1
|
Tyagi W, Das S. Temporal regulation of acetylation status determines PARP1 role in DNA damage response and metabolic homeostasis. SCIENCE ADVANCES 2024; 10:eado7720. [PMID: 39423262 PMCID: PMC11488539 DOI: 10.1126/sciadv.ado7720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1) is an abundant nuclear protein involved in DNA repair, chromatin structure, and transcription. However, the regulation of its different functions remains poorly understood. Here, we report the role of PARP1 acetylation status in modulating its DNA repair and transactivation functions. We demonstrate that histone deacetylase 5 (HDAC5) determines PARP1 acetylation at Lys498 and Lys521 sites. HDAC5-mediated deacetylation at Lys498 site regulates PARP1 DNA damage response and facilitates efficient recruitment of DNA repair factors at damaged sites, thereby promoting cell survival. Additionally, HDAC5-mediated deacetylation at Lys521 site promotes PARP1 coactivator function, resulting in induction of proliferative and metabolic genes in an activating transcription factor 4-dependent manner. Thus, PARP1 induces metabolic adaptation to spur malignant phenotype. Our studies in mouse tumor models suggest that pharmacological inhibition of PARP1 enzymatic activity does not block tumor progression robustly as transactivation function remains unperturbed. These findings provide key mechanistic insights into PARP1 regulation and expand its role in tumor development.
Collapse
Affiliation(s)
- Witty Tyagi
- Molecular Oncology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi-110067, India
| | - Sanjeev Das
- Molecular Oncology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi-110067, India
| |
Collapse
|
2
|
Lin M, Zhou W, Wang Y, Ye J, Jiang T, Han S, Zhu F, Ye M, Fang Z. HDAC5 deacetylates c-Myc and facilitates cell cycle progression in hepatocellular carcinoma cells. Cell Signal 2024; 124:111386. [PMID: 39243916 DOI: 10.1016/j.cellsig.2024.111386] [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: 05/26/2024] [Revised: 08/09/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Histone deacetylase 5 (HDAC5) is an enzyme that deacetylates lysine residues on the N-terminal of histones and other proteins. It has been reported that HDAC5 deacetylates p53, the critical factor regulating cell cycle, in response to cellular stress, but the transcriptional products haven't been identified. Herein, we used p53 signaling pathway qPCR-chip to determine how HDAC5-mediated deacetylation of p53 affects cell cycle. However, validation using immunoblotting analysis revealed that acetylation of p53 at K120 impacted little to the expression of the genes identified using the qPCR-chip, indicating HDAC5 might deacetylate some other proteins to facilitate cell cycle via transactivating the differentially expressed genes determined by the qPCR-chip. The subsequent assays demonstrated that HDAC5 deacetylated c-Myc at K143 and K157 to facilitate the transactivation of CDK1, CDK4, and CDC25C, promoting cell cycle progression of hepatocellular carcinoma (HCC). This study shows that HDAC5 plays important roles in modulating deacetylation of c-Myc and regulating cell cycle progression, and it proves that LMK-235, the inhibitor targeting HDAC5 potentially serves as a drug for combating HCC via promoting acetylation of c-Myc at K143 and K157.
Collapse
Affiliation(s)
- Min Lin
- Central Laboratory, Sanmen People's Hospital, Sanmen 317100, China.
| | - Weihua Zhou
- Department of Pathology, Sanmen People's Hospital, Sanmen 317100, China.
| | - Yizhang Wang
- Central Laboratory, Sanmen People's Hospital, Sanmen 317100, China.
| | - Jiangwei Ye
- Department of General Surgery, Sanmen People's Hospital, No. 15 Taihe Road, Hairun Street, Sanmen 317100, China.
| | - TingJia Jiang
- Department of Pathology, Sanmen People's Hospital, Sanmen 317100, China.
| | - Shanshan Han
- Central Laboratory, Sanmen People's Hospital, Sanmen 317100, China.
| | - Fengjiao Zhu
- Central Laboratory, Sanmen People's Hospital, Sanmen 317100, China.
| | - Ming Ye
- Department of General Surgery, Sanmen People's Hospital, No. 15 Taihe Road, Hairun Street, Sanmen 317100, China.
| | - Zejun Fang
- Central Laboratory, Sanmen People's Hospital, Sanmen 317100, China.
| |
Collapse
|
3
|
Talukdar P, Pal S, Biswas D. Post-translational modification-dependent oligomerization switch in regulation of global transcription and DNA damage repair during genotoxic stress. Nat Commun 2024; 15:4128. [PMID: 38750015 PMCID: PMC11096357 DOI: 10.1038/s41467-024-48530-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
Mechanisms of functional cross-talk between global transcriptional repression and efficient DNA damage repair during genotoxic stress are poorly known. In this study, using human AF9 as representative of Super Elongation Complex (SEC) components, we delineate detailed mechanisms of these processes. Mechanistically, we describe that Poly-Serine domain-mediated oligomerization is pre-requisite for AF9 YEATS domain-mediated TFIID interaction-dependent SEC recruitment at the promoter-proximal region for release of paused RNA polymerase II. Interestingly, during genotoxic stress, CaMKII-mediated phosphorylation-dependent nuclear export of AF9-specific deacetylase HDAC5 enhances concomitant PCAF-mediated acetylation of K339 residue. This causes monomerization of AF9 and reduces TFIID interaction for transcriptional downregulation. Furthermore, the K339 acetylation-dependent enhanced AF9-DNA-PKc interaction leads to phosphorylation at S395 residue which reduces AF9-SEC interaction resulting in transcriptional downregulation and efficient repair of DNA damage. After repair, nuclear re-entry of HDAC5 reduces AF9 acetylation and restores its TFIID and SEC interaction to restart transcription.
Collapse
Affiliation(s)
| | - Sujay Pal
- CSIR-Indian Institute of Chemical Biology, Kolkata, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Debabrata Biswas
- CSIR-Indian Institute of Chemical Biology, Kolkata, 700032, India.
| |
Collapse
|
4
|
Lee SM, Cho J, Choi S, Kim DH, Ryu JW, Kim I, Woo DC, Sung YH, Jeong JY, Baek IJ, Pack CG, Rho JK, Lee SW, Ha CH. HDAC5-mediated exosomal Maspin and miR-151a-3p as biomarkers for enhancing radiation treatment sensitivity in hepatocellular carcinoma. Biomater Res 2023; 27:134. [PMID: 38102691 PMCID: PMC10725039 DOI: 10.1186/s40824-023-00467-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Tumor-derived exosomes are critical elements of the cell-cell communication response to various stimuli. This study aims to reveal that the histone deacetylase 5 (HDAC5) and p53 interaction upon radiation in hepatocellular carcinoma intricately regulates the secretion and composition of exosomes. METHODS We observed that HDAC5 and p53 expression were significantly increased by 2 Gy and 4 Gy radiation exposure in HCC. Normal- and radiation-derived exosomes released by HepG2 were purified to investigate the exosomal components. RESULTS We found that in the radiation-derived exosome, exosomal Maspin was notably increased. Maspin is known as an anti-angiogenic gene. The expression of Maspin was regulated at the cellular level by HDAC5, and it was elaborately regulated and released in the exosome. Radiation-derived exosome treatment caused significant inhibition of angiogenesis in HUVECs and mouse aortic tissues. Meanwhile, we confirmed that miR-151a-3p was significantly reduced in the radiation-derived exosome through exosomal miRNA sequencing, and three HCC-specific exosomal miRNAs were also decreased. In particular, miR-151a-3p induced an anti-apoptotic response by inhibiting p53, and it was shown to induce EMT and promote tumor growth by regulating p53-related tumor progression genes. In the HCC xenograft model, radiation-induced exosome injection significantly reduced angiogenesis and tumor size. CONCLUSIONS Our present findings demonstrated HDAC5 is a vital gene of the p53-mediated release of exosomes resulting in tumor suppression through anti-cancer exosomal components in response to radiation. Finally, we highlight the important role of exosomal Maspin and mi-151a-3p as a biomarker in enhancing radiation treatment sensitivity. Therapeutic potential of HDAC5 through p53-mediated exosome modulation in radiation treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Seung Min Lee
- Department of Biochemistry and Molecular Biology and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Jeongin Cho
- Department of Biochemistry and Molecular Biology and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Sujin Choi
- Department of Biochemistry and Molecular Biology and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Dong Ha Kim
- Department of Biochemistry and Molecular Biology and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Je-Won Ryu
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Inki Kim
- Department of Pharmacology, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Cheol Woo
- Department of Biomedical Engineering, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young Hoon Sung
- Department of Cell and Genetic Engineering, Asan Medical Center, Asan Institute for Life Sciences University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin-Yong Jeong
- Department of Microbiology, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In-Jeoung Baek
- Department of Cell and Genetic Engineering, Asan Medical Center, Asan Institute for Life Sciences University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chan-Gi Pack
- Department of Biomedical Engineering, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Kyung Rho
- Department of Biochemistry and Molecular Biology and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Sang-Wook Lee
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| | - Chang Hoon Ha
- Department of Biochemistry and Molecular Biology and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| |
Collapse
|
5
|
Martin SD, Connor T, Sanigorski A, McEwen KA, Henstridge DC, Nijagal B, De Souza D, Tull DL, Meikle PJ, Kowalski GM, Bruce CR, Gregorevic P, Febbraio MA, Collier FM, Walder KR, McGee SL. Class IIa HDACs inhibit cell death pathways and protect muscle integrity in response to lipotoxicity. Cell Death Dis 2023; 14:787. [PMID: 38040704 PMCID: PMC10692215 DOI: 10.1038/s41419-023-06319-5] [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: 08/01/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/03/2023]
Abstract
Lipotoxicity, the accumulation of lipids in non-adipose tissues, alters the metabolic transcriptome and mitochondrial metabolism in skeletal muscle. The mechanisms involved remain poorly understood. Here we show that lipotoxicity increased histone deacetylase 4 (HDAC4) and histone deacetylase 5 (HDAC5), which reduced the expression of metabolic genes and oxidative metabolism in skeletal muscle, resulting in increased non-oxidative glucose metabolism. This metabolic reprogramming was also associated with impaired apoptosis and ferroptosis responses, and preserved muscle cell viability in response to lipotoxicity. Mechanistically, increased HDAC4 and 5 decreased acetylation of p53 at K120, a modification required for transcriptional activation of apoptosis. Redox drivers of ferroptosis derived from oxidative metabolism were also reduced. The relevance of this pathway was demonstrated by overexpression of loss-of-function HDAC4 and HDAC5 mutants in skeletal muscle of obese db/db mice, which enhanced oxidative metabolic capacity, increased apoptosis and ferroptosis and reduced muscle mass. This study identifies HDAC4 and HDAC5 as repressors of skeletal muscle oxidative metabolism, which is linked to inhibition of cell death pathways and preservation of muscle integrity in response to lipotoxicity.
