1
|
Wang C, Liu ZP. Diffusion-based generation of gene regulatory networks from scRNA-seq data with DigNet. Genome Res 2025; 35:340-354. [PMID: 39694856 PMCID: PMC11874984 DOI: 10.1101/gr.279551.124] [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: 05/06/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
A gene regulatory network (GRN) intricately encodes the interconnectedness of identities and functionalities of genes within cells, ultimately shaping cellular specificity. Despite decades of endeavors, reverse engineering of GRNs from gene expression profiling data remains a profound challenge, particularly when it comes to reconstructing cell-specific GRNs that are tailored to precise cellular and genetic contexts. Here, we propose a discrete diffusion generation model, called DigNet, capable of generating corresponding GRNs from high-throughput single-cell RNA sequencing (scRNA-seq) data. DigNet embeds the network generation process into a multistep recovery procedure with Markov properties. Each intermediate step has a specific model to recover a portion of the gene regulatory architectures. It thus can ensure compatibility between global network structures and regulatory modules through the unique multistep diffusion procedure. Furthermore, through iMetacell integration and non-Euclidean discrete space modeling, DigNet is robust to the presence of noise in scRNA-seq data and the sparsity of GRNs. Benchmark evaluation results against more than a dozen state-of-the-art network inference methods demonstrate that DigNet achieves superior performance across various single-cell GRN reconstruction experiments. Furthermore, DigNet provides unique insights into the immune response in breast cancer, derived from differential gene regulation identified in T cells. As an open-source software, DigNet offers a powerful and effective tool for generating cell-specific GRNs from scRNA-seq data.
Collapse
Affiliation(s)
- Chuanyuan Wang
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Zhi-Ping Liu
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| |
Collapse
|
2
|
Song H, Guo Z, Xie K, Liu X, Yang X, Shen R, Wang D. Crotonylation of MCM6 enhances chemotherapeutics sensitivity of breast cancer via inducing DNA replication stress. Cell Prolif 2025; 58:e13759. [PMID: 39477811 PMCID: PMC11839194 DOI: 10.1111/cpr.13759] [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: 04/19/2024] [Revised: 09/03/2024] [Accepted: 09/14/2024] [Indexed: 02/21/2025] Open
Abstract
Breast cancer is associated with high morbidity and mortality, which are closely influenced by protein post-translational modifications (PTMs). Lysine crotonylation (Kcr) serves as a newly identified PTM type that plays a role in various biological processes; however, its involvement in breast cancer progression remains unclear. Minichromosome maintenance 6 (MCM6) is a critical component of DNA replication and has been previous confirmed to exhibit a significant role in tumorigenesis. Despite this, a comprehensive analysis of MCM6, particularly regarding its modifications in breast cancer is lacking. In this study, we found MCM6 is upregulated in breast invasive carcinoma (BRCA) and is associated with poorer overall survival by regulating the DNA damage repair mechanisms. Furthermore, MCM6-knockdown resulted in decreased cell proliferation and inhibited the DNA replication, leading to DNA replication stress and sustained DNA damage, thereby enhancing the chemotherapeutic sensitivity of breast cancer. Additionally, SIRT7-mediated crotonylation of MCM6 at K599 (MCM6-K599cr) was significantly upregulated in response to DNA replication stress, primarily due to the disassemebly of the MCM2-7 complex and regulated by RNF8-mediated ubiquitination. Concurrently, kaempferol, which acts as a regulator of SIRT7, was found to enhance the Kcr level of MCM6, reducing tumour weight, particular when combined with paclitaxel, highlighting its potential chemotherapeutic target for BRCA therapy.
Collapse
Affiliation(s)
- Haoyun Song
- School of Basic Medical SciencesLanzhou UniversityGansuChina
| | - Zhao Guo
- School of Basic Medical SciencesLanzhou UniversityGansuChina
| | - Kun Xie
- School of Basic Medical SciencesLanzhou UniversityGansuChina
| | - Xiangwen Liu
- School of Basic Medical SciencesLanzhou UniversityGansuChina
| | - Xuguang Yang
- School of Basic Medical SciencesLanzhou UniversityGansuChina
| | - Rong Shen
- School of Basic Medical SciencesLanzhou UniversityGansuChina
| | - Degui Wang
- School of Basic Medical SciencesLanzhou UniversityGansuChina
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal TumorLanzhouGansu ProvinceChina
| |
Collapse
|
3
|
Campagna R, Mazzanti L, Pompei V, Alia S, Vignini A, Emanuelli M. The Multifaceted Role of Endothelial Sirt1 in Vascular Aging: An Update. Cells 2024; 13:1469. [PMID: 39273039 PMCID: PMC11394039 DOI: 10.3390/cells13171469] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
NAD+-dependent deacetylase sirtuin-1 (Sirt1) belongs to the sirtuins family, known to be longevity regulators, and exerts a key role in the prevention of vascular aging. By aging, the expression levels of Sirt1 decline with a severe impact on vascular function, such as the rise of endothelial dysfunction, which in turn promotes the development of cardiovascular diseases. In this context, the impact of Sirt1 activity in preventing endothelial senescence is particularly important. Given the key role of Sirt1 in counteracting endothelial senescence, great efforts have been made to deepen the knowledge about the intricate cross-talks and interactions of Sirt1 with other molecules, in order to set up possible strategies to boost Sirt1 activity to prevent or treat vascular aging. The aim of this review is to provide a proper background on the regulation and function of Sirt1 in the vascular endothelium and to discuss the recent advances regarding the therapeutic strategies of targeting Sirt1 to counteract vascular aging.
Collapse
Affiliation(s)
- Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Laura Mazzanti
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
- Fondazione Salesi, Ospedale G. Salesi, 60100 Ancona, Italy
| | - Veronica Pompei
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Sonila Alia
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Arianna Vignini
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
- Research Center of Health Education and Health Promotion, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| |
Collapse
|
4
|
Liu Y, Liang M, Chen K, Wang L, Yang Y, Li Q, Lian B, Zhuo T, Huang J. A novel entity of HIPK2::YAP1 pulmonary fibromatosis. BMC Pulm Med 2024; 24:223. [PMID: 38714933 PMCID: PMC11075317 DOI: 10.1186/s12890-024-03026-5] [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: 11/14/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Pulmonary fibromatosis (PF) is a specific variant of fibromatosis, which is rarely reported occurring in the lung. PF with HIPK2-YAP1 fusion was a novel entity. CASE PRESENTATION In this report, a 66-year-old male with PF had been smoking over 40 years. Multiple cords and small nodules in both lungs had been detected in a health examination two years earlier at our hospital. But approximately twofold enlarged in the lingual segment of the upper lobe in the left lung were disclosed in this year. Immunohistochemical analysis demonstrated that the vimentin and β-Catenin were positive in the largest nodule. After underwent a DNA/RNA panel next-generation sequencing (NGS), missense mutations and HIPK2-YAP1 fusion were found in this sample. Ultimately, the case diagnosis as PF with HIPK2-YAP1 fusion after multidisciplinary treatment. Currently, the patient is doing well and recurrence-free at 14 months post-surgery. CONCLUSIONS It's difficult for patients with complex morphology to make accurate diagnosis solely based on morphology and immunohistochemistry. But molecular detection is an effective method for further determining pathological subtypes.
Collapse
Affiliation(s)
- Yuqiang Liu
- Department of Proctology, Honliv Hospital, Xinxiang, 453400, Henan Province, China
| | - Meng Liang
- Department of Academic Affairs, Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Kai Chen
- Department of Respiratory Medicine, Yongcheng People Hospital, Yongcheng, 476600, Henan Province, China
| | - Lucas Wang
- Precision Medicine Center, Guangzhou Huayin Health Medical Group Co., Ltd, No.33 Binhe Road, Huangpu District, Guangzhou, 510700, Guangdong Province, China
| | - Yaxian Yang
- Precision Medicine Center, Guangzhou Huayin Health Medical Group Co., Ltd, No.33 Binhe Road, Huangpu District, Guangzhou, 510700, Guangdong Province, China
| | - Qi Li
- Precision Medicine Center, Guangzhou Huayin Health Medical Group Co., Ltd, No.33 Binhe Road, Huangpu District, Guangzhou, 510700, Guangdong Province, China
| | - Bin Lian
- Precision Medicine Center, Guangzhou Huayin Health Medical Group Co., Ltd, No.33 Binhe Road, Huangpu District, Guangzhou, 510700, Guangdong Province, China
| | - Tongxu Zhuo
- Precision Medicine Center, Guangzhou Huayin Health Medical Group Co., Ltd, No.33 Binhe Road, Huangpu District, Guangzhou, 510700, Guangdong Province, China
| | - Jian Huang
- Precision Medicine Center, Guangzhou Huayin Health Medical Group Co., Ltd, No.33 Binhe Road, Huangpu District, Guangzhou, 510700, Guangdong Province, China.
- Department of Pathological Diagnosis and Research Center, the Affiliated Hospital of Guangdong Medical University, No.57 South Renmin Avenue, Xiashan District, Zhanjiang, 524001, Guangdong Province, China.
| |
Collapse
|
5
|
Krzysiak TC, Choi Y, Kim YJ, Yang Y, DeHaven C, Thompson L, Ponticelli R, Mermigos MM, Thomas L, Marquez A, Sipula I, Kemper JK, Jurczak M, Thomas G, Gronenborn AM. Inhibitory protein-protein interactions of the SIRT1 deacetylase are choreographed by post-translational modification. Protein Sci 2024; 33:e4938. [PMID: 38533551 PMCID: PMC10966392 DOI: 10.1002/pro.4938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/15/2023] [Accepted: 02/07/2024] [Indexed: 03/28/2024]
Abstract
Regulation of SIRT1 activity is vital to energy homeostasis and plays important roles in many diseases. We previously showed that insulin triggers the epigenetic regulator DBC1 to prime SIRT1 for repression by the multifunctional trafficking protein PACS-2. Here, we show that liver DBC1/PACS-2 regulates the diurnal inhibition of SIRT1, which is critically important for insulin-dependent switch in fuel metabolism from fat to glucose oxidation. We present the x-ray structure of the DBC1 S1-like domain that binds SIRT1 and an NMR characterization of how the SIRT1 N-terminal region engages DBC1. This interaction is inhibited by acetylation of K112 of DBC1 and stimulated by the insulin-dependent phosphorylation of human SIRT1 at S162 and S172, catalyzed sequentially by CK2 and GSK3, resulting in the PACS-2-dependent inhibition of nuclear SIRT1 enzymatic activity and translocation of the deacetylase in the cytoplasm. Finally, we discuss how defects in the DBC1/PACS-2-controlled SIRT1 inhibitory pathway are associated with disease, including obesity and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Troy C. Krzysiak
- Department of Structural BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Present address:
College of PharmacyDaegu Catholic UniversityGyeongsanRepublic of Korea
| | - You‐Jin Choi
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- College of Pharmacy and Research Institute of Pharmaceutical SciencesSeoul National UniversitySeoulRepublic of Korea
- Present address:
College of PharmacyDaegu Catholic UniversityGyeongsanRepublic of Korea
| | - Yong Joon Kim
- Department of Structural BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Yunhan Yang
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Christopher DeHaven
- Department of Structural BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Lariah Thompson
- Department of Structural BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Ryan Ponticelli
- Department of Structural BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Mara M. Mermigos
- Department of Structural BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Laurel Thomas
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Andrea Marquez
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Ian Sipula
- Department of Medicine, Division of Endocrinology and MetabolismUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Jongsook Kim Kemper
- Department of Molecular and Integrative PhysiologyUniversity of Illinois at UrbanaUrbanaIllinoisUSA
| | - Michael Jurczak
- Department of Medicine, Division of Endocrinology and MetabolismUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Gary Thomas
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Angela M. Gronenborn
- Department of Structural BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
6
|
Chen X, Wang Z, Li C, Zhang Z, Lu S, Wang X, Liang Q, Zhu X, Pan C, Wang Q, Ji Z, Wang Y, Piao M, Chi G, Ge P. SIRT1 activated by AROS sensitizes glioma cells to ferroptosis via induction of NAD+ depletion-dependent activation of ATF3. Redox Biol 2024; 69:103030. [PMID: 38181705 PMCID: PMC10791567 DOI: 10.1016/j.redox.2024.103030] [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: 11/14/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024] Open
Abstract
Ferroptosis is a type of programmed cell death resulting from iron overload-dependent lipid peroxidation, and could be promoted by activating transcription factor 3 (ATF3). SIRT1 is an enzyme accounting for removing acetylated lysine residues from target proteins by consuming NAD+, but its role remains elusive in ferroptosis and activating ATF3. In this study, we found SIRT1 was activated during the process of RSL3-induced glioma cell ferroptosis. Moreover, the glioma cell death was aggravated by SIRT1 activator SRT2183, but suppressed by SIRT inhibitor EX527 or when SIRT1 was silenced with siRNA. These indicated SIRT1 sensitized glioma cells to ferroptosis. Furthermore, we found SIRT1 promoted RSL3-induced expressional upregulation and nuclear translocation of ATF3. Silence of ATF3 with siRNA attenuated RSL3-induced increases of ferrous iron and lipid peroxidation, downregulation of SLC7A11 and GPX4 and depletion of cysteine and GSH. Thus, SIRT1 promoted glioma cell ferroptosis by inducting ATF3 activation. Mechanistically, ATF3 activation was reinforced when RSL3-induced decline of NAD+ was aggravated by FK866 that could inhibit NAD + synthesis via salvage pathway, but suppressed when intracellular NAD+ was maintained at higher level by supplement of exogenous NAD+. Notably, the NAD + decline caused by RSL3 was enhanced when SIRT1 was further activated by SRT2183, but attenuated when SIRT1 activation was inhibited by EX527. These indicated SIRT1 promoted ATF3 activation via consumption of NAD+. Finally, we found RSL3 activated SIRT1 by inducing reactive oxygen species-dependent upregulation of AROS. Together, our study revealed SIRT1 activated by AROS sensitizes glioma cells to ferroptosis via activation of ATF3-dependent inhibition of SLC7A11 and GPX4.
