1
|
Wang J, Shao F, Yu QX, Ye L, Wusiman D, Wu R, Tuo Z, Wang Z, Li D, Cho WC, Wei W, Feng D. The Common Hallmarks and Interconnected Pathways of Aging, Circadian Rhythms, and Cancer: Implications for Therapeutic Strategies. RESEARCH (WASHINGTON, D.C.) 2025; 8:0612. [PMID: 40046513 PMCID: PMC11880593 DOI: 10.34133/research.0612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/17/2025]
Abstract
The intricate relationship between cancer, circadian rhythms, and aging is increasingly recognized as a critical factor in understanding the mechanisms underlying tumorigenesis and cancer progression. Aging is a well-established primary risk factor for cancer, while disruptions in circadian rhythms are intricately associated with the tumorigenesis and progression of various tumors. Moreover, aging itself disrupts circadian rhythms, leading to physiological changes that may accelerate cancer development. Despite these connections, the specific interplay between these processes and their collective impact on cancer remains inadequately explored in the literature. In this review, we systematically explore the physiological mechanisms of circadian rhythms and their influence on cancer development. We discuss how core circadian genes impact tumor risk and prognosis, highlighting the shared hallmarks of cancer and aging such as genomic instability, cellular senescence, and chronic inflammation. Furthermore, we examine the interplay between circadian rhythms and aging, focusing on how this crosstalk contributes to tumorigenesis, tumor proliferation, and apoptosis, as well as the impact on cellular metabolism and genomic stability. By elucidating the common pathways linking aging, circadian rhythms, and cancer, this review provides new insights into the pathophysiology of cancer and identifies potential therapeutic strategies. We propose that targeting the circadian regulation of cancer hallmarks could pave the way for novel treatments, including chronotherapy and antiaging interventions, which may offer important benefits in the clinical management of cancer.
Collapse
Affiliation(s)
- Jie Wang
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Fanglin Shao
- Department of Rehabilitation,
The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qing Xin Yu
- Department of Pathology,
Ningbo Clinical Pathology Diagnosis Center, Ningbo, Zhejiang 315211, China
- Department of Pathology,
Ningbo Medical Centre Lihuili Hospital, Ningbo, Zhejiang 315040, China
| | - Luxia Ye
- Department of Public Research Platform,
Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47906, USA
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Zhouting Tuo
- Department of Urological Surgery, Daping Hospital, Army Medical Center of PLA,
Army Medical University, Chongqing, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People’s Hospital,
University of Electronic Science and Technology of China, Chengdu, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - William C. Cho
- Department of Clinical Oncology,
Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital,
Sichuan University, Chengdu 610041, China
- Division of Surgery and Interventional Science,
University College London, London W1W 7TS, UK
| |
Collapse
|
2
|
Mohan N, Johnson GS, Tovar Perez JE, Dashwood WM, Rajendran P, Dashwood RH. Alternative splicing of BAZ1A in colorectal cancer disrupts the DNA damage response and increases chemosensitization. Cell Death Dis 2024; 15:570. [PMID: 39112459 PMCID: PMC11306231 DOI: 10.1038/s41419-024-06954-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
Bromodomain Adjacent to Zinc Finger Domain 1A (BAZ1A) is a critical regulator of chromatin remodeling. We sought to clarify the roles of BAZ1A in the etiology of colorectal cancer, including the mechanisms of its alternatively spliced variants. Public databases were examined and revealed high BAZ1A expression in the majority of colorectal cancer patients, which was corroborated in a panel of human colon cancer cell lines. BAZ1A silencing reduced cell viability and increased markers of DNA damage, apoptosis, and senescence, along with the downregulation of Wnt/β-catenin signaling. The corresponding molecular changes resulted in tumor growth inhibition when BAZ1A-knockout cells were implanted into nude mice. In rescue experiments, a short isoform of BAZ1A that was associated with alternative splicing by the DBIRD complex failed to restore DNA repair activity in colon cancer cells and maintained chemosensitivity to phleomycin treatment, unlike the full-length BAZ1A. A working model proposes that a buried domain in the N-terminus of the BAZ1A short isoform lacks the ability to access linker DNA, thereby disrupting the activity of the associated chromatin remodeling complexes. Given the current interest in RNA splicing deregulation and cancer etiology, additional mechanistic studies are warranted with new lead compounds targeting BAZ1A, and other members of the BAZ family, with a view to improved therapeutic interventions.
Collapse
Affiliation(s)
- Nivedhitha Mohan
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA
| | - Gavin S Johnson
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA
- CRISPR Therapeutics, South Boston, MA, USA
| | | | | | - Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA.
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX, USA.
| | - Roderick H Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA.
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX, USA.
| |
Collapse
|
3
|
Lugano D, Barrett L, Westerheide SD, Kee Y. Multifaceted roles of CCAR family proteins in the DNA damage response and cancer. Exp Mol Med 2024; 56:59-65. [PMID: 38172598 PMCID: PMC10834508 DOI: 10.1038/s12276-023-01139-1] [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: 06/05/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 01/05/2024] Open
Abstract
The cell cycle apoptosis regulator (CCAR) family of proteins consists of two proteins, CCAR1 and CCAR2, that play a variety of roles in cellular physiology and pathology. These multidomain proteins are able to perform multiple interactions and functions, playing roles in processes such as stress responses, metabolism, and the DNA damage response. The evolutionary conservation of CCAR family proteins allows their study in model organisms such as Caenorhabditis elegans, where a role for CCAR in aging was revealed. This review particularly highlights the multifaceted roles of CCAR family proteins and their implications in the DNA damage response and in cancer biology.
Collapse
Affiliation(s)
- D Lugano
- Department of Molecular Biosciences, College of Arts and Sciences, University of South Florida, Tampa, FL, 33647, USA
| | - L Barrett
- Department of Molecular Biosciences, College of Arts and Sciences, University of South Florida, Tampa, FL, 33647, USA
| | - S D Westerheide
- Department of Molecular Biosciences, College of Arts and Sciences, University of South Florida, Tampa, FL, 33647, USA
| | - Y Kee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno-Joongang-daero, Dalseong-gun, Daegu, 42988, Republic of Korea.
| |
Collapse
|
4
|
Wang Y, Wang JM, Xiao Y, Hu XB, Zheng SY, Fu JL, Zhang L, Gan YW, Liang XM, Li DWC. SUMO1-regulated DBC1 promotes p53-dependent stress-induced apoptosis of lens epithelial cells. Aging (Albany NY) 2023; 15:8812-8832. [PMID: 37683133 PMCID: PMC10522365 DOI: 10.18632/aging.205001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/20/2023] [Indexed: 09/10/2023]
Abstract
Deleted in breast cancer 1 (DBC1) was initially identified from a homozygously deleted region in human chromosome 8p21. It has been well established that DBC1 plays a dual role during cancer development. Depending on the physiological context, it can promote or inhibit tumorigenesis. Whether it plays a role in lens pathogenesis remains elusive. In the present study, we demonstrated that DBC1 is highly expressed in lens epithelial cells from different vertebrates and in retina pigment epithelial cells as well. Moreover, DBC1 is SUMOylated through SUMO1 conjugation at K591 residue in human and mouse lens epithelial cells. The SUMOylated DBC1 is localized in the nucleus and plays an essential role in promoting stress-induced apoptosis. Silence of DBC1 attenuates oxidative stress-induced apoptosis. In contrast, overexpression of DBC1 enhances oxidative stress-induced apoptosis, and this process depends on p53. Mechanistically, DBC1 interacts with p53 to regulate its phosphorylation status at multiple sites and the SUMOylation of DBC1 enhances its interaction with p53. Together, our results identify that DBC1 is an important regulator mediating stress-induced apoptosis in lens, and thus participates in control of lens cataractogenesis.
Collapse
Affiliation(s)
- Yan Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Jing-Miao Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Yuan Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Xue-Bin Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Shu-Yu Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Jia-Ling Fu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Lan Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Yu-Wen Gan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - Xing-Miao Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| | - David Wan-Cheng Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, China
| |
Collapse
|
5
|
Kim HJ, Moon SJ, Kim JH. Mechanistic insights into the dual role of CCAR2/DBC1 in cancer. Exp Mol Med 2023; 55:1691-1701. [PMID: 37524873 PMCID: PMC10474295 DOI: 10.1038/s12276-023-01058-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/21/2023] [Accepted: 05/17/2023] [Indexed: 08/02/2023] Open
Abstract
Cell cycle and apoptosis regulator 2 (CCAR2), also known as deleted in breast cancer 1 (DBC1), has been recently identified as a master regulator of transcriptional processes and plays diverse roles in physiology and pathophysiology, including as a regulator of apoptosis, DNA repair, metabolism, and tumorigenesis. CCAR2 functions as a coregulator of various transcription factors and a critical regulator of numerous epigenetic modifiers. Based on its ability to stimulate apoptosis by activating and stabilizing p53, CCAR2 was initially considered to be a tumor suppressor. However, an increasing number of studies have shown that CCAR2 also functions as a tumor-promoting coregulator by activating oncogenic transcription factors and regulating the enzymatic activity of epigenetic modifiers, indicating that CCAR2 may play a dual role in cancer progression by acting as a tumor suppressor and tumor promoter. Here, we review recent progress in understanding the dual tumor-suppressing and oncogenic roles of CCAR2 in cancer. We discuss CCAR2 domain structures, its interaction partners, and the molecular mechanisms by which it regulates the activities of transcription factors and epigenetic modifiers.
Collapse
Affiliation(s)
- Hwa Jin Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Sue Jin Moon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Jeong Hoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea.
| |
Collapse
|
6
|
Rasti G, Becker M, Vazquez BN, Espinosa-Alcantud M, Fernández-Duran I, Gámez-García A, Ianni A, Gonzalez J, Bosch-Presegué L, Marazuela-Duque A, Guitart-Solanes A, Segura-Bayona S, Bech-Serra JJ, Scher M, Serrano L, Shankavaram U, Erdjument-Bromage H, Tempst P, Reinberg D, Olivella M, Stracker T, de la Torre C, Vaquero A. SIRT1 regulates DNA damage signaling through the PP4 phosphatase complex. Nucleic Acids Res 2023; 51:6754-6769. [PMID: 37309898 PMCID: PMC10359614 DOI: 10.1093/nar/gkad504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 05/24/2023] [Accepted: 06/08/2023] [Indexed: 06/14/2023] Open
Abstract
The Sirtuin family of NAD+-dependent enzymes plays an important role in maintaining genome stability upon stress. Several mammalian Sirtuins have been linked directly or indirectly to the regulation of DNA damage during replication through Homologous recombination (HR). The role of one of them, SIRT1, is intriguing as it seems to have a general regulatory role in the DNA damage response (DDR) that has not yet been addressed. SIRT1-deficient cells show impaired DDR reflected in a decrease in repair capacity, increased genome instability and decreased levels of γH2AX. Here we unveil a close functional antagonism between SIRT1 and the PP4 phosphatase multiprotein complex in the regulation of the DDR. Upon DNA damage, SIRT1 interacts specifically with the catalytical subunit PP4c and promotes its inhibition by deacetylating the WH1 domain of the regulatory subunits PP4R3α/β. This in turn regulates γH2AX and RPA2 phosphorylation, two key events in the signaling of DNA damage and repair by HR. We propose a mechanism whereby during stress, SIRT1 signaling ensures a global control of DNA damage signaling through PP4.
Collapse
Affiliation(s)
- George Rasti
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Via de l’Hospitalet, 199-203, 08908 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Maximilian Becker
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Via de l’Hospitalet, 199-203, 08908 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Berta N Vazquez
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Via de l’Hospitalet, 199-203, 08908 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Maria Espinosa-Alcantud
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Via de l’Hospitalet, 199-203, 08908 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Irene Fernández-Duran
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Andrés Gámez-García
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Alessandro Ianni
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231Bad Nauheim, Germany
| | - Jessica Gonzalez
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Via de l’Hospitalet, 199-203, 08908 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Laia Bosch-Presegué
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Via de l’Hospitalet, 199-203, 08908 L’Hospitalet de Llobregat, Barcelona, Spain
- Tissue Repair and Regeneration Laboratory (TR2Lab), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IrisCC). Experimental Sciences and Methodology Department. Faculty of Health Sciences and Welfare (FCSB), University of Vic - Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Anna Marazuela-Duque
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Via de l’Hospitalet, 199-203, 08908 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Anna Guitart-Solanes
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Sandra Segura-Bayona
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Current affiliation: The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Joan-Josep Bech-Serra
- Proteomic Unit, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Spain
| | - Michael Scher
- Howard Hughes Medical Institute, Division of Nucleic Acids Enzymology, Department of Biochemistry, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, NJ08854, USA
| | - Lourdes Serrano
- Department of Science, BMCC, The City University of New York (CUNY), 199 Chambers Street N699P, New Yirk, NY10007, USA
| | - Uma Shankavaram
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD20892, USA
| | - Hediye Erdjument-Bromage
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY10065, USA
- Department of Cell Biology, New York University School of Medicine, New York, NY10016, USA
| | - Paul Tempst
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY10065, USA
| | - Danny Reinberg
- Howard Hughes Medical Institute, Division of Nucleic Acids Enzymology, Department of Biochemistry, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, NJ08854, USA
- Howard Hughes Medical Institute, Department of Biochemistry, New York University School of Medicine, New York, NY10016, USA
| | - Mireia Olivella
- Bioinfomatics and Medical Statistics Group, Faculty of Science, Technology and Engineering. University of Vic-Central University of Catalonia, Vic, Spain
| | - Travis H Stracker
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD20892, USA
| | - Carolina de la Torre
- Proteomic Unit, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Spain
| | - Alejandro Vaquero
- Chromatin Biology Laboratory, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Barcelona, Spain
- Chromatin Biology Laboratory, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Via de l’Hospitalet, 199-203, 08908 L’Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
7
|
García-Fernández P, Reinhold C, Üçeyler N, Sommer C. Local Inflammatory Mediators Involved in Neuropathic Pain. Int J Mol Sci 2023; 24:ijms24097814. [PMID: 37175520 PMCID: PMC10178336 DOI: 10.3390/ijms24097814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Polyneuropathy (PNP) is a term to describe diseases of the peripheral nervous system, 50% of which present with neuropathic pain. In some types of PNP, pain is restricted to the skin distally in the leg, suggesting a local regulatory process leading to pain. In this study, we proposed a pro-inflammatory pathway mediated by NF-κB that might be involved in the development of pain in patients with painful PNP. To test this hypothesis, we have collected nerve and skin samples from patients with different etiologies and levels of pain. We performed RT-qPCR to analyze the gene expression of the proposed inflammatory pathway components in sural nerve and in distal and proximal skin samples. In sural nerve, we showed a correlation of TLR4 and TNFα to neuropathic pain, and an upregulation of TNFα in patients with severe pain. Patients with an inflammatory PNP also presented a lower expression of TRPV1 and SIRT1. In distal skin, we found a reduced expression of TLR4 and miR-146-5p, in comparison to proximal skin. Our findings thus support our hypothesis of local inflammatory processes involved in pain in PNP, and further show disturbed anti-inflammatory pathways involving TRPV1 and SIRT1 in inflammatory PNP.
Collapse
Affiliation(s)
| | - Colette Reinhold
- Department of Neurology, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Nurcan Üçeyler
- Department of Neurology, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital of Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
8
|
Zhao R, Cai K, Yang JJ, Zhou Q, Cao W, Xiang J, Shen YH, Cheng LL, Zang WD, Lin Y, Yuan YY, Xu W, Tao H, Zhao SM, Zhao JY. Nuclear ATR lysine-tyrosylation protects against heart failure by activating DNA damage response. Cell Rep 2023; 42:112400. [PMID: 37071536 DOI: 10.1016/j.celrep.2023.112400] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 01/12/2023] [Accepted: 03/31/2023] [Indexed: 04/19/2023] Open
Abstract
Dysregulated amino acid increases the risk for heart failure (HF) via unclear mechanisms. Here, we find that increased plasma tyrosine and phenylalanine levels are associated with HF. Increasing tyrosine or phenylalanine by high-tyrosine or high-phenylalanine chow feeding exacerbates HF phenotypes in transverse aortic constriction and isoproterenol infusion mice models. Knocking down phenylalanine dehydrogenase abolishes the effect of phenylalanine, indicating that phenylalanine functions by converting to tyrosine. Mechanistically, tyrosyl-tRNA synthetase (YARS) binds to ataxia telangiectasia and Rad3-related gene (ATR), catalyzes lysine tyrosylation (K-Tyr) of ATR, and activates the DNA damage response (DDR) in the nucleus. Increased tyrosine inhibits the nuclear localization of YARS, inhibits the ATR-mediated DDR, accumulates DNA damage, and elevates cardiomyocyte apoptosis. Enhancing ATR K-Tyr by overexpressing YARS, restricting tyrosine, or supplementing tyrosinol, a structural analog of tyrosine, promotes YARS nuclear localization and alleviates HF in mice. Our findings implicate facilitating YARS nuclear translocation as a potential preventive and/or interfering measure against HF.
Collapse
Affiliation(s)
- Rui Zhao
- Obstetrics & Gynecology Hospital of Fudan University, Zhongshan Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ke Cai
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jing-Jing Yang
- Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, and Cardiovascular Research Center, Anhui Medical University, Hefei 230601, China
| | - Qian Zhou
- Obstetrics & Gynecology Hospital of Fudan University, Zhongshan Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China; Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Wei Cao
- Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, and Cardiovascular Research Center, Anhui Medical University, Hefei 230601, China
| | - Jie Xiang
- Obstetrics & Gynecology Hospital of Fudan University, Zhongshan Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Yi-Hui Shen
- Obstetrics & Gynecology Hospital of Fudan University, Zhongshan Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Lei-Lei Cheng
- Obstetrics & Gynecology Hospital of Fudan University, Zhongshan Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Wei-Dong Zang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yan Lin
- Obstetrics & Gynecology Hospital of Fudan University, Zhongshan Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Yi-Yuan Yuan
- Obstetrics & Gynecology Hospital of Fudan University, Zhongshan Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Wei Xu
- Obstetrics & Gynecology Hospital of Fudan University, Zhongshan Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Hui Tao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, and Cardiovascular Research Center, Anhui Medical University, Hefei 230601, China.
| | - Shi-Min Zhao
- Obstetrics & Gynecology Hospital of Fudan University, Zhongshan Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China.
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, and Cardiovascular Research Center, Anhui Medical University, Hefei 230601, China; School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
9
|
Zhu H, Zhang Y, Zhu Y. MiR-342-5p protects neurons from cerebral ischemia induced-apoptosis through regulation of Akt/NF-κB pathways by targeting CCAR2. J Stroke Cerebrovasc Dis 2023; 32:106901. [PMID: 36434857 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/24/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES Ischemic stroke causes high morbidity, mortality and health burden in the world. MiR-342-5p was associated with Alzheimer's disease and cardio-protection. Herein, we aimed to reveal effects of miR-342-5p on cerebral ischemia injury as well as novel targets for stroke. MATERIALS AND METHODS AgomiR-342-5p was intracerebroventricularly injected into the middle cerebral artery occlusion (MCAO) mouse models to evaluate functions of miR-342-5p on cerebral ischemia. RT-qPCR and western blot assays were used to evaluate genes expression. Oxygen-glucose deprivation (OGD) was used as an in vitro model for ischemia. Viability and apoptosis ratio of neurons was evaluated by CCK-8, LDH release detection, and flow cytometry. The potential targets of miR-342-5p were predicted by Targetscan, and their interaction was confirmed by luciferase assay. RESULTS The intervention of miR-342-5p effectively attenuated ischemic injury in MCAO mice. MiR-342-5p overexpression could protect neurons against OGD-induced injury, as revealed by increased cell viability and BCL2 expression, and decreased LDH release, apoptosis ratio, and BAX expression in OGD-induced neurons. Mechanically, miR-342-5p could directly bound with CCAR2 to inhibit its expression. Overexpressing CARR2 aggravated the OGD-induced injury of neurons, which was partly restrained by overexpressing miR-342-5p reversed. Furthermore, miR-342-5p/CARR2 axis regulates Akt/NF-κB signaling pathway in vitro as well as in vivo cerebral ischemia models. CONCLUSIONS MiR-342-5p inhibited neuron apoptosis by regulating Akt/NF-kB signaling pathway via CCAR2 suppression. Our findings revealed the neuroprotection of miR-342-5p in cerebral ischemia.
Collapse
Affiliation(s)
- Haochun Zhu
- Department of Neurology, General Hospital of Hebi Coal Industry Group Co., Ltd., No. 84, Hongqi Street, Hebi, Henan 458000, China.
| | - Yanhua Zhang
- Department of Neurology, General Hospital of Hebi Coal Industry Group Co., Ltd., No. 84, Hongqi Street, Hebi, Henan 458000, China.
| | - Yanling Zhu
- Department of Neurology, General Hospital of Hebi Coal Industry Group Co., Ltd., No. 84, Hongqi Street, Hebi, Henan 458000, China.
| |
Collapse
|
10
|
Erichsen L, Adjaye J. Crosstalk between age accumulated DNA-damage and the SIRT1-AKT-GSK3ß axis in urine derived renal progenitor cells. Aging (Albany NY) 2022; 14:8179-8204. [PMID: 36170022 PMCID: PMC9648809 DOI: 10.18632/aging.204300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/08/2022] [Indexed: 12/02/2022]
Abstract
The aging process is manifested by a multitude of inter-linked biological processes. These processes contribute to genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, de-regulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. The mammalian ortholog of the yeast silent information regulator (Sir2) SIRT1 is a NAD+-dependent class III histone deacetylase and has been recognized to be involved in many of the forementioned processes. Furthermore, the physiological activity of several Sirtuin family members has been connected to the regulation of life span of lower organisms (Caenorhabditis elegans and Drosophila melanogaster) as well as mammals. In the present study, we provide evidence that SIX2-positive urine derived renal progenitor cells-UdRPCs isolated directly from human urine show typical hallmarks of aging. This includes the subsequent transcriptional downregulation of SIRT1 and its downstream targets AKT and GSK3ß with increased donor age. This transcriptional downregulation is accompanied by an increase in DNA damage and transcriptional levels of several cell cycle inhibitors such as P16. We provide evidence that the renal progenitor transcription factor SIX2 binds to the coding sequence of SIRT1. Furthermore, we show that the SIRT1 promoter region is methylation sensitive and becomes methylated during aging, dividing them into SIRT1-high and -low expressing UdRPCs. Our results highlight the importance of SIRT1 in DNA damage repair recognition in UdRPCs and the control of differentiation by regulating the activation of GSK3β through AKT.
Collapse
Affiliation(s)
- Lars Erichsen
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine- University Düsseldorf, Düsseldorf 40225, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine- University Düsseldorf, Düsseldorf 40225, Germany
| |
Collapse
|
11
|
Liu Q, Luo Q, Feng J, Zhao Y, Ma B, Cheng H, Zhao T, Lei H, Mu C, Chen L, Meng Y, Zhang J, Long Y, Su J, Chen G, Li Y, Hu G, Liao X, Chen Q, Zhu Y. Hypoxia-induced proteasomal degradation of DBC1 by SIAH2 in breast cancer progression. eLife 2022; 11:81247. [PMID: 35913115 PMCID: PMC9377797 DOI: 10.7554/elife.81247] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/19/2022] [Indexed: 11/26/2022] Open
Abstract
DBC1 has been characterized as a key regulator of physiological and pathophysiological activities, such as DNA damage, senescence, and tumorigenesis. However, the mechanism by which the functional stability of DBC1 is regulated has yet to be elucidated. Here, we report that the ubiquitination-mediated degradation of DBC1 is regulated by the E3 ubiquitin ligase SIAH2 and deubiquitinase OTUD5 under hypoxic stress. Mechanistically, hypoxia promoted DBC1 to interact with SIAH2 but not OTUD5, resulting in the ubiquitination and subsequent degradation of DBC1 through the ubiquitin–proteasome pathway. SIAH2 knockout inhibited tumor cell proliferation and migration, which could be rescued by double knockout of SIAH2/CCAR2. Human tissue microarray analysis further revealed that the SIAH2/DBC1 axis was responsible for tumor progression under hypoxic stress. These findings define a key role of the hypoxia-mediated SIAH2-DBC1 pathway in the progression of human breast cancer and provide novel insights into the metastatic mechanism of breast cancer.
Collapse
Affiliation(s)
- Qiangqiang Liu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Qian Luo
- College of Life Sciences, Nankai University, Tianjin, China
| | - Jianyu Feng
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yanping Zhao
- School of Statistics and Data Science, Nankai University, Tianjin, China
| | - Biao Ma
- College of Life Sciences, Nankai University, Tianjin, China
| | | | - Tian Zhao
- College of Life Sciences, Nankai University, Tianjin, China
| | - Hong Lei
- College of Life Sciences, Nankai University, Tianjin, China
| | - Chenglong Mu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Linbo Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yuanyuan Meng
- College of Life Sciences, Nankai University, Tianjin, China
| | - Jiaojiao Zhang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yijia Long
- College of Life Sciences, Nankai University, Tianjin, China
| | - Jingyi Su
- College of Life Sciences, Nankai University, Tianjin, China
| | - Guo Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yanjun Li
- College of Life Sciences, Nankai University, Tianjin, China
| | - Gang Hu
- School of Statistics and Data Science, Nankai University, Tianjin, China
| | - Xudong Liao
- College of Life Sciences, Nankai University, Tianjin, China
| | - Quan Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yushan Zhu
- College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
12
|
Sarwar Z, Nabi N, Bhat SA, Gillani SQ, Reshi I, Un Nisa M, Adelmant G, Marto J, Andrabi S. Interaction of DBC1 with polyoma small T antigen promotes its degradation and negatively regulates tumorigenesis. J Biol Chem 2021; 298:101496. [PMID: 34921839 PMCID: PMC8784333 DOI: 10.1016/j.jbc.2021.101496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 11/09/2021] [Accepted: 12/10/2021] [Indexed: 12/05/2022] Open
Abstract
Deleted in Breast Cancer 1 (DBC1) is an important metabolic sensor. Previous studies have implicated DBC1 in various cellular functions, notably cell proliferation, apoptosis, histone modification, and adipogenesis. However, current reports about the role of DBC1 in tumorigenesis are controversial and designate DBC1 alternatively as a tumor suppressor or a tumor promoter. In the present study, we report that polyoma small T antigen (PyST) associates with DBC1 in mammalian cells, and this interaction leads to the posttranslational downregulation of DBC1 protein levels. When coexpressed, DBC1 overcomes PyST-induced mitotic arrest and promotes the exit of cells from mitosis. Using both transient and stable modes of PyST expression, we also show that cellular DBC1 is subjected to degradation by LKB1, a tumor suppressor and cellular energy sensor kinase, in an AMP kinase-independent manner. Moreover, LKB1 negatively regulates the phosphorylation as well as activity of the prosurvival kinase AKT1 through DBC1 and its downstream pseudokinase substrate, Tribbles 3 (TRB3). Using both transient transfection and stable cell line approaches as well as soft agar assay, we demonstrate that DBC1 has oncogenic potential. In conclusion, our study provides insight into a novel signaling axis that connects LKB1, DBC1, TRB3, and AKT1. We propose that the LKB1–DBC1–AKT1 signaling paradigm may have an important role in the regulation of cell cycle and apoptosis and consequently tumorigenesis.
Collapse
Affiliation(s)
- Zarka Sarwar
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006
| | - Nusrat Nabi
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006
| | - Sameer Ahmed Bhat
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006
| | | | - Irfana Reshi
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006
| | - Misbah Un Nisa
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006
| | - Guillaume Adelmant
- Blais Proteomics Centre, Dana Farber Cancer Institute, Harvard University, Boston, USA
| | - Jarrod Marto
- Blais Proteomics Centre, Dana Farber Cancer Institute, Harvard University, Boston, USA
| | - Shaida Andrabi
- Department of Biochemistry, University of Kashmir, Srinagar, India, 190006.
| |
Collapse
|
13
|
Yan P, Li Z, Xiong J, Geng Z, Wei W, Zhang Y, Wu G, Zhuang T, Tian X, Liu Z, Liu J, Sun K, Chen F, Zhang Y, Zeng C, Huang Y, Zhang B. LARP7 ameliorates cellular senescence and aging by allosterically enhancing SIRT1 deacetylase activity. Cell Rep 2021; 37:110038. [PMID: 34818543 DOI: 10.1016/j.celrep.2021.110038] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/24/2021] [Accepted: 11/01/2021] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is associated with pleiotropic physiopathological processes, including aging and age-related diseases. The persistent DNA damage is a major stress leading to senescence, but the underlying molecular link remains elusive. Here, we identify La Ribonucleoprotein 7 (LARP7), a 7SK RNA binding protein, as an aging antagonist. DNA damage-mediated Ataxia Telangiectasia Mutated (ATM) activation triggers the extracellular shuttling and downregulation of LARP7, which dampens SIRT1 deacetylase activity, enhances p53 and NF-κB (p65) transcriptional activity by augmenting their acetylation, and thereby accelerates cellular senescence. Deletion of LARP7 leads to senescent cell accumulation and premature aging in rodent model. Furthermore, we show this ATM-LARP7-SIRT1-p53/p65 senescence axis is active in vascular senescence and atherogenesis, and preventing its activation substantially alleviates senescence and atherogenesis. Together, this study identifies LARP7 as a gatekeeper of senescence, and the altered ATM-LARP7-SIRT1-p53/p65 pathway plays an important role in DNA damage response (DDR)-mediated cellular senescence and atherosclerosis.
Collapse
Affiliation(s)
- Pengyi Yan
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Zixuan Li
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Junhao Xiong
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Zilong Geng
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Weiting Wei
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Yan Zhang
- Renji-Med Clinical Stem Cell Research Center, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gengze Wu
- Department of Cardiology, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Tao Zhuang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Xiaoyu Tian
- School of Biomedical Sciences, Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Zhijie Liu
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology and Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Sun
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Fengyuan Chen
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Chunyu Zeng
- Department of Cardiology, Chongqing Institute of Cardiology, Chongqing Cardiovascular Clinical Research Center, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Bing Zhang
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China.
| |
Collapse
|
14
|
Li BS, Jin AL, Zhou Z, Seo JH, Choi BM. DRG2 Accelerates Senescence via Negative Regulation of SIRT1 in Human Diploid Fibroblasts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7301373. [PMID: 34777693 PMCID: PMC8580627 DOI: 10.1155/2021/7301373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/04/2021] [Indexed: 11/21/2022]
Abstract
Accumulating evidence suggests that developmentally regulated GTP-binding protein 2 (DRG2), an evolutionarily conserved GTP-binding protein, plays an important role in regulating cell growth, inflammation, and mitochondria dynamics. However, the effect of DRG2 in aging remains unclear. In this study, we found that endogenous DRG2 protein expression is upregulated in oxidative stress-induced premature senescence models and tissues of aged mice. Ectopic expression of DRG2 significantly promoted senescence-associated β-galactosidase (SA-β-gal) activity and inhibited cell growth, concomitant with increase in levels of acetyl (ac)-p53 (Lys382), ac-nuclear factor-kB (NF-κB) p65 (Lys310), p21 Waf1/Cip1 , and p16 Ink4a and a decrease in cyclin D1. In this process, reactive oxygen species (ROS) and phosphorylation of H2A histone family member X (H2A.X), forming γ-H2A.X, were enhanced. Mechanistically, ectopic expression of DRG2 downregulated Sirtuin-1 (SIRT1), resulting in augmented acetylation of p53 and NF-κB p65. Additionally, DRG2 knockdown significantly abolished oxidative stress-induced premature senescence. Our results provide a possible molecular mechanism for investigation of cellular senescence and aging regulated by DRG2.
Collapse
Affiliation(s)
- Bing Si Li
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Ai Lin Jin
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - ZiQi Zhou
- Department of Herbology, Wonkwang University School of Korean Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jae Ho Seo
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
- Sarcopenia Total Solution Center, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Byung-Min Choi
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| |
Collapse
|
15
|
Halasa M, Adamczuk K, Adamczuk G, Afshan S, Stepulak A, Cybulski M, Wawruszak A. Deacetylation of Transcription Factors in Carcinogenesis. Int J Mol Sci 2021; 22:11810. [PMID: 34769241 PMCID: PMC8583941 DOI: 10.3390/ijms222111810] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/25/2021] [Indexed: 02/07/2023] Open
Abstract
Reversible Nε-lysine acetylation/deacetylation is one of the most common post-translational modifications (PTM) of histones and non-histone proteins that is regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs). This epigenetic process is highly involved in carcinogenesis, affecting histone and non-histone proteins' properties and their biological functions. Some of the transcription factors, including tumor suppressors and oncoproteins, undergo this modification altering different cell signaling pathways. HDACs deacetylate their targets, which leads to either the upregulation or downregulation of proteins involved in the regulation of cell cycle and apoptosis, ultimately influencing tumor growth, invasion, and drug resistance. Therefore, epigenetic modifications are of great clinical importance and may constitute a new therapeutic target in cancer treatment. This review is aimed to present the significance of HDACs in carcinogenesis through their influence on functions of transcription factors, and therefore regulation of different signaling pathways, cancer progression, and metastasis.
Collapse
Affiliation(s)
- Marta Halasa
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, Witolda Chodźki 1 St., 20-093 Lublin, Poland; (M.H.); (K.A.); (A.S.); (M.C.)
| | - Kamila Adamczuk
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, Witolda Chodźki 1 St., 20-093 Lublin, Poland; (M.H.); (K.A.); (A.S.); (M.C.)
| | - Grzegorz Adamczuk
- Independent Medical Biology Unit, Medical University of Lublin, Kazimierza Jaczewskiego 8b St., 20-090 Lublin, Poland;
| | - Syeda Afshan
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku and Turku University Hospital, 20520 Turku, Finland;
| | - Andrzej Stepulak
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, Witolda Chodźki 1 St., 20-093 Lublin, Poland; (M.H.); (K.A.); (A.S.); (M.C.)
| | - Marek Cybulski
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, Witolda Chodźki 1 St., 20-093 Lublin, Poland; (M.H.); (K.A.); (A.S.); (M.C.)
| | - Anna Wawruszak
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, Witolda Chodźki 1 St., 20-093 Lublin, Poland; (M.H.); (K.A.); (A.S.); (M.C.)
| |
Collapse
|
16
|
Storchova R, Burdova K, Palek M, Medema RH, Macurek L. A novel assay for screening WIP1 phosphatase substrates in nuclear extracts. FEBS J 2021; 288:6035-6051. [PMID: 33982878 DOI: 10.1111/febs.15965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 11/30/2022]
Abstract
Upon exposure to genotoxic stress, cells activate DNA damage response (DDR) that coordinates DNA repair with a temporal arrest in the cell cycle progression. DDR is triggered by activation of ataxia telangiectasia mutated/ataxia telangiectasia and Rad3-related protein kinases that phosphorylate multiple targets including tumor suppressor protein tumor suppressor p53 (p53). In addition, DNA damage can activate parallel stress response pathways [such as mitogen-activated protein kinase p38 alpha (p38)/MAPK-activated protein kinase 2 (MK2) kinases] contributing to establishing the cell cycle arrest. Wild-type p53-induced phosphatase 1 (WIP1) controls timely inactivation of DDR and is needed for recovery from the G2 checkpoint by counteracting the function of p53. Here, we developed a simple in vitro assay for testing WIP1 substrates in nuclear extracts. Whereas we did not detect any activity of WIP1 toward p38/MK2, we confirmed p53 as a substrate of WIP1. Inhibition or inactivation of WIP1 in U2OS cells increased phosphorylation of p53 at S15 and potentiated its acetylation at K382. Further, we identified Deleted in breast cancer gene 1 (DBC1) as a new substrate of WIP1 but surprisingly, depletion of DBC1 did not interfere with the ability of WIP1 to regulate p53 acetylation. Instead, we have found that WIP1 activity suppresses p53-K382 acetylation by inhibiting the interaction between p53 and the acetyltransferase p300. Newly established phosphatase assay allows an easy comparison of WIP1 ability to dephosphorylate various proteins and thus contributes to identification of its physiological substrates.
Collapse
Affiliation(s)
- Radka Storchova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Kamila Burdova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Matous Palek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - René H Medema
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
17
|
Zhang H, Xiao X, Wei W, Huang C, Wang M, Wang L, He Y, Sun J, Jiang Y, Jiang G, Zhang X. CircLIFR synergizes with MSH2 to attenuate chemoresistance via MutSα/ATM-p73 axis in bladder cancer. Mol Cancer 2021; 20:70. [PMID: 33874956 PMCID: PMC8054397 DOI: 10.1186/s12943-021-01360-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cisplatin (CDDP) has become a standard-of-care treatment for muscle-invasive bladder cancer (MIBC), while chemoresistance remains a major challenge. Accumulating evidence indicates that circular RNAs (circRNAs) are discrete functional entities. However, the regulatory functions as well as complexities of circRNAs in modulating CDDP-based chemotherapy in bladder cancer are yet to be well revealed. METHODS Through analyzing the expression profile of circRNAs in bladder cancer tissues, RNA FISH, circRNA pull-down assay, mass spectrometry analysis and RIP, circLIFR was identified and its interaction with MSH2 was confirmed. The effects of circLIFR and MSH2 on CDDP-based chemotherapy were explored by flow cytometry and rescue experiments. Co-IP and Western blot were used to investigate the molecular mechanisms underlying the functions of circLIFR and MSH2. Biological implications of circLIFR and MSH2 in bladder cancer were implemented in tumor xenograft models and PDX models. RESULTS CircLIFR was downregulated in bladder cancer and expression was positively correlated with favorable prognosis. Moreover, circLIFR synergizing with MSH2, which was a mediator of CDDP sensitivity in bladder cancer cells, positively modulated sensitivity to CDDP in vitro and in vivo. Mechanistically, circLIFR augmented the interaction between MutSα and ATM, ultimately contributing to stabilize p73, which triggered to apoptosis. Importantly, MIBC with high expression of circLIFR and MSH2 was more sensitive to CDDP-based chemotherapy in tumor xenograft models and PDX models. CONCLUSIONS CircLIFR could interact with MSH2 to positively modulate CDDP-sensitivity through MutSα/ATM-p73 axis in bladder cancer. CircLIFR and MSH2 might be act as promising therapeutic targets for CDDP-resistant bladder cancer.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xingyuan Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenjie Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chao Huang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Miao Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liang Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuanqiao He
- Department of Laboratory Animal Science, Nanchang University, Nanchang, 330006, China
| | - Jiayin Sun
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yangkai Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guosong Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
18
|
Wang M, Lin H. Understanding the Function of Mammalian Sirtuins and Protein Lysine Acylation. Annu Rev Biochem 2021; 90:245-285. [PMID: 33848425 DOI: 10.1146/annurev-biochem-082520-125411] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein lysine acetylation is an important posttranslational modification that regulates numerous biological processes. Targeting lysine acetylation regulatory factors, such as acetyltransferases, deacetylases, and acetyl-lysine recognition domains, has been shown to have potential for treating human diseases, including cancer and neurological diseases. Over the past decade, many other acyl-lysine modifications, such as succinylation, crotonylation, and long-chain fatty acylation, have also been investigated and shown to have interesting biological functions. Here, we provide an overview of the functions of different acyl-lysine modifications in mammals. We focus on lysine acetylation as it is well characterized, and principles learned from acetylation are useful for understanding the functions of other lysine acylations. We pay special attention to the sirtuins, given that the study of sirtuins has provided a great deal of information about the functions of lysine acylation. We emphasize the regulation of sirtuins to illustrate that their regulation enables cells to respond to various signals and stresses.
Collapse
Affiliation(s)
- Miao Wang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA;
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA; .,Howard Hughes Medical Institute, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
19
|
Fan G, Sun L, Meng L, Hu C, Wang X, Shi Z, Hu C, Han Y, Yang Q, Cao L, Zhang X, Zhang Y, Song X, Xia S, He B, Zhang S, Wang C. The ATM and ATR kinases regulate centrosome clustering and tumor recurrence by targeting KIFC1 phosphorylation. Nat Commun 2021; 12:20. [PMID: 33397932 PMCID: PMC7782532 DOI: 10.1038/s41467-020-20208-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 11/18/2020] [Indexed: 12/31/2022] Open
Abstract
Drug resistance and tumor recurrence are major challenges in cancer treatment. Cancer cells often display centrosome amplification. To maintain survival, cancer cells achieve bipolar division by clustering supernumerary centrosomes. Targeting centrosome clustering is therefore considered a promising therapeutic strategy. However, the regulatory mechanisms of centrosome clustering remain unclear. Here we report that KIFC1, a centrosome clustering regulator, is positively associated with tumor recurrence. Under DNA damaging treatments, the ATM and ATR kinases phosphorylate KIFC1 at Ser26 to selectively maintain the survival of cancer cells with amplified centrosomes via centrosome clustering, leading to drug resistance and tumor recurrence. Inhibition of KIFC1 phosphorylation represses centrosome clustering and tumor recurrence. This study identified KIFC1 as a prognostic tumor recurrence marker, and revealed that tumors can acquire therapeutic resistance and recurrence via triggering centrosome clustering under DNA damage stresses, suggesting that blocking KIFC1 phosphorylation may open a new vista for cancer therapy.
Collapse
Affiliation(s)
- Guangjian Fan
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Lianhui Sun
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Ling Meng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, 271000, Shandong, China
| | - Chen Hu
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Xing Wang
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Zhan Shi
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Congli Hu
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Yang Han
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Qingqing Yang
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, College of Translational Medicine, China Medical University, 110000, Shenyang, China
| | - Xiaohong Zhang
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R., Detroit, MI, 48201, USA
| | - Yan Zhang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Xianmin Song
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Shujie Xia
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine; Institute of Urology, Shanghai Jiao Tong University, 200080, Shanghai, China
| | - Baokun He
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Shengping Zhang
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China.
| | - Chuangui Wang
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China.
| |
Collapse
|
20
|
Xu D, Liu L, Zhao Y, Yang L, Cheng J, Hua R, Zhang Z, Li Q. Melatonin protects mouse testes from palmitic acid-induced lipotoxicity by attenuating oxidative stress and DNA damage in a SIRT1-dependent manner. J Pineal Res 2020; 69:e12690. [PMID: 32761924 DOI: 10.1111/jpi.12690] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/23/2020] [Accepted: 08/01/2020] [Indexed: 12/13/2022]
Abstract
Palmitic acid (PA), the main component of dietary saturated fat, has been known to increase in patients with obesity, and PA-induced lipotoxicity may contribute to obesity-related male infertility. Melatonin has beneficial effects on reproductive processes; however, the effect and the underlying molecular mechanism of melatonin's involvement in PA-induced cytotoxicity in the testes are poorly understood. Our findings showed that lipotoxicity was observed in mouse testes after long-term PA treatment and that melatonin therapy restored spermatogenesis and fertility in these males. Moreover, melatonin therapy suppressed PA-induced apoptosis by modulating apoptosis-associated proteins such as Bcl2, Bax, C-Caspase3, C-Caspase12, and CHOP in type B spermatogonial stem cells. Changes in the expression of endoplasmic reticulum (ER) stress markers (p-IRE1, p-PERK, ATF4) and intracellular Ca2+ levels showed that melatonin relieved PA-induced ER stress. Mechanistically, melatonin stimulated the expression and nuclear translocation of SIRT1 through its receptors and prevented PA-induced ROS production and mitochondrial dysfunction via SIRT1 signaling pathway. Furthermore, melatonin promoted SIRT1-mediated p53 deacetylation, thereby relieving G2/M arrest in response to PA-stimulated DNA damage. Collectively, these findings indicate that melatonin protects the testes from PA-induced lipotoxicity through the activation of SIRT1, which alleviates oxidative stress, ER stress, mitochondrial dysfunction, and DNA damage.
Collapse
Affiliation(s)
- Dejun Xu
- College of Animal Science and Technology, Southwest University, Chongqing, China
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Lingbin Liu
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yongju Zhao
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Li Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jianyong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Rongmao Hua
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zelin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Qingwang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
21
|
Johnson GS, Rajendran P, Dashwood RH. CCAR1 and CCAR2 as gene chameleons with antagonistic duality: Preclinical, human translational, and mechanistic basis. Cancer Sci 2020; 111:3416-3425. [PMID: 33403784 PMCID: PMC7540973 DOI: 10.1111/cas.14579] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/04/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Cell Cycle and Apoptosis Regulator 1 (CCAR1) and Cell Cycle and Apoptosis Regulator 2 (CCAR2) have emerged as key players in physiology and pathophysiology, with critical roles in the DNA damage response, nuclear receptor function, and Wnt signaling, among other activities. Contradictory reports exist on the functional duality of CCAR1 and CCAR2 as either tumor promoters or suppressors, suggesting that CCAR1 and CCAR2 have the hallmarks of gene chameleons. We review herein the mechanistic, preclinical, and human translational findings for CCAR1 and CCAR2, based on available RNA and protein expression data from human studies, The Cancer Genome Atlas (TCGA) data mining, gene knockout mouse models, and cell-based assays. Multiple factors contribute to the divergent activities of CCAR1 and CCAR2, including tissue type, mutation/genetic background, protein-protein interactions, dynamic regulation via posttranslational modifications, and alternative RNA splicing. An array of protein partners interact with CCAR1 and CCAR2 in the context of tumor promotion and suppression, including β-catenin, androgen receptor, p21Cip1/Waf1, tumor protein p53 (p53), sirtuin 1, and histone deacetylase 3. Genetic changes frequently found in cancer, such as TP53 mutation, also serve as critical determinants of survival outcomes in cancer patients. This review seeks to provide the impetus for further investigation into CCAR1 and CCAR2 as potential master regulators of metabolism, aging, and cancer.
Collapse
Affiliation(s)
- Gavin S. Johnson
- Center for Epigenetics & Disease PreventionTexas A&M Health Science CenterHoustonTXUSA
| | - Praveen Rajendran
- Center for Epigenetics & Disease PreventionTexas A&M Health Science CenterHoustonTXUSA
| | - Roderick H. Dashwood
- Center for Epigenetics & Disease PreventionTexas A&M Health Science CenterHoustonTXUSA
- Department of Translational Medical Sciences, Texas A&M College of MedicineTexas A&M UniversityHouston CampusTXUSA
- Department of Clinical Cancer PreventionThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| |
Collapse
|
22
|
Akande OE, Damle PK, Pop M, Sherman NE, Szomju BB, Litovchick LV, Grossman SR. DBC1 Regulates p53 Stability via Inhibition of CBP-Dependent p53 Polyubiquitination. Cell Rep 2020; 26:3323-3335.e4. [PMID: 30893604 PMCID: PMC6478392 DOI: 10.1016/j.celrep.2019.02.076] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/21/2018] [Accepted: 02/20/2019] [Indexed: 01/10/2023] Open
Abstract
The control of p53 protein stability is critical to its tumor suppressor functions. The CREB binding protein (CBP) transcriptional co-activator co-operates with MDM2 to maintain normally low physiological p53 levels in cells via exclusively cytoplasmic E4 polyubiquitination activity. Using mass spectrometry to identify nuclear and cytoplasmic CBP-interacting proteins that regulate compartmentalized CBP E4 activity, we identified deleted in breast cancer 1 (DBC1) as a stoichiometric CBP-interacting protein that negatively regulates CBP-dependent p53 polyubiquitination, stabilizes p53, and augments p53-dependent apoptosis. TCGA analysis demonstrated that solid tumors often retain wild-type p53 alleles in conjunction with DBC1 loss, supporting the hypothesis that DBC1 is selected for disruption during carcinogenesis as a surrogate for p53 functional loss. Because DBC1 maintains p53 stability in the nucleus, where p53 exerts its tumor-suppressive transcriptional function, replacement of DBC1 functionality in DBC1-deleted tumors might enhance p53 function and chemosensitivity for therapeutic benefit. Akande et al. describe DBC1 as a negative regulator of CBP p53-directed ubiquitin ligase activity that maintains nuclear p53 stability. DBC1 loss may be selected in tumors to inactivate the p53 pathway. Because expression of DBC1 enhances p53-dependent apoptosis, restoration of DBC1 in null tumors may be of therapeutic value.
Collapse
Affiliation(s)
- Oluwatoyin E Akande
- Department of Internal Medicine, Virginia Commonwealth University (VCU), Richmond, VA, USA; VCU Massey Cancer Center, VCU, Richmond, VA, USA
| | - Priyadarshan K Damle
- Department of Internal Medicine, Virginia Commonwealth University (VCU), Richmond, VA, USA; VCU Massey Cancer Center, VCU, Richmond, VA, USA
| | | | - Nicholas E Sherman
- W.M. Keck Biomedical Mass Spectrometry Laboratory, University of Virginia, Charlottesville, VA, USA
| | - Barbara B Szomju
- Department of Internal Medicine, Virginia Commonwealth University (VCU), Richmond, VA, USA; VCU Massey Cancer Center, VCU, Richmond, VA, USA
| | - Larisa V Litovchick
- Department of Internal Medicine, Virginia Commonwealth University (VCU), Richmond, VA, USA; VCU Massey Cancer Center, VCU, Richmond, VA, USA
| | - Steven R Grossman
- Department of Internal Medicine, Virginia Commonwealth University (VCU), Richmond, VA, USA; VCU Massey Cancer Center, VCU, Richmond, VA, USA.
| |
Collapse
|
23
|
Liu J, Meng C, Li C, Tang K, Tang H, Liao J. Deleted in Breast Cancer 1 as a Novel Prognostic Biomarker for Digestive System Cancers: A Meta-Analysis. J Cancer 2019; 10:1633-1641. [PMID: 31205519 PMCID: PMC6548013 DOI: 10.7150/jca.26935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 01/19/2019] [Indexed: 11/06/2022] Open
Abstract
Deleted in Breast Cancer 1 (DBC1/CCAR2) is a regulatory protein involved in cell survival and cancer progression. Herein, we focused on summarizing the overall prognostic value of DBC1 for digestive system cancers. Therefore, we conducted a meta-analysis based on 9 studies with 2391 patients to generated combined hazard ratios (HR) or odds ratio (OR) with its 95% confidence intervals (CI) for overall survival (OS) and clinicopathological features. Positive DBC1 expression was significantly associated with poor OS of digestive system cancers (pooled HR=1.650, 95% CI=1.087-2.504, P<0.019). Stratified analysis also verified the potential prognostic prediction of DBC1 in some subgroups, such as digestive tract cancers (pooled HR=1.685, 95% CI=1.013-2.802, P=0.044), univariate analysis method (pooled HR=2.077, 95%CI=1.221-3.533, P=0.007), publication date within five years (pooled HR=1.609, 95%CI=1.097-2.358, P =0.015), study sample size smaller than 200 (pooled HR=2.304, 95%CI=1.716-3.093, P<0.001) and cutoff value for positive tumor cells more than 50% (pooled HR=1.944, 95% CI=1.479-2.556, P<0.001). Additionally, in terms of the association between DBC1 expression and clinicopathological characteristics, DBC1 expression was correlated to age (pooled OR=0.596, 95%CI =0.467-0.761, P<0.001), WHO classification (pooled OR =3.780, 95% CI=2.303-6.205, P <0.001), Lauren classification (pooled OR=2.000, 95%CI =1.492-2.680, P<0.001), and lymph node metastasis (pooled OR=0.405, 95%CI=0.203-0.806, P=0.010). In conclusion, DBC1 could not only be an independent prognostic factor for survival of patients with digestive system cancer, but might also be a novel target for cancer therapy.
Collapse
Affiliation(s)
- Jingting Liu
- Department of Emergency, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, Zhejiang, 310020, China
| | - Chunyan Meng
- Department of General Surgery, Zhejiang Hospital, 12 Lingyin Road, Zhejiang 310013, China
| | - Changcan Li
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine , 100 Haining Road, Shanghai 200080, China
| | - Kaifeng Tang
- Department of General Surgery, Zhejiang Hospital, 12 Lingyin Road, Zhejiang 310013, China
| | - Hongchao Tang
- Department of General Surgery, Zhejiang Hospital, 12 Lingyin Road, Zhejiang 310013, China
| | - Jianhua Liao
- Department of General Surgery, Zhejiang Hospital, 12 Lingyin Road, Zhejiang 310013, China
| |
Collapse
|
24
|
Sekhar SC, Venkatesh J, Cheriyan VT, Muthu M, Levi E, Assad H, Meister P, Undyala VV, Gauld JW, Rishi AK. A H2AX⁻CARP-1 Interaction Regulates Apoptosis Signaling Following DNA Damage. Cancers (Basel) 2019; 11:cancers11020221. [PMID: 30769864 PMCID: PMC6406907 DOI: 10.3390/cancers11020221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/07/2019] [Accepted: 02/10/2019] [Indexed: 12/16/2022] Open
Abstract
Cell Cycle and Apoptosis Regulatory Protein (CARP-1/CCAR1) is a peri-nuclear phosphoprotein that regulates apoptosis via chemotherapeutic Adriamycin (doxorubicin) and a novel class of CARP-1 functional mimetic (CFM) compounds. Although Adriamycin causes DNA damage, data from Comet assays revealed that CFM-4.16 also induced DNA damage. Phosphorylation of histone 2AX (γH2AX) protein is involved in regulating DNA damage repair and apoptosis signaling. Adriamycin or CFM-4.16 treatments inhibited cell growth and caused elevated CARP-1 and γH2AX in human breast (HBC) and cervical cancer (HeLa) cells. In fact, a robust nuclear or peri-nuclear co-localization of CARP-1 and γH2AX occurred in cells undergoing apoptosis. Knock-down of CARP-1 diminished γH2AX, their co-localization, and apoptosis in CFM-4.16- or Adriamycin-treated cells. We found that CARP-1 directly binds with H2AX, and H2AX interacted with CARP-1, but not CARP-1 (Δ600–652) mutant. Moreover, cells expressing CARP-1 (Δ600–652) mutant were resistant to apoptosis, and had diminished levels of γH2AX, when compared with cells expressing wild-type CARP-1. Mutagenesis studies revealed that H2AX residues 1–35 harbored a CARP-1-binding epitope, while CARP-1 amino acids 636–650 contained an H2AX-interacting epitope. Surface plasmon resonance studies revealed that CARP-1 (636–650) peptide bound with H2AX (1–35) peptide with a dissociation constant (Kd) of 127 nM. Cells expressing enhanced GFP (EGFP)-tagged H2AX (1–35) peptide or EGFP-tagged CARP-1 (636–650) peptide were resistant to inhibition by Adriamycin or CFM-4.16. Treatment of cells with transactivator of transcription (TAT)-tagged CARP-1 (636–650) peptide resulted in a moderate, statistically significant abrogation of Adriamycin-induced growth inhibition of cancer cells. Our studies provide evidence for requirement of CARP-1 interaction with H2AX in apoptosis signaling by Adriamycin and CFM compounds.
Collapse
Affiliation(s)
- Sreeja C Sekhar
- John D. Dingell Veterans Administration Medical Center, Detroit, MI 48201, USA.
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201, USA.
| | - Jaganathan Venkatesh
- John D. Dingell Veterans Administration Medical Center, Detroit, MI 48201, USA.
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201, USA.
| | - Vino T Cheriyan
- John D. Dingell Veterans Administration Medical Center, Detroit, MI 48201, USA.
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201, USA.
| | - Magesh Muthu
- John D. Dingell Veterans Administration Medical Center, Detroit, MI 48201, USA.
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201, USA.
| | - Edi Levi
- John D. Dingell Veterans Administration Medical Center, Detroit, MI 48201, USA.
| | - Hadeel Assad
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201, USA.
| | - Paul Meister
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada.
| | - Vishnu V Undyala
- Cardiovascular Research Institute, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| | - James W Gauld
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada.
| | - Arun K Rishi
- John D. Dingell Veterans Administration Medical Center, Detroit, MI 48201, USA.
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201, USA.
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI 48201, USA.
| |
Collapse
|
25
|
Li L, Feng R, Fei S, Cao J, Zhu Q, Ji G, Zhou J. NANOGP8 expression regulates gastric cancer cell progression by transactivating DBC1 in gastric cancer MKN-45 cells. Oncol Lett 2018; 17:555-563. [PMID: 30655801 DOI: 10.3892/ol.2018.9595] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 07/23/2018] [Indexed: 02/06/2023] Open
Abstract
NANOGP8 is one of the NANOG pseudogenes and is expressed together with NANOG in multiple tumor tissues and cell lines. The biological functions of NANOGP8 in progression of gastric cancer are unclear. In the present study, the role of NANOGP8 was investigated in gastric cancer cells. The gathered data demonstrated that NANOG expression in both mRNA and protein was elevated in gastric cancer cell lines relative to a normal gastric epithelial cell line. Downregulation of NANOGP8 inhibited cell proliferation and increased apoptosis in human gastric carcinoma cell lines. Furthermore, silencing of NANOGP8 suppressed tumor growth in vivo. Interestingly, it was identified that deleted in breast cancer 1 (DBC1) expression was also markedly downregulated following NANOGP8 knockdown. DNA microarray and dual-luciferase assays further indicated that NANOGP8 may bind to the DBC1 promoter region and regulate DBC1 expression. Therefore, the gathered data provided evidence that NANOGP8 contributes to progression of gastric cancer via DBC1 and may have potential translational significance.
Collapse
Affiliation(s)
- Li Li
- Department of Molecular Cell Biology and Toxicology, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.,Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Ru Feng
- Department of Geriatrics, Suqian People's Hospital Drum Tower Hospital Group, Suqian, Jiangsu 223800, P.R. China
| | - Sujuan Fei
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Jiang Cao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Qinqin Zhu
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Guozhong Ji
- Department of Medical Examination Center and Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Jianwei Zhou
- Department of Molecular Cell Biology and Toxicology, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
26
|
Sankunny M, Eng C. KLLN-mediated DNA damage-induced apoptosis is associated with regulation of p53 phosphorylation and acetylation in breast cancer cells. Cell Death Discov 2018; 4:31. [PMID: 30245854 PMCID: PMC6134104 DOI: 10.1038/s41420-018-0094-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/26/2018] [Accepted: 07/26/2018] [Indexed: 12/13/2022] Open
Abstract
KLLN is a target of p53 involved in S-phase cell cycle regulation deemed necessary and sufficient for p53-mediated apoptosis. Germline promoter hypermethylation of KLLN is associated with a cancer-predisposition syndrome, Cowden syndrome. KLLN’s DNA-binding ability is associated with transcription regulation and maintenance of genomic stability. Here, we report on KLLN’s role in DNA damage response (DDR) mediated through apoptosis in breast cells with and without a cancer phenotype. KLLN expression was upregulated after doxorubicin-induced DNA damage and this upregulation can be abrogated using RNAi-mediated gene silencing. Silencing KLLN after doxorubicin treatment effected DDR shown by decreased γ-H2AX foci and expression, and apoptosis assessed by decreased frequency of apoptotic nuclei and decreased expression of definitive markers of apoptosis. Contrary to expectations, there was no change in cell cycle regulation after KLLN silencing. These results were observed in breast cells with wildtype and mutant p53. At early timepoints after doxorubicin treatment, knocking down KLLN resulted in decreased Ser15-phosphorylation of p53 but not Thr68-phosphorylation of CHK2 or the phosphorylation of upstream regulators such as ATM and ATR. Interestingly, a second pathway for p53 activation was also affected by knockdown of KLLN. After doxorubicin treatment, Thr454-phosphorylation of DBC1, required to inhibit deacetylation of p53 by SIRT1, was decreased and therefore acetylation of p53 was also decreased with KLLN knockdown. Therefore, our observations suggest that KLLN’s role in DNA damage-induced apoptosis is likely independent of p53 and is associated with a two-pronged regulation of p53 activation.
Collapse
Affiliation(s)
- Madhav Sankunny
- 1Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195 USA.,2Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195 USA
| | - Charis Eng
- 1Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195 USA.,2Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195 USA.,3Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195 USA.,4Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106 USA.,5Germline High Risk Focus Group, CASE Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106 USA
| |
Collapse
|
27
|
TSPYL2 is a novel regulator of SIRT1 and p300 activity in response to DNA damage. Cell Death Differ 2018; 26:918-931. [PMID: 30050056 DOI: 10.1038/s41418-018-0168-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 06/13/2018] [Accepted: 07/04/2018] [Indexed: 02/05/2023] Open
Abstract
Protein acetylation and deacetylation events are finely regulated by lysine-acetyl-transferases and lysine-deacetylases and constitute an important tool for the activation or inhibition of specific cellular pathways. One of the most important lysine-acetyl-transferases is p300, which is involved in the regulation of gene expression, cell growth, DNA repair, differentiation, apoptosis, and tumorigenesis. A well-known target of p300 is constituted by the tumor suppressor protein p53, which plays a critical role in the maintenance of genomic stability and whose activity is known to be controlled by post-translational modifications, among which acetylation. p300 activity toward p53 is negatively regulated by the NAD-dependent deacetylase SIRT1, which deacetylates p53 preventing its transcriptional activation and the induction of p53-dependent apoptosis. However, the mechanisms responsible for p53 regulation by p300 and SIRT1 are still poorly understood. Here we identify the nucleosome assembly protein TSPY-Like 2 (TSPYL2, also known as TSPX, DENTT, and CDA1) as a novel regulator of SIRT1 and p300 function. We demonstrate that, upon DNA damage, TSPYL2 inhibits SIRT1, disrupting its association with target proteins, and promotes p300 acetylation and activation, finally stimulating p53 acetylation and p53-dependent cell death. Indeed, in response to DNA damage, cells silenced for TSPYL2 were found to be defective in p53 activation and apoptosis induction and these events were shown to be dependent on SIRT1 and p300 function. Collectively, our results shed new light on the regulation of p53 acetylation and activation and reveal a novel TSPYL2 function with important implications in cancerogenesis.
Collapse
|
28
|
Magni M, Buscemi G, Zannini L. Cell cycle and apoptosis regulator 2 at the interface between DNA damage response and cell physiology. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 776:1-9. [DOI: 10.1016/j.mrrev.2018.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 01/06/2023]
|
29
|
Li B, He X, Zhuang M, Niu B, Wu C, Mu H, Tang F, Cui Y, Liu W, Zhao B, Peng S, Li G, Hua J. Melatonin Ameliorates Busulfan-Induced Spermatogonial Stem Cell Oxidative Apoptosis in Mouse Testes. Antioxid Redox Signal 2018; 28:385-400. [PMID: 28027652 DOI: 10.1089/ars.2016.6792] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIMS Many men endure immunosuppressive or anticancer treatments that contain alkylating agents before the age of sexual maturity, especially the increasing number of preadolescent males who undergo busulfan treatment for myeloablative conditioning before hematopoietic stem cell transplantation. Before sperm production, there are no sperm available for cryopreservation. Thus, it is necessary to identify a solution to ameliorate the busulfan-induced damage of spermatogonial stem cells (SSCs). RESULTS In this study, we demonstrated that melatonin relieved the previously described SSC loss and apoptosis in mouse testes. Melatonin increased the expression of manganese superoxide dismutase (MnSOD), which regulated the production of busulfan-induced reactive oxygen species (ROS). Moreover, melatonin promoted sirtuin type 1 (SIRT1) expression. SIRT1 participated in the deacetylation of p53, which promotes p53 ubiquitin degradation. Decreased concentrations of deacetylated p53 resulted in spermatogonial cell resistance to apoptosis. Acute T cell leukemia cell assay demonstrated that melatonin does not affect busulfan-induced cancer cell apoptosis and ROS. INNOVATION The current evidence suggests that melatonin may alleviate the side effects of alkylating drugs, such as busulfan. CONCLUSION Melatonin promoted MnSOD and SIRT1 expression, which successfully ameliorated busulfan-induced SSC apoptosis caused by high concentrations of ROS and p53. Antioxid. Redox Signal. 28, 385-400.
Collapse
Affiliation(s)
- Bo Li
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Xin He
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Mengru Zhuang
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Bowen Niu
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Chongyang Wu
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Hailong Mu
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Furong Tang
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Yanhua Cui
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Weishuai Liu
- 3 Department of Pathology, Yangling Demonstration Zone Hospital , Yangling, Shaanxi, China
| | - Baoyu Zhao
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Sha Peng
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| | - Guangpeng Li
- 2 Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University , Hohhot, China
| | - Jinlian Hua
- 1 College of Veterinary Medicine, Shaanxi Centre of Stem Cell Engineering and Technology, Northwest A&F University , Yangling, Shaanxi, China
| |
Collapse
|
30
|
Kim W, Cheon MG, Kim JE. Mitochondrial CCAR2/DBC1 is required for cell survival against rotenone-induced mitochondrial stress. Biochem Biophys Res Commun 2017; 485:782-789. [PMID: 28254432 DOI: 10.1016/j.bbrc.2017.02.131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 02/26/2017] [Indexed: 11/24/2022]
Abstract
CCAR2 (cell cycle and apoptosis regulator protein 2; formerly DBC1, deleted in breast cancer 1) functions in diverse cellular processes including responses to genotoxic and metabolic stresses. However, its role in the mitochondrial stress response has not been fully elucidated. To investigate how CCAR2 regulates stress response, we purified CCAR2-containing complexes. Interestingly, the results revealed that CCAR2 localized to the mitochondria, and also bound Hsp60 (heat shock protein 60), a mitochondrial chaperone. The binding of CCAR2 to Hsp60 increased following rotenone-induced mitochondrial stress. The deficiencies in CCAR2 and Hsp60 also disrupted the mitochondrial membrane potential, thereby promoting apoptosis following mitochondrial stress. In summary, the CCAR2-Hsp60 complex promoted cell survival during mitochondrial stress-induced apoptosis. These data suggest that CCAR2 is critical for maintaining mitochondrial homeostasis in response to stress.
Collapse
Affiliation(s)
- Wootae Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Min Gyeong Cheon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja-Eun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmacology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
31
|
Restelli M, Magni M, Ruscica V, Pinciroli P, De Cecco L, Buscemi G, Delia D, Zannini L. A novel crosstalk between CCAR2 and AKT pathway in the regulation of cancer cell proliferation. Cell Death Dis 2016; 7:e2453. [PMID: 27809307 PMCID: PMC5260903 DOI: 10.1038/cddis.2016.359] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/29/2016] [Accepted: 10/04/2016] [Indexed: 02/06/2023]
Abstract
Human CCAR2 has recently emerged as having a pivotal role in the DNA damage response, promoting apoptosis and repair of heterochromatic DNA breaks. However, less is known about the function of CCAR2 in tumor formation and cancer progression. Here, we demonstrate, for the first time, that CCAR2 loss inhibits the proliferation of cancer cells, but preserves the growth of normal cells. Investigating the mechanisms responsible for this differential effect, we found that CCAR2 depletion specifically impairs the activation of AKT pathway in cancer cells, but not in normal cells, by reducing AKT phosphorylation on Ser473. This effect is achieved through the transcriptional upregulation of TRB3 gene and accumulation of TRB3 protein, which then binds to and inhibits the phosphorylation and activation of AKT. The defective activation of AKT finally results in reduced GSK3β phosphorylation, prevention of G1/S transition and inhibition of cancer cell growth. These results establish an important role for CCAR2 in cancer cells proliferation and could shed new light on novel therapeutic strategies against cancer, devoid of detrimental side effects.
Collapse
Affiliation(s)
- Michela Restelli
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Martina Magni
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Vincenzo Ruscica
- Department of Biochemistry, Max Planck Institute for Developmental Biology, Tubingen, Germany
| | - Patrizia Pinciroli
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Loris De Cecco
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Giacomo Buscemi
- Department of Biosciences, University of Milan, via Celoria 26, Milan 20133, Italy
| | - Domenico Delia
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| | - Laura Zannini
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
| |
Collapse
|
32
|
Best SA, Nwaobasi AN, Schmults CD, Ramsey MR. CCAR2 Is Required for Proliferation and Tumor Maintenance in Human Squamous Cell Carcinoma. J Invest Dermatol 2016; 137:506-512. [PMID: 27725203 DOI: 10.1016/j.jid.2016.09.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 09/21/2016] [Accepted: 09/26/2016] [Indexed: 02/09/2023]
Abstract
CCAR2 is a widely expressed protein involved in the regulation of a variety of transcriptional complexes. High expression of CCAR2 correlates with poor outcomes in many human tumor types such as squamous cell carcinoma (SCC). Paradoxically, loss of Ccar2 in the mouse results in an increased tumor burden, suggesting that CCAR2 may in fact function as a tumor suppressor. This tumor suppressor function is dependent on p53, a protein that is inactivated in the vast majority of SCC tumors, leaving the role of CCAR2 in p53-null tumors unclear. We sought to identify p53-independent CCAR2 functions in SCC and to examine its role in tumorigenesis. We found that CCAR2 is highly overexpressed in p53-deficient SCC cell lines compared with normal primary keratinocytes due to increased protein stability. We identify a role for CCAR2 in promoting the stability of the transcription factors RFX1 and CREB1, which are both required for proliferation. Finally, we show that CCAR2 is required for proliferation in vitro and in established SCC tumors in vivo. Our data suggest an important role for CCAR2 in maintaining cell cycle progression and promoting SCC tumorigenesis.
Collapse
Affiliation(s)
- Sarah A Best
- Brigham and Women's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Department of Dermatology, Boston, Massachusetts, USA
| | - Amy N Nwaobasi
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Chrysalyne D Schmults
- Brigham and Women's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Department of Dermatology, Boston, Massachusetts, USA
| | - Matthew R Ramsey
- Brigham and Women's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Department of Dermatology, Boston, Massachusetts, USA.
| |
Collapse
|
33
|
Ochodnicka-Mackovicova K, Bahjat M, Maas C, van der Veen A, Bloedjes TA, de Bruin AM, van Andel H, Schrader CE, Hendriks RW, Verhoeyen E, Bende RJ, van Noesel CJM, Guikema JEJ. The DNA Damage Response Regulates RAG1/2 Expression in Pre-B Cells through ATM-FOXO1 Signaling. THE JOURNAL OF IMMUNOLOGY 2016; 197:2918-29. [PMID: 27559048 DOI: 10.4049/jimmunol.1501989] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 07/20/2016] [Indexed: 01/01/2023]
Abstract
The recombination activating gene (RAG) 1 and RAG2 protein complex introduces DNA breaks at Tcr and Ig gene segments that are required for V(D)J recombination in developing lymphocytes. Proper regulation of RAG1/2 expression safeguards the ordered assembly of Ag receptors and the development of lymphocytes, while minimizing the risk for collateral damage. The ataxia telangiectasia mutated (ATM) kinase is involved in the repair of RAG1/2-mediated DNA breaks and prevents their propagation. The simultaneous occurrence of RAG1/2-dependent and -independent DNA breaks in developing lymphocytes exposed to genotoxic stress increases the risk for aberrant recombinations. In this study, we assessed the effect of genotoxic stress on RAG1/2 expression in pre-B cells and show that activation of the DNA damage response resulted in the rapid ATM-dependent downregulation of RAG1/2 mRNA and protein expression. We show that DNA damage led to the loss of FOXO1 binding to the enhancer region of the RAG1/2 locus (Erag) and provoked FOXO1 cleavage. We also show that DNA damage caused by RAG1/2 activity in pre-B cells was able to downmodulate RAG1/2 expression and activity, confirming the existence of a negative feedback regulatory mechanism. Our data suggest that pre-B cells are endowed with a protective mechanism that reduces the risk for aberrant recombinations and chromosomal translocations when exposed to DNA damage, involving the ATM-dependent regulation of FOXO1 binding to the Erag enhancer region.
Collapse
Affiliation(s)
- Katarina Ochodnicka-Mackovicova
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Mahnoush Bahjat
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Chiel Maas
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Amélie van der Veen
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Timon A Bloedjes
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Alexander M de Bruin
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Harmen van Andel
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Carol E Schrader
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, 3000 CA Rotterdam, the Netherlands
| | - Els Verhoeyen
- Centre International de Recherche en Infectiologie, Virus Enveloppés, Vecteurs et Réponses Innées Équipe, INSERM U1111, CNRS, UMR5308, Université de Lyon-1, École Normale Supérieure de Lyon, 69007 Lyon, France; and INSERM, U1065, Centre de Médecine Moléculaire, Équipe 3, 06204 Nice, France
| | - Richard J Bende
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Carel J M van Noesel
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Jeroen E J Guikema
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Lymphoma and Myeloma Center Amsterdam, 1105 AZ Amsterdam, the Netherlands;
| |
Collapse
|
34
|
López-Saavedra A, Gómez-Cabello D, Domínguez-Sánchez MS, Mejías-Navarro F, Fernández-Ávila MJ, Dinant C, Martínez-Macías MI, Bartek J, Huertas P. A genome-wide screening uncovers the role of CCAR2 as an antagonist of DNA end resection. Nat Commun 2016; 7:12364. [PMID: 27503537 PMCID: PMC4980490 DOI: 10.1038/ncomms12364] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/27/2016] [Indexed: 01/29/2023] Open
Abstract
There are two major and alternative pathways to repair DNA double-strand breaks: non-homologous end-joining and homologous recombination. Here we identify and characterize novel factors involved in choosing between these pathways; in this study we took advantage of the SeeSaw Reporter, in which the repair of double-strand breaks by homology-independent or -dependent mechanisms is distinguished by the accumulation of green or red fluorescence, respectively. Using a genome-wide human esiRNA (endoribonuclease-prepared siRNA) library, we isolate genes that control the recombination/end-joining ratio. Here we report that two distinct sets of genes are involved in the control of the balance between NHEJ and HR: those that are required to facilitate recombination and those that favour NHEJ. This last category includes CCAR2/DBC1, which we show inhibits recombination by limiting the initiation and the extent of DNA end resection, thereby acting as an antagonist of CtIP. A DNA double strand break can be repaired through either the non-homologous end-joining or the homologous recombination pathways. Here the authors conduct a genome-wide screen and identify a role for CCAR2 in pathway choice by regulating DNA end resection by CtIP.
Collapse
Affiliation(s)
- Ana López-Saavedra
- Departamento de Genética, Universidad de Sevilla, 41080 Sevilla, Spain.,Department of Regenerative Medicine, Centro Andaluz de Biología Molecular y Medicina Regenerativa, 41092 Sevilla, Spain
| | - Daniel Gómez-Cabello
- Department of Regenerative Medicine, Centro Andaluz de Biología Molecular y Medicina Regenerativa, 41092 Sevilla, Spain
| | - María Salud Domínguez-Sánchez
- Department of Regenerative Medicine, Centro Andaluz de Biología Molecular y Medicina Regenerativa, 41092 Sevilla, Spain
| | - Fernando Mejías-Navarro
- Departamento de Genética, Universidad de Sevilla, 41080 Sevilla, Spain.,Department of Regenerative Medicine, Centro Andaluz de Biología Molecular y Medicina Regenerativa, 41092 Sevilla, Spain
| | - María Jesús Fernández-Ávila
- Department of Regenerative Medicine, Centro Andaluz de Biología Molecular y Medicina Regenerativa, 41092 Sevilla, Spain
| | - Christoffel Dinant
- Genome Integrity Unit, Danish Cancer Society Research Centre, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - María Isabel Martínez-Macías
- Departamento de Genética, Universidad de Sevilla, 41080 Sevilla, Spain.,Department of Regenerative Medicine, Centro Andaluz de Biología Molecular y Medicina Regenerativa, 41092 Sevilla, Spain
| | - Jiri Bartek
- Genome Integrity Unit, Danish Cancer Society Research Centre, Strandboulevarden 49, 2100 Copenhagen, Denmark.,Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Pablo Huertas
- Departamento de Genética, Universidad de Sevilla, 41080 Sevilla, Spain.,Department of Regenerative Medicine, Centro Andaluz de Biología Molecular y Medicina Regenerativa, 41092 Sevilla, Spain
| |
Collapse
|
35
|
Subramaniyan B, Jagadeesan K, Ramakrishnan S, Mathan G. Targeting the interaction of Aurora kinases and SIRT1 mediated by Wnt signaling pathway in colorectal cancer: A critical review. Biomed Pharmacother 2016; 82:413-24. [DOI: 10.1016/j.biopha.2016.05.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 05/18/2016] [Accepted: 05/18/2016] [Indexed: 12/22/2022] Open
|
36
|
Acetylation at lysine 71 inactivates superoxide dismutase 1 and sensitizes cancer cells to genotoxic agents. Oncotarget 2016; 6:20578-91. [PMID: 26008972 PMCID: PMC4653027 DOI: 10.18632/oncotarget.3987] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/10/2015] [Indexed: 01/13/2023] Open
Abstract
Cancer cells are characterized by a high dependency on antioxidant enzymes to cope with the elevated rates of reactive oxygen species (ROS). Impairing antioxidant capacity in cancer cells disturbs the ROS homeostasis and exposes cancer cells to massive oxidative stress. In this study, we have discovered that superoxide dismutase 1 (SOD1), a major player in maintaining the cellular redox status, was acetylated at lysine 71. This acetylation, which was primarily deacetylated by Sirtuin 1 (SIRT1), suppressed the enzymatic activity of SOD1 via disrupting its association with copper chaperone for SOD1 (CCS). More importantly, genotoxic agