis during mitosis and the development of targeted deactivating agents. Bioorg Chem 2024; 153:107818. [PMID: 39288633 DOI: 10.1016/j.bioorg.2024.107818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/19/2024]
Abstract
Epigenetics, particularly deacetylation, plays a critical role in tumorigenesis as many carcinogens are under tight control by post-translational modification. HDAC6, an important and special histone deacetylase (HDAC) family member, has been indicated to increase carcinogenesis through various functions. Recent studies demonstrated the effects of HDAC6 inhibitors in mitotic arrest, however, detailed mechanisms still remain unknown. Herein, we review and summarize HDAC6-associated proteins that have been implicated in important roles in mitosis. We also discuss the development of medicinal agents targeting HDAC6.
Collapse
Affiliation(s)
- Jie Peng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Hongyan Liu
- The People's Hospital of Zhaoyuan City, No. 168 Yingbin Road, Zhaoyuan 265400, Shandong Province, PR China
| | - Yujing Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jingqian Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Qianlong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Wenjia Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Haoqian Niu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Haoyu Xue
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jie Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jingde Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
15
|
Ferreira-Silva GÁ, Rodrigues DA, Pressete CG, Caixeta ES, Gamero AMC, Miyazawa M, Hanemann JAC, Fraga CAM, Aissa AF, Ionta M. Selective inhibition of HDAC6 by N-acylhydrazone derivative reduces the proliferation and induces senescence in carcinoma hepatocellular cells. Toxicol In Vitro 2024; 99:105884. [PMID: 38945376 DOI: 10.1016/j.tiv.2024.105884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
Hepatocellular carcinoma (HCC) is a significant contributor to cancer-related deaths globally. Systemic therapy is the only treatment option for HCC at an advanced stage, with limited therapeutic response. In this study, we evaluated the antitumor potential of four N-acylhydrazone (NAH) derivatives, namely LASSBio-1909, 1911, 1935, and 1936, on HCC cell lines. We have previously demonstrated that the aforementioned NAH derivatives selectively inhibit histone deacetylase 6 (HDAC6) in lung cancer cells, but their effects on HCC cells have not been explored. Thus, the present study aimed to evaluate the effects of NAH derivatives on the proliferative behavior of HCC cells. LASSBio-1911 was the most cytotoxic compound against HCC cells, however its effects were minimal on normal cells. Our results showed that LASSBio-1911 inhibited HDAC6 in HCC cells leading to cell cycle arrest and decreased cell proliferation. There was also an increase in the frequency of cells in mitosis onset, which was associated with disturbing mitotic spindle formation. These events were accompanied by elevated levels of CDKN1A mRNA, accumulation of CCNB1 protein, and sustained ERK1 phosphorylation. Furthermore, LASSBio-1911 induced DNA damage, resulting in senescence and/or apoptosis. Our findings indicate that selective inhibition of HDAC6 may provide an effective therapeutic strategy for the treatment of advanced HCC, including tumor subtypes with integrated viral genome. Further, in vivo studies are required to validate the antitumor effect of LASSBio-1911 on liver cancer.
Collapse
Affiliation(s)
| | - Daniel Alencar Rodrigues
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, CCS, Rio de Janeiro, RJ, Brazil
| | | | | | - Angel Mauricio Castro Gamero
- Human Genetics Laboratory, Institute of Natural Science, Federal University of Alfenas, zip-code 37130-001, Alfenas, MG, Brazil
| | - Marta Miyazawa
- School of Dentistry, Federal University of Alfenas, 37130-001 MG, Brazil
| | | | - Carlos Alberto Manssour Fraga
- Laboratory of Evaluation and Synthesis of Bioactive Substances (LASSBio), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, CCS, Rio de Janeiro, RJ, Brazil
| | - Alexandre Ferro Aissa
- Institute of Biomedical Sciences, Federal University of Alfenas, MG 37130-001, Brazil.
| | - Marisa Ionta
- Institute of Biomedical Sciences, Federal University of Alfenas, MG 37130-001, Brazil.
| |
Collapse
|
16
|
Mao C, Li S, Che J, Liu D, Mao X, Rao H. The ubiquitin ligase UBR4 and the deubiquitylase USP5 modulate the stability of DNA mismatch repair protein MLH1. J Biol Chem 2024; 300:107592. [PMID: 39032648 PMCID: PMC11375253 DOI: 10.1016/j.jbc.2024.107592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/29/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024] Open
Abstract
MLH1 plays a critical role in DNA mismatch repair and genome maintenance. MLH1 deficiency promotes cancer development and progression, but the mechanism underlying MLH1 regulation remains enigmatic. In this study, we demonstrated that MLH1 protein is degraded by the ubiquitin-proteasome system and have identified vital cis-elements and trans-factors involved in MLH1 turnover. We found that the region encompassing the amino acids 516 to 650 is crucial for MLH1 degradation. The mismatch repair protein PMS2 may shield MLH1 from degradation as it binds to the MLH1 segment key to its turnover. Furthermore, we have identified the E3 ubiquitin ligase UBR4 and the deubiquitylase USP5, which oppositely modulate MLH1 stability. In consistence, UBR4 or USP5 deficiency affects the cellular response to nucleotide analog 6-TG, supporting their roles in regulating mismatch repair. Our study has revealed important insights into the regulatory mechanisms underlying MLH1 proteolysis, critical to DNA mismatch repair related diseases.
Collapse
Affiliation(s)
- Chenyu Mao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Siqi Li
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jun Che
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Dongzhou Liu
- Department of Rheumatology and Immunology, Shenzhen People's Hospital, Shenzhen, Guangdong, China; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xinliang Mao
- Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Hai Rao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China; Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
17
|
Seane EN, Nair S, Vandevoorde C, Joubert A. Mechanistic Sequence of Histone Deacetylase Inhibitors and Radiation Treatment: An Overview. Pharmaceuticals (Basel) 2024; 17:602. [PMID: 38794172 PMCID: PMC11124271 DOI: 10.3390/ph17050602] [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: 03/27/2024] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Histone deacetylases inhibitors (HDACis) have shown promising therapeutic outcomes in haematological malignancies such as leukaemia, multiple myeloma, and lymphoma, with disappointing results in solid tumours when used as monotherapy. As a result, combination therapies either with radiation or other deoxyribonucleic acid (DNA) damaging agents have been suggested as ideal strategy to improve their efficacy in solid tumours. Numerous in vitro and in vivo studies have demonstrated that HDACis can sensitise malignant cells to both electromagnetic and particle types of radiation by inhibiting DNA damage repair. Although the radiosensitising ability of HDACis has been reported as early as the 1990s, the mechanisms of radiosensitisation are yet to be fully understood. This review brings forth the various protocols used to sequence the administration of radiation and HDACi treatments in the different studies. The possible contribution of these various protocols to the ambiguity that surrounds the mechanisms of radiosensitisation is also highlighted.
Collapse
Affiliation(s)
- Elsie Neo Seane
- Department of Radiography, School of Health Care Sciences, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
- Department of Medical Imaging and Therapeutic Sciences, Faculty of Health and Wellness, Cape Peninsula University of Technology, Cape Town 7530, South Africa
- Radiation Biophysics Division, Separate Sector Cyclotron (SSC) Laboratory, iThemba LABS, Cape Town 7131, South Africa;
| | - Shankari Nair
- Radiation Biophysics Division, Separate Sector Cyclotron (SSC) Laboratory, iThemba LABS, Cape Town 7131, South Africa;
| | - Charlot Vandevoorde
- GSI Helmholtz Centre for Heavy Ion Research, Department of Biophysics, 64291 Darmstadt, Germany;
| | - Anna Joubert
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa;
| |
Collapse
|
18
|
Li S, Wu W, Yang B, Liu Z, Duan X, Sun X, Liu H, Zhang S, Zhou Y, Wu W. Histone deacetylase 6 suppression of renal tubular epithelial cell promotes interstitial mineral deposition via alpha-tubulin acetylation. Cell Signal 2024; 116:111057. [PMID: 38242268 DOI: 10.1016/j.cellsig.2024.111057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/04/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
Randall's plaque (RP) is derived from interstitial mineral deposition and is highly prevalent in renal calcium oxalate (CaOx) stone disease, which is predictive of recurrence. This study shows that histone deacetylase 6 (HDAC6) levels are suppressed in renal tubular epithelial cells in RP samples, in kidney tissues of hyperoxaluria rats, and in hyper-oxalate-treated or mineralized cultured renal tubular epithelial (MDCK) cells in vitro. Mineral deposition in MDCK cells was exacerbated by HDAC6 inhibition but alleviated by HDAC6 overexpression. Surprisingly, the expression of some osteogenic-associated proteins, were not increased along with the increasing of mineral deposition, and result of single-cell RNA sequencing of renal papillae samples revealed that epithelial cells possess lower calcific activity, suggesting that osteogenic-transdifferentiation may not have actually occurred in tubular epithelial cells despite mineral deposition. The initial mineral depositions facilitated by HDAC6 inhibitor were localized in extracellular dome rather than inside the cells, moreover, suppression of HDAC6 significantly increased the calcium content of co-cultured renal interstitial fibroblasts (NRK49F) and enhanced mineral deposition of indirectly co-cultured NRK49F cells, suggesting that HDAC6 may influence trans-MDCK monolayer secretion of mineral. Further experiments revealed that this regulatory role was partially alpha-tubulinLys40 acetylation dependent. Collectively, these results suggest that hyper-oxalate exposure led to HDAC6 suppression in renal tubular epithelial cells, which may contribute to interstitial mineral deposition by promoting alpha-tubulinLys40 acetylation. Therapeutic agents that influence HDAC6 activity may be beneficial in preventing RP and CaOx stone formation.
Collapse
Affiliation(s)
- Shujue Li
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute Of Urology, Guangzhou, Guangdong 510230, China; Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Wenzheng Wu
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Baotong Yang
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Zezhen Liu
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute Of Urology, Guangzhou, Guangdong 510230, China
| | - Xiaolu Duan
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute Of Urology, Guangzhou, Guangdong 510230, China
| | - Xinyuan Sun
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute Of Urology, Guangzhou, Guangdong 510230, China
| | - Hongxing Liu
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute Of Urology, Guangzhou, Guangdong 510230, China
| | - Shike Zhang
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Yuhao Zhou
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Wenqi Wu
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Urology, Guangdong Engineering Research Center of Urinary Minimally invasive surgery Robot and Intelligent Equipment, Guangzhou Institute Of Urology, Guangzhou, Guangdong 510230, China; Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, China.
| |
Collapse
|
19
|
Du D, Yang Y, Zhang Y, Wang G, Chen L, Guan X, Rasmussen LJ, Liu D. MRE11A: a novel negative regulator of human DNA mismatch repair. Cell Mol Biol Lett 2024; 29:37. [PMID: 38486171 PMCID: PMC10938699 DOI: 10.1186/s11658-024-00547-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/08/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND DNA mismatch repair (MMR) is a highly conserved pathway that corrects DNA replication errors, the loss of which is attributed to the development of various types of cancers. Although well characterized, MMR factors remain to be identified. As a 3'-5' exonuclease and endonuclease, meiotic recombination 11 homolog A (MRE11A) is implicated in multiple DNA repair pathways. However, the role of MRE11A in MMR is unclear. METHODS Initially, short-term and long-term survival assays were used to measure the cells' sensitivity to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Meanwhile, the level of apoptosis was also determined by flow cytometry after MNNG treatment. Western blotting and immunofluorescence assays were used to evaluate the DNA damage within one cell cycle after MNNG treatment. Next, a GFP-heteroduplex repair assay and microsatellite stability test were used to measure the MMR activities in cells. To investigate the mechanisms, western blotting, the GFP-heteroduplex repair assay, and chromatin immunoprecipitation were used. RESULTS We show that knockdown of MRE11A increased the sensitivity of HeLa cells to MNNG treatment, as well as the MNNG-induced DNA damage and apoptosis, implying a potential role of MRE11 in MMR. Moreover, we found that MRE11A was largely recruited to chromatin and negatively regulated the DNA damage signals within the first cell cycle after MNNG treatment. We also showed that knockdown of MRE11A increased, while overexpressing MRE11A decreased, MMR activity in HeLa cells, suggesting that MRE11A negatively regulates MMR activity. Furthermore, we show that recruitment of MRE11A to chromatin requires MLH1 and that MRE11A competes with PMS2 for binding to MLH1. This decreases PMS2 levels in whole cells and on chromatin, and consequently comprises MMR activity. CONCLUSIONS Our findings reveal that MRE11A is a negative regulator of human MMR.
Collapse
Affiliation(s)
- Demin Du
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yueyan Yang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuanyuan Zhang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guanxiong Wang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Liying Chen
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiaowei Guan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Dekang Liu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
20
|
Lin Y, Jin X. Effect of ubiquitin protease system on DNA damage response in prostate cancer (Review). Exp Ther Med 2024; 27:33. [PMID: 38125344 PMCID: PMC10731405 DOI: 10.3892/etm.2023.12321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/26/2023] [Indexed: 12/23/2023] Open
Abstract
Genomic instability is an essential hallmark of cancer, and cellular DNA damage response (DDR) defects drive tumorigenesis by disrupting genomic stability. Several studies have identified abnormalities in DDR-associated genes, and a dysfunctional ubiquitin-proteasome system (UPS) is the most common molecular event in metastatic castration-resistant prostate cancer (PCa). For example, mutations in Speckle-type BTB/POZ protein-Ser119 result in DDR downstream target activation deficiency. Skp2 excessive upregulation inhibits homologous recombination repair and promotes cell growth and migration. Abnormally high expression of a deubiquitination enzyme, ubiquitin-specific protease 12, stabilizes E3 ligase MDM2, which further leads to p53 degradation, causing DDR interruption and genomic instability. In the present review, the basic pathways of DDR, UPS dysfunction, and its induced DDR alterations mediated by genomic instability, and especially the potential application of UPS and DDR alterations as biomarkers and therapeutic targets in PCa treatment, were described.
Collapse
Affiliation(s)
- Yan Lin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Oncology, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
21
|
Wang L, Yang S, Xue Y, Bo T, Xu J, Wang W. Mismatch Repair Protein Msh6 Tt Is Necessary for Nuclear Division and Gametogenesis in Tetrahymena thermophila. Int J Mol Sci 2023; 24:17619. [PMID: 38139447 PMCID: PMC10743813 DOI: 10.3390/ijms242417619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023] Open
Abstract
DNA mismatch repair (MMR) improves replication accuracy by up to three orders of magnitude. The MutS protein in E. coli or its eukaryotic homolog, the MutSα (Msh2-Msh6) complex, recognizes base mismatches and initiates the mismatch repair mechanism. Msh6 is an essential protein for assembling the heterodimeric complex. However, the function of the Msh6 subunit remains elusive. Tetrahymena undergoes multiple DNA replication and nuclear division processes, including mitosis, amitosis, and meiosis. Here, we found that Msh6Tt localized in the macronucleus (MAC) and the micronucleus (MIC) during the vegetative growth stage and starvation. During the conjugation stage, Msh6Tt only localized in MICs and newly developing MACs. MSH6Tt knockout led to aberrant nuclear division during vegetative growth. The MSH6TtKO mutants were resistant to treatment with the DNA alkylating agent methyl methanesulfonate (MMS) compared to wild type cells. MSH6Tt knockout affected micronuclear meiosis and gametogenesis during the conjugation stage. Furthermore, Msh6Tt interacted with Msh2Tt and MMR-independent factors. Downregulation of MSH2Tt expression affected the stability of Msh6Tt. In addition, MSH6Tt knockout led to the upregulated expression of several MSH6Tt homologs at different developmental stages. Msh6Tt is involved in macronuclear amitosis, micronuclear mitosis, micronuclear meiosis, and gametogenesis in Tetrahymena.
Collapse
Affiliation(s)
- Lin Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China; (L.W.); (S.Y.); (Y.X.); (T.B.)
| | - Sitong Yang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China; (L.W.); (S.Y.); (Y.X.); (T.B.)
| | - Yuhuan Xue
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China; (L.W.); (S.Y.); (Y.X.); (T.B.)
| | - Tao Bo
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China; (L.W.); (S.Y.); (Y.X.); (T.B.)
- Shanxi Key Laboratory of Biotechnology, Taiyuan 030006, China
| | - Jing Xu
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China; (L.W.); (S.Y.); (Y.X.); (T.B.)
- School of Life Science, Shanxi University, Taiyuan 030006, China
| | - Wei Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China; (L.W.); (S.Y.); (Y.X.); (T.B.)
- Shanxi Key Laboratory of Biotechnology, Taiyuan 030006, China
| |
Collapse
|
22
|
Duda J, Thomas SN. Interactions of Histone Deacetylase 6 with DNA Damage Repair Factors Strengthen its Utility as a Combination Drug Target in High-Grade Serous Ovarian Cancer. ACS Pharmacol Transl Sci 2023; 6:1924-1933. [PMID: 38107255 PMCID: PMC10723650 DOI: 10.1021/acsptsci.3c00215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 12/19/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is the deadliest gynecologic malignancy in women. The low survival rate is largely due to drug resistance. Approximately 80% of patients who initially respond to treatment relapse and become drug-resistant. The lack of effective second-line therapeutics remains a substantial challenge for BRCA-1/2 wild-type HGSOC patients. Histone Deacetylases (HDACs) are promising targets in HGSOC treatment; however, the mechanism and efficacy of HDAC inhibitors are understudied in HGSOC. In order to consider HDACs as a treatment target, an improved understanding of their function within HGSOC is required. This includes elucidating HDAC6-specific protein-protein interactions. In this study, we carried out substrate trapping followed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) to elucidate HDAC6 catalytic domain (CD)-specific interactors in the context of BRCA-1/2 wild-type HGSOC. Overall, this study identified new HDAC6 substrates that may be unique to HGSOC. The HDAC6-CD1 mutant condition contained the largest number of significant proteins compared to the CD2 mutant and the CD1/2 mutant conditions, suggesting the HDAC6-CD1 domain has catalytic activity that is independent of CD2. Among the identified substrates were proteins involved in DNA damage repair including PARP proteins. These findings further justify the use of HDAC inhibitors as a combination treatment with platinum chemotherapy agents and PARP inhibitors in HGSOC.
Collapse
Affiliation(s)
- Jolene
M. Duda
- Department
of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Stefani N. Thomas
- Department
of Laboratory Medicine and Pathology, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
23
|
Wu F, Muskat NH, Dvilansky I, Koren O, Shahar A, Gazit R, Elia N, Arbely E. Acetylation-dependent coupling between G6PD activity and apoptotic signaling. Nat Commun 2023; 14:6208. [PMID: 37798264 PMCID: PMC10556143 DOI: 10.1038/s41467-023-41895-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023] Open
Abstract
Lysine acetylation has been discovered in thousands of non-histone human proteins, including most metabolic enzymes. Deciphering the functions of acetylation is key to understanding how metabolic cues mediate metabolic enzyme regulation and cellular signaling. Glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme in the pentose phosphate pathway, is acetylated on multiple lysine residues. Using site-specifically acetylated G6PD, we show that acetylation can activate (AcK89) and inhibit (AcK403) G6PD. Acetylation-dependent inactivation is explained by structural studies showing distortion of the dimeric structure and active site of G6PD. We provide evidence for acetylation-dependent K95/97 ubiquitylation of G6PD and Y503 phosphorylation, as well as interaction with p53 and induction of early apoptotic events. Notably, we found that the acetylation of a single lysine residue coordinates diverse acetylation-dependent processes. Our data provide an example of the complex roles of acetylation as a posttranslational modification that orchestrates the regulation of enzymatic activity, posttranslational modifications, and apoptotic signaling.
Collapse
Affiliation(s)
- Fang Wu
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Natali H Muskat
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Inbar Dvilansky
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Omri Koren
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Anat Shahar
- Macromolecular Crystallography Research Center (MCRC), Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Roi Gazit
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Natalie Elia
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Eyal Arbely
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel.
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel.
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel.
| |
Collapse
|
24
|
Qiu L, Xu W, Lu X, Chen F, Chen Y, Tian Y, Zhu Q, Liu X, Wang Y, Pei XH, Xu X, Zhang J, Zhu WG. The HDAC6-RNF168 axis regulates H2A/H2A.X ubiquitination to enable double-strand break repair. Nucleic Acids Res 2023; 51:9166-9182. [PMID: 37503842 PMCID: PMC10516627 DOI: 10.1093/nar/gkad631] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/24/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023] Open
Abstract
Histone deacetylase 6 (HDAC6) mediates DNA damage signaling by regulating the mismatch repair and nucleotide excision repair pathways. Whether HDAC6 also mediates DNA double-strand break (DSB) repair is unclear. Here, we report that HDAC6 negatively regulates DSB repair in an enzyme activity-independent manner. In unstressed cells, HDAC6 interacts with H2A/H2A.X to prevent its interaction with the E3 ligase RNF168. Upon sensing DSBs, RNF168 rapidly ubiquitinates HDAC6 at lysine 116, leading to HDAC6 proteasomal degradation and a restored interaction between RNF168 and H2A/H2A.X. H2A/H2A.X is ubiquitinated by RNF168, precipitating the recruitment of DSB repair factors (including 53BP1 and BRCA1) to chromatin and subsequent DNA repair. These findings reveal novel regulatory machinery based on an HDAC6-RNF168 axis that regulates the H2A/H2A.X ubiquitination status. Interfering with this axis might be leveraged to disrupt a key mechanism of cancer cell resistance to genotoxic damage and form a potential therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Lingyu Qiu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Wenchao Xu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xiaopeng Lu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Feng Chen
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Yongcan Chen
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Yuan Tian
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Qian Zhu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xiangyu Liu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Yongqing Wang
- Division of Rheumatology and Immunology, University of Toledo Medical Center, 3120 Glendale Avenue, Toledo 43614, OH, USA
| | - Xin-Hai Pei
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xingzhi Xu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Cell Biology and Medical Genetics, Shenzhen University Medical School, Shenzhen 518055, China
| | - Jun Zhang
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Wei-Guo Zhu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, China
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
25
|
Qu M, Zhang H, Cheng P, Wubshet AK, Yin X, Wang X, Sun Y. Histone deacetylase 6's function in viral infection, innate immunity, and disease: latest advances. Front Immunol 2023; 14:1216548. [PMID: 37638049 PMCID: PMC10450946 DOI: 10.3389/fimmu.2023.1216548] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
In the family of histone-deacetylases, histone deacetylase 6 (HDAC6) stands out. The cytoplasmic class IIb histone deacetylase (HDAC) family is essential for many cellular functions. It plays a crucial and debatable regulatory role in innate antiviral immunity. This review summarises the current state of our understanding of HDAC6's structure and function in light of the three mechanisms by which it controls DNA and RNA virus infection: cytoskeleton regulation, host innate immune response, and autophagy degradation of host or viral proteins. In addition, we summed up how HDAC6 inhibitors are used to treat a wide range of diseases, and how its upstream signaling plays a role in the antiviral mechanism. Together, the findings of this review highlight HDAC6's importance as a new therapeutic target in antiviral immunity, innate immune response, and some diseases, all of which offer promising new avenues for the development of drugs targeting the immune response.
Collapse
Affiliation(s)
- Min Qu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Huijun Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengyuan Cheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Ashenafi Kiros Wubshet
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Department of Basic and Diagnostic Sciences, College of Veterinary Science, Mekelle University, Mekelle, Tigray, Ethiopia
| | - Xiangping Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yuefeng Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
26
|
Wang L, Xue Y, Yang S, Bo T, Xu J, Wang W. Mismatch Repair Protein Msh2 Is Necessary for Macronuclear Stability and Micronuclear Division in Tetrahymena thermophila. Int J Mol Sci 2023; 24:10559. [PMID: 37445734 DOI: 10.3390/ijms241310559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Mismatch repair (MMR) is a conserved mechanism that is primarily responsible for the repair of DNA mismatches during DNA replication. Msh2 forms MutS heterodimer complexes that initiate the MMR in eukaryotes. The function of Msh2 is less clear under different chromatin structures. Tetrahymena thermophila contains a transcriptionally active macronucleus (MAC) and a transcriptionally silent micronucleus (MIC) in the same cytoplasm. Msh2 is localized in the MAC and MIC during vegetative growth. Msh2 is localized in the perinuclear region around the MIC and forms a spindle-like structure as the MIC divides. During the early conjugation stage, Msh2 is localized in the MIC and disappears from the parental MAC. Msh2 is localized in the new MAC and new MIC during the late conjugation stage. Msh2 also forms a spindle-like structure with a meiotic MIC and mitotic gametic nucleus. MSH2 knockdown inhibits the division of MAC and MIC during vegetative growth and affects cellular proliferation. MSH2 knockdown mutants are sensitive to cisplatin treatment. MSH2 knockdown also affects micronuclear meiosis and gametogenesis during sexual development. Furthermore, Msh2 interacts with MMR-dependent and MMR-independent factors. Therefore, Msh2 is necessary for macronuclear stability, as well as micronuclear mitosis and meiosis in Tetrahymena.
Collapse
Affiliation(s)
- Lin Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Yuhuan Xue
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Sitong Yang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Tao Bo
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
- Shanxi Key Laboratory of Biotechnology, Taiyuan 030006, China
| | - Jing Xu
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
- School of Life Science, Shanxi University, Taiyuan 030006, China
| | - Wei Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
- Shanxi Key Laboratory of Biotechnology, Taiyuan 030006, China
| |
Collapse
|
27
|
He Q, Yu C, Li Y, Hao P, Mai H, Guo R, Zhong G, Zhang K, Wong C, Chen Q, Chen Y. ERRα contributes to HDAC6-induced chemoresistance of osteosarcoma cells. Cell Biol Toxicol 2023; 39:813-825. [PMID: 34524571 DOI: 10.1007/s10565-021-09651-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/03/2021] [Indexed: 10/20/2022]
Abstract
Chemotherapy resistance is an important problem for clinical therapy of osteosarcoma (OS). The potential effects of histone deacetylases (HDACs) on OS chemoresistance are studied. The expression of HDACs in OS cells resistance to doxorubicin (Dox) and cisplatin (CDDP) is checked. Among 11 members of HDACs, levels of HDAC6 are significantly upregulated in OS cells resistance to Dox and CDDP. Inhibition of HDAC6 via its specific inhibitor ACY1215 restores chemosensitivity of OS-resistant cells. Further, HDAC6 directly binds with estrogen-related receptors alpha (ERRα) to regulate its acetylation and protein stability. Inhibition of ERRα further strengthens ACY1215-increased chemosensitivity of OS-resistant cells. Mechanistically, K129 acetylation is the key residue for HDAC6-regulated protein levels of ERRα. Collectively, we find that ERRα contributes to HDAC6-induced chemoresistance of OS cells. Inhibition of HDAC6/ERRα axis might be a potential approach to overcome chemoresistance and improve therapy efficiency for OS treatment. 1. HDAC6 was significantly upregulated in Dox and CDDP resistant OS cells; 2. Inhibition of HDAC6 can restore chemosensitivity of OS cells; 3. HDAC6 binds with ERRα at K129 to decrease its acetylation and increase protein stability; 4. ERRα contributes to HDAC6-induced chemoresistance of OS cells.
Collapse
Affiliation(s)
- Qing He
- Department of Surgical Intensive Care Unit, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Changzhi Yu
- Department of Chinese Traditional Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yang Li
- Pediatric Hematology and Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Peng Hao
- Department of Surgical Intensive Care Unit, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hantao Mai
- Department of Surgical Intensive Care Unit, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ruilian Guo
- Department of Surgical Intensive Care Unit, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Guifang Zhong
- Department of Surgical Intensive Care Unit, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kelin Zhang
- Department of Surgical Intensive Care Unit, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chipiu Wong
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, China
| | - Qian Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, China
| | - Yantao Chen
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, China.
| |
Collapse
|
28
|
Li Z, Duan P, Qiu R, Fang L, Fang P, Xiao S. HDAC6 Degrades nsp8 of Porcine Deltacoronavirus through Deacetylation and Ubiquitination to Inhibit Viral Replication. J Virol 2023; 97:e0037523. [PMID: 37133375 PMCID: PMC10231189 DOI: 10.1128/jvi.00375-23] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/17/2023] [Indexed: 05/04/2023] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging swine enteropathogenic coronavirus that has the potential to infect humans. Histone deacetylase 6 (HDAC6) is a unique type IIb cytoplasmic deacetylase with both deacetylase activity and ubiquitin E3 ligase activity, which mediates a variety of cellular processes by deacetylating histone and nonhistone substrates. In this study, we found that ectopic expression of HDAC6 significantly inhibited PDCoV replication, while the reverse effects could be observed after treatment with an HDAC6-specific inhibitor (tubacin) or knockdown of HDAC6 expression by specific small interfering RNA. Furthermore, we demonstrated that HDAC6 interacted with viral nonstructural protein 8 (nsp8) in the context of PDCoV infection, resulting in its proteasomal degradation, which was dependent on the deacetylation activity of HDAC6. We further identified the key amino acid residues lysine 46 (K46) and K58 of nsp8 as acetylation and ubiquitination sites, respectively, which were required for HDAC6-mediated degradation. Through a PDCoV reverse genetics system, we confirmed that recombinant PDCoV with a mutation at either K46 or K58 exhibited resistance to the antiviral activity of HDAC6, thereby exhibiting higher replication compared with wild-type PDCoV. Collectively, these findings contribute to a better understanding of the function of HDAC6 in regulating PDCoV infection and provide new strategies for the development of anti-PDCoV drugs. IMPORTANCE As an emerging enteropathogenic coronavirus with zoonotic potential, porcine deltacoronavirus (PDCoV) has sparked tremendous attention. Histone deacetylase 6 (HDAC6) is a critical deacetylase with both deacetylase activity and ubiquitin E3 ligase activity and is extensively involved in many important physiological processes. However, little is known about the role of HDAC6 in the infection and pathogenesis of coronaviruses. Our present study demonstrates that HDAC6 targets PDCoV-encoded nonstructural protein 8 (nsp8) for proteasomal degradation through the deacetylation at the lysine 46 (K46) and the ubiquitination at K58, suppressing viral replication. Recombinant PDCoV with a mutation at K46 and/or K58 of nsp8 displayed resistance to the antiviral activity of HDAC6. Our work provides significant insights into the role of HDAC6 in regulating PDCoV infection, opening avenues for the development of novel anti-PDCoV drugs.
Collapse
Affiliation(s)
- Zhuang Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Panpan Duan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Runhui Qiu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Liurong Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Puxian Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Shaobo Xiao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
29
|
Park JC, Kim YJ, Han JH, Kim D, Park MJ, Kim J, Jang HK, Bae S, Cha HJ. MutSα and MutSβ as size-dependent cellular determinants for prime editing in human embryonic stem cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:914-922. [PMCID: PMC10280094 DOI: 10.1016/j.omtn.2023.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 05/10/2023] [Indexed: 06/22/2023]
Abstract
Precise genome editing in human pluripotent stem cells (hPSCs) has potential applications in isogenic disease modeling and ex vivo stem cell therapy, necessitating diverse genome editing tools. However, unlike differentiated somatic cells, hPSCs have unique cellular properties that maintain genome integrity, which largely determine the overall efficiency of an editing tool. Considering the high demand for prime editors (PEs), it is imperative to characterize the key molecular determinants of PE outcomes in hPSCs. Through homozygous knockout (KO) of MMR pathway key proteins MSH2, MSH3, and MSH6, we reveal that MutSα and MutSβ determine PE efficiency in an editing size-dependent manner. Notably, MSH2 perturbation disrupted both MutSα and MutSβ complexes, dramatically escalating PE efficiency from base mispair to 10 bases, up to 50 folds. Similarly, impaired MutSα by MSH6 KO improved editing efficiency from single to three base pairs, while defective MutSβ by MSH3 KO heightened efficiency from three to 10 base pairs. Thus, the size-dependent effect of MutSα and MutSβ on prime editing implies that MMR is a vital PE efficiency determinant in hPSCs and highlights the distinct roles of MutSα and MutSβ in its outcome.
Collapse
Affiliation(s)
- Ju-Chan Park
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yun-Jeong Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Jun Hee Han
- Department of Chemistry, Hanyang University, Seoul, Republic of Korea
| | - Dayeon Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Mihn Jeong Park
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Jumee Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Hyeon-Ki Jang
- Division of Chemical Engineering and Bioengineering, College of Art Culture and Engineering, Kangwon National University, Chuncheon, South Korea
| | - Sangsu Bae
- College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyuk-Jin Cha
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
30
|
Zhu Y, Feng M, Wang B, Zheng Y, Jiang D, Zhao L, Mamun MAA, Kang H, Nie H, Zhang X, Guo N, Qin S, Wang N, Liu H, Gao Y. New insights into the non-enzymatic function of HDAC6. Biomed Pharmacother 2023; 161:114438. [PMID: 37002569 DOI: 10.1016/j.biopha.2023.114438] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/11/2023] Open
Abstract
Histone deacetylase 6 (HDAC6) is a class IIb histone deacetylase that contains two catalytic domains and a zinc-finger ubiquitin binding domain (ZnF-UBP) domain. The deacetylation function of HDAC6 has been extensively studied with common substrates such as α-tubulin, cortactin, and Hsp90. Apart from its deacetylase activity, HDAC6 ZnF-UBP binds to unanchored ubiquitin of specific sequences and serves as a carrier for transporting aggregated proteins. As a result, aggresomes are formed and protein degradation is facilitated by the autophagy-lysosome pathway. This HDAC6-dependent microtubule transport can be used by cells to assemble and activate inflammasomes, which play a critical role in immune regulation. Even viruses can benefit from the carrier of HDAC6 to assist in uncoating their surfaces during their infection cycle. However, HDAC6 is also capable of blocking virus invasion and replication in a non-enzymatic manner. Given these non-enzymatic functions, HDAC6 is closely associated with various diseases, including neurodegeneration, inflammasome-associated diseases, cancer, and viral infections. Small molecule inhibitors targeting the ubiquitin binding pocket of HDAC6 have been investigated. In this review, we focus on mechanisms in non-enzymatic functions of HDAC6 and discuss the rationality and prospects of therapeutic strategies by intervening the activation of HDAC6 ZnF-UBP in concrete diseases.
Collapse
Affiliation(s)
- Yuanzai Zhu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Mengkai Feng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Bo Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
| | - Yichao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Dandan Jiang
- Department of Pharmacy, People's Hospital of Henan Province, Zhengzhou University, Henan 450001, China
| | - Lijuan Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - M A A Mamun
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Huiqin Kang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Haiqian Nie
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Xiya Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ningjie Guo
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Shangshang Qin
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ning Wang
- The School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
31
|
Xu W, Yan P, Zhou Z, Yao J, Pan H, Jiang L, Bo Z, Ni B, Sun M, Gao S, Huan C. HDAC6 Triggers the ATM-Dependent DNA Damage Response To Promote PRV Replication. Microbiol Spectr 2023; 11:e0213222. [PMID: 36951571 PMCID: PMC10101138 DOI: 10.1128/spectrum.02132-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 03/01/2023] [Indexed: 03/24/2023] Open
Abstract
Pseudorabies virus (PRV) infection is modulated by various cellular host factors. In this study, we investigated the role of histone deacetylase 6 (HDAC6) in this process. We determined HDAC6 expression in vitro and performed gene knockout, pharmacological inhibition analyses, immunofluorescence assays, and statistical analyses. We found that the pharmacological and genetic inhibition of HDAC6 significantly decreased PRV replication, whereas its overexpression promoted PRV replication. Additionally, we demonstrated that PRV infection can induce the phosphorylation of histone H2AX and lead to DNA damage response (DDR), and the ataxia telangiectasia mutated (ATM) inhibitor KU55933 inhibits DDR and PRV infection. Mechanistically, the HDAC6 inhibitor tubacin and HDAC6 knockout can decrease DDR. The results of this study suggested that HDAC6 may be a crucial factor in PRV-induced ATM-dependent DDR to promote PRV replication. IMPORTANCE Pseudorabies virus (PRV) is a member of the subfamily Alphaherpesvirinae of the family Herpesviridae. PRV infection in swine can lead to high morbidity and mortality of swine, causing huge economic losses. In particular, PRV variants can cause severe damage to the nervous and respiratory systems of humans, revealing that PRV may be a potential zoonotic pathogen. Vaccines for PRV have been developed that can delay or reduce the epidemic, but they currently cannot eliminate this disease completely. Therefore, studies should investigate new targets for the prevention and control of PRV infection. In this study, we demonstrated that HDAC6 can induce ataxia telangiectasia mutated-dependent DNA damage response to foster PRV replication, indicating that HDAC6 is a therapeutic target for PRV infection.
Collapse
Affiliation(s)
- Weiyin Xu
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China
| | - Ping Yan
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China
| | - Ziyan Zhou
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China
| | - Jingting Yao
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China
| | - Haochun Pan
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China
| | - Luyao Jiang
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China
| | - Zongyi Bo
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Bo Ni
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Mingxia Sun
- Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Song Gao
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China
| | - Changchao Huan
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China
| |
Collapse
|
32
|
Abildgaard AB, Nielsen SV, Bernstein I, Stein A, Lindorff-Larsen K, Hartmann-Petersen R. Lynch syndrome, molecular mechanisms and variant classification. Br J Cancer 2023; 128:726-734. [PMID: 36434153 PMCID: PMC9978028 DOI: 10.1038/s41416-022-02059-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/27/2022] Open
Abstract
Patients with the heritable cancer disease, Lynch syndrome, carry germline variants in the MLH1, MSH2, MSH6 and PMS2 genes, encoding the central components of the DNA mismatch repair system. Loss-of-function variants disrupt the DNA mismatch repair system and give rise to a detrimental increase in the cellular mutational burden and cancer development. The treatment prospects for Lynch syndrome rely heavily on early diagnosis; however, accurate diagnosis is inextricably linked to correct clinical interpretation of individual variants. Protein variant classification traditionally relies on cumulative information from occurrence in patients, as well as experimental testing of the individual variants. The complexity of variant classification is due to (1) that variants of unknown significance are rare in the population and phenotypic information on the specific variants is missing, and (2) that individual variant testing is challenging, costly and slow. Here, we summarise recent developments in high-throughput technologies and computational prediction tools for the assessment of variants of unknown significance in Lynch syndrome. These approaches may vastly increase the number of interpretable variants and could also provide important mechanistic insights into the disease. These insights may in turn pave the road towards developing personalised treatment approaches for Lynch syndrome.
Collapse
Affiliation(s)
- Amanda B Abildgaard
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Sofie V Nielsen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Inge Bernstein
- Department of Surgical Gastroenterology, Aalborg University Hospital, Aalborg, Denmark
- Institute of Clinical Medicine, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
| | - Amelie Stein
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kresten Lindorff-Larsen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Rasmus Hartmann-Petersen
- The Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
33
|
Fenton NM, Nguyen TB, Sharpe LJ, Brown AJ. Refining sugar's involvement in cholesterol synthesis. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159266. [PMID: 36528253 DOI: 10.1016/j.bbalip.2022.159266] [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: 08/02/2022] [Revised: 11/03/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022]
Abstract
Glucose metabolism and cholesterol synthesis are often regarded in isolation. Increasing evidence not only links these pathways but also suggests glucose catabolism regulates cholesterol synthesis. Uptake of glucose increases cholesterol production. However, the precise mechanism by which this occurs is not fully understood and is likely to involve many aspects of cellular pathways participating in energy sensing, cholesterol regulation, and synthesis. Here, we review some interesting links between cholesterol synthesis and glucose metabolism. Given glucose breakdown produces energy (both via glycolysis and its products through oxidative phosphorylation), and considering cholesterol synthesis is an energetically demanding process, it would seem logical that glucose metabolism impacts cholesterol synthesis. The energy sensing kinase AMPK carefully monitors energy supply to induce or suppress cholesterol synthesis as needed. Akt, activated by the insulin signalling cascade, regulates key transcription factors involved in lipid metabolism. The insulin signalling pathway also activates machinery involved in the deubiquitination of a key cholesterol synthesis enzyme. Moreover, glucose metabolites, acetyl-CoA, and GlcNAc are substrates for protein acetylation and N-glycosylation, respectively, and can stabilise proteins involved in cholesterol synthesis. As glucose and cholesterol dysregulation are both associated with numerous diseases, understanding the mechanisms of how glucose metabolism and cholesterol synthesis intersect may offer new avenues for therapeutics that make use of these findings.
Collapse
Affiliation(s)
- Nicole M Fenton
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Tina B Nguyen
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
34
|
Shukla S, Komarek J, Novakova Z, Nedvedova J, Ustinova K, Vankova P, Kadek A, Uetrecht C, Mertens H, Barinka C. In-solution structure and oligomerization of human histone deacetylase 6 - an integrative approach. FEBS J 2023; 290:821-836. [PMID: 36062318 DOI: 10.1111/febs.16616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 07/08/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023]
Abstract
Human histone deacetylase 6 (HDAC6) is a structurally unique, multidomain protein implicated in a variety of physiological processes including cytoskeletal remodelling and the maintenance of cellular homeostasis. Our current understanding of the HDAC6 structure is limited to isolated domains, and a holistic picture of the full-length protein structure, including possible domain interactions, is missing. Here, we used an integrative structural biology approach to build a solution model of HDAC6 by combining experimental data from several orthogonal biophysical techniques complemented by molecular modelling. We show that HDAC6 is best described as a mosaic of folded and intrinsically disordered domains that in-solution adopts an ensemble of conformations without any stable interactions between structured domains. Furthermore, HDAC6 forms dimers/higher oligomers in a concentration-dependent manner, and its oligomerization is mediated via the positively charged N-terminal microtubule-binding domain. Our findings provide the first insights into the structure of full-length human HDAC6 and can be used as a basis for further research into structure function and physiological studies of this unique deacetylase.
Collapse
Affiliation(s)
- Shivam Shukla
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic.,Department of Physical Chemistry, Faculty of Natural Science, Charles University, Prague, Czech Republic
| | - Jan Komarek
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Zora Novakova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Jana Nedvedova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Kseniya Ustinova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Pavla Vankova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Alan Kadek
- Leibniz Institute of Virology (LIV), Hamburg, Germany.,European XFEL GmbH, Schenefeld, Germany
| | - Charlotte Uetrecht
- Leibniz Institute of Virology (LIV), Hamburg, Germany.,European XFEL GmbH, Schenefeld, Germany.,Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron (DESY), Hamburg, Germany.,Department of Health Sciences and Biomedicine, School of Life Sciences, University of Siegen, Germany
| | - Haydyn Mertens
- European Molecular Biology Laboratory (EMBL)-Hamburg Outstation, c/o DESY, Germany
| | - Cyril Barinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
35
|
Ming H, Li B, Jiang J, Qin S, Nice EC, He W, Lang T, Huang C. Protein degradation: expanding the toolbox to restrain cancer drug resistance. J Hematol Oncol 2023; 16:6. [PMID: 36694209 PMCID: PMC9872387 DOI: 10.1186/s13045-023-01398-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 01/01/2023] [Indexed: 01/25/2023] Open
Abstract
Despite significant progress in clinical management, drug resistance remains a major obstacle. Recent research based on protein degradation to restrain drug resistance has attracted wide attention, and several therapeutic strategies such as inhibition of proteasome with bortezomib and proteolysis-targeting chimeric have been developed. Compared with intervention at the transcriptional level, targeting the degradation process seems to be a more rapid and direct strategy. Proteasomal proteolysis and lysosomal proteolysis are the most critical quality control systems responsible for the degradation of proteins or organelles. Although proteasomal and lysosomal inhibitors (e.g., bortezomib and chloroquine) have achieved certain improvements in some clinical application scenarios, their routine application in practice is still a long way off, which is due to the lack of precise targeting capabilities and inevitable side effects. In-depth studies on the regulatory mechanism of critical protein degradation regulators, including E3 ubiquitin ligases, deubiquitylating enzymes (DUBs), and chaperones, are expected to provide precise clues for developing targeting strategies and reducing side effects. Here, we discuss the underlying mechanisms of protein degradation in regulating drug efflux, drug metabolism, DNA repair, drug target alteration, downstream bypass signaling, sustaining of stemness, and tumor microenvironment remodeling to delineate the functional roles of protein degradation in drug resistance. We also highlight specific E3 ligases, DUBs, and chaperones, discussing possible strategies modulating protein degradation to target cancer drug resistance. A systematic summary of the molecular basis by which protein degradation regulates tumor drug resistance will help facilitate the development of appropriate clinical strategies.
Collapse
Affiliation(s)
- Hui Ming
- West China School of Basic Medical Sciences and Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Bowen Li
- West China School of Basic Medical Sciences and Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Jingwen Jiang
- West China School of Basic Medical Sciences and Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Siyuan Qin
- West China School of Basic Medical Sciences and Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Weifeng He
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Army Military Medical University, Chongqing, 400038, China.
| | - Tingyuan Lang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, 400030, People's Republic of China. .,Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People's Republic of China.
| | - Canhua Huang
- West China School of Basic Medical Sciences and Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
36
|
Krämer OH, Diehl T, Roos WP. Assessing the Effect of Histone Deacetylase Inhibitors on DNA Double-Strand Break Repair by Nonhomologous End Joining. Methods Mol Biol 2023; 2589:293-302. [PMID: 36255632 DOI: 10.1007/978-1-0716-2788-4_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
This book chapter describes a plasmid-based reporter method, first described by Bennardo et al. (2008) that we use in our laboratory for determining the activity of the repair of DNA double-strand breaks by nonhomologous end joining. This method can be used to measure the impact of epigenetic modifiers of the histone deacetylase family on this DNA repair pathway by flow cytometry.
Collapse
Affiliation(s)
- Oliver H Krämer
- Institute of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Tamara Diehl
- Institute of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Wynand P Roos
- Institute of Toxicology, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
37
|
Sun D, Zhang J, Dong G, He S, Sheng C. Blocking Non-enzymatic Functions by PROTAC-Mediated Targeted Protein Degradation. J Med Chem 2022; 65:14276-14288. [DOI: 10.1021/acs.jmedchem.2c01159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Donghuan Sun
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Jing Zhang
- Department of Pathology, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai 200003, China
| | - Guoqiang Dong
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Shipeng He
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| |
Collapse
|
38
|
Li W, Wang Z, Wang H, Zhang J, Wang X, Xing S, Chen S. IQGAP3 in clear cell renal cell carcinoma contributes to drug resistance and genome stability. PeerJ 2022; 10:e14201. [PMID: 36275458 PMCID: PMC9586079 DOI: 10.7717/peerj.14201] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 09/19/2022] [Indexed: 01/24/2023] Open
Abstract
Background Clear cell renal clear cell carcinoma (ccRCC) is resistant to most chemotherapeutic drugs and the molecular mechanisms have not been fully revealed. Genomic instability and the abnormal activation of bypass DNA repair pathway is the potential cause of tumor resistance to radiotherapy and chemotherapy. IQ-motif GTPase activating protein 3 (IQGAP3) regulates cell migration and intercellular adhesion. This study aims to analysis the effects of IQGAP3 expression on cell survival, genome stability and clinical prognosis in ccRCC. Methods Multiple bioinformatics analysis based on TCGA database and IHC analysis on clinical specimens were included. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot (WB) were used to determine protein expression level. MTT assay and 3D spheroid cell growth assay were used to assess cell proliferation and drug resistance in RNAi transfected ccRCC cells. Cell invasion capacity was evaluated by transwell assay. The influence of IQGAP3 on genome instability was revealed by micronuclei number and γ H2AX recruitment test. Results The highly expressed IQGAP3 in multiple subtypes of renal cell carcinoma has a clear prognostic value. Deletion of IQGAP3 inhibits cell growth in 3D Matrigel. IQGAP3 depletion lso increases accumulated DNA damage, and improves cell sensitivity to ionizing radiation and chemotherapeutic drugs. Therefore, targeting DNA damage repair function of IQGAP3 in tumorigenesis can provide ideas for the development of new targets for early diagnosis.
Collapse
Affiliation(s)
- Wen Li
- Health Science Center, School of Medicine, Shenzhen University, Shenzhen, Guangdong, China,Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong, China
| | - Zhifeng Wang
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan, China
| | - Hanlin Wang
- Health Science Center, School of Medicine, Shenzhen University, Shenzhen, Guangdong, China
| | - Jian Zhang
- Department of Pharmacy, Health Science Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Xiaobin Wang
- Health Science Center, School of Medicine, Shenzhen University, Shenzhen, Guangdong, China,Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong, China
| | - Shaojun Xing
- Health Science Center, School of Medicine, Shenzhen University, Shenzhen, Guangdong, China,Marshall Laboratory of Biomedical Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - Si Chen
- Health Science Center, School of Medicine, Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
39
|
Norcia LF, Watanabe EM, Hamamoto Filho PT, Hasimoto CN, Pelafsky L, de Oliveira WK, Sassaki LY. Polycystic Liver Disease: Pathophysiology, Diagnosis and Treatment. Hepat Med 2022; 14:135-161. [PMID: 36200122 PMCID: PMC9528914 DOI: 10.2147/hmer.s377530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022] Open
Abstract
Polycystic liver disease (PLD) is a clinical condition characterized by the presence of more than 10 cysts in the liver. It is a rare disease Of genetic etiology that presents as an isolated disease or assoc\iated with polycystic kidney disease. Ductal plate malformation, ciliary dysfunction, and changes in cell signaling are the main factors involved in its pathogenesis. Most patients with PLD are asymptomatic, but in 2-5% of cases the disease has disabling symptoms and a significant reduction in quality of life. The diagnosis is based on family history of hepatic and/or renal polycystic disease, clinical manifestations, patient age, and polycystic liver phenotype shown on imaging examinations. PLD treatment has evolved considerably in the last decades. Somatostatin analogues hold promise in controlling disease progression, but liver transplantation remains a unique curative treatment modality.
Collapse
Affiliation(s)
- Luiz Fernando Norcia
- Department of Surgery, São Paulo State University (Unesp), Medical School, Botucatu, São Paulo, Brazil
| | - Erika Mayumi Watanabe
- Department of Radiology, São Paulo State University (Unesp), Medical School, Botucatu, São Paulo, Brazil
| | - Pedro Tadao Hamamoto Filho
- Department of Neurology, Psychology and Psychiatry, São Paulo State University (Unesp), Medical School, Botucatu, São Paulo, Brazil
| | - Claudia Nishida Hasimoto
- Department of Surgery, São Paulo State University (Unesp), Medical School, Botucatu, São Paulo, Brazil
| | - Leonardo Pelafsky
- Department of Surgery, São Paulo State University (Unesp), Medical School, Botucatu, São Paulo, Brazil
| | - Walmar Kerche de Oliveira
- Department of Surgery, São Paulo State University (Unesp), Medical School, Botucatu, São Paulo, Brazil
| | - Ligia Yukie Sassaki
- Department of Internal Medicine, São Paulo State University (Unesp), Medical School, Botucatu, São Paulo, Brazil
| |
Collapse
|
40
|
Liu N, Zhou L, Lin G, Hu Y, Jiao Y, Wang Y, Liu J, Yang S, Yao S. HDAC inhibitors improve CRISPR-Cas9 mediated prime editing and base editing. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:36-46. [PMID: 35784015 PMCID: PMC9207553 DOI: 10.1016/j.omtn.2022.05.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 05/27/2022] [Indexed: 01/19/2023]
Abstract
Recent advances in CRISPR-Cas9 techniques, especially the discovery of base and prime editing, have significantly improved our ability to make precise changes in the genome. We hypothesized that modulating certain endogenous pathway cells could improve the action of those editing tools in mammalian cells. We established a reporter system in which a small fragment was integrated into the genome by prime editing (PE). With this system, we screened an in-house small-molecule library and identified a group of histone deacetylase inhibitors (HDACi) increasing prime editing. We also found that HDACi increased the efficiency of both cytosine base editing (CBE) and adenine base editing (ABE). Moreover, HDACi increased the purity of cytosine base editor products, which was accompanied by an upregulation of the acetylation of uracil DNA glycosylase (UNG) and UNG inhibitor (UGI) and an enhancement of their interaction. In summary, our work demonstrated that HDACi improves Cas9-mediated prime editing and base editing.
Collapse
Affiliation(s)
- Nan Liu
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Renmin Nanlu 17, Chengdu 610041, Sichuan, China
| | - Lifang Zhou
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Renmin Nanlu 17, Chengdu 610041, Sichuan, China
| | - Guifeng Lin
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Renmin Nanlu 17, Chengdu 610041, Sichuan, China
| | - Yun Hu
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Renmin Nanlu 17, Chengdu 610041, Sichuan, China
| | - Yaoge Jiao
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Renmin Nanlu 17, Chengdu 610041, Sichuan, China
| | - Yanhong Wang
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Renmin Nanlu 17, Chengdu 610041, Sichuan, China
| | - Jingming Liu
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Renmin Nanlu 17, Chengdu 610041, Sichuan, China
| | - Shengyong Yang
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Renmin Nanlu 17, Chengdu 610041, Sichuan, China
| | - Shaohua Yao
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Renmin Nanlu 17, Chengdu 610041, Sichuan, China
| |
Collapse
|
41
|
Firnau MB, Brieger A. CK2 and the Hallmarks of Cancer. Biomedicines 2022; 10:1987. [PMID: 36009534 PMCID: PMC9405757 DOI: 10.3390/biomedicines10081987] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a leading cause of death worldwide. Casein kinase 2 (CK2) is commonly dysregulated in cancer, impacting diverse molecular pathways. CK2 is a highly conserved serine/threonine kinase, constitutively active and ubiquitously expressed in eukaryotes. With over 500 known substrates and being estimated to be responsible for up to 10% of the human phosphoproteome, it is of significant importance. A broad spectrum of diverse types of cancer cells has been already shown to rely on disturbed CK2 levels for their survival. The hallmarks of cancer provide a rationale for understanding cancer's common traits. They constitute the maintenance of proliferative signaling, evasion of growth suppressors, resisting cell death, enabling of replicative immortality, induction of angiogenesis, the activation of invasion and metastasis, as well as avoidance of immune destruction and dysregulation of cellular energetics. In this work, we have compiled evidence from the literature suggesting that CK2 modulates all hallmarks of cancer, thereby promoting oncogenesis and operating as a cancer driver by creating a cellular environment favorable to neoplasia.
Collapse
Affiliation(s)
| | - Angela Brieger
- Department of Internal Medicine I, Biomedical Research Laboratory, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
42
|
Tseng CC, Huang SY, Tsai HP, Wu CW, Hsieh TH. HDAC6 is a prognostic biomarker that mediates IL-13 expression to regulate macrophage polarization through AP-1 in oral squamous cell carcinoma. Sci Rep 2022; 12:10513. [PMID: 35732647 PMCID: PMC9217956 DOI: 10.1038/s41598-022-14052-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/31/2022] [Indexed: 01/17/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common malignant tumor worldwide that is characterized by abnormal lesions or malignant hyperplasia of soft and hard tissues in the oral cavity. Previous research has found that HDAC6 may be a potential therapeutic target for cancer patients and has the ability to regulate immune cells. However, the mechanism of HDAC6 in OSCC pathogenesis is unclear. We collected clinical samples and analyzed the level of HDAC6 in OSCC patients. The results showed that in the high HDAC6 expression group, HDAC6 expression was positively correlated with the grade of OSCC (R = 0.182, P = 0.036) and that this group had a 3.248-fold increase in the mortality risk compared with the low HDAC6 expression group (P = 0.003). Survival analysis also identified a correlation between the expression of HDAC6 and overall survival in OSCC patients, and it was found that the expression of HDAC6 was inversely correlated with survival (P ≤ 0.001). In addition, we found that HDAC6 induced IL-13 expression through AP-1, resulting in M2 polarization of macrophages. Together, these results demonstrate that the level of HDAC6 may be a useful prognostic biomarker and offer a novel immune cell-related therapeutic strategy of targeting IL-13 in OSCC.
Collapse
Affiliation(s)
- Chung-Chih Tseng
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan.,Department of Dentistry, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, 81342, Taiwan
| | - Shi-Ying Huang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, 361021, China
| | - Hung-Pei Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chia-Wei Wu
- Department of Medical Research, E-Da Hospital/E-Da Cancer Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Tsung-Hua Hsieh
- Department of Medical Research, E-Da Hospital/E-Da Cancer Hospital, I-Shou University, Kaohsiung, 82445, Taiwan.
| |
Collapse
|
43
|
Wang M, Zhou C, Yu L, Kong D, Ma W, Lv B, Wang Y, Wu W, Zhou M, Cui G. Upregulation of MDH1 acetylation by HDAC6 inhibition protects against oxidative stress-derived neuronal apoptosis following intracerebral hemorrhage. Cell Mol Life Sci 2022; 79:356. [PMID: 35678904 PMCID: PMC11073123 DOI: 10.1007/s00018-022-04341-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/18/2022] [Accepted: 04/30/2022] [Indexed: 11/25/2022]
Abstract
Oxidative stress impairs functional recovery after intracerebral hemorrhage (ICH). Histone deacetylase 6 (HDAC6) plays an important role in the initiation of oxidative stress. However, the function of HDAC6 in ICH and the underlying mechanism of action remain elusive. We demonstrated here that HDAC6 knockout mice were resistant to oxidative stress following ICH, as assessed by the MDA and NADPH/NADP+ assays and ROS detection. HDAC6 deficiency also resulted in reduced neuronal apoptosis and lower expression levels of apoptosis-related proteins. Further mechanistic studies showed that HDAC6 bound to malate dehydrogenase 1 (MDH1) and mediated-MDH1 deacetylation on the lysine residues at position 121 and 298. MDH1 acetylation was inhibited in HT22 cells that were challenged with ICH-related damaging agents (Hemin, Hemoglobin, and Thrombin), but increased when HDAC6 was inhibited, suggesting an interplay between HDAC6 and MDH1. The acetylation-mimetic mutant, but not the acetylation-resistant mutant, of MDH1 protected neurons from oxidative injury. Furthermore, HDAC6 inhibition failed to alleviate brain damage after ICH when MDH1 was knockdown. Taken together, our study showed that HDAC6 inhibition protects against brain damage during ICH through MDH1 acetylation.
Collapse
Affiliation(s)
- Miao Wang
- Department of Geriatrics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China.
- Department of Neurology, Xuzhou First People's Hospital, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chao Zhou
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Lu Yu
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Delian Kong
- Department of Neurology, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Neurology, Xuzhou First People's Hospital, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Weijing Ma
- Department of Neurology, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bingchen Lv
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Yan Wang
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Weifeng Wu
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Mingyue Zhou
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Guiyun Cui
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China.
| |
Collapse
|
44
|
Masyuk AI, Masyuk TV, Trussoni CE, Pirius NE, LaRusso NF. Autophagy promotes hepatic cystogenesis in polycystic liver disease by depletion of cholangiocyte ciliogenic proteins. Hepatology 2022; 75:1110-1122. [PMID: 34942041 PMCID: PMC9035076 DOI: 10.1002/hep.32298] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUNDS AND AIMS Polycystic liver disease (PLD) is characterized by defective cholangiocyte cilia that regulate progressive growth of hepatic cysts. Because formation of primary cilia is influenced by autophagy through degradation of proteins involved in ciliogenesis, we hypothesized that ciliary defects in PLD cholangiocytes (PLDCs) originate from autophagy-mediated depletion of ciliogenic proteins ADP-ribosylation factor-like protein 3 (ARL3) and ADP-ribosylation factor-like protein 13B (ARL13B) and ARL-dependent mislocation of a ciliary-localized bile acid receptor, Takeda G-protein-coupled receptor 5 (TGR5), the activation of which enhances hepatic cystogenesis (HCG). The aims here were to determine whether: (1) ciliogenesis is impaired in PLDC, is associated with increased autophagy, and involves autophagy-mediated depletion of ARL3 and ARL13B; (2) depletion of ARL3 and ARL13B in PLDC cilia impacts ciliary localization of TGR5; and (3) pharmacological inhibition of autophagy re-establishes cholangiocyte cilia and ciliary localization of ARL3, ARL3B, and TGR5 and reduces HCG. APPROACH AND RESULTS By using liver tissue from healthy persons and patients with PLD, in vitro and in vivo models of PLD, and in vitro models of ciliogenesis, we demonstrated that, in PLDCs: ciliogenesis is impaired; autophagy is enhanced; ARL3 and ARL13B are ubiquitinated by HDAC6, depleted in cilia, and present in autophagosomes; depletion of ARL3 and ARL13B impacts ciliary localization of TGR5; and pharmacological inhibition of autophagy with mefloquine and verteporfin re-establishes cholangiocyte cilia and ciliary localization of ARL3, ARL13B, and TGR5 and reduces HCG. CONCLUSIONS The intersection between autophagy, defective cholangiocyte cilia, and enhanced HCG contributes to PLD progression and can be considered a target for therapeutic interventions.
Collapse
Affiliation(s)
- Anatoliy I. Masyuk
- Mayo Clinic College of Medicine and Science, 200 First Street, SW Rochester, Minnesota 55905, USA
| | - Tatyana V. Masyuk
- Mayo Clinic College of Medicine and Science, 200 First Street, SW Rochester, Minnesota 55905, USA
| | - Christy E. Trussoni
- Mayo Clinic College of Medicine and Science, 200 First Street, SW Rochester, Minnesota 55905, USA
| | - Nicholas E. Pirius
- Mayo Clinic College of Medicine and Science, 200 First Street, SW Rochester, Minnesota 55905, USA
| | - Nicholas F. LaRusso
- Mayo Clinic College of Medicine and Science, 200 First Street, SW Rochester, Minnesota 55905, USA
| |
Collapse
|
45
|
Natural Bioactive Compounds Targeting Histone Deacetylases in Human Cancers: Recent Updates. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27082568. [PMID: 35458763 PMCID: PMC9027183 DOI: 10.3390/molecules27082568] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/13/2022]
Abstract
Cancer is a complex pathology that causes a large number of deaths worldwide. Several risk factors are involved in tumor transformation, including epigenetic factors. These factors are a set of changes that do not affect the DNA sequence, while modifying the gene’s expression. Histone modification is an essential mark in maintaining cellular memory and, therefore, loss of this mark can lead to tumor transformation. As these epigenetic changes are reversible, the use of molecules that can restore the functions of the enzymes responsible for the changes is therapeutically necessary. Natural molecules, mainly those isolated from medicinal plants, have demonstrated significant inhibitory properties against enzymes related to histone modifications, particularly histone deacetylases (HDACs). Flavonoids, terpenoids, phenolic acids, and alkaloids exert significant inhibitory effects against HDAC and exhibit promising epi-drug properties. This suggests that epi-drugs against HDAC could prevent and treat various human cancers. Accordingly, the present study aimed to evaluate the pharmacodynamic action of different natural compounds extracted from medicinal plants against the enzymatic activity of HDAC.
Collapse
|
46
|
Chen X, Zhang G, Li P, Yu J, Kang L, Qin B, Wang Y, Wu J, Wang Y, Zhang J, Qin M, Guan H. SYVN1-mediated ubiquitination and degradation of MSH3 promotes the apoptosis of lens epithelial cells. FEBS J 2022; 289:5682-5696. [PMID: 35334159 DOI: 10.1111/febs.16447] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/24/2021] [Accepted: 03/24/2022] [Indexed: 12/29/2022]
Abstract
The pathology of age-related cataract (ARC) mainly involves the misfolding and aggregation of proteins, especially oxidative damage repair proteins, in the lens, induced by ultraviolet-B (UVB). MSH3, as a key member of the mismatch repair family, primarily maintains genome stability. However, the function of MSH3 and the mechanism by which cells maintain MSH3 proteostasis during cataractogenesis remains unknown. In the present study, the protein expression levels of MSH3 were found to be attenuated in ARC specimens and SRA01/04 cells under UVB exposure. The ectopic expression of MSH3 notably impeded UVB-induced apoptosis, whereas the knockdown of MSH3 promoted apoptosis. Protein half-life assay revealed that UVB irradiation accelerated the decline of MSH3 by ubiquitination and degradation. Subsequently, we found that E3 ubiquitin ligase synoviolin (SYVN1) interacted with MSH3 and promoted its ubiquitination and degradation. Of note, the expression and function of SYVN1 were contrary to those of MSH3 and SYVN1 regulated MSH3 protein degradation via the ubiquitin-proteasome pathway and the autophagy-lysosome pathway. Based on these findings, we propose a mechanism for ARC pathogenesis that involves SYVN1-mediated degradation of MSH3 via the ubiquitin-proteasome pathway and the autophagy-lysosome pathway, and suggest that interventions targeting SYVN1 might be a potential therapeutic strategy for ARC.
Collapse
Affiliation(s)
- Xiaojuan Chen
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Guowei Zhang
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Pengfei Li
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Jianfeng Yu
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Lihua Kang
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Bai Qin
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Ying Wang
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Jian Wu
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Yong Wang
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Junfang Zhang
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Miaomiao Qin
- Eye Institute, Affiliated Hospital of Nantong University, China
| | - Huaijin Guan
- Eye Institute, Affiliated Hospital of Nantong University, China
| |
Collapse
|
47
|
Abstract
In mammalian cells, genomic DNA is packaged with histone proteins and condensed into chromatin. To gain access to the DNA, chromatin remodelling is required that is enhanced through histone post-translational modifications, which subsequently stimulate processes including DNA repair and transcription. Histone acetylation is one of the most well understood modifications and is controlled by histone acetyltransferases (HATs) and histone deacetylases (HDACs). These enzymes play critical roles in normal cellular functioning, and the dysregulation of HDAC expression in particular has been linked with the development of a number of different cancer types. Conversely, tumour cell killing following radiotherapy is triggered through DNA damage and HDACs can help co-ordinate the cellular DNA damage response which promotes radioresistance. Consequently, HDAC inhibitors have been investigated as potential radiosensitizers in vitro and in vivo to improve the efficacy or radiotherapy in specific tumour types. In this review, we provide an up-to-date summary of HDACs and their cellular functions, including in DNA damage repair. We also review evidence demonstrating that HDAC inhibitors can effectively enhance tumour radiosensitisation, and which therefore show potential for translation into the clinic for cancer patient benefit.
Collapse
|
48
|
Guervilly JH, Blin M, Laureti L, Baudelet E, Audebert S, Gaillard PH. SLX4 dampens MutSα-dependent mismatch repair. Nucleic Acids Res 2022; 50:2667-2680. [PMID: 35166826 PMCID: PMC8934664 DOI: 10.1093/nar/gkac075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
The tumour suppressor SLX4 plays multiple roles in the maintenance of genome stability, acting as a scaffold for structure-specific endonucleases and other DNA repair proteins. It directly interacts with the mismatch repair (MMR) protein MSH2 but the significance of this interaction remained unknown until recent findings showing that MutSβ (MSH2-MSH3) stimulates in vitro the SLX4-dependent Holliday junction resolvase activity. Here, we characterize the mode of interaction between SLX4 and MSH2, which relies on an MSH2-interacting peptide (SHIP box) that drives interaction of SLX4 with both MutSβ and MutSα (MSH2-MSH6). While we show that this MSH2 binding domain is dispensable for the well-established role of SLX4 in interstrand crosslink repair, we find that it mediates inhibition of MutSα-dependent MMR by SLX4, unravelling an unanticipated function of SLX4.
Collapse
Affiliation(s)
- Jean-Hugues Guervilly
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm, CNRS, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Marion Blin
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm, CNRS, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Luisa Laureti
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm, CNRS, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Emilie Baudelet
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm, CNRS, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Stéphane Audebert
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm, CNRS, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Pierre-Henri Gaillard
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm, CNRS, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
49
|
Lysine Acetylation, Cancer Hallmarks and Emerging Onco-Therapeutic Opportunities. Cancers (Basel) 2022; 14:cancers14020346. [PMID: 35053509 PMCID: PMC8773583 DOI: 10.3390/cancers14020346] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/21/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Several histone deacetylase inhibitors have been approved by FDA for cancer treatment. Intensive efforts have been devoted to enhancing its anti-cancer efficacy by combining it with various other agents. Yet, no guideline is available to assist in the choice of candidate drugs for combination towards optimal solutions for different clinical problems. Thus, it is imperative to characterize the primary cancer hallmarks that lysine acetylation is associated with and gain knowledge on the key cancer features that each combinatorial onco-therapeutic modality targets to aid in the combinatorial onco-therapeutic design. Cold atmospheric plasma represents an emerging anti-cancer modality via manipulating cellular redox level and has been demonstrated to selectively target several cancer hallmarks. This review aims to delineate the intrinsic connections between lysine acetylation and cancer properties, and forecast opportunities histone deacetylase inhibitors may have when combined with cold atmospheric plasma as novel precision onco-therapies. Abstract Acetylation, a reversible epigenetic process, is implicated in many critical cellular regulatory systems including transcriptional regulation, protein structure, activity, stability, and localization. Lysine acetylation is the most prevalent and intensively investigated among the diverse acetylation forms. Owing to the intrinsic connections of acetylation with cell metabolism, acetylation has been associated with metabolic disorders including cancers. Yet, relatively little has been reported on the features of acetylation against the cancer hallmarks, even though this knowledge may help identify appropriate therapeutic strategies or combinatorial modalities for the effective treatment and resolution of malignancies. By examining the available data related to the efficacy of lysine acetylation against tumor cells and elaborating the primary cancer hallmarks and the associated mechanisms to target the specific hallmarks, this review identifies the intrinsic connections between lysine acetylation and cancer hallmarks and proposes novel modalities that can be combined with HDAC inhibitors for cancer treatment with higher efficacy and minimum adverse effects.
Collapse
|
50
|
Quaas CE, Long DT. Targeting (de)acetylation: A Diversity of Mechanism and Disease. COMPREHENSIVE PHARMACOLOGY 2022:469-492. [DOI: 10.1016/b978-0-12-820472-6.00076-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|