Collapse
Affiliation(s)
- Xi Chen
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhenchuan Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen Li
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhao Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun, 130021, China
| | - Shan Lu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Xuanzhong Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Qi Liang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaoxi Zhu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chengliang Pan
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Qingxuan Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhilin Ji
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Yubo Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Meihua Piao
- Department of Anesthesiology, First Hospital of Jilin University, Changchun, 130021, China
| | - Guangfan Chi
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Pengfei Ge
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
7
|
Ha JH, Lee BW, Yi DH, Lee SJ, Kim WI, Pak SW, Kim HY, Kim SH, Shin IS, Kim JC, Lee IC. Particulate matter-mediated oxidative stress induces airway inflammation and pulmonary dysfunction through TXNIP/NF-κB and modulation of the SIRT1-mediated p53 and TGF-β/Smad3 pathways in mice. Food Chem Toxicol 2024; 183:114201. [PMID: 38013002 DOI: 10.1016/j.fct.2023.114201] [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: 09/07/2023] [Revised: 11/01/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023]
Abstract
Exposure to particulate matter is currently recognized as a serious aggravating factor of respiratory diseases. In this study, we investigated the effects of particulate matter (PM) on the respiratory system in BALB/c mice and NCI-H292 cells. PM (0, 2.5, 5 and 20 mg/kg) was administered to mice by intra-tracheal instillation for 7 days. After a 7 day-repeated treatment of PM, we evaluated inflammatory cytokines/cell counts in bronchoalveolar lavage fluid (BALF) and conducted pulmonary histology and functional test. We also investigated the role of TXNIP/NF-κB and SIRT1-mediated p53 and TGF-β/Smad3 pathways in PM-induced airway inflammation and pulmonary dysfunction. PM caused a significant increase in pro-inflammatory cytokines, inflammatory cell counts in bronchoalveolar lavage fluid. PM-mediated oxidative stress down-regulated thioredoxin-1 and up-regulated thioredoxin-interacting protein and activation of nuclear factor-kappa B in the lung tissue and PM-treated NCI-H292 cells. PM suppressed sirtuin1 protein levels and increased p53 acetylation in PM-exposed mice and PM-treated NCI-H292 cells. In addition, PM caused inflammatory cell infiltration and the thickening of alveolar walls by exacerbating the inflammatory response in the lung tissue. PM increased levels of transforming growth factor-β, phosphorylation of Smad3 and activation of α-smooth muscle actin, and collagen type1A2 in PM-exposed mice and PM-treated NCI-H292 cells. In pulmonary function tests, PM exposure impaired pulmonary function resembling pulmonary fibrosis, characterized by increased resistance and elastance of the respiratory system, and resistance, elastance, and damping of lung tissues, whereas decreased compliance of the respiratory system, forced expired volume and forced vital capacity. Overall, PM-mediated oxidative stress caused airway inflammation and pulmonary dysfunction with pulmonary fibrosis via TXNIP pathway/NF-κB activation and modulation of the SIRT1-mediated TGF-β/Smad3 pathways. The results of this study can provide fundamental data on the potential adverse effects and underlying mechanism of pulmonary fibrosis caused by PM exposure as a public health concern. Due to the potential toxicity of PM, people with respiratory disease must be careful with PM exposure.
Collapse
Affiliation(s)
- Ji-Hye Ha
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea; College of Veterinary Medicine and BK21 FOUR Program, Chungnam National University, Daejeon, Republic of Korea
| | - Ba-Wool Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Da-Hye Yi
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Se-Jin Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Woong-Il Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - So-Won Pak
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Hyeon-Young Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, Republic of Korea
| | - Sung-Hwan Kim
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, Republic of Korea
| | - In-Sik Shin
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea.
| | - In-Chul Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea.
| |
Collapse
|
8
|
Müller A, Weyerhäuser P, Berte N, Jonin F, Lyubarskyy B, Sprang B, Kantelhardt SR, Salinas G, Opitz L, Schulz-Schaeffer W, Giese A, Kim EL. Concurrent Activation of Both Survival-Promoting and Death-Inducing Signaling by Chloroquine in Glioblastoma Stem Cells: Implications for Potential Risks and Benefits of Using Chloroquine as Radiosensitizer. Cells 2023; 12:cells12091290. [PMID: 37174691 PMCID: PMC10177603 DOI: 10.3390/cells12091290] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Lysosomotropic agent chloroquine was shown to sensitize non-stem glioblastoma cells to radiation in vitro with p53-dependent apoptosis implicated as one of the underlying mechanisms. The in vivo outcomes of chloroquine or its effects on glioblastoma stem cells have not been previously addressed. This study undertakes a combinatorial approach encompassing in vitro, in vivo and in silico investigations to address the relationship between chloroquine-mediated radiosensitization and p53 status in glioblastoma stem cells. Our findings reveal that chloroquine elicits antagonistic impacts on signaling pathways involved in the regulation of cell fate via both transcription-dependent and transcription-independent mechanisms. Evidence is provided that transcriptional impacts of chloroquine are primarily determined by p53 with chloroquine-mediated activation of pro-survival mevalonate and p21-DREAM pathways being the dominant response in the background of wild type p53. Non-transcriptional effects of chloroquine are conserved and converge on key cell fate regulators ATM, HIPK2 and AKT in glioblastoma stem cells irrespective of their p53 status. Our findings indicate that pro-survival responses elicited by chloroquine predominate in the context of wild type p53 and are diminished in cells with transcriptionally impaired p53. We conclude that p53 is an important determinant of the balance between pro-survival and pro-death impacts of chloroquine and propose that p53 functional status should be taken into consideration when evaluating the efficacy of glioblastoma radiosensitization by chloroquine.
Collapse
Affiliation(s)
- Andreas Müller
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Patrick Weyerhäuser
- Institute of Toxicology, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Nancy Berte
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Fitriasari Jonin
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Bogdan Lyubarskyy
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Bettina Sprang
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Sven Rainer Kantelhardt
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Gabriela Salinas
- NGS Integrative Genomics Core Unit (NIG), Institute for Human Genetics, University Medical Centre, 37075 Göttingen, Germany
| | - Lennart Opitz
- Functional Genomics Center Zurich, ETH Zurich, University of Zurich, 8092 Zurich, Switzerland
| | | | - Alf Giese
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| | - Ella L Kim
- Experimental Neurooncology Group, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany
| |
Collapse
|
9
|
Zhao P, Malik S. The phosphorylation to acetylation/methylation cascade in transcriptional regulation: how kinases regulate transcriptional activities of DNA/histone-modifying enzymes. Cell Biosci 2022; 12:83. [PMID: 35659740 PMCID: PMC9164400 DOI: 10.1186/s13578-022-00821-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/27/2022] [Indexed: 11/30/2022] Open
Abstract
Transcription factors directly regulate gene expression by recognizing and binding to specific DNA sequences, involving the dynamic alterations of chromatin structure and the formation of a complex with different kinds of cofactors, like DNA/histone modifying-enzymes, chromatin remodeling factors, and cell cycle factors. Despite the significance of transcription factors, it remains unclear to determine how these cofactors are regulated to cooperate with transcription factors, especially DNA/histone modifying-enzymes. It has been known that DNA/histone modifying-enzymes are regulated by post-translational modifications. And the most common and important modification is phosphorylation. Even though various DNA/histone modifying-enzymes have been classified and partly explained how phosphorylated sites of these enzymes function characteristically in recent studies. It still needs to find out the relationship between phosphorylation of these enzymes and the diseases-associated transcriptional regulation. Here this review describes how phosphorylation affects the transcription activity of these enzymes and other functions, including protein stability, subcellular localization, binding to chromatin, and interaction with other proteins.
Collapse
|
10
|
Insights into Regulators of p53 Acetylation. Cells 2022; 11:cells11233825. [PMID: 36497084 PMCID: PMC9737083 DOI: 10.3390/cells11233825] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
The tumor suppressor p53 is a transcription factor that regulates the expression of dozens of target genes and diverse physiological processes. To precisely regulate the p53 network, p53 undergoes various post-translational modifications and alters the selectivity of target genes. Acetylation plays an essential role in cell fate determination through the activation of p53. Although the acetylation of p53 has been examined, the underlying regulatory mechanisms remain unclear and, thus, have attracted the interest of researchers. We herein discuss the role of acetylation in the p53 pathway, with a focus on p53 acetyltransferases and deacetylases. We also review recent findings on the regulators of these enzymes to understand the mode of p53 acetylation from a broader perspective.
Collapse
|
11
|
Liebl MC, Hofmann TG. Regulating the p53 Tumor Suppressor Network at PML Biomolecular Condensates. Cancers (Basel) 2022; 14:4549. [PMID: 36230470 PMCID: PMC9558958 DOI: 10.3390/cancers14194549] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
By forming specific functional entities, nuclear biomolecular condensates play an important function in guiding biological processes. PML biomolecular condensates, also known as PML nuclear bodies (NBs), are macro-molecular sub-nuclear organelles involved in central biological processes, including anti-viral response and cell fate control upon genotoxic stress. PML condensate formation is stimulated upon cellular stress, and relies on protein-protein interactions establishing a PML protein meshwork capable of recruiting the tumor suppressor p53, along with numerous modifiers of p53, thus balancing p53 posttranslational modifications and activity. This stress-regulated process appears to be controlled by liquid-liquid phase separation (LLPS), which may facilitate regulated protein-unmixing of p53 and its regulators into PML nuclear condensates. In this review, we summarize and discuss the molecular mechanisms underlying PML nuclear condensate formation, and how these impact the biological function of p53 in driving the cell death and senescence responses. In addition, by using an in silico approach, we identify 299 proteins which share PML and p53 as binding partners, thus representing novel candidate proteins controlling p53 function and cell fate decision-making at the level of PML nuclear biocondensates.
Collapse
Affiliation(s)
| | - Thomas G. Hofmann
- Institute of Toxicology, University Medical Center Mainz, Johannes Gutenberg University, 55131 Mainz, Germany
| |
Collapse
|
12
|
Lacroix E, Audas TE. Keeping up with the condensates: The retention, gain, and loss of nuclear membrane-less organelles. Front Mol Biosci 2022; 9:998363. [PMID: 36203874 PMCID: PMC9530788 DOI: 10.3389/fmolb.2022.998363] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/19/2022] [Indexed: 12/04/2022] Open
Abstract
In recent decades, a growing number of biomolecular condensates have been identified in eukaryotic cells. These structures form through phase separation and have been linked to a diverse array of cellular processes. While a checklist of established membrane-bound organelles is present across the eukaryotic domain, less is known about the conservation of membrane-less subcellular structures. Many of these structures can be seen throughout eukaryotes, while others are only thought to be present in metazoans or a limited subset of species. In particular, the nucleus is a hub of biomolecular condensates. Some of these subnuclear domains have been found in a broad range of organisms, which is a characteristic often attributed to essential functionality. However, this does not always appear to be the case. For example, the nucleolus is critical for ribosomal biogenesis and is present throughout the eukaryotic domain, while the Cajal bodies are believed to be similarly conserved, yet these structures are dispensable for organismal survival. Likewise, depletion of the Drosophila melanogaster omega speckles reduces viability, despite the apparent absence of this domain in higher eukaryotes. By reviewing primary research that has analyzed the presence of specific condensates (nucleoli, Cajal bodies, amyloid bodies, nucleolar aggresomes, nuclear speckles, nuclear paraspeckles, nuclear stress bodies, PML bodies, omega speckles, NUN bodies, mei2 dots) in a cross-section of organisms (e.g., human, mouse, D. melanogaster, Caenorhabditis elegans, yeast), we adopt a human-centric view to explore the emergence, retention, and absence of a subset of nuclear biomolecular condensates. This overview is particularly important as numerous biomolecular condensates have been linked to human disease, and their presence in additional species could unlock new and well characterized model systems for health research.
Collapse
Affiliation(s)
- Emma Lacroix
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Timothy E. Audas
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- *Correspondence: Timothy E. Audas,
| |
Collapse
|
13
|
Balasubramanian S, Perumal E. A systematic review on fluoride-induced epigenetic toxicity in mammals. Crit Rev Toxicol 2022; 52:449-468. [PMID: 36422650 DOI: 10.1080/10408444.2022.2122771] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Fluoride, one of the global groundwater contaminants, is ubiquitous in our day-to-day life from various natural and anthropogenic sources. Numerous in vitro, in vivo, and epidemiological studies are conducted to understand the effect of fluoride on biological systems. A low concentration of fluoride is reported to increase oral health, whereas chronic exposure to higher concentrations causes fluoride toxicity (fluorosis). It includes dental fluorosis, skeletal fluorosis, and fluoride toxicity in soft tissues. The mechanism of fluoride toxicity has been reviewed extensively. However, epigenetic regulation in fluoride toxicity has not been reviewed. This systematic review summarizes the current knowledge regarding fluoride-induced epigenetic toxicity in the in vitro, in vivo, and epidemiological studies in mammalian systems. We examined four databases for the association between epigenetics and fluoride exposure. Out of 932 articles (as of 31 March 2022), 39 met our inclusion criteria. Most of the studies focused on different genes, and overall, preliminary evidence for epigenetic regulation of fluoride toxicity was identified. We further highlight the need for epigenome studies rather than candidate genes and provide recommendations for future research. Our results indicate a correlation between fluoride exposure and epigenetic processes. Further studies are warranted to elucidate and confirm the mechanism of epigenetic alterations mediated fluoride toxicity.
Collapse
Affiliation(s)
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| |
Collapse
|
14
|
Aengenvoort J, Sekeres M, Proksch P, Fritz G. Targeting Mechanisms of the DNA Damage Response (DDR) and DNA Repair by Natural Compounds to Improve cAT-Triggered Tumor Cell Death. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113567. [PMID: 35684504 PMCID: PMC9182506 DOI: 10.3390/molecules27113567] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/11/2022] [Accepted: 05/26/2022] [Indexed: 12/20/2022]
Abstract
Recently, we identified secalonic acid F (SA), 5-epi-nakijiquinone Q (NQ) and 5-epi-ilimaquinone (IQ) as natural compounds (NC) affecting mechanisms of the DNA damage response (DDR). Here, we further characterized their effects on DDR, DNA repair and cytotoxicity if used in mono- and co-treatment with conventional anticancer therapeutics (cAT) (cisplatin (Cis), doxorubicin (Doxo)) in vitro. All three NC influence the phosphorylation level of selected DDR-related factors (i.e., pCHK1, pKAP1, pP53, pRPA32) in mono- and/or co-treatment. Both SA and NQ attenuate the Cis- and Doxo-induced G2/M-phase arrest and effectively stimulate caspase-mediated apoptosis. Notably, SA impacts DNA repair as reflected by enhanced steady-state levels of Cis-(1,2-GpG)-DNA adducts and Doxo-induced DNA double-strand breaks (DSB). Moreover, SA decreased the mRNA and protein expression of the homologous recombination (HR)-related DSB repair factors RAD51 and BRCA1. Both SA and NQ promote Cis- and Doxo-induced cytotoxicity in an additive to synergistic manner (CI ≤ 1.0). Summarizing, we conclude that SA promotes cAT-driven caspase-dependent cell death by interfering with DSB repair and DDR-related checkpoint control mechanisms. Hence, SA is considered as the most promising lead compound to evaluate its therapeutic window in forthcoming pre-clinical in vivo studies.
Collapse
Affiliation(s)
- Jana Aengenvoort
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany; (J.A.); (M.S.)
| | - Marlena Sekeres
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany; (J.A.); (M.S.)
| | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine University Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany;
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany; (J.A.); (M.S.)
- Correspondence: ; Tel.: +49-211-8113022
| |
Collapse
|
15
|
Prevention of anticancer therapy-induced neurotoxicity: putting DNA damage in perspective. Neurotoxicology 2022; 91:1-10. [PMID: 35487345 DOI: 10.1016/j.neuro.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 11/24/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a severe side effect of conventional cancer therapeutics (cAT) that significantly impacts the quality of life of tumor patients. The molecular mechanisms of CIPN are incompletely understood and there are no effective preventive or therapeutic measures available to date. Here, we present a brief overview of the current knowledge about mechanisms underlying CIPN and discuss DNA damage-related stress responses as feasible targets for the prevention of CIPN. In addition, we discuss that the nematode Caenorhabditis elegans is a useful 3R-conform model organism to further elucidate molecular mechanisms of CIPN and to identify novel lead compounds protecting from cAT-triggered neuropathy.
Collapse
|
16
|
Yang Y, Liu Y, Wang Y, Chao Y, Zhang J, Jia Y, Tie J, Hu D. Regulation of SIRT1 and Its Roles in Inflammation. Front Immunol 2022; 13:831168. [PMID: 35359990 PMCID: PMC8962665 DOI: 10.3389/fimmu.2022.831168] [Citation(s) in RCA: 250] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/15/2022] [Indexed: 12/28/2022] Open
Abstract
The silent information regulator sirtuin 1 (SIRT1) protein, a highly conserved NAD+-dependent deacetylase belonging to the sirtuin family, is a post-translational regulator that plays a role in modulating inflammation. SIRT1 affects multiple biological processes by deacetylating a variety of proteins including histones and non-histone proteins. Recent studies have revealed intimate links between SIRT1 and inflammation, while alterations to SIRT1 expression and activity have been linked to inflammatory diseases. In this review, we summarize the mechanisms that regulate SIRT1 expression, including upstream activators and suppressors that operate on the transcriptional and post-transcriptional levels. We also summarize factors that influence SIRT1 activity including the NAD+/NADH ratio, SIRT1 binding partners, and post-translational modifications. Furthermore, we underscore the role of SIRT1 in the development of inflammation by commenting on the proteins that are targeted for deacetylation by SIRT1. Finally, we highlight the potential for SIRT1-based therapeutics for inflammatory diseases.
Collapse
Affiliation(s)
- Yunshu Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yang Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yunwei Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yongyi Chao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jinxin Zhang
- Department of Emergency, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yanhui Jia
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jun Tie
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
17
|
Begum MK, Konja D, Singh S, Chlopicki S, Wang Y. Endothelial SIRT1 as a Target for the Prevention of Arterial Aging: Promises and Challenges. J Cardiovasc Pharmacol 2021; 78:S63-S77. [PMID: 34840264 DOI: 10.1097/fjc.0000000000001154] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 09/25/2021] [Indexed: 12/15/2022]
Abstract
ABSTRACT SIRT1, a member of the sirtuin family of longevity regulators, possesses potent activities preventing vascular aging. The expression and function of SIRT1 in endothelial cells are downregulated with age, in turn causing early vascular aging and predisposing various vascular abnormalities. Overexpression of SIRT1 in the vascular endothelium prevents aging-associated endothelial dysfunction and senescence, thus the development of hypertension and atherosclerosis. Numerous efforts have been directed to increase SIRT1 signaling as a potential strategy for different aging-associated diseases. However, the complex mechanisms underlying the regulation of SIRT1 have posed a significant challenge toward the design of specific and effective therapeutics. This review aimed to provide a summary on the regulation and function of SIRT1 in the vascular endothelium and to discuss the different approaches targeting this molecule for the prevention and treatment of age-related cardiovascular and cerebrovascular diseases.
Collapse
Affiliation(s)
- Musammat Kulsuma Begum
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Sandeep Singh
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland; and
- Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology
- The Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
18
|
Lee YH, Kim SJ, Fang X, Song NY, Kim DH, Suh J, Na HK, Kim KO, Baek JH, Surh YJ. JNK-mediated Ser27 phosphorylation and stabilization of SIRT1 promote growth and progression of colon cancer through deacetylation-dependent activation of Snail. Mol Oncol 2021; 16:1555-1571. [PMID: 34826187 PMCID: PMC8978515 DOI: 10.1002/1878-0261.13143] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/31/2021] [Accepted: 11/25/2021] [Indexed: 01/14/2023] Open
Abstract
Sirtuin 1 (SIRT1), an NAD+ -dependent histone/protein deacetylase, has multifaceted functions in various biological events such as inflammation, aging, and energy metabolism. The role of SIRT1 in carcinogenesis, however, is still under debate. Recent studies have indicated that aberrant overexpression of SIRT1 is correlated with metastasis and poor prognosis in several types of malignancy, including colorectal cancer. In the present study, we found that both SIRT1 and SIRT1 phosphorylated on serine 27 were coordinately upregulated in colon cancer patients' tissues and human colon cancer cell lines. This prompted us to investigate a role of phospho-SIRT1 in the context of colon cancer progression. A phosphorylation-defective mutant form of SIRT1, in which serine 27 was substituted by alanine (SIRT1-S27A), exhibited lower protein stability compared to that of wild-type SIRT1. Notably, human colon cancer (HCT-116) cells harboring the SIRT1-S27A mutation showed decreased cell proliferation and reduced capability to form xenograft tumor in athymic nude mice, which was accompanied by diminished transcriptional activity of Snail. HCT-116 cells carrying SIRT1-S27A were less capable of deacetylating the Snail protein, with a concomitant decrease in the levels of interleukin (IL)-6 and IL-8 mRNA transcripts. Taken together, these observations suggest that SIRT1 stabilized through phosphorylation on serine 27 exerts oncogenic effects at least partly through deacetylation-dependent activation of Snail and subsequent transcription of IL-6 and IL-8 in human colon cancer cells.
Collapse
Affiliation(s)
- Yeon-Hwa Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Su-Jung Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Xizhu Fang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Na-Young Song
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, South Korea
| | - Jinyoung Suh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul, South Korea
| | - Kyung-Ok Kim
- Gachon Medical Research Institute, Gil Medical Center, Gachon University, Incheon, Korea
| | - Jeong-Heum Baek
- Division of Colon and Rectal Surgery, Department of Surgery, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Young-Joon Surh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea.,Cancer Research Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
19
|
Visser H, Thomas AD. MicroRNAs and the DNA damage response: How is cell fate determined? DNA Repair (Amst) 2021; 108:103245. [PMID: 34773895 DOI: 10.1016/j.dnarep.2021.103245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/25/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022]
Abstract
It is becoming clear that the DNA damage response orchestrates an appropriate response to a given level of DNA damage, whether that is cell cycle arrest and repair, senescence or apoptosis. It is plausible that the alternative regulation of the DNA damage response (DDR) plays a role in deciding cell fate following damage. MicroRNAs (miRNAs) are associated with the transcriptional regulation of many cellular processes. They have diverse functions, affecting, presumably, all aspects of cell biology. Many have been shown to be DNA damage inducible and it is conceivable that miRNA species play a role in deciding cell fate following DNA damage by regulating the expression and activation of key DDR proteins. From a clinical perspective, miRNAs are attractive targets to improve cancer patient outcomes to DNA-damaging chemotherapy. However, cancer tissue is known to be, or to become, well adapted to DNA damage as a means of inducing chemoresistance. This frequently results from an altered DDR, possibly owing to miRNA dysregulation. Though many studies provide an overview of miRNAs that are dysregulated within cancerous tissues, a tangible, functional association is often lacking. While miRNAs are well-documented in 'ectopic biology', the physiological significance of endogenous miRNAs in the context of the DDR requires clarification. This review discusses miRNAs of biological relevance and their role in DNA damage response by potentially 'fine-tuning' the DDR towards a particular cell fate in response to DNA damage. MiRNAs are thus potential therapeutic targets/strategies to limit chemoresistance, or improve chemotherapeutic efficacy.
Collapse
Affiliation(s)
- Hartwig Visser
- Centre for Research in Biosciences, University of the West of England, Frenchay Campus, Bristol BS16 1QY, United Kingdom
| | - Adam D Thomas
- Centre for Research in Biosciences, University of the West of England, Frenchay Campus, Bristol BS16 1QY, United Kingdom.
| |
Collapse
|
20
|
Abstract
Sirtuin1 is a nutrient-sensitive class III histone deacetylase which is a well-known regulator of organismal lifespan. It has been extensively studied for its role in metabolic regulation as well. Along with its involvement in ageing and metabolism, Sirtuin1 directly deacetylates many critical proteins controlling cardiovascular pathophysiology. Studies using conditional expression and deletion of Sirtuin1 have revealed that it functions in a highly tissue/organ-specific manner. In the vasculature, Sirtuin1 controls endothelial homoeostasis by governing the expression of inflammatory mediators, oxidants and essential transcription factors. Adding to this complexity, Sirtuin1 expression and/or function is also governed by some of these target proteins. Therefore, the importance of better understanding the organ and tissue specificity of Sirtuin1 is highly desirable. Considering the huge volume of research done in this field, this review focuses on Sirtuin1 targets regulating vascular endothelial function. Here, we summarize the discovery of Sirtuin1 as a transcription controller and the further identification of direct target proteins involved in the vascular physiology. Overall, this review presents a holistic picture of the complex cross-talk involved in the molecular regulation of vascular physiology by Sirtuin1.
Collapse
Affiliation(s)
- Jitendra Kumar
- François M. Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Santosh Kumar
- François M. Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
21
|
Inhibition of the DSB repair protein RAD51 potentiates the cytotoxic efficacy of doxorubicin via promoting apoptosis-related death pathways. Cancer Lett 2021; 520:361-373. [PMID: 34389435 DOI: 10.1016/j.canlet.2021.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 01/04/2023]
Abstract
The anthracycline derivative doxorubicin (Doxo) induces DNA double-strand breaks (DSBs) by inhibition of DNA topoisomerase type II. Defective mismatch repair (MMR) contributes to Doxo resistance and has been reported for colon and mammary carcinomas. Here, we investigated the outcome of pharmacological inhibition of various DNA repair-related mechanisms on Doxo-induced cytotoxicity employing MMR-deficient HCT-116 colon carcinoma cells. Out of different inhibitors tested (i.e. HDACi, PARPi, MRE11i, RAD52i, RAD51i), we identified the RAD51-inhibitor B02 as the most powerful compound to synergistically increase Doxo-induced cytotoxicity. B02-mediated synergism rests on pleiotropic mechanisms, including pronounced G2/M arrest, damage to mitochondria and caspase-driven apoptosis. Of note, B02 also promotes the cytotoxicity of oxaliplatin and 5-fluoruracil (5-FU) in HCT-116 cells and, furthermore, also increases Doxo-induced cytotoxicity in MMR-proficient colon and mammary carcinoma cells. Summarizing, pharmacological inhibition of RAD51 is suggested to synergistically increase the cytotoxic efficacy of various types of conventional anticancer drugs in different tumor entities. Hence, pre-clinical in vivo studies are preferable to determine the therapeutic window of B02 in a clinically oriented therapeutic regimen.
Collapse
|
22
|
Hai R, He L, Shu G, Yin G. Characterization of Histone Deacetylase Mechanisms in Cancer Development. Front Oncol 2021; 11:700947. [PMID: 34395273 PMCID: PMC8360675 DOI: 10.3389/fonc.2021.700947] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/05/2021] [Indexed: 02/01/2023] Open
Abstract
Over decades of studies, accumulating evidence has suggested that epigenetic dysregulation is a hallmark of tumours. Post-translational modifications of histones are involved in tumour pathogenesis and development mainly by influencing a broad range of physiological processes. Histone deacetylases (HDACs) and histone acetyltransferases (HATs) are pivotal epigenetic modulators that regulate dynamic processes in the acetylation of histones at lysine residues, thereby influencing transcription of oncogenes and tumour suppressor genes. Moreover, HDACs mediate the deacetylation process of many nonhistone proteins and thus orchestrate a host of pathological processes, such as tumour pathogenesis. In this review, we elucidate the functions of HDACs in cancer.
Collapse
Affiliation(s)
- Rihan Hai
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.,School of Basic Medical Sciences, Central South University, Changsha, China
| | - Liuer He
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.,School of Basic Medical Sciences, Central South University, Changsha, China
| | - Guang Shu
- School of Basic Medical Sciences, Central South University, Changsha, China
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
23
|
Wang M, Lin H. Understanding the Function of Mammalian Sirtuins and Protein Lysine Acylation. Annu Rev Biochem 2021; 90:245-285. [PMID: 33848425 DOI: 10.1146/annurev-biochem-082520-125411] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein lysine acetylation is an important posttranslational modification that regulates numerous biological processes. Targeting lysine acetylation regulatory factors, such as acetyltransferases, deacetylases, and acetyl-lysine recognition domains, has been shown to have potential for treating human diseases, including cancer and neurological diseases. Over the past decade, many other acyl-lysine modifications, such as succinylation, crotonylation, and long-chain fatty acylation, have also been investigated and shown to have interesting biological functions. Here, we provide an overview of the functions of different acyl-lysine modifications in mammals. We focus on lysine acetylation as it is well characterized, and principles learned from acetylation are useful for understanding the functions of other lysine acylations. We pay special attention to the sirtuins, given that the study of sirtuins has provided a great deal of information about the functions of lysine acylation. We emphasize the regulation of sirtuins to illustrate that their regulation enables cells to respond to various signals and stresses.
Collapse
Affiliation(s)
- Miao Wang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA;
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA; .,Howard Hughes Medical Institute, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
24
|
Liebl MC, Moehlenbrink J, Becker H, Raddatz G, Abdeen SK, Aqeilan RI, Lyko F, Hofmann TG. DAZAP2 acts as specifier of the p53 response to DNA damage. Nucleic Acids Res 2021; 49:2759-2776. [PMID: 33591310 PMCID: PMC7969023 DOI: 10.1093/nar/gkab084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/13/2021] [Accepted: 01/28/2021] [Indexed: 12/30/2022] Open
Abstract
The DNA damage-responsive tumor suppressors p53 and HIPK2 are well established regulators of cell fate decision-making and regulate the cellular sensitivity to DNA-damaging drugs. Here, we identify Deleted in Azoospermia-associated protein 2 (DAZAP2), a small adaptor protein, as a novel regulator of HIPK2 and specifier of the DNA damage-induced p53 response. Knock-down or genetic deletion of DAZAP2 strongly potentiates cancer cell chemosensitivity both in cells and in vivo using a mouse tumour xenograft model. In unstressed cells, DAZAP2 stimulates HIPK2 polyubiquitination and degradation through interplay with the ubiquitin ligase SIAH1. Upon DNA damage, HIPK2 site-specifically phosphorylates DAZAP2, which terminates its HIPK2-degrading function and triggers its re-localization to the cell nucleus. Interestingly, nuclear DAZAP2 interacts with p53 and specifies target gene expression through modulating a defined subset of p53 target genes. Furthermore, our results suggest that DAZAP2 co-occupies p53 response elements to specify target gene expression. Collectively, our findings propose DAZAP2 as novel regulator of the DNA damage-induced p53 response that controls cancer cell chemosensitivity.
Collapse
Affiliation(s)
- Magdalena C Liebl
- Institute of Toxicology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Jutta Moehlenbrink
- Institute of Toxicology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Huong Becker
- Institute of Toxicology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Günter Raddatz
- Division of Epigenetics, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Suhaib K Abdeen
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Rami I Aqeilan
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel.,Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Frank Lyko
- Division of Epigenetics, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Thomas G Hofmann
- Institute of Toxicology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
25
|
Liu L, Xia G, Li P, Wang Y, Zhao Q. Sirt-1 Regulates Physiological Process and Exerts Protective Effects against Oxidative Stress. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5542545. [PMID: 33834065 PMCID: PMC8012122 DOI: 10.1155/2021/5542545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND Recent studies suggest a correlation between the reduced Sirt-1 expression with Alzheimer's diseases (AD) and depression, respectively, suggesting a possible pathogenic role of the altered Sirt-1 expression in neuronal degenerative diseases, such as AD and depression. However, the molecular mechanisms underlying how Sirt-1 reduction impairs neuronal functions remain unknown. METHODS We used the SK-N-SH neuroblastoma cells to study the role of Sirt-1 expression on physiological roles in neuronal cells. Gain of Sirt-1 was achieved by transiently transfecting Sirt-1 expression plasmid. Sirt-1-specific shRNA was used to elucidate the role of Sirt-1 loss of function. CCK-8 (Cell Counting Kit-8) assay and flow cytometry were used to evaluate cell proliferation. Semiquantitative western blotting was used to detect relative protein levels. A further luciferase reporter gene assay was employed to examine the effect of Sirt-1 expression on the transcriptional activity of p53. RT-qPCR was used to determine the mRNA levels of p21, Bax, and Bcl-2, which were the downstream target genes of p53. RESULTS Sirt-1 suppressed the p53 downstream gene p21 transcription, while shRNA-mediated Sirt-1 knockdown resulted in a significant increase in p21 expression, implying a possibility that Sirt-1 promotes neuron proliferation through suppressing p53 transcriptional activity. The mRNA and protein levels of p53 were not affected by the altered Sirt-1 expression, suggesting that Sirt-1 regulates the transcriptional regulatory activity of p53 rather than p53 expression. Indeed, we further confirmed that Sirt-1 appeared to inhibit p53 transcriptional activity by attenuating its acetylation and resulted in a decrease of p53's binding to the p21 promoter. Overexpressed Sirt-1 scavenged reactive oxygen species (ROS) production in SK-N-SH with H2O2. Knockdown of Sirt-1 presented opposite effect; the addition of EX527 (Sirt-1 inhibitor) increased ROS accumulation. CONCLUSIONS Oxidative stress induces Sirt-1 in neuron cells, and Sirt-1 promotes proliferation in SK-N-SH cells, which protects them from oxidative stress-induced cell death, potentially via suppressing the transcriptional activity of p53. These results provide a molecular explanation underlying how the reduced Sirt-1 potentially causes the AD and depression-related diseases, supporting the idea that Sirt-1 can possibly be used as a diagnostic biomarker and/or therapeutic drug target for the AD and depression-related diseases.
Collapse
Affiliation(s)
- Lei Liu
- Department of Mental Health and Psychiatry, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China 215006
- Department of Psychiatry, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China 550025
- Department of Psychiatry, Zaozhuang Mental Health Center, Zaozhuang, Shandong, China 277103
| | - Guangyuan Xia
- Department of Psychiatry, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China 550025
- College Students' Mental Health Education and Counseling Center, Guizhou Medical University, Guiyang, Guizhou, China 550004
| | - Peifan Li
- Department of Psychiatry, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China 550025
| | - Yiming Wang
- Department of Psychiatry, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China 550025
- College Students' Mental Health Education and Counseling Center, Guizhou Medical University, Guiyang, Guizhou, China 550004
| | - Qian Zhao
- Department of Nuclear Medicine, General Hospital of Ningxia Medical University, Ningxia, Ningxia Hui Autonomous Region, China 750004
| |
Collapse
|
26
|
Chen M, Chen Z, Huang D, Sun C, Xie J, Chen T, Zhao X, Huang Y, Li D, Wu B, Wu D. Myricetin inhibits TNF-α-induced inflammation in A549 cells via the SIRT1/NF-κB pathway. Pulm Pharmacol Ther 2021; 65:102000. [PMID: 33601000 DOI: 10.1016/j.pupt.2021.102000] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/27/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Although myricetin exerts anti-inflammation, anti-cancer, and anti-oxidation effects, the relationship between myricetin and tumor necrosis factor alpha (TNF-α) -stimulated inflammation in A549 cells remains unclear. This study sought to assess whether myricetin has an anti-inflammatory effect on TNF-α-induced A549 cells and clarify the potential mechanisms. METHODS Cell viability was examined with a Cell Counting Kit-8, and cytokine levels were determined by enzyme-linked immunosorbent assay and reverse transcription-quantitative PCR. Potential mechanisms were further explored by western blotting, immunofluorescence, and SIRT1 activity assays. RESULTS In A549 cells, TNF-α stimulation upregulated the production of interleukin-6 (IL-6) and interleukin-8 (IL-8). Moreover, TNF-α activated the nuclear factor-κB (NF-κB) pathway, as confirmed by IκB-α degradation, and phosphorylation and nuclear migration of NF-κB p65. However, pretreatment with myricetin significantly attenuated the observed responses triggered by TNF-α. Mechanistically, myricetin strongly increased the deacetylase activity through decreasing phosphorylation, but not expression, of sirtuin-1 (SIRT1) in TNF-α-stimulated A549 cells. Myricetin-mediated SIRT1 activation was further evidenced by the decreased acetylation of NF-κB p65 and p53. Subsequently, all of these concurrent changes were reversed by the addition of salermide (SIRT1 inhibitor), illustrating the critical role of SIRT1 in mediation of anti-inflammatory processes by myricetin. CONCLUSIONS Myricetin, an enhancer of SIRT1, inhibited TNF-α-induced NF-κB activation in A549 cells, therefore, reducing their inflammatory response. Our findings provide insight for novel therapies for inflammation-related diseases, such as asthma and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Min Chen
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Ziyu Chen
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Dan Huang
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Chaoqun Sun
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Jinye Xie
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Tingting Chen
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xuanna Zhao
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yujie Huang
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Dongming Li
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Bin Wu
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Dong Wu
- Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
27
|
Wang S, Yang Y, He X, Yang L, Wang J, Xia S, Liu D, Liu S, Yang L, Liu W, Duan H. Cdk5-Mediated Phosphorylation of Sirt1 Contributes to Podocyte Mitochondrial Dysfunction in Diabetic Nephropathy. Antioxid Redox Signal 2021; 34:171-190. [PMID: 32660255 DOI: 10.1089/ars.2020.8038] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Aims: Mitochondrial dysfunction contributes to podocyte injury, which is the leading cause of proteinuria in diabetic nephropathy (DN). In this study, we explored the role of cyclin-dependent kinase 5 (Cdk5) in mitochondrial dysfunction of podocytes under diabetic conditions. Results: Our results showed that the expression and activity of Cdk5 were significantly upregulated in vivo and in vitro under diabetic conditions, accompanied by the downregulation of synaptopodin and nephrin, as well as structural and functional mitochondrial dysfunction. Inhibition of Cdk5 with roscovitine or dominant-negative Cdk5 led to the attenuation of podocyte injury by upregulating synaptopodin and nephrin. The inhibition of Cdk5 also ameliorated mitochondrial dysfunction by decreasing reactive oxygen species levels and cytochrome c release, while increasing adenosine triphosphate production. Sirt1, an NAD+-dependent deacetylase, was decreased in podocytes with high glucose (HG) treatment; however, its phosphorylation level at S47 was significantly upregulated. We demonstrated that HG levels cause overactive Cdk5 to phosphorylate Sirt1 at S47. Suppression of Cdk5 reduced Sirt1 phosphorylation levels and mutation of S47 to nonphosphorable alanine (S47A), significantly attenuated podocyte injury and mitochondrial dysfunction in diabetic condition in vivo and in vitro. Innovation and Conclusion: Our study has demonstrated the role of Cdk5 in regulating mitochondrial function through Sirt1 phosphorylation and thus can potentially be a new therapeutic target for DN treatment. IRB number: 20190040. Antioxid. Redox Signal. 34, 171-190.
Collapse
Affiliation(s)
- Shuo Wang
- Key Laboratory of Kidney Diseases of Hebei Province, Department of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Yakun Yang
- Key Laboratory of Kidney Diseases of Hebei Province, Department of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Xingyu He
- Key Laboratory of Kidney Diseases of Hebei Province, Department of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Lin Yang
- Department of Nephrology and Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianrong Wang
- Department of Nephrology and Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shunjie Xia
- Key Laboratory of Kidney Diseases of Hebei Province, Department of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Dan Liu
- Key Laboratory of Kidney Diseases of Hebei Province, Department of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Shuxia Liu
- Key Laboratory of Kidney Diseases of Hebei Province, Department of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Li Yang
- Department of Cardiac Ultrasound, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wei Liu
- Key Laboratory of Kidney Diseases of Hebei Province, Department of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Huijun Duan
- Key Laboratory of Kidney Diseases of Hebei Province, Department of Pathology, Hebei Medical University, Shijiazhuang, China.,Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
28
|
Zhou S, Qiao YM, Liu YG, Liu D, Hu JM, Liao J, Li M, Guo Y, Fan LP, Li LY, Zhao M. Bone marrow derived mesenchymal stem cells pretreated with erythropoietin accelerate the repair of acute kidney injury. Cell Biosci 2020; 10:130. [PMID: 33292452 PMCID: PMC7667799 DOI: 10.1186/s13578-020-00492-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 10/30/2020] [Indexed: 12/18/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) represent a promising treatment option for acute kidney injury (AKI). The main drawbacks of MSCs therapy, including the lack of specific homing after systemic infusion and early cell death in the inflammatory microenvironment, directly affect the therapeutic efficacy of MSCs. Erythropoietin (EPO)-preconditioning of MSCs promotes their therapeutic effect, however, the underlying mechanism remains unknown. In this study, we sought to investigate the efficacy and mechanism of EPO in bone marrow derived mesenchymal stem cells (BMSCs) for AKI treatment. Results We found that incubation of BMSCs with ischemia/reperfusion(I/R)-induced AKI kidney homogenate supernatant (KHS) caused apoptosis in BMSCs, which was decreased by EPO pretreatment, indicating that EPO protected the cells from apoptosis. Further, we showed that EPO up-regulated silent information regulator 1 (SIRT1) and Bcl-2 expression and down-regulated p53 expression. This effect was partially reversed by SIRT1 siRNA intervention. The anti-apoptotic effect of EPO in pretreated BMSCs may be mediated through the SIRT1 pathway. In a rat AKI model, 24 h after intravenous infusion, GFP-BMSCs were predominantly located in the lungs. However, EPO pretreatment reduced the lung entrapment of BMSCs and increased their distribution in the target organs. AKI rats infused with EPO-BMSCs had significantly lower levels of serum IL-1β and TNF-α, and a significantly higher level of IL-10 as compared to rats infused with untreated BMSCs. The administration of EPO-BMSCs after reperfusion reduced serum creatinine, blood urea nitrogen, and pathological scores in I/R-AKI rats more effectively than BMSCs treatment did. Conclusions Our data suggest that EPO pretreatment enhances the efficacy of BMSCs to improve the renal function and pathological presentation of I/R-AKI rats.
Collapse
Affiliation(s)
- Song Zhou
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Yu-Ming Qiao
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Yong-Guang Liu
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Ding Liu
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Jian-Min Hu
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Jun Liao
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Min Li
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Ying Guo
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Li-Pei Fan
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Liu-Yang Li
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Ming Zhao
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Haizhu District, Guangzhou, 510280, Guangdong Province, China.
| |
Collapse
|
29
|
Haas J, Bloesel D, Bacher S, Kracht M, Schmitz ML. Chromatin Targeting of HIPK2 Leads to Acetylation-Dependent Chromatin Decondensation. Front Cell Dev Biol 2020; 8:852. [PMID: 32984337 PMCID: PMC7490299 DOI: 10.3389/fcell.2020.00852] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 08/10/2020] [Indexed: 11/13/2022] Open
Abstract
The protein kinase homeodomain-interacting protein kinase 2 (HIPK2) plays an important role in development and in the response to external cues. The kinase associates with an exceptionally large number of different transcription factors and chromatin regulatory proteins to direct distinct gene expression programs. In order to investigate the function of HIPK2 for chromatin compaction, HIPK2 was fused to the DNA-binding domains of Gal4 or LacI, thus allowing its specific targeting to binding sites for these transcription factors that were integrated in specific chromosome loci. Tethering of HIPK2 resulted in strong decompaction of euchromatic and heterochromatic areas. HIPK2-mediated heterochromatin decondensation started already 4 h after its chromatin association and required the functionality of its SUMO-interacting motif. This process was paralleled by disappearance of the repressive H3K27me3 chromatin mark, recruitment of the acetyltransferases CBP and p300 and increased histone acetylation at H3K18 and H4K5. HIPK2-mediated chromatin decompaction was strongly inhibited in the presence of a CBP/p300 inhibitor and completely blocked by the BET inhibitor JQ1, consistent with a causative role of acetylations for this process. Chromatin tethering of HIPK2 had only a minor effect on basal transcription, while it strongly boosted estrogen-triggered gene expression by acting as a transcriptional cofactor.
Collapse
Affiliation(s)
- Jana Haas
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany.,Member of the German Center for Lung Research, Giessen, Germany
| | - Daniel Bloesel
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany.,Member of the German Center for Lung Research, Giessen, Germany
| | - Susanne Bacher
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany.,Member of the German Center for Lung Research, Giessen, Germany
| | - Michael Kracht
- Member of the German Center for Lung Research, Giessen, Germany.,Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University, Giessen, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany.,Member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
30
|
Abstract
![]()
The cellular outcomes of chemical
exposure are as much about the
cellular response to the chemical as it is an effect of the chemical. We are growing in our understanding
of the genotoxic interaction between chemistry and biology. For example,
recent data has revealed the biological basis for mutation induction
curves for a methylating chemical, which has been shown to be dependent
on the repair capacity of the cells. However, this is just one end
point in the toxicity pathway from chemical exposure to cell death.
Much remains to be known in order for us to predict how cells will
respond to a certain dose. Methylating agents, a subset of alkylating
agents, are of particular interest, because of the variety of adverse
genetic end points that can result, not only at increasing doses,
but also over time. For instance, methylating agents are mutagenic,
their potency, for this end point, is determined by the cellular repair
capacity of an enzyme called methylguanine DNA-methyltransferase (MGMT)
and its ability to repair the induceed methyl adducts. However, methyl
adducts can become clastogenic. Erroneous biological processing will
convert mutagenic adducts to clastogenic events in the form of double
strand breaks (DSBs). How the cell responds to DSBs is via a cascade
of protein kinases, which is called the DNA damage response (DDR),
which will determine if the damage is repaired effectively, via homologous
recombination, or with errors, via nonhomologous end joining, or whether
the cell dies via apoptosis or enters senescence. The fate of cells
may be determined by the extent of damage and the resulting strength
of DDR signaling. Therefore, thresholds of damage may exist that determine
cell fate. Such thresholds would be dependent on each of the repair
and response mechanisms that these methyl adducts stimulate. The molecular
mechanism of how methyl adducts kill cells is still to be fully resolved.
If we are able to quantify each of these thresholds of damage for
a given cell, then we can ascertain, of the many adducts that are
induced, what proportion of them are mutagenic, what proportion are
clastogenic, and how many of these clastogenic events are toxic. This
review examines the possibility of dose and damage thresholds for
methylating agents, from the perspective of the underlying evolutionary
mechanisms that may be accountable.
Collapse
Affiliation(s)
- Adam D Thomas
- Centre for Research in Biosciences, University of the West of England, Frenchay Campus, Bristol BS16 1QY, United Kingdom
| |
Collapse
|
31
|
Regulation of histone deacetylase activities and functions by phosphorylation and its physiological relevance. Cell Mol Life Sci 2020; 78:427-445. [PMID: 32683534 DOI: 10.1007/s00018-020-03599-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 12/31/2022]
Abstract
Histone deacetylases (HDACs) are conserved enzymes that regulate many cellular processes by catalyzing the removal of acetyl groups from lysine residues on histones and non-histone proteins. As appropriate for proteins that occupy such an essential biological role, HDAC activities and functions are in turn highly regulated. Overwhelming evidence suggests that the dysregulation of HDACs plays a major role in many human diseases. The regulation of HDACs is achieved by multiple different mechanisms, including posttranslational modifications. One of the most common posttranslational modifications on HDACs is reversible phosphorylation. Many HDAC phosphorylations are context-dependent, occurring in specific tissues or as a consequence of certain stimuli. Additionally, whereas phosphorylation can regulate some HDACs in a non-specific manner, many HDAC phosphorylations result in specific consequences. Although some of these modifications support normal HDAC function, aberrations can contribute to disease development. Here we review and critically evaluate how reversible phosphorylation activates or deactivates HDACs and, thereby, regulates their many functions under various cellular and physiological contexts.
Collapse
|
32
|
Liang L, Xie R, Lu R, Ma R, Wang X, Wang F, Liu B, Wu S, Wang Y, Zhang H. Involvement of homodomain interacting protein kinase 2-c-Jun N-terminal kinase/c-Jun cascade in the long-term synaptic toxicity and cognition impairment induced by neonatal Sevoflurane exposure. J Neurochem 2020; 154:372-388. [PMID: 31705656 PMCID: PMC7496229 DOI: 10.1111/jnc.14910] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/23/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022]
Abstract
Sevoflurane is one of the most widely used anesthetics with recent concerns rising about its pediatric application. The synaptic toxicity and mechanisms underlying its long‐term cognition impairment remain unclear. In this study, we investigated the expression and roles of homeodomain interacting protein kinase 2 (HIPK2), a stress activating kinase involved in neuronal survival and synaptic plasticity, and its downstream c‐Jun N‐terminal kinase (JNK)/c‐Jun signaling in the long‐term toxicity of neonatal Sevoflurane exposure. Our data showed that neonatal Sevoflurane exposure results in impairment of memory, enhancement of anxiety, less number of excitatory synapses and lower levels of synaptic proteins in the hippocampus of adult rats without significant changes of hippocampal neuron numbers. Up‐regulation of HIPK2 and JNK/c‐Jun was observed in hippocampal granular neurons shortly after Sevoflurane exposure and persisted to adult. 5‐((6‐Oxo‐5‐(6‐(piperazin‐1‐yl)pyridin‐3‐yl)‐1,6‐dihydropyridin‐3‐yl)methylene)thiazolidine‐2,4‐dione trifluoroacetate, antagonist of HIPK2, could significantly rescue the cognition impairment, decrease in long‐term potentiation, reduction in spine density and activation of JNK/c‐Jun induced by Sevoflurane. JNK antagonist SP600125 partially restored synapse development and cognitive function without affecting the expression of HIPK2. These data, in together, revealed a novel role of HIPK2‐JNK/c‐Jun signaling in the long‐term synaptic toxicity and cognition impairment of neonatal Sevoflurane exposure, indicating HIPK2‐JNK/c‐Jun cascade as a potential target for reducing the synaptic toxicity of Sevoflurane. ![]()
Cover Image for this issue: doi: 10.1111/jnc.14757.
Collapse
Affiliation(s)
- Lirong Liang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Rougang Xie
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Rui Lu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Ruixue Ma
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Xiaoxia Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Fengjuan Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Bing Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Shengxi Wu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Yazhou Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| | - Hui Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research, Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, P. R. China
| |
Collapse
|
33
|
van Stuijvenberg J, Proksch P, Fritz G. Targeting the DNA damage response (DDR) by natural compounds. Bioorg Med Chem 2020; 28:115279. [PMID: 31980363 DOI: 10.1016/j.bmc.2019.115279] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 12/09/2019] [Accepted: 12/15/2019] [Indexed: 12/31/2022]
Abstract
Natural compounds (NC) are an important source of anticancer drugs. The genomic DNA of tumor cells is a major target of conventional anticancer therapeutics (cAT). DNA damage elicits a complex stress response programme termed DNA damage response (DDR), with the PI3-like kinase ATM and ATR being the key regulators. Since the DDR coordinates mechanisms of DNA repair and apoptosis, hence regulating the balance between death and survival, it is an attractive target of novel anticancer strategies. The aim of the study was to identify natural compounds derived from endophytic fungi, lichens, marine sponges or plants that interfere with mechanisms of the DDR. To this end, the cytotoxic and DDR modulating potency of 296 natural compounds, used alone or in combination with the cAT cisplatin (Cis) and doxorubicin (Doxo) was investigated by fluorescence-based analysis of the ATM/ATR-catalyzed S139 phosphorylation of histone 2AX (γH2AX), a surrogate marker of DNA damage-triggered DDR. After initial screening, a total of ten natural compounds were identified that were toxic in pancreatic carcinoma cells and activated the DDR on their own and/or promoted the DDR if used in combination with cAT. Their mode of action was shown to be independent of drug transport mechanisms. Based on their chemical structures, DDR modulatory activity and published data we suggest the marine NC 5-epi-nakijiquinone Q and 5-epi-ilimaquinone as well as the fungal compound secalonic acid F as most promising NC-based drug candidates for future synthesis of DDR-modulating chemical derivatives and their preclinical in vitro and in vivo testing.
Collapse
Affiliation(s)
- Jana van Stuijvenberg
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine University Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
34
|
Ling H, Peng L, Wang J, Rahhal R, Seto E. Histone Deacetylase SIRT1 Targets Plk2 to Regulate Centriole Duplication. Cell Rep 2019; 25:2851-2865.e3. [PMID: 30517871 DOI: 10.1016/j.celrep.2018.11.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 09/04/2018] [Accepted: 11/02/2018] [Indexed: 11/17/2022] Open
Abstract
The protein deacetylase SIRT1 (Sirtuin 1) regulates many cellular processes, including cell-cycle progression, DNA damage response, and metabolism. Although the centrosome is a key regulator of cell-cycle progression and genome stability, little is known concerning SIRT1 controlled centrosome-associated events. Here we report that the centrosome protein Plk2 is acetylated and undergoes deacetylation by SIRT1. Acetylation protects Plk2 from ubiquitination, and SIRT1-mediated deacetylation promotes ubiquitin-dependent degradation of Plk2. SIRT1 controls centriole duplication by temporally modulating centrosomal Plk2 levels. AURKA phosphorylates SIRT1 and promotes the SIRT1-Plk2 interaction in mitosis. In early-mid G1, phosphorylated SIRT1 deacetylates and promotes Plk2 degradation. In late G1, SIRT1 is hypophosphorylated and its affinity to Plk2 is decreased, resulting in a rapid accumulation of centrosomal Plk2, which contributes to the timely initiation of centriole duplication. Collectively, our findings uncover a critical role of SIRT1 in centriole duplication and provide a mechanistic insight into SIRT1-mediated centrosome-associated functions.
Collapse
Affiliation(s)
- Hongbo Ling
- George Washington University Cancer Center, Washington, DC 20052, USA; Department of Biochemistry & Molecular Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
| | - Lirong Peng
- George Washington University Cancer Center, Washington, DC 20052, USA; Department of Biochemistry & Molecular Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
| | - Jianbo Wang
- Department of Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Raneen Rahhal
- George Washington University Cancer Center, Washington, DC 20052, USA; Department of Biochemistry & Molecular Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
| | - Edward Seto
- George Washington University Cancer Center, Washington, DC 20052, USA; Department of Biochemistry & Molecular Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA.
| |
Collapse
|
35
|
Liebl MC, Hofmann TG. Cell Fate Regulation upon DNA Damage: p53 Serine 46 Kinases Pave the Cell Death Road. Bioessays 2019; 41:e1900127. [PMID: 31621101 DOI: 10.1002/bies.201900127] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/16/2019] [Indexed: 12/20/2022]
Abstract
Mild and massive DNA damage are differentially integrated into the cellular signaling networks and, in consequence, provoke different cell fate decisions. After mild damage, the tumor suppressor p53 directs the cellular response to cell cycle arrest, DNA repair, and cell survival, whereas upon severe damage, p53 drives the cell death response. One posttranslational modification of p53, phosphorylation at Serine 46, selectively occurs after severe DNA damage and is envisioned as a marker of the cell death response. However, the molecular mechanism of action of the p53 Ser46 phospho-isomer, the molecular timing of this phosphorylation event, and its activating effects on apoptosis and ferroptosis still await exploration. In this essay, the current body of evidence on the molecular function of this deadly p53 mark, its evolutionary conservation, and the regulation of the key players of this response, the p53 Serine 46 kinases, are reviewed and dissected.
Collapse
Affiliation(s)
- Magdalena C Liebl
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg, University Mainz, Obere Zahlbacher Str. 67, 55131, Mainz, Germany
| | - Thomas G Hofmann
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg, University Mainz, Obere Zahlbacher Str. 67, 55131, Mainz, Germany
| |
Collapse
|
36
|
Lu P, Hogan-Cann AD, Kamboj A, Roy Chowdhury SK, Aghanoori MR, Fernyhough P, Anderson CM. Poly(ADP-ribose) polymerase-1 inhibits mitochondrial respiration by suppressing PGC-1α activity in neurons. Neuropharmacology 2019; 160:107755. [PMID: 31487495 DOI: 10.1016/j.neuropharm.2019.107755] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/30/2019] [Accepted: 08/31/2019] [Indexed: 12/23/2022]
Abstract
Poly(ADP-ribose) polymerase-1 (PARP1) is a ubiquitous nuclear enzyme that regulates DNA repair and genomic stability. In oxidative genotoxic conditions, PARP1 activity is enhanced significantly, leading to excessive depletion of nicotinamide adenine dinucleotide (NAD+) and mitochondrial dysfunction. We hypothesized that PARP1-induced NAD+ depletion inhibits NAD+-dependent sirtuin deacetylase activity, thereby interfering with the mitochondrial regulator, peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α). The DNA alkylator, N'-Nitro-N-nitroso-N-methylguanidine (MNNG), induced NAD+ depletion, inhibited sirtuin deacetylase activity and enhanced acetylation of PGC-1α. This was associated with reduced interaction between PGC-1α and nuclear respiratory factor 1 (NRF-1), which is a nuclear transcription factor that drives mitochondrial replication by regulating mitochondrial transcription factor A (TFAM). MNNG also reduced binding of NRF-1 to the tfam upstream promoter region and reduced TFAM mRNA, mitochondrial DNA copy number and respiratory function. MNNG effects were mitigated by PARP1 inhibition and genetic loss of function, by enhancing intracellular NAD+ levels, and with sirtuin (SIRT1) gain of function, supporting a mechanism dependent on PARP1 activity, NAD+-depletion and SIRT1 inhibition. This and other work from our group supports a destructive sequelae of events related to PARP1-induced sirtuin inhibition and sirtuin-mediated regulation of transcription.
Collapse
Affiliation(s)
- Ping Lu
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Winnipeg Health Sciences Centre, Winnipeg, R3E 0Z3, Canada; Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E 0W3, Canada
| | - Adam D Hogan-Cann
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Winnipeg Health Sciences Centre, Winnipeg, R3E 0Z3, Canada; Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E 0W3, Canada
| | - Amit Kamboj
- Division of Neurodegenerative Disorders, St. Boniface Albrechtsen Research Centre, Winnipeg, R2H 2A6, Canada
| | - Subir K Roy Chowdhury
- Division of Neurodegenerative Disorders, St. Boniface Albrechtsen Research Centre, Winnipeg, R2H 2A6, Canada
| | - Mohamad-Reza Aghanoori
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E 0W3, Canada; Division of Neurodegenerative Disorders, St. Boniface Albrechtsen Research Centre, Winnipeg, R2H 2A6, Canada
| | - Paul Fernyhough
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E 0W3, Canada; Division of Neurodegenerative Disorders, St. Boniface Albrechtsen Research Centre, Winnipeg, R2H 2A6, Canada
| | - Christopher M Anderson
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Winnipeg Health Sciences Centre, Winnipeg, R3E 0Z3, Canada; Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E 0W3, Canada.
| |
Collapse
|
37
|
p53 at the Crossroads between Different Types of HDAC Inhibitor-Mediated Cancer Cell Death. Int J Mol Sci 2019; 20:ijms20102415. [PMID: 31096697 PMCID: PMC6567317 DOI: 10.3390/ijms20102415] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022] Open
Abstract
Cancer is a complex genetic and epigenetic-based disease that has developed an armada of mechanisms to escape cell death. The deregulation of apoptosis and autophagy, which are basic processes essential for normal cellular activity, are commonly encountered during the development of human tumors. In order to assist the cancer cell in defeating the imbalance between cell growth and cell death, histone deacetylase inhibitors (HDACi) have been employed to reverse epigenetically deregulated gene expression caused by aberrant post-translational protein modifications. These interfere with histone acetyltransferase- and deacetylase-mediated acetylation of both histone and non-histone proteins, and thereby exert a wide array of HDACi-stimulated cytotoxic effects. Key determinants of HDACi lethality that interfere with cellular growth in a multitude of tumor cells are apoptosis and autophagy, which are either mutually exclusive or activated in combination. Here, we compile known molecular signals and pathways involved in the HDACi-triggered induction of apoptosis and autophagy. Currently, the factors that determine the mode of HDACi-elicited cell death are mostly unclear. Correspondingly, we also summarized as yet established intertwined mechanisms, in particular with respect to the oncogenic tumor suppressor protein p53, that drive the interplay between apoptosis and autophagy in response to HDACi. In this context, we also note the significance to determine the presence of functional p53 protein levels in the cancer cell. The confirmation of the context-dependent function of autophagy will pave the way to improve the benefit from HDACi-mediated cancer treatment.
Collapse
|
38
|
Kreiner G, Sönmez A, Liss B, Parlato R. Integration of the Deacetylase SIRT1 in the Response to Nucleolar Stress: Metabolic Implications for Neurodegenerative Diseases. Front Mol Neurosci 2019; 12:106. [PMID: 31110473 PMCID: PMC6499230 DOI: 10.3389/fnmol.2019.00106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/09/2019] [Indexed: 01/10/2023] Open
Abstract
Understanding underlying mechanisms of neurodegenerative diseases is fundamental to develop effective therapeutic intervention. Yet they remain largely elusive, but metabolic, and transcriptional dysregulation are common events. Sirtuin 1 (SIRT1) is a nicotinamide adenine dinucleotide (NAD+)-dependent lysine deacetylase, regulating transcription, and critical for the cellular adaptations to metabolic stress. SIRT1 regulates the transcription of ribosomal RNA (rRNA), connecting the energetic state with cell growth and function. The activity of the transcription initiation factor-IA (TIF-IA) is important for the transcriptional regulation of ribosomal DNA (rDNA) genes in the nucleolus, and is also sensitive to changes in the cellular energetic state. Moreover, TIF-IA is responsive to nutrient-deprivation, neurotrophic stimulation, and oxidative stress. Hence, both SIRT1 and TIF-IA connect changes in cellular stress with transcriptional regulation and metabolic adaptation. Moreover, they finely tune the activity of the transcription factor p53, maintain mitochondrial function, and oxidative stress responses. Here we reviewed and discussed evidence that SIRT1 and TIF-IA are regulated by shared pathways and their activities preserve neuronal homeostasis in response to metabolic stressors. We provide evidence that loss of rDNA transcription due to altered TIF-IA function alters SIRT1 expression and propose a model of interdependent feedback mechanisms. An imbalance of this signaling might be a critical common event in neurodegenerative diseases. In conclusion, we provide a novel perspective for the prediction of the therapeutic benefits of the modulation of SIRT1- and nucleolar-dependent pathways in metabolic and neurodegenerative diseases.
Collapse
Affiliation(s)
- Grzegorz Kreiner
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Aynur Sönmez
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,New College, Oxford University, Oxford, United Kingdom
| | - Rosanna Parlato
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
39
|
Cao L, Yang G, Gao S, Jing C, Montgomery RR, Yin Y, Wang P, Fikrig E, You F. HIPK2 is necessary for type I interferon-mediated antiviral immunity. Sci Signal 2019; 12:12/573/eaau4604. [PMID: 30890658 DOI: 10.1126/scisignal.aau4604] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Precise control of interferons (IFNs) is crucial to maintain immune homeostasis. Here, we demonstrated that homeodomain-interacting protein kinase 2 (HIPK2) was required for the production of type I IFNs in response to RNA virus infection. HIPK2 deficiency markedly impaired IFN production in macrophages after vesicular stomatitis virus (VSV) infection, and HIPK2-deficient mice were more susceptible to lethal VSV disease than were wild-type mice. After VSV infection, HIPK2 was cleaved by active caspases, which released a hyperactive, N-terminal fragment that translocated to the nucleus and further augmented antiviral responses. In part, HIPK2 interacted with ELF4 and promoted its phosphorylation at Ser369, which enabled Ifn-b transcription. In addition, HIPK2 production was stimulated by type I IFNs to further enhance antiviral immunity. These data suggest that the kinase activity and nuclear localization of HIPK2 are essential for the production of type I IFNs.
Collapse
Affiliation(s)
- Lili Cao
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Guang Yang
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 208022, USA.,Department of Parasitology, Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, No. 601, Huangpu Avenue West, Guangzhou, Guangdong 510632, China
| | - Shandian Gao
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 208022, USA
| | - Chunxia Jing
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Ruth R Montgomery
- Section of Rheumatology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Penghua Wang
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 208022, USA.,Department of Immunology, University of Connecticut Health Center, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 208022, USA. .,Howard Hughes Medical Institute, Chevy Chase, MA 20815, USA
| | - Fuping You
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
40
|
|
41
|
SIRT1 suppresses p53-dependent apoptosis by modulation of p21 in osteoblast-like MC3T3-E1 cells exposed to fluoride. Toxicol In Vitro 2019; 57:28-38. [PMID: 30738887 DOI: 10.1016/j.tiv.2019.02.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/23/2019] [Accepted: 02/06/2019] [Indexed: 02/07/2023]
Abstract
Fluoride is very crucial for development of teeth and bones. Excessive fluoride, however, causes damage to teeth and bones resulting in serious public health problem. SIRT1 regulates physiological and pathological processes such as apoptosis and cell cycle. Although SIRT1 inhibits p53-mediated transactivation, how SIRT1 regulates p53 in fluorosis remains unclear. This study aims to investigate the involvement of SIRT1 in fluoride-induced cell cycle arrest and apoptosis in MC3T3-E1 cells and the underlying mechanism. Cell apoptosis was determined using Annexin V-FITC/PI dual staining, cell cycle detected with PI staining, intracellular ROS levels measured with DCFH-DA probe, and apoptosis-related protein expressions determined using Western blotting. Results showed that there was a promotion in apoptosis rate, intracellular ROS levels, the ratio of Bax/Bcl-2, protein expression (Cyt c, Caspase-3, p53, Ac-p53 and p21) and blockage of S phase after cells were exposed to NaF. Afterwards, the influence of SIRT1 on apoptosis was explored after SRT1720 (SIRT1 activator) and Ex-527 (SIRT1 inhibitor) was introduced. Results indicated that SRT1720 in combination with fluoride significantly decreased the intracellular ROS levels, the protein expression of Caspase-3, Ac-p53 and p21 and alleviated apoptosis, while it was reversed by Ex-527. Collectively, SIRT1 plays an essential role in protection against fluoride-induced oxidative stress and mitochondria-dependent apoptosis in MC3T3-E1 cells. The SIRT1/p53/p21 pathway may be a potential therapeutic target for fluorosis.
Collapse
|
42
|
Epigenetic Targeting of Autophagy via HDAC Inhibition in Tumor Cells: Role of p53. Int J Mol Sci 2018; 19:ijms19123952. [PMID: 30544838 PMCID: PMC6321134 DOI: 10.3390/ijms19123952] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022] Open
Abstract
Tumor development and progression is the consequence of genetic as well as epigenetic alterations of the cell. As part of the epigenetic regulatory system, histone acetyltransferases (HATs) and deacetylases (HDACs) drive the modification of histone as well as non-histone proteins. Derailed acetylation-mediated gene expression in cancer due to a delicate imbalance in HDAC expression can be reversed by histone deacetylase inhibitors (HDACi). Histone deacetylase inhibitors have far-reaching anticancer activities that include the induction of cell cycle arrest, the inhibition of angiogenesis, immunomodulatory responses, the inhibition of stress responses, increased generation of oxidative stress, activation of apoptosis, autophagy eliciting cell death, and even the regulation of non-coding RNA expression in malignant tumor cells. However, it remains an ongoing issue how tumor cells determine to respond to HDACi treatment by preferentially undergoing apoptosis or autophagy. In this review, we summarize HDACi-mediated mechanisms of action, particularly with respect to the induction of cell death. There is a keen interest in assessing suitable molecular factors allowing a prognosis of HDACi-mediated treatment. Addressing the results of our recent study, we highlight the role of p53 as a molecular switch driving HDACi-mediated cellular responses towards one of both types of cell death. These findings underline the importance to determine the mutational status of p53 for an effective outcome in HDACi-mediated tumor therapy.
Collapse
|
43
|
Liu D, Zhong L, Yuan Z, Yao J, Zhong P, Liu J, Yao S, Zhao Y, Liu L, Chen M, Li L, Liu B. miR-382-5p modulates the ATRA-induced differentiation of acute promyelocytic leukemia by targeting tumor suppressor PTEN. Cell Signal 2018; 54:1-9. [PMID: 30453015 DOI: 10.1016/j.cellsig.2018.11.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 01/03/2023]
Abstract
In acute promyelocytic leukemia (APL), all-trans retinoic acid (ATRA) treatment induces granulocytic differentiation and maturation. MicroRNAs play pivotal roles in formation of the leukemic phenotype. Previously, microRNA-382-5p (miR-382-5p) was upregulated in acute myeloid leukemia (AML) with t(15;17). In the present study, we found that miR-382-5p expression was elevated with ATRA-induced differentiation of APL. To investigate the potential functional role of miR-382-5p in APL differentiation, an APL cell line was transfected with miR-382-5p mimics, inhibitors, or negative control (NC). The results showed in APL cell line NB4 that miR-382-5p downregulation upon ATRA treatment was a key event in the drug response. Mechanistic investigations revealed that miR-382-5p targeted the ATRA-regulated tumor suppressor gene PTEN through direct binding to its 3' UTR. Enforced expression of miR-382-5p or specific PTEN inhibitors inhibited ATRA-induced granulocytic differentiation via regulation of the cell cycle regulator cyclinD1. Conversely, PTEN overexpression promoted differentiation and enhanced sensitivity of NB4 cell line to physiological levels of ATRA. Finally, we found that PTEN overexpression restored PML nuclear bodies (NBs). Taken together, these results demonstrated that up-regulated miR-382-5p in NB4 cell line inhibited granulocytic differentiation through the miR-382-5p/PTEN axis, uncovering PTEN as a critical element in the granulocytic differentiation program induced by ATRA in APL.
Collapse
Affiliation(s)
- Dongdong Liu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China; Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Liang Zhong
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zhen Yuan
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Juanjuan Yao
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Pengqiang Zhong
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Junmei Liu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Shifei Yao
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Yi Zhao
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Lu Liu
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Min Chen
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Lianwen Li
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Beizhong Liu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China; Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
44
|
Wang W, Li F, Xu Y, Wei J, Zhang Y, Yang H, Gao B, Yu G, Fang D. JAK1-mediated Sirt1 phosphorylation functions as a negative feedback of the JAK1-STAT3 pathway. J Biol Chem 2018; 293:11067-11075. [PMID: 29789426 DOI: 10.1074/jbc.ra117.001387] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 05/08/2018] [Indexed: 11/06/2022] Open
Abstract
The type III NAD-dependent histone deacetylase Sirt1 plays important roles in a variety of pathobiological functions through targeting either the acetylated histones or transcription factors. However, the molecular mechanisms underlying how the Sirt1 functions are regulated remain vague. Herein we identified that the Janus kinase 1 (JAK1) interacts with Sirt1 and catalyzes its phosphorylation at the tyrosine residues of 280 and 301, both of which are highly conserved and located in the histone deacetylase catalytic domain of Sirt1. IL-6 stimulation enhanced Sirt1 interaction with JAK1 and JAK1-mediated Sirt1 phosphorylation. Interestingly, JAK1-mediated Sirt1 phosphorylation did not alter Sirt1 deacetylase catalytic activity, but instead it is required for Sirt1 interaction with the downstream transcription factor STAT3. JAK1-mediated phosphorylation enhanced Sirt1 suppression of STAT3 acetylation and transcriptional activity. As a consequence, Sirt1 activation attenuates IL-6 activity in protecting cancer cells from chemotherapeutic drug-induced apoptosis. Our studies identify JAK1 as a previously unappreciated tyrosine kinase of Sirt1 and reveal a novel negative feedback of the JAK1-STAT3 pathway.
Collapse
Affiliation(s)
- Wenhui Wang
- From the Department of Oncology, Wei Fang People's Hospital, Wei Fang, Shandong Province, 261041 China.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Fei Li
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Yuanming Xu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Juncheng Wei
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Yana Zhang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and.,Department of Otolaryngology-Head and Neck Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623 China
| | - Heeyoung Yang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Beixue Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| | - Guohua Yu
- From the Department of Oncology, Wei Fang People's Hospital, Wei Fang, Shandong Province, 261041 China,
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, and
| |
Collapse
|
45
|
Lallemand-Breitenbach V, de Thé H. PML nuclear bodies: from architecture to function. Curr Opin Cell Biol 2018; 52:154-161. [PMID: 29723661 DOI: 10.1016/j.ceb.2018.03.011] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/06/2018] [Accepted: 03/30/2018] [Indexed: 12/11/2022]
Abstract
PML nuclear bodies are nucleated by the PML protein, which polymerizes into spherical shells where it concentrates many unrelated partner proteins. Emerging data has connected PML bodies to post-translational control, notably conjugation by SUMOs. High concentrations of SUMO-bound proteins were proposed to condense into liquid-like droplets and such phase transition may occur within NBs. Many stress pathways modulate NB formation and recent findings have directly implicated PML in oxidative stress response in vivo. PML may also undergo SUMO-dependent ubiquitination/degradation. We highlight recent advances linking PML to partner degradation and other adaptative post-translational modifications in the context of chromatin remodeling, telomere biology, senescence or viral infections.
Collapse
Affiliation(s)
- Valérie Lallemand-Breitenbach
- INSERM U944-CNRS UMR 7212, Equipe labellisée par la Ligue Nationale contre le Cancer, France; Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, 1 Avenue Claude Vellefaux, 75475 Paris cedex 10, France; Collège de France, PSL Research University, 11 Place Marcelin Berthelot, 75231 Paris Cedex 05, France.
| | - Hugues de Thé
- INSERM U944-CNRS UMR 7212, Equipe labellisée par la Ligue Nationale contre le Cancer, France; Université Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis, 1 Avenue Claude Vellefaux, 75475 Paris cedex 10, France; Collège de France, PSL Research University, 11 Place Marcelin Berthelot, 75231 Paris Cedex 05, France; Service de Biochimie, Hôpital St. Louis, AP-HP, Paris, France
| |
Collapse
|
46
|
Kino T. GR-regulating Serine/Threonine Kinases: New Physiologic and Pathologic Implications. Trends Endocrinol Metab 2018; 29:260-270. [PMID: 29501228 DOI: 10.1016/j.tem.2018.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 12/17/2022]
Abstract
Glucocorticoid hormones, end products of the hypothalamic-pituitary-adrenal axis, virtually influence all human functions both in a basal homeostatic condition and under stress. The glucocorticoid receptor (GR), a nuclear hormone receptor superfamily protein, mediates these actions of glucocorticoids by acting as a ligand-dependent transcription factor. Because glucocorticoid actions are diverse and strong, many biological pathways adjust them in local tissues by targeting the GR signaling pathway as part of the regulatory loop coordinating complex human functions. Phosphorylation of GR protein by serine/threonine kinases is one of the major regulatory mechanisms for this communication. In this review, recent progress in research investigating GR phosphorylation by these kinases is discussed, along with the possible physiologic and pathophysiologic implications.
Collapse
Affiliation(s)
- Tomoshige Kino
- Department of Human Genetics, Division of Translational Medicine, Sidra Medical and Research Center, Doha 26999, Qatar.
| |
Collapse
|
47
|
Zullo A, Simone E, Grimaldi M, Musto V, Mancini FP. Sirtuins as Mediator of the Anti-Ageing Effects of Calorie Restriction in Skeletal and Cardiac Muscle. Int J Mol Sci 2018; 19:E928. [PMID: 29561771 PMCID: PMC5979282 DOI: 10.3390/ijms19040928] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/14/2018] [Accepted: 03/20/2018] [Indexed: 12/17/2022] Open
Abstract
Fighting diseases and controlling the signs of ageing are the major goals of biomedicine. Sirtuins, enzymes with mainly deacetylating activity, could be pivotal targets of novel preventive and therapeutic strategies to reach such aims. Scientific proofs are accumulating in experimental models, but, to a minor extent, also in humans, that the ancient practice of calorie restriction could prove an effective way to prevent several degenerative diseases and to postpone the detrimental signs of ageing. In the present review, we summarize the evidence about the central role of sirtuins in mediating the beneficial effects of calorie restriction in skeletal and cardiac muscle since these tissues are greatly damaged by diseases and advancing years. Moreover, we entertain the possibility that the identification of sirtuin activators that mimic calorie restriction could provide the benefits without the inconvenience of this dietary style.
Collapse
Affiliation(s)
- Alberto Zullo
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
- CEINGE Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy.
| | - Emanuela Simone
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| | - Maddalena Grimaldi
- Department of Pediatric Oncology and Hematology, Charité University Hospital, 13353 Berlin, Germany.
| | - Vincenzina Musto
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| | | |
Collapse
|
48
|
Matt S, Hofmann TG. Crosstalk between p53 modifiers at PML bodies. Mol Cell Oncol 2018; 5:e1074335. [PMID: 30250876 DOI: 10.1080/23723556.2015.1074335] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 07/13/2015] [Accepted: 07/13/2015] [Indexed: 10/23/2022]
Abstract
Tumor protein p53 (TP53, best known as p53), the most frequently mutated tumor suppressor in cancer, plays a central role in cell fate decisions induced by DNA damage. Regulation of p53 activity by post-translational modifications has been linked to promyelocytic leukemia nuclear bodies (PML-NBs), where p53 encounters many of its regulators. Recent evidence implies that crosstalk between p53 regulators at the PML-NB shapes post-translational modifications and function of p53.
Collapse
Affiliation(s)
- Sonja Matt
- German Cancer Research Center (dkfz), Cellular Senescence Group, DKFZ-ZMBH Alliance, Heidelberg Germany
| | - Thomas G Hofmann
- German Cancer Research Center (dkfz), Cellular Senescence Group, DKFZ-ZMBH Alliance, Heidelberg Germany
| |
Collapse
|
49
|
Song J, Yang B, Jia X, Li M, Tan W, Ma S, Shi X, Feng L. Distinctive Roles of Sirtuins on Diabetes, Protective or Detrimental? Front Endocrinol (Lausanne) 2018; 9:724. [PMID: 30559718 PMCID: PMC6284472 DOI: 10.3389/fendo.2018.00724] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/15/2018] [Indexed: 12/21/2022] Open
Abstract
Dysregulation of metabolic pathways leads to type 2 diabetes, characteristic of high glucose concentration caused by insulin resistance. The histone deacetylases sirtuins exhibit remarkable enzymatic activities. Accumulating evidence indicates that sirtuins can be pharmacologically activated to ameliorate diabetes. Here, we evaluated different roles of sirtuins (SIRT1-SIRT7) in diabetes progression and described their involvement in metabolic pathways of skeletal muscle, adipose tissue and liver. The nuclear sirtuins, SIRT1, SIRT6, and SIRT7, regulate the activity of key transcription factors and cofactors in almost all tissues with the cellular responses to energy demands. The mitochondrial sirtuins, SIRT3, SIRT4, and SIRT5, regulate the activity of mitochondrial enzymes in response to fasting and calorie restriction. Moreover, genetic polymorphisms of SIRT1 and SIRT2 have been reported to associate with diabetes development. It's worth noting that SIRT1, SIRT2, SIRT3, and SIRT6 are positive regulators of insulin resistance in most cases. In the opposite, SIRT4 and SIRT7 inhibit insulin secretion and fatty acid oxidation. Identification of SIRT1 activators for diabetes has gained wide attention, such as metformin, resveratrol, and resveratrol derivatives. Randomized, prospective, and large-scale clinical trials are warrant to uncover the responsibilities of SIRTs modulators on diabetes progress.
Collapse
Affiliation(s)
- Jie Song
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
- Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bing Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mingyu Li
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Tan
- Affiliated Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shitang Ma
- Life and Health college, Anhui Science and Technology University, Fengyang, China
| | - Xinhong Shi
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liang Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
- *Correspondence: Liang Feng
| |
Collapse
|
50
|
Suzuki M, Ikeda A, Bartlett JD. Sirt1 overexpression suppresses fluoride-induced p53 acetylation to alleviate fluoride toxicity in ameloblasts responsible for enamel formation. Arch Toxicol 2017; 92:1283-1293. [PMID: 29185024 DOI: 10.1007/s00204-017-2135-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 12/21/2022]
Abstract
Low-dose fluoride is an effective caries prophylactic, but high-dose fluoride is an environmental health hazard that causes skeletal and dental fluorosis. Treatments to prevent fluorosis and the molecular pathways responsive to fluoride exposure remain to be elucidated. Previously we showed that fluoride activates SIRT1 as an adaptive response to protect cells. Here, we demonstrate that fluoride induced p53 acetylation (Ac-p53) [Lys379], which is a SIRT1 deacetylation target, in ameloblast-derived LS8 cells in vitro and in enamel organ in vivo. Here we assessed SIRT1 function on fluoride-induced Ac-p53 formation using CRISPR/Cas9-mediated Sirt1 knockout (LS8Sirt/KO) cells or CRISPR/dCas9/SAM-mediated Sirt1 overexpressing (LS8Sirt1/over) cells. NaF (5 mM) induced Ac-p53 formation and increased cell cycle arrest via Cdkn1a/p21 expression in Wild-type (WT) cells. However, fluoride-induced Ac-p53 was suppressed by the SIRT1 activator resveratrol (50 µM). Without fluoride, Ac-p53 persisted in LS8Sirt/KO cells, whereas it decreased in LS8Sirt1/over. Fluoride-induced Ac-p53 formation was also suppressed in LS8Sirt1/over cells. Compared to WT cells, fluoride-induced Cdkn1a/p21 expression was elevated in LS8Sirt/KO and these cells were more susceptible to fluoride-induced growth inhibition. In contrast, LS8Sirt1/over cells were significantly more resistant. In addition, fluoride-induced cytochrome-c release and caspase-3 activation were suppressed in LS8Sirt1/over cells. Fluoride induced expression of the DNA double strand break marker γH2AX in WT cells and this was augmented in LS8Sirt1/KO cells, but was attenuated in LS8Sirt1/over cells. Our results suggest that SIRT1 deacetylates Ac-p53 to mitigate fluoride-induced cell growth inhibition, mitochondrial damage, DNA damage and apoptosis. This is the first report implicating Ac-p53 in fluoride toxicity.
Collapse
Affiliation(s)
- Maiko Suzuki
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA
| | - Atsushi Ikeda
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA
| | - John D Bartlett
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|