Collapse
Affiliation(s)
- Sheree D Martin
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
| | - Timothy Connor
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
| | - Andrew Sanigorski
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
| | - Kevin A McEwen
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
| | - Darren C Henstridge
- College of Health and Medicine, School of Health Sciences, University of Tasmania, Launceston, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Brunda Nijagal
- Metabolomics Australia, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - David De Souza
- Metabolomics Australia, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dedreia L Tull
- Metabolomics Australia, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Greg M Kowalski
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
- Institute of Physical Activity and Nutrition (IPAN) and School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, 3216, Australia
| | - Clinton R Bruce
- Institute of Physical Activity and Nutrition (IPAN) and School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, 3216, Australia
| | - Paul Gregorevic
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | | | - Ken R Walder
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia
| | - Sean L McGee
- Institute for Mental and Physical Heath and Clinical Translation (IMPACT) and Metabolic Research Unit, School of Medicine, Deakin University, Geelong, VIC, 3216, Australia.
| |
Collapse
|
6
|
Sharma S, Tyagi W, Tamang R, Das S. HDAC5 modulates SATB1 transcriptional activity to promote lung adenocarcinoma. Br J Cancer 2023; 129:586-600. [PMID: 37400677 PMCID: PMC10421875 DOI: 10.1038/s41416-023-02341-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/29/2023] [Accepted: 06/20/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Dysregulation of histone deacetylases has been linked to diverse cancers. HDAC5 is a histone deacetylase belonging to Class IIa family of histone deacetylases. Limited substrate repertoire restricts the understanding of molecular mechanisms underlying its role in tumorigenesis. METHODS We employed a biochemical screen to identify SATB1 as HDAC5-interacting protein. Coimmunoprecipitation and deacetylation assay were performed to validate SATB1 as a HDAC5 substrate. Proliferation, migration assay and xenograft studies were performed to determine the effect of HDAC5-SATB1 interaction on tumorigenesis. RESULTS Here we report that HDAC5 binds to and deacetylates SATB1 at the conserved lysine 411 residue. Furthermore, dynamic regulation of acetylation at this site is determined by TIP60 acetyltransferase. We also established that HDAC5-mediated deacetylation is critical for SATB1-dependent downregulation of key tumor suppressor genes. Deacetylated SATB1 also represses SDHA-induced epigenetic remodeling and anti-proliferative transcriptional program. Thus, SATB1 spurs malignant phenotype in a HDAC5-dependent manner. CONCLUSIONS Our study highlights the pivotal role of HDAC5 in tumorigenesis. Our findings provide key insights into molecular mechanisms underlying SATB1 promoted tumor growth and metastasis.
Collapse
Affiliation(s)
- Shalakha Sharma
- Molecular Oncology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Witty Tyagi
- Molecular Oncology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Rohini Tamang
- Molecular Oncology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Sanjeev Das
- Molecular Oncology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| |
Collapse
|
7
|
HDAC6 promotes aggressive development of liver cancer by improving egfr mRNA stability. Neoplasia 2022; 35:100845. [PMID: 36334332 PMCID: PMC9640351 DOI: 10.1016/j.neo.2022.100845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022] Open
|
8
|
Dang F, Wei W. Targeting the acetylation signaling pathway in cancer therapy. Semin Cancer Biol 2022; 85:209-218. [PMID: 33705871 PMCID: PMC8423867 DOI: 10.1016/j.semcancer.2021.03.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/22/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022]
Abstract
Acetylation represents one of the major post-translational protein modifications, which introduces an acetyl functional group into amino acids such as the lysine residue to yield an acetate ester bond, neutralizing its positive charge. Regulation of protein functions by acetylation occurs in multiple ways, such as affecting protein stability, activity, localization, and interaction with other proteins or DNA. It has been well documented that the recruitment of histone acetyltransferases (HATs) and histone deacetylases (HDACs) to the transcriptional machinery can modulate histone acetylation status, which is directly involved in the dynamic regulation of genes controlling cell proliferation and division. Dysregulation of gene expression is involved in tumorigenesis and aberrant activation of histone deacetylases has been reported in several types of cancer. Moreover, there is growing body of evidence showing that acetylation is widely involved in non-histone proteins to impact their roles in various cellular processes including tumorigenesis. As such, small molecular compounds inhibiting HAT or HDAC enzymatic activities have been developed and investigated for therapeutic purpose. Here we review the recent progress in our understanding of protein acetylation and discuss the therapeutic potential of targeting the acetylation signaling pathway in cancer.
Collapse
Affiliation(s)
- Fabin Dang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
9
|
Cao X, Wang X, Zhang W, Xia G, Zhang L, Wen Z, He J, Wang Z, Huang J, Wu S. WNT10A induces apoptosis of senescent synovial resident stem cells through Wnt/calcium pathway-mediated HDAC5 phosphorylation in OA joints. Bone 2021; 150:116006. [PMID: 34000432 DOI: 10.1016/j.bone.2021.116006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/01/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
Recently, the accumulation of senescent cells (SnCs) within joints was found to promote osteoarthritis (OA) progression. Our previous study found that Wnt proteins, especially Wnt10a, have marked effects on cellular senescence and joint health. However, the effect of WNT10A on SnCs in OA joints remains unknown. In this study, we confirmed that the synovium was the first and most marked site of SnC accumulation in the OA joint, and synovial resident mesenchymal stem cells (SMSCs) seemed to be the main source of these SnCs. In synovium samples from OA patients, WNT10A level inversely correlated with the extent of SnCs accumulation. Therefore, we further explored the possible regulatory role and mechanism of WNT10A in intraarticular senescent SMSCs. In brief, we confirmed that WNT10A could specifically clear these senescent OA-SMSCs in vitro experiments and naturally occurring OA models via proapoptotic effects. Mechanistically, WNT10A activated noncanonical Wnt/calcium signaling in senescent OA-SMSCs, which in turn induced histone deacetylase 5 (HDAC5) phosphorylation and nuclear export via its downstream Ca2+/calmodulin-dependent protein kinase II (CaMKII) to regulate cell fate. The regulation of this pathway significantly improved the regenerative microenvironment of OA, exhibiting its potential as a novel clinical disease-modifying OA drugs (DMOADs) target.
Collapse
Affiliation(s)
- Xu Cao
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, China
| | - Xinxing Wang
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, China
| | - Wenxiu Zhang
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, China
| | - Guang Xia
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, China
| | - Lina Zhang
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, China
| | - Zi Wen
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, China
| | - Jinshen He
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, China
| | - Zili Wang
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, China
| | - Junjie Huang
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, China..
| | - Song Wu
- Department of Orthopaedics of the 3rd Xiangya Hospital, Central South University, China..
| |
Collapse
|
10
|
Toro TB, Swanier JS, Bezue JA, Broussard CG, Watt TJ. Lysine Deacetylase Substrate Selectivity: A Dynamic Ionic Interaction Specific to KDAC8. Biochemistry 2021; 60:2524-2536. [PMID: 34357750 DOI: 10.1021/acs.biochem.1c00384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lysine acetylation and deacetylation are critical for regulation of many cellular proteins. Despite the importance of this cycle, it is unclear how lysine deacetylase (KDAC) family members discriminate between acetylated proteins to react with a discrete set of substrates. Potential short-range interactions between KDAC8 and a known biologically relevant peptide substrate were identified using molecular dynamics (MD) simulations. Activity assays with a panel of peptides derived from this substrate supported a putative ionic interaction between arginine at the -1 substrate position and KDAC8 D101. Additional assays and MD simulations confirmed this novel interaction, which promotes deacetylation of substrates. Verification that a negatively charged residue at the 101 position is necessary for the ionic interaction and observed reactivity with the substrates was performed using KDAC8 derivatives. Notably, this interaction is specific to KDAC8, as KDAC1 and KDAC6 do not form this interaction and each KDAC has a different specificity profile with the peptide substrates, even though all KDACs could potentially form ionic interactions. When reacted with a panel of putative human KDAC substrates, KDAC8 preferentially deacetylated substrates containing an arginine at the -1 position. KDAC8 D101-R(-1) is a specific enzyme-substrate interaction that begins to explain how KDACs discriminate between potential substrates and how different KDAC family members can react with different subsets of acetylated proteins in cells. This multi-pronged approach will be extended to identify other critical interactions for KDAC8 substrate binding and determine critical interactions for other KDACs.
Collapse
Affiliation(s)
- Tasha B Toro
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| | - Jordan S Swanier
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| | - Jada A Bezue
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| | - Christian G Broussard
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| | - Terry J Watt
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| |
Collapse
|
11
|
Impairment of human terminal erythroid differentiation by histone deacetylase 5 deficiency. Blood 2021; 138:1615-1627. [PMID: 34036344 DOI: 10.1182/blood.2020007401] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 04/22/2021] [Indexed: 11/20/2022] Open
Abstract
Histone deacetylases (HDACs) are a group of enzymes catalyzing the removal of acetyl groups from histone and non-histone proteins. HDACs have been shown to play diverse functions in a wide range of biological processes. However, their roles in mammalian erythropoiesis remain to be fully defined. We show here that of the eleven classic HDAC family members, six of them (HDAC 1,2,3 and HDAC 5,6,7) are expressed in human erythroid cells with HDAC5 most significantly up regulated during terminal erythroid differentiation. Knockdown of HDAC5 by either shRNA or siRNA in human CD34+ cells followed by erythroid cell culture led to increased apoptosis, decreased chromatin condensation, and impaired enucleation of erythroblasts. Biochemical analyses revealed that HDAC5 deficiency resulted in activation of p53 in association with increased acetylation of p53. Furthermore, while acetylation of histone 4 (H4) is decreased during normal terminal erythroid differentiation, HDAC5 deficiency led to increased acetylation of H4 (K12) in late stage erythroblasts. This increased acetylation was accompanied by decreased chromatin condensation, implying a role for H4 (K12) deacetylation in chromatin condensation. ATAC-seq and RNA-seq analyses revealed that HDAC5 knockdown leads to increased chromatin accessibility genome wide and global changes in gene expression. Moreover, pharmacological inhibition of HDAC5 by the inhibitor LMK235 also led to increased H4 acetylation, impaired chromatin condensation and enucleation. Taken together, our findings have uncovered previously unrecognized roles and molecular mechanisms of action for HDAC5 in human erythropoiesis. These results may provide insights into understanding the anemia associated with HDAC inhibitor treatment.
Collapse
|
12
|
Falcicchio M, Ward JA, Macip S, Doveston RG. Regulation of p53 by the 14-3-3 protein interaction network: new opportunities for drug discovery in cancer. Cell Death Discov 2020; 6:126. [PMID: 33298896 PMCID: PMC7669891 DOI: 10.1038/s41420-020-00362-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/02/2020] [Accepted: 10/23/2020] [Indexed: 01/17/2023] Open
Abstract
Most cancers evolve to disable the p53 pathway, a key tumour suppressor mechanism that prevents transformation and malignant cell growth. However, only ~50% exhibit inactivating mutations of p53, while in the rest its activity is suppressed by changes in the proteins that modulate the pathway. Therefore, restoring p53 activity in cells in which it is still wild type is a highly attractive therapeutic strategy that could be effective in many different cancer types. To this end, drugs can be used to stabilise p53 levels by modulating its regulatory pathways. However, despite the emergence of promising strategies, drug development has stalled in clinical trials. The need for alternative approaches has shifted the spotlight to the 14-3-3 family of proteins, which strongly influence p53 stability and transcriptional activity through direct and indirect interactions. Here, we present the first detailed review of how 14-3-3 proteins regulate p53, with special emphasis on the mechanisms involved in their binding to different members of the pathway. This information will be important to design new compounds that can reactivate p53 in cancer cells by influencing protein-protein interactions. The intricate relationship between the 14-3-3 isoforms and the p53 pathway suggests that many potential drug targets for p53 reactivation could be identified and exploited to design novel antineoplastic therapies with a wide range of applications.
Collapse
Affiliation(s)
- Marta Falcicchio
- Leicester Institute for Structural and Chemical Biology, University of Leicester, University Road, Leicester, LE1 7RH, UK
- School of Chemistry, University of Leicester, University Road, Leicester, LE1 7RH, UK
| | - Jake A Ward
- Leicester Institute for Structural and Chemical Biology, University of Leicester, University Road, Leicester, LE1 7RH, UK
- Mechanisms of Cancer and Ageing Lab, Department of Molecular and Cell Biology, University of Leicester, University Road, Leicester, LE1 7RH, UK
| | - Salvador Macip
- Mechanisms of Cancer and Ageing Lab, Department of Molecular and Cell Biology, University of Leicester, University Road, Leicester, LE1 7RH, UK.
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain.
| | - Richard G Doveston
- Leicester Institute for Structural and Chemical Biology, University of Leicester, University Road, Leicester, LE1 7RH, UK.
- School of Chemistry, University of Leicester, University Road, Leicester, LE1 7RH, UK.
| |
Collapse
|
13
|
Feng YC, Liu XY, Teng L, Ji Q, Wu Y, Li JM, Gao W, Zhang YY, La T, Tabatabaee H, Yan XG, Jamaluddin MFB, Zhang D, Guo ST, Scott RJ, Liu T, Thorne RF, Zhang XD, Jin L. c-Myc inactivation of p53 through the pan-cancer lncRNA MILIP drives cancer pathogenesis. Nat Commun 2020; 11:4980. [PMID: 33020477 PMCID: PMC7536215 DOI: 10.1038/s41467-020-18735-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
The functions of the proto-oncoprotein c-Myc and the tumor suppressor p53 in controlling cell survival and proliferation are inextricably linked as “Yin and Yang” partners in normal cells to maintain tissue homeostasis: c-Myc induces the expression of ARF tumor suppressor (p14ARF in human and p19ARF in mouse) that binds to and inhibits mouse double minute 2 homolog (MDM2) leading to p53 activation, whereas p53 suppresses c-Myc through a combination of mechanisms involving transcriptional inactivation and microRNA-mediated repression. Nonetheless, the regulatory interactions between c-Myc and p53 are not retained by cancer cells as is evident from the often-imbalanced expression of c-Myc over wildtype p53. Although p53 repression in cancer cells is frequently associated with the loss of ARF, we disclose here an alternate mechanism whereby c-Myc inactivates p53 through the actions of the c-Myc-Inducible Long noncoding RNA Inactivating P53 (MILIP). MILIP functions to promote p53 polyubiquitination and turnover by reducing p53 SUMOylation through suppressing tripartite-motif family-like 2 (TRIML2). MILIP upregulation is observed amongst diverse cancer types and is shown to support cell survival, division and tumourigenicity. Thus our results uncover an inhibitory axis targeting p53 through a pan-cancer expressed RNA accomplice that links c-Myc to suppression of p53. c-Myc and p53 operate in a negative feedback manner to maintain cellular homeostasis. Here, the authors report a long noncoding RNA, MILIP as a downstream target of c-Myc and that MILIP represses p53 to support tumorigenicity.
Collapse
Affiliation(s)
- Yu Chen Feng
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Xiao Ying Liu
- Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Liu Teng
- Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Qiang Ji
- Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Yongyan Wu
- Department of Otolaryngology, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, the first affiliated hospital, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jin Ming Li
- Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Wei Gao
- Department of Otolaryngology, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, the first affiliated hospital, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yuan Yuan Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Ting La
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Hessam Tabatabaee
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Xu Guang Yan
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - M Fairuz B Jamaluddin
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Didi Zhang
- Department of Orthopaedics, John Hunter Hospital, Hunter New England Health, Newcastle, 2305, NSW, Australia
| | - Su Tang Guo
- Department of Molecular Biology, Shanxi Cancer Hospital and Institute, Taiyuan, 030013, Shanxi, China
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, 2750, NSW, Australia
| | - Rick F Thorne
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia.,Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Xu Dong Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, 2308, NSW, Australia. .,Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China.
| | - Lei Jin
- Translational Research Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450053, Henan, China. .,School of Medicine and Public Health, The University of Newcastle, Newcastle, 2308, NSW, Australia.
| |
Collapse
|
14
|
Li G, Tian Y, Zhu WG. The Roles of Histone Deacetylases and Their Inhibitors in Cancer Therapy. Front Cell Dev Biol 2020; 8:576946. [PMID: 33117804 PMCID: PMC7552186 DOI: 10.3389/fcell.2020.576946] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022] Open
Abstract
Genetic mutations and abnormal gene regulation are key mechanisms underlying tumorigenesis. Nucleosomes, which consist of DNA wrapped around histone cores, represent the basic units of chromatin. The fifth amino group (Nε) of histone lysine residues is a common site for post-translational modifications (PTMs), and of these, acetylation is the second most common. Histone acetylation is modulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs), and is involved in the regulation of gene expression. Over the past two decades, numerous studies characterizing HDACs and HDAC inhibitors (HDACi) have provided novel and exciting insights concerning their underlying biological mechanisms and potential anti-cancer treatments. In this review, we detail the diverse structures of HDACs and their underlying biological functions, including transcriptional regulation, metabolism, angiogenesis, DNA damage response, cell cycle, apoptosis, protein degradation, immunity and other several physiological processes. We also highlight potential avenues to use HDACi as novel, precision cancer treatments.
Collapse
Affiliation(s)
- Guo Li
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Yuan Tian
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Wei-Guo Zhu
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
- Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
15
|
Toro TB, Watt TJ. Critical review of non-histone human substrates of metal-dependent lysine deacetylases. FASEB J 2020; 34:13140-13155. [PMID: 32862458 DOI: 10.1096/fj.202001301rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/29/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022]
Abstract
Lysine acetylation is a posttranslational modification that occurs on thousands of human proteins, most of which are cytoplasmic. Acetylated proteins are involved in numerous cellular processes and human diseases. Therefore, how the acetylation/deacetylation cycle is regulated is an important question. Eleven metal-dependent lysine deacetylases (KDACs) have been identified in human cells. These enzymes, along with the sirtuins, are collectively responsible for reversing lysine acetylation. Despite several large-scale studies which have characterized the acetylome, relatively few of the specific acetylated residues have been matched to a proposed KDAC for deacetylation. To understand the function of lysine acetylation, and its association with diseases, specific KDAC-substrate pairs must be identified. Identifying specific substrates of a KDAC is complicated both by the complexity of assaying relevant activity and by the non-catalytic interactions of KDACs with cellular proteins. Here, we discuss in vitro and cell-based experimental strategies used to identify KDAC-substrate pairs and evaluate each for the purpose of directly identifying non-histone substrates of metal-dependent KDACs. We propose criteria for a combination of reproducible experimental approaches that are necessary to establish a direct enzymatic relationship. This critical analysis of the literature identifies 108 proposed non-histone substrate-KDAC pairs for which direct experimental evidence has been reported. Of these, five pairs can be considered well-established, while another thirteen pairs have both cell-based and in vitro evidence but lack independent replication and/or sufficient cell-based evidence. We present a path forward for evaluating the remaining substrate leads and reliably identifying novel KDAC substrates.
Collapse
Affiliation(s)
- Tasha B Toro
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| | - Terry J Watt
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| |
Collapse
|
16
|
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: 7.8] [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
|
17
|
Xu CL, Sang B, Liu GZ, Li JM, Zhang XD, Liu LX, Thorne RF, Wu M. SENEBLOC, a long non-coding RNA suppresses senescence via p53-dependent and independent mechanisms. Nucleic Acids Res 2020; 48:3089-3102. [PMID: 32030426 PMCID: PMC7102969 DOI: 10.1093/nar/gkaa063] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/06/2020] [Accepted: 01/21/2020] [Indexed: 01/07/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as important biological tuners. Here, we reveal the role of an uncharacterized lncRNA we call SENEBLOC that is expressed by both normal and transformed cells under homeostatic conditions. SENEBLOC was shown to block the induction of cellular senescence through dual mechanisms that converge to repress the expression of p21. SENEBLOC facilitates the association of p53 with MDM2 by acting as a scaffold to promote p53 turnover and decrease p21 transactivation. Alternatively, SENEBLOC was shown to affect epigenetic silencing of the p21 gene promoter through regulation of HDAC5. Thus SENEBLOC drives both p53-dependent and p53-independent mechanisms that contribute to p21 repression. Moreover, SENEBLOC was shown to be involved in both oncogenic and replicative senescence, and from the perspective of senolytic agents we show that the antagonistic actions of rapamycin on senescence are dependent on SENEBLOC expression.
Collapse
Affiliation(s)
- Cheng Lin Xu
- CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Centre for Excellence in Molecular Cell Science, School of Life Sciences and First Affiliated Hospital of University of Science and Technology of China, Hefei 230027, China
| | - Ben Sang
- CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Centre for Excellence in Molecular Cell Science, School of Life Sciences and First Affiliated Hospital of University of Science and Technology of China, Hefei 230027, China
| | - Guang Zhi Liu
- Key Laboratory of Stem Cell Differentiation & Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China
| | - Jin Ming Li
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China
| | - Xu Dong Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW 2308, Australia
| | - Lian Xin Liu
- CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Centre for Excellence in Molecular Cell Science, School of Life Sciences and First Affiliated Hospital of University of Science and Technology of China, Hefei 230027, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China
- School of Environmental & Life Sciences, University of Newcastle, NSW 2258, Australia
| | - Mian Wu
- CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Centre for Excellence in Molecular Cell Science, School of Life Sciences and First Affiliated Hospital of University of Science and Technology of China, Hefei 230027, China
- Key Laboratory of Stem Cell Differentiation & Modification, School of Clinical Medicine, Henan University, Zhengzhou 450003, China
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China
| |
Collapse
|
18
|
Phase I studies of vorinostat with ixazomib or pazopanib imply a role of antiangiogenesis-based therapy for TP53 mutant malignancies. Sci Rep 2020; 10:3080. [PMID: 32080210 PMCID: PMC7033174 DOI: 10.1038/s41598-020-58366-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022] Open
Abstract
We performed two phase I trials of the histone deacetylase inhibitor vorinostat combined with either the vascular endothelial growth factor inhibitor pazopanib (NCT01339871) or the proteasome inhibitor ixazomib (NCT02042989) in patients with metastatic TP53 mutant solid tumors. Both trials followed a 3 + 3 dose-escalation design allowing for a dose expansion cohort of up to 14 additional patients with a specific tumor type. Patients had to have a confirmed TP53 mutation to be enrolled in NCT02042989. Among patients enrolled in NCT01339871, TP53 mutation status was determined for those for whom tumor specimens were available. The results of NCT01339871 were reported previously. Common treatment-related adverse events in NCT02042989 included anemia, thrombocytopenia, fatigue, nausea, vomiting, and diarrhea. Compared with patients with metastatic TP53 hotspot mutant solid tumors who were treated with ixazomib and vorinostat (n = 59), those who were treated with pazopanib and vorinostat (n = 11) had a significantly higher rate of clinical benefit, defined as stable disease lasting ≥6 months or an objective response (3.4% vs. 45%; p < 0.001), a significantly longer median progression-free survival duration (1.7 months [95% confidence interval (CI), 1.1–2.3] vs. 3.5 months [95% CI, 1.7–5.2]; p = 0.002), and a longer median overall survival duration (7.3 months [95% CI, 4.8–9.8] vs. 12.7 months [95% CI, 7.1–18.3]; p = 0.24). Our two phase I trials provide preliminary evidence supporting the use of antiangiogenisis-based therapy in patients with metastatic TP53 mutant solid tumors, especially in those with metastatic sarcoma or metastatic colorectal cancer.
Collapse
|
19
|
Bowen ME, McClendon J, Long HK, Sorayya A, Van Nostrand JL, Wysocka J, Attardi LD. The Spatiotemporal Pattern and Intensity of p53 Activation Dictates Phenotypic Diversity in p53-Driven Developmental Syndromes. Dev Cell 2019; 50:212-228.e6. [PMID: 31178404 PMCID: PMC6650355 DOI: 10.1016/j.devcel.2019.05.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 03/14/2019] [Accepted: 05/03/2019] [Indexed: 12/11/2022]
Abstract
Inappropriate activation of the p53 transcription factor contributes to numerous developmental syndromes characterized by distinct constellations of phenotypes. How p53 drives exquisitely specific sets of symptoms in diverse syndromes, however, remains enigmatic. Here, we deconvolute the basis of p53-driven developmental syndromes by leveraging an array of mouse strains to modulate the spatial expression pattern, temporal profile, and magnitude of p53 activation during embryogenesis. We demonstrate that inappropriate p53 activation in the neural crest, facial ectoderm, anterior heart field, and endothelium induces distinct spectra of phenotypes. Moreover, altering the timing and degree of p53 hyperactivation substantially affects the phenotypic outcomes. Phenotypes are associated with p53-driven cell-cycle arrest or apoptosis, depending on the cell type, with gene expression programs, rather than extent of mitochondrial priming, largely governing the specific response. Together, our findings provide a critical framework for decoding the role of p53 as a mediator of diverse developmental syndromes.
Collapse
Affiliation(s)
- Margot E Bowen
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jacob McClendon
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hannah K Long
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aryo Sorayya
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jeanine L Van Nostrand
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institue, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
20
|
Avrahami EM, Levi S, Zajfman E, Regev C, Ben-David O, Arbely E. Reconstitution of Mammalian Enzymatic Deacylation Reactions in Live Bacteria Using Native Acylated Substrates. ACS Synth Biol 2018; 7:2348-2354. [PMID: 30207693 PMCID: PMC6198279 DOI: 10.1021/acssynbio.8b00314] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Lysine deacetylases
(KDACs) are enzymes that catalyze the hydrolysis
of acyl groups from acyl-lysine residues. The recent identification
of thousands of putative acylation sites, including specific acetylation
sites, created an urgent need for biochemical methodologies aimed
at better characterizing KDAC-substrate specificity and evaluating
KDACs activity. To address this need, we utilized genetic code expansion
technology to coexpress site-specifically acylated substrates with
mammalian KDACs, and study substrate recognition and deacylase activity
in live Escherichia coli. In this system the bacterial
cell serves as a “biological test tube” in which the
incubation of a single mammalian KDAC and a potential peptide or full-length
acylated substrate transpires. We report novel deacetylation activities
of Zn2+-dependent deacetylases and sirtuins in bacteria.
We also measure the deacylation of propionyl-, butyryl-, and crotonyl-lysine,
as well as novel deacetylation of Lys310-acetylated RelA by SIRT3,
SIRT5, SIRT6, and HDAC8. This study highlights the importance of native
interactions to KDAC-substrate recognition and deacylase activity.
Collapse
Affiliation(s)
- Emanuel M. Avrahami
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Shahar Levi
- Department of Chemistry and The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Eyal Zajfman
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Clil Regev
- Department of Chemistry and The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Oshrit Ben-David
- Department of Chemistry and The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Eyal Arbely
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Department of Chemistry and The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
21
|
Heterochromatin protects retinal pigment epithelium cells from oxidative damage by silencing p53 target genes. Proc Natl Acad Sci U S A 2018; 115:E3987-E3995. [PMID: 29622681 PMCID: PMC5924883 DOI: 10.1073/pnas.1715237115] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Oxidative stress (OS)-induced retinal pigment epithelium (RPE) cell apoptosis is critically implicated in the pathogenesis of age-related macular degeneration (AMD), a leading cause of blindness in the elderly. Heterochromatin, a compact and transcriptional inert chromatin structure, has been recently shown to be dynamically regulated in response to stress stimuli. The functional mechanism of heterochromatin on OS exposure is unclear, however. Here we show that OS increases heterochromatin formation both in vivo and in vitro, which is essential for protecting RPE cells from oxidative damage. Mechanistically, OS-induced heterochromatin selectively accumulates at p53-regulated proapoptotic target promoters and inhibits their transcription. Furthermore, OS-induced desumoylation of p53 promotes p53-heterochromatin interaction and regulates p53 promoter selection, resulting in the locus-specific recruitment of heterochromatin and transcription repression. Together, our findings demonstrate a protective function of OS-induced heterochromatin formation in which p53 desumoylation-guided promoter selection and subsequent heterochromatin recruitment play a critical role. We propose that targeting heterochromatin provides a plausible therapeutic strategy for the treatment of AMD.
Collapse
|
22
|
Szymula A, Palermo RD, Bayoumy A, Groves IJ, Ba abdullah M, Holder B, White RE. Epstein-Barr virus nuclear antigen EBNA-LP is essential for transforming naïve B cells, and facilitates recruitment of transcription factors to the viral genome. PLoS Pathog 2018; 14:e1006890. [PMID: 29462212 PMCID: PMC5834210 DOI: 10.1371/journal.ppat.1006890] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 03/02/2018] [Accepted: 01/21/2018] [Indexed: 12/11/2022] Open
Abstract
The Epstein-Barr virus (EBV) nuclear antigen leader protein (EBNA-LP) is the first viral latency-associated protein produced after EBV infection of resting B cells. Its role in B cell transformation is poorly defined, but it has been reported to enhance gene activation by the EBV protein EBNA2 in vitro. We generated EBNA-LP knockout (LPKO) EBVs containing a STOP codon within each repeat unit of internal repeat 1 (IR1). EBNA-LP-mutant EBVs established lymphoblastoid cell lines (LCLs) from adult B cells at reduced efficiency, but not from umbilical cord B cells, which died approximately two weeks after infection. Adult B cells only established EBNA-LP-null LCLs with a memory (CD27+) phenotype. Quantitative PCR analysis of virus gene expression after infection identified both an altered ratio of the EBNA genes, and a dramatic reduction in transcript levels of both EBNA2-regulated virus genes (LMP1 and LMP2) and the EBNA2-independent EBER genes in the first 2 weeks. By 30 days post infection, LPKO transcription was the same as wild-type EBV. In contrast, EBNA2-regulated cellular genes were induced efficiently by LPKO viruses. Chromatin immunoprecipitation revealed that EBNA2 and the host transcription factors EBF1 and RBPJ were delayed in their recruitment to all viral latency promoters tested, whereas these same factors were recruited efficiently to several host genes, which exhibited increased EBNA2 recruitment. We conclude that EBNA-LP does not simply co-operate with EBNA2 in activating gene transcription, but rather facilitates the recruitment of several transcription factors to the viral genome, to enable transcription of virus latency genes. Additionally, our findings suggest that EBNA-LP is essential for the survival of EBV-infected naïve B cells. Epstein-Barr virus (EBV) infects almost everyone. Once infected, people harbor the virus for life, shedding it in saliva. Infection of children is asymptomatic, but a first infection during adolescence or adulthood can cause glandular fever (infectious mononucleosis). EBV is also implicated in several different cancers. EBV infection of B cells (antibody-producing immune cells) can drive them to replicate almost indefinitely (‘transformation’), generating cell lines. We have investigated the role of an EBV protein (EBNA-LP) which is thought to support gene activation by the essential virus protein EBNA2. We have made an EBV in which the EBNA-LP gene has been disrupted. This virus (LPKO) shows several properties. 1. It is reduced in its ability to transform B cells; 2. ‘Naïve’ B cells (those whose antibodies have not adapted to fight infections) die two weeks after LPKO infection; 3. Some virus genes fail to turn on immediately after LPKO infection. 4. Binding of EBNA2 and various cellular factors to these genes is delayed. 5. EBNA-LP does not affect EBNA2-targeted cellular genes in the same way. This shows that EBNA-LP is more important in naïve B cells, and that it helps to turn on virus genes, but not cell genes.
Collapse
MESH Headings
- Adult
- B-Lymphocytes/pathology
- B-Lymphocytes/virology
- Cell Transformation, Viral/genetics
- Cells, Cultured
- Epstein-Barr Virus Infections/complications
- Epstein-Barr Virus Infections/genetics
- Epstein-Barr Virus Infections/pathology
- Female
- Gene Expression Regulation, Viral
- Genome, Viral
- HEK293 Cells
- Herpesvirus 4, Human/genetics
- Humans
- Infant, Newborn
- Leukemia, B-Cell/genetics
- Leukemia, B-Cell/pathology
- Leukemia, B-Cell/virology
- Pregnancy
- Promoter Regions, Genetic
- Protein Binding/genetics
- Transcription Factors/metabolism
- Viral Proteins/physiology
Collapse
Affiliation(s)
- Agnieszka Szymula
- Section of Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Richard D. Palermo
- Section of Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Amr Bayoumy
- Section of Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Ian J. Groves
- Section of Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Mohammed Ba abdullah
- Section of Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Beth Holder
- Section of Pediatrics, Department of Medicine, Imperial College London, London, United Kingdom
| | - Robert E. White
- Section of Virology, Department of Medicine, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Shi X, Pi L, Zhou S, Li X, Min F, Wang S, Liu Z, Wu J. Activation of Sirtuin 1 Attenuates High Glucose-Induced Neuronal Apoptosis by Deacetylating p53. Front Endocrinol (Lausanne) 2018; 9:274. [PMID: 29892266 PMCID: PMC5985296 DOI: 10.3389/fendo.2018.00274] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 05/09/2018] [Indexed: 12/04/2022] Open
Abstract
Diabetes mellitus (DM) has been proven to be a key risk factor for cognitive impairment. Previous studies have implicated hippocampal neuronal apoptosis in diabetes-related cognitive impairment. However, the underlying mechanism remains unknown. Sirtuin 1 (SIRT1) is a protein deacetylase depended on nicotinamide adenine dinucleotide. Furthermore, it is indispensable in normal learning and memory. Whether SIRT1 is taken part in diabetes-induced neuronal apoptosis and thus involve in the development of diabetic cognitive impairment is still not clear. To address this issue, we examined the possible role of SIRT1 in hippocampal neuronal apoptosis in streptozotocin-induced diabetic mice. Furthermore, the possible mechanism was investigated in high glucose-induced SH-SY5Y cells. We found that downregulation of the activity and expression of SIRT1 was associated with increased hippocampal neuronal apoptosis in mice. In vitro, cell apoptosis induced by high glucose which was accompanied by a downregulation of SIRT1 and an increased acetylation of p53. On the contrary, activation of SIRT1 using its agonist resveratrol ameliorated cell apoptosis via deacetylating p53. Our data suggest that high concentration of glucose can induce neuronal apoptosis through downregulation of SIRT1 and increased acetylation of p53, which likely contribute to the development of cognitive impairment in diabetes.
Collapse
Affiliation(s)
- Xiajie Shi
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Linhua Pi
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Shanlei Zhou
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Li
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Fangyuan Min
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Shan Wang
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, VA, United States
| | - Jing Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Jing Wu,
| |
Collapse
|
24
|
Li N, Yuan Q, Cao XL, Zhang Y, Min ZL, Xu SQ, Yu ZJ, Cheng J, Zhang C, Hu XM. Opposite effects of HDAC5 and p300 on MRTF-A-related neuronal apoptosis during ischemia/reperfusion injury in rats. Cell Death Dis 2017; 8:e2624. [PMID: 28230854 PMCID: PMC5386465 DOI: 10.1038/cddis.2017.16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 12/23/2016] [Accepted: 12/28/2016] [Indexed: 01/22/2023]
Abstract
Our recent study has revealed that the myocardin-related transcription factor-A (MRTF-A) is involved in the apoptosis of cortical neurons induced by ischemia/reperfusion (I/R). Histone deacetylase 5 (HDAC5) and histone acetyltransferase p300 (P300) are two well-known regulators for transcription factors; however, their roles in MRTF-A-related effect on neuronal injuries during I/R are still unclear. In this study, in a model rat cerebral I/R injury via middle cerebral artery occlusion and reperfusion, we found that the expression and activity of HDAC5 was upregulated, whereas p300 and MRTF-A were downregulated both in expression and activity during I/R. Their expression changes and the interaction of the MRTF-A with HDAC5 or p300 were further verified by double immunofluorescence and co-immunoprecipitation. In cultured neuronal apoptosis model induced by H2O2, MRTF-A exhibited an anti-apoptotic effect by enhancing the transcription of Bcl-2 and Mcl-1 via CArG box binding. MRTF-A-induced anti-apoptotic effect was effectively inhibited by HDAC5, but was significantly enhanced by p300. The results suggest that both HDAC5 and p300 are involved in MRTF-A-mediated effect on neuronal apoptosis during ischemia/reperfusion injury, but with opposite effects.
Collapse
Affiliation(s)
- Na Li
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Drug Research Base of Cardiovascular and Cerebral Vascular, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Qiong Yuan
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Drug Research Base of Cardiovascular and Cerebral Vascular, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Xiao-Lu Cao
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Drug Research Base of Cardiovascular and Cerebral Vascular, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Ying Zhang
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Drug Research Base of Cardiovascular and Cerebral Vascular, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Zhen-Li Min
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Drug Research Base of Cardiovascular and Cerebral Vascular, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Shi-Qiang Xu
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Drug Research Base of Cardiovascular and Cerebral Vascular, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Zhi-Jun Yu
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Drug Research Base of Cardiovascular and Cerebral Vascular, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Jing Cheng
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Drug Research Base of Cardiovascular and Cerebral Vascular, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| | - Chunxiang Zhang
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Drug Research Base of Cardiovascular and Cerebral Vascular, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Department of Biomedical Engineering, School of Medicine and School of Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xia-Min Hu
- Department of Pharmacy, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China.,Drug Research Base of Cardiovascular and Cerebral Vascular, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province 430065, China
| |
Collapse
|
25
|
Lazo PA. Reverting p53 activation after recovery of cellular stress to resume with cell cycle progression. Cell Signal 2017; 33:49-58. [PMID: 28189587 DOI: 10.1016/j.cellsig.2017.02.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 01/23/2017] [Accepted: 02/06/2017] [Indexed: 11/17/2022]
Abstract
The activation of p53 in response to different types of cellular stress induces several protective reactions including cell cycle arrest, senescence or cell death. These protective effects are a consequence of the activation of p53 by specific phosphorylation performed by several kinases. The reversion of the cell cycle arrest, induced by p53, is a consequence of the phosphorylated and activated p53, which triggers its own downregulation and that of its positive regulators. The different down-regulatory processes have a sequential and temporal order of events. The mechanisms implicated in p53 down-regulation include phosphatases, deacetylases, and protein degradation by the proteasome or autophagy, which also affect different p53 protein targets and functions. The necessary first step is the dephosphorylation of p53 to make it available for interaction with mdm2 ubiquitin-ligase, which requires the activation of phosphatases targeting both p53 and p53-activating kinases. In addition, deacetylation of p53 is required to make lysine residues accessible to ubiquitin ligases. The combined action of these downregulatory mechanisms brings p53 protein back to its basal levels, and cell cycle progression can resume if cells have overcome the stress or damage situation. The specific targeting of these down-regulatory mechanisms can be exploited for therapeutic purposes in cancers harbouring wild-type p53.
Collapse
Affiliation(s)
- Pedro A Lazo
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain.
| |
Collapse
|
26
|
Lee E, Choi SY, Bin BH, Kim NH, Kim KH, Choi DH, Han J, Choi H, Lee AY, Lee TR, Cho EG. Interferon-inducible T-cell alpha chemoattractant (ITAC) induces the melanocytic migration and hypopigmentation through destabilizing p53 via histone deacetylase 5: a possible role of ITAC in pigment-related disorders. Br J Dermatol 2016; 176:127-137. [PMID: 27436825 DOI: 10.1111/bjd.14878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2016] [Indexed: 11/26/2022]
Abstract
BACKGROUND Cell migration plays a major role in the immune response and in tumorigenesis. Interferon-inducible T-cell alpha chemoattractant (ITAC) elicits a strong chemotactic response from immune cells. OBJECTIVES To examine the effect of ITAC on melanocyte migration and pigmentation and its involvement in related disorders, and to investigate potential key players in these processes. METHODS Human melanocytes or melanoma cells were treated with ITAC and a migration assay was carried out. Global gene expression analysis was performed to find genes regulated by ITAC treatment. The function of key players involved in ITAC-induced cellular processes was addressed using knockdown or overexpression experiments in combination with ITAC treatment. ITAC expression in the inflammation-associated hypopigmentary disorder, vitiligo, was examined. RESULTS Among CXCR3 ligands, only ITAC induced melanocyte migration. ITAC treatment upregulated the expression of histone deacetylase 5 (HDAC5) and downregulated that of p53, a known target of HDAC5. Through knockdown or overexpression of HDAC5 and p53, we confirmed that HDAC5 mediates ITAC-induced migration by decreasing levels of p53 via deacetylation. In addition, ITAC treatment could decrease pigmentation in a p53- and HDAC5-dependent manner. Finally, the increased migration of human melanoma cells by ITAC treatment and the increased ITAC expression in the epidermis of vitiligo skin were verified. CONCLUSIONS This study provides in vitro evidence for the migratory and hypopigmentation effects of ITAC on melanocytic cells, gives translational insights into the roles of ITAC in pathological conditions, and suggests that HDAC5 and its substrate p53 are potent targets for regulating ITAC-induced cellular processes.
Collapse
Affiliation(s)
- E Lee
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Republic of Korea
| | - S-Y Choi
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Republic of Korea
| | - B-H Bin
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Republic of Korea
| | - N-H Kim
- Department of Dermatology, Dongguk University Ilsan Hospital, Goyang-si, Gyeonggi-do, Republic of Korea
| | - K H Kim
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Republic of Korea
| | - D-H Choi
- Gyeonggi Bio Center, Gyeonggi Institute of Science & Technology Promotion, Suwon-si, Gyeonggi-do, Republic of Korea
| | - J Han
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Republic of Korea
| | - H Choi
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Republic of Korea
| | - A-Y Lee
- Department of Dermatology, Dongguk University Ilsan Hospital, Goyang-si, Gyeonggi-do, Republic of Korea
| | - T R Lee
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Republic of Korea
| | - E-G Cho
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
27
|
Structural Basis for p53 Lys120-Acetylation-Dependent DNA-Binding Mode. J Mol Biol 2016; 428:3013-25. [DOI: 10.1016/j.jmb.2016.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/09/2016] [Accepted: 06/14/2016] [Indexed: 01/20/2023]
|
28
|
Functional interaction of histone deacetylase 5 (HDAC5) and lysine-specific demethylase 1 (LSD1) promotes breast cancer progression. Oncogene 2016; 36:133-145. [PMID: 27212032 PMCID: PMC5121103 DOI: 10.1038/onc.2016.186] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 03/21/2016] [Accepted: 04/14/2016] [Indexed: 02/07/2023]
Abstract
We have previously demonstrated that crosstalk between lysine-specific demethylase 1 (LSD1) and histone deacetylases (HDACs) facilitates breast cancer proliferation. However, the underlying mechanisms are largely unknown. Here, we report that expression of HDAC5 and LSD1 proteins were positively correlated in human breast cancer cell lines and tissue specimens of primary breast tumors. Protein expression of HDAC5 and LSD1 was significantly increased in primary breast cancer specimens in comparison with matched-normal adjacent tissues. Using HDAC5 deletion mutants and co-immunoprecipitation studies, we showed that HDAC5 physically interacted with the LSD1 complex through its domain containing nuclear localization sequence and phosphorylation sites. Although the in vitro acetylation assays revealed that HDAC5 decreased LSD1 protein acetylation, small interfering RNA (siRNA)-mediated HDAC5 knockdown did not alter the acetylation level of LSD1 in MDA-MB-231 cells. Overexpression of HDAC5 stabilized LSD1 protein and decreased the nuclear level of H3K4me1/me2 in MDA-MB-231 cells, whereas loss of HDAC5 by siRNA diminished LSD1 protein stability and demethylation activity. We further demonstrated that HDAC5 promoted the protein stability of USP28, a bona fide deubiquitinase of LSD1. Overexpression of USP28 largely reversed HDAC5-KD-induced LSD1 protein degradation, suggesting a role of HDAC5 as a positive regulator of LSD1 through upregulation of USP28 protein. Depletion of HDAC5 by shRNA hindered cellular proliferation, induced G1 cell cycle arrest, and attenuated migration and colony formation of breast cancer cells. A rescue study showed that increased growth of MDA-MB-231 cells by HDAC5 overexpression was reversed by concurrent LSD1 depletion, indicating that tumor-promoting activity of HDAC5 is an LSD1 dependent function. Moreover, overexpression of HDAC5 accelerated cellular proliferation and promoted acridine mutagen ICR191-induced transformation of MCF10A cells. Taken together, these results suggest that HDAC5 is critical in regulating LSD1 protein stability through post-translational modification, and the HDAC5-LSD1 axis has an important role in promoting breast cancer development and progression.
Collapse
|
29
|
Abstract
Histone posttranslational modifications represent a versatile set of epigenetic marks involved not only in dynamic cellular processes, such as transcription and DNA repair, but also in the stable maintenance of repressive chromatin. In this article, we review many of the key and newly identified histone modifications known to be deregulated in cancer and how this impacts function. The latter part of the article addresses the challenges and current status of the epigenetic drug development process as it applies to cancer therapeutics.
Collapse
Affiliation(s)
- James E Audia
- Constellation Pharmaceuticals, Cambridge, Massachusetts 02142
| | - Robert M Campbell
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana 46285
| |
Collapse
|
30
|
Yun T, Yu K, Yang S, Cui Y, Wang Z, Ren H, Chen S, Li L, Liu X, Fang M, Jiang X. Acetylation of p53 Protein at Lysine 120 Up-regulates Apaf-1 Protein and Sensitizes the Mitochondrial Apoptotic Pathway. J Biol Chem 2016; 291:7386-95. [PMID: 26851285 DOI: 10.1074/jbc.m115.706341] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Indexed: 11/06/2022] Open
Abstract
The p53 tumor suppressor controls cell growth, metabolism, and death by regulating the transcription of various target genes. The target-specific transcriptional activity of p53 is highly regulated. Here we demonstrate that acetylation of p53 at Lys-120 up-regulates its transcriptional activity toward Apaf-1, a core component in the mitochondrial apoptotic pathway, and thus sensitizes caspase activation and apoptosis. We found that histone deacetylase (HDAC) inhibitors, including butyrate, augment Lys-120 acetylation of p53 and thus Apaf-1 expression by inhibiting HDAC1. In p53-null cells, transfection of wild-type but not K120R mutant p53 can restore the p53-dependent sensitivity to butyrate. Strikingly, transfection of acetylation-mimicking K120Q mutant p53 is sufficient to up-regulates Apaf-1 in a manner independent of butyrate treatment. Therefore, HDAC inhibitors can induce p53 acetylation at lysine 120, which in turn enhances mitochondrion-mediated apoptosis through transcriptional up-regulation of Apaf-1.
Collapse
Affiliation(s)
- Tao Yun
- From the Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Division of Cell Biology, School of Life Sciences, the Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Kaiwen Yu
- Institute of Analytical Chemistry and Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, and
| | - ShuangShuang Yang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Yiheyuan Avenue 5, Haidian District, Beijing 100875, China
| | - Yifan Cui
- From the Peking-Tsinghua Center for Life Sciences, Division of Cell Biology, School of Life Sciences
| | - Zixi Wang
- From the Peking-Tsinghua Center for Life Sciences, Division of Cell Biology, School of Life Sciences
| | - Huiyu Ren
- From the Peking-Tsinghua Center for Life Sciences, Division of Cell Biology, School of Life Sciences
| | - She Chen
- the National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China, and
| | - Lin Li
- the National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China, and
| | - Xiaoyun Liu
- Institute of Analytical Chemistry and Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, and
| | - Min Fang
- From the Peking-Tsinghua Center for Life Sciences, Division of Cell Biology, School of Life Sciences,
| | - Xuejun Jiang
- the Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
31
|
Ray A, Rana S, Banerjee D, Mitra A, Datta R, Naskar S, Sarkar S. Improved bioavailability of targeted Curcumin delivery efficiently regressed cardiac hypertrophy by modulating apoptotic load within cardiac microenvironment. Toxicol Appl Pharmacol 2015; 290:54-65. [PMID: 26612707 DOI: 10.1016/j.taap.2015.11.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 01/30/2023]
Abstract
Cardiomyocyte apoptosis acts as a prime modulator of cardiac hypertrophy leading to heart failure, a major cause of human mortality worldwide. Recent therapeutic interventions have focussed on translational applications of diverse pharmaceutical regimes among which, Curcumin (from Curcuma longa) is known to have an anti-hypertrophic potential but with limited pharmacological efficacies due to low aqueous solubility and poor bioavailability. In this study, Curcumin encapsulated by carboxymethyl chitosan (CMC) nanoparticle conjugated to a myocyte specific homing peptide was successfully delivered in bioactive form to pathological myocardium for effective regression of cardiac hypertrophy in a rat (Rattus norvegicus) model. Targeted nanotization showed higher cardiac bioavailability of Curcumin at a low dose of 5 mg/kg body weight compared to free Curcumin at 35 mg/kg body weight. Moreover, Curcumin/CMC-peptide treatment during hypertrophy significantly improved cardiac function by downregulating expression of hypertrophy marker genes (ANF, β-MHC), apoptotic mediators (Bax, Cytochrome-c) and activity of apoptotic markers (Caspase 3 and PARP); whereas free Curcumin in much higher dose showed minimal improvement during compromised cardiac function. Targeted Curcumin treatment significantly lowered p53 expression and activation in diseased myocardium via inhibited interaction of p53 with p300-HAT. Thus attenuated acetylation of p53 facilitated p53 ubiquitination and reduced the apoptotic load in hypertrophied cardiomyocytes; thereby limiting cardiomyocytes' need to enter the regeneration cycle during hypertrophy. This study elucidates for the first time an efficient targeted delivery regimen for Curcumin and also attributes towards probable mechanistic insight into its therapeutic potential as a cardio-protective agent for regression of cardiac hypertrophy.
Collapse
Affiliation(s)
- Aramita Ray
- Genetics and Molecular Cardiology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700 019, West Bengal, India.
| | - Santanu Rana
- Genetics and Molecular Cardiology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700 019, West Bengal, India.
| | - Durba Banerjee
- Genetics and Molecular Cardiology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700 019, West Bengal, India.
| | - Arkadeep Mitra
- Genetics and Molecular Cardiology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700 019, West Bengal, India.
| | - Ritwik Datta
- Genetics and Molecular Cardiology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700 019, West Bengal, India.
| | - Shaon Naskar
- Genetics and Molecular Cardiology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700 019, West Bengal, India.
| | - Sagartirtha Sarkar
- Genetics and Molecular Cardiology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700 019, West Bengal, India.
| |
Collapse
|
32
|
Abdullah A, Sane S, Freeling JL, Wang H, Zhang D, Rezvani K. Nucleocytoplasmic Translocation of UBXN2A Is Required for Apoptosis during DNA Damage Stresses in Colon Cancer Cells. J Cancer 2015; 6:1066-78. [PMID: 26516353 PMCID: PMC4615341 DOI: 10.7150/jca.12134] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 08/02/2015] [Indexed: 12/12/2022] Open
Abstract
The subcellular localization, expression level, and activity of anti-cancer proteins alter in response to intrinsic and extrinsic cellular stresses to reverse tumor progression. The purpose of this study is to determine whether UBXN2A, an activator of the p53 tumor suppressor protein, has different subcellular compartmentalization in response to the stress of DNA damage. We measured trafficking of the UBXN2A protein in response to two different DNA damage stresses, UVB irradiation and the genotoxic agent Etoposide, in colon cancer cell lines. Using a cytosol-nuclear fractionation technique followed by western blot and immunofluorescence staining, we monitored and quantitated UBXN2A and p53 proteins as well as p53's downstream apoptotic pathway. We showed that the anti-cancer protein UBXN2A acts in the early phase of cell response to two different DNA damage stresses, being induced to translocate into the cytoplasm in a dose- and time-dependent manner. UVB-induced cytoplasmic UBXN2A binds to mortalin-2 (mot-2), a known oncoprotein in colon tumors. UVB-dependent upregulation of UBXN2A in the cytoplasm decreases p53 binding to mot-2 and activates apoptotic events in colon cancer cells. In contrast, the shRNA-mediated depletion of UBXN2A leads to significant reduction in apoptosis in colon cancer cells exposed to UVB and Etoposide. Leptomycin B (LMB), which was able to block UBXN2A nuclear export following Etoposide treatment, sustained p53-mot-2 interaction and had partially antagonistic effects with Etoposide on cell apoptosis. The present study shows that nucleocytoplasmic translocation of UBXN2A in response to stresses is necessary for its anti-cancer function in the cytoplasm. In addition, LMB-dependent suppression of UBXN2A's translocation to the cytoplasm upon stress allows the presence of an active mot-2 oncoprotein in the cytoplasm, resulting in p53 sequestration as well as activation of other mot-2-dependent growth promoting pathways.
Collapse
Affiliation(s)
- Ammara Abdullah
- 1. Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD 57069, USA
| | - Sanam Sane
- 1. Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD 57069, USA
| | - Jessica L Freeling
- 1. Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD 57069, USA
| | - Hongmin Wang
- 1. Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD 57069, USA
| | - Dong Zhang
- 2. Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Northern Blvd., P.O. Box 8000, Old Westbury, NY 11568-8000, USA
| | - Khosrow Rezvani
- 1. Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD 57069, USA
| |
Collapse
|
33
|
Kikuchi S, Suzuki R, Ohguchi H, Yoshida Y, Lu D, Cottini F, Jakubikova J, Bianchi G, Harada T, Gorgun G, Tai YT, Richardson PG, Hideshima T, Anderson KC. Class IIa HDAC inhibition enhances ER stress-mediated cell death in multiple myeloma. Leukemia 2015; 29:1918-27. [PMID: 25801913 DOI: 10.1038/leu.2015.83] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/05/2015] [Accepted: 03/12/2015] [Indexed: 01/14/2023]
Abstract
Histone deacetylase (HDAC) inhibitors have been extensively investigated as therapeutic agents in cancer. However, the biological role of class IIa HDACs (HDAC4, 5, 7 and 9) in cancer cells, including multiple myeloma (MM), remains unclear. Recent studies show HDAC4 interacts with activating transcription factor 4 (ATF4) and inhibits activation of endoplasmic reticulum (ER) stress-associated proapoptotic transcription factor C/EBP homologous protein (CHOP). In this study, we hypothesized that HDAC4 knockdown and/or inhibition could enhance apoptosis in MM cells under ER stress condition by upregulating ATF4, followed by CHOP. HDAC4 knockdown showed modest cell growth inhibition; however, it markedly enhanced cytotoxicity induced by either tunicamycin or carfilzomib (CFZ), associated with upregulating ATF4 and CHOP. For pharmacological inhibition of HDAC4, we employed a novel and selective class IIa HDAC inhibitor TMP269, alone and in combination with CFZ. As with HDAC4 knockdown, TMP269 significantly enhanced cytotoxicity induced by CFZ in MM cell lines, upregulating ATF4 and CHOP and inducing apoptosis. Conversely, enhanced cytotoxicity was abrogated by ATF4 knockdown, confirming that ATF4 has a pivotal role mediating cytotoxicity in this setting. These results provide the rationale for novel treatment strategies combining class IIa HDAC inhibitors with ER stressors, including proteasome inhibitors, to improve patient outcome in MM.
Collapse
Affiliation(s)
- S Kikuchi
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - R Suzuki
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - H Ohguchi
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Y Yoshida
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - D Lu
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - F Cottini
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - J Jakubikova
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - G Bianchi
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - T Harada
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - G Gorgun
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Y-T Tai
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - P G Richardson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - T Hideshima
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - K C Anderson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
34
|
Fu S, Hou MM, Naing A, Janku F, Hess K, Zinner R, Subbiah V, Hong D, Wheler J, Piha-Paul S, Tsimberidou A, Karp D, Araujo D, Kee B, Hwu P, Wolff R, Kurzrock R, Meric-Bernstam F. Phase I study of pazopanib and vorinostat: a therapeutic approach for inhibiting mutant p53-mediated angiogenesis and facilitating mutant p53 degradation. Ann Oncol 2015; 26:1012-1018. [PMID: 25669829 DOI: 10.1093/annonc/mdv066] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/29/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND We carried out a phase I trial of the vascular endothelial growth factor inhibitor pazopanib and the histone deacetylase inhibitor vorinostat to determine the safety and efficacy. Because these agents are known to target factors activated by TP53 mutation and facilitate mutant p53 degradation, a subgroup analysis may be interesting in patients with TP53 mutant malignancies. PATIENTS AND METHODS Patients with advanced solid tumors (n = 78) were enrolled following a 3 + 3 design, with dose expansion for those with responsive tumors. Hotspot TP53 mutations were tested when tumor specimens were available. RESULTS Adverse events of ≥grade 3 included thrombocytopenia, neutropenia, fatigue, hypertension, diarrhea and vomiting. Overall, the treatment produced stable disease for at least 6 months or partial response (SD ≥6 months/PR) in 19% of the patients, median progression-free survival (PFS) of 2.2 months, and median overall survival (OS) of 8.9 months. In patients with detected hotspot TP53 mutant advanced solid tumors (n = 11), the treatment led to a 45% rate of SD ≥6 months/PR (1 PR and 3 SD ≥6 months), median PFS of 3.5 months, and median OS of 12.7 months, compared favorably with the results for patients with undetected hotspot TP53 mutations (n = 25): 16% (1 PR and 3 SD ≥6 months, P = 0.096), 2.0 months (P = 0.042), and 7.4 months (P = 0.1), respectively. CONCLUSION The recommended phase II dosage was oral pazopanib at 600 mg daily plus oral vorinostat at 300 mg daily. The preliminary evidence supports further evaluation of the combination in cancer patients with mutated TP53, especially in those with metastatic sarcoma or metastatic colorectal cancer. CLINICAL TRIAL REGISTRATION www.clinicaltrials.gov, NCT01339871.
Collapse
Affiliation(s)
- S Fu
- Departments of Investigational Cancer Therapeutics.
| | - M M Hou
- Departments of Investigational Cancer Therapeutics; Division of Hematology-Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - A Naing
- Departments of Investigational Cancer Therapeutics
| | - F Janku
- Departments of Investigational Cancer Therapeutics
| | | | - R Zinner
- Departments of Investigational Cancer Therapeutics
| | - V Subbiah
- Departments of Investigational Cancer Therapeutics
| | - D Hong
- Departments of Investigational Cancer Therapeutics
| | - J Wheler
- Departments of Investigational Cancer Therapeutics
| | - S Piha-Paul
- Departments of Investigational Cancer Therapeutics
| | | | - D Karp
- Departments of Investigational Cancer Therapeutics
| | | | - B Kee
- GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | | | - R Wolff
- GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - R Kurzrock
- University of California San Diego, Moores Cancer Center, La Jolla, USA
| | | |
Collapse
|
35
|
Wei JY, Lu QN, Li WM, He W. Intracellular translocation of histone deacetylase 5 regulates neuronal cell apoptosis. Brain Res 2015; 1604:15-24. [PMID: 25661252 DOI: 10.1016/j.brainres.2015.01.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 01/08/2015] [Accepted: 01/23/2015] [Indexed: 01/07/2023]
Abstract
Histone deacetylase 5 (HDAC5) undergoes signal-dependent shuttling between the nucleus and cytoplasm, which is regulated in part by calcium/calmodulin-dependent kinase (CaMK)-mediated phosphorylation. Here, we report that HDAC5 regulates the survival of cortical neurons in pathological conditions. HDAC5 was evenly localized to the nucleus and cytoplasm in cultured cortical neurons. However, in response to 50μM NMDA conditions that induced neuronal cell apoptosis, nuclear-distributed HDAC5 was rapidly phosphorylated and translocated into cytoplasm of the cultured cortical neurons. Treatment with a CaMKII inhibitor KN93 suppressed HDAC5 phosphorylation and nuclear translocation induced by NMDA, whereas constitutively active CaMKIIα (T286D) stimulated HDAC5 nuclear export. Importantly, we showed that ectopic expression of nuclear-localized HDAC5 in cortical neurons suppressed NMDA-induced apoptosis. Finally, inactivation of HDAC5 by treatment with the class II-specific HDAC inhibitor trichostatin A (TSA) promoted NMDA-induced neuronal cell apoptosis. Altogether, these data identify HDAC5 and its intracellular translocation as key effectors of multiple pathways that regulate neuronal cell apoptosis.
Collapse
Affiliation(s)
- Jia-Yi Wei
- Department of Developmental Biology, Key Lab of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 92 Beier Road, Heping District, Shenyang 110001, China
| | - Qiu-Nan Lu
- Department of Developmental Biology, Key Lab of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 92 Beier Road, Heping District, Shenyang 110001, China
| | - Wan-Ming Li
- Department of Developmental Biology, Key Lab of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 92 Beier Road, Heping District, Shenyang 110001, China
| | - Wei He
- Department of Developmental Biology, Key Lab of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 92 Beier Road, Heping District, Shenyang 110001, China.
| |
Collapse
|
36
|
Tang BL. Class II HDACs and neuronal regeneration. J Cell Biochem 2015; 115:1225-33. [PMID: 24604703 DOI: 10.1002/jcb.24802] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/16/2014] [Indexed: 02/03/2023]
Abstract
The vastly more superior regenerative capacity of the axons of peripheral nerves over central nervous system (CNS) neurons has been partly attributed to the former's intrinsic capacity to initiate and sustain the functionality of a new growth cone. Growth cone generation involves a myriad of processes that centers around the organization of microtubule bundles. Histone deacetylases (HDACs) modulate a wide range of key neuronal processes such as neural progenitor differentiation, learning and memory, neuronal death, and degeneration. HDAC inhibitors have been shown to be beneficial in attenuating neuronal death and promoting neurite outgrowth and axonal regeneration. Recent advances have provided insights on how manipulating HDAC activities, particularly the type II HDACs 5 and 6, which deacetylate tubulin, may benefit axonal regeneration. These advances are discussed herein.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Medical Drive, Singapore, 117597, Singapore
| |
Collapse
|
37
|
Kumari R, Kohli S, Das S. p53 regulation upon genotoxic stress: intricacies and complexities. Mol Cell Oncol 2014; 1:e969653. [PMID: 27308356 DOI: 10.4161/23723548.2014.969653] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/02/2014] [Accepted: 09/02/2014] [Indexed: 12/11/2022]
Abstract
p53, the revered savior of genomic integrity, receives signals from diverse stress sensors and strategizes to maintain cellular homeostasis. However, the predominance of p53 overshadows the fact that this herculean task is no one-man show; rather, there is a huge army of regulators that reign over p53 at various levels to avoid an unnecessary surge in its levels and sculpt it dynamically to favor one cellular outcome over another. This governance starts right at the time of p53 translation, which is gated by proteins that bind to p53 mRNA and keep a stringent check on p53 protein levels. The same effect is also achieved by ubiquitylases and deubiquitylases that fine-tune p53 turnover and miRNAs that modulate p53 levels, adding precision to this entire scheme. In addition, extensive covalent modifications and differential protein interactions allow p53 to trigger a tailor-made response for a given circumstance. To magnify the marvel, these various tiers of regulation operate simultaneously and in various combinations. In this review, we have tried to provide a glimpse into this bewildering labyrinth. We believe that further studies will result in a better understanding of p53 regulation and that new insights will help unravel many aspects of cancer biology.
Collapse
Affiliation(s)
- Rajni Kumari
- Molecular Oncology Laboratory; National Institute of Immunology ; New Delhi, India
| | - Saishruti Kohli
- Molecular Oncology Laboratory; National Institute of Immunology ; New Delhi, India
| | - Sanjeev Das
- Molecular Oncology Laboratory; National Institute of Immunology ; New Delhi, India
| |
Collapse
|
38
|
LIU QUAN, ZHENG JIEMIN, CHEN JIAKANG, YAN XIANLEI, CHEN HONGMOU, NONG WEIXIA, HUANG HEQING. Histone deacetylase 5 promotes the proliferation of glioma cells by upregulation of Notch 1. Mol Med Rep 2014; 10:2045-50. [DOI: 10.3892/mmr.2014.2395] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 05/14/2014] [Indexed: 11/05/2022] Open
|
39
|
Abstract
In this issue of Molecular Cell, Sen et al. (2013) identify HDAC5 as a deacetylase with specificity for the K120 site of p53, a key residue of p53 that undergoes acetylation after DNA damage and regulates target gene selection.
Collapse
Affiliation(s)
- Sam Sulgi Kim
- Department of Biology, York University, Toronto M3J 1P3, Ontario, Canada
| | | |
Collapse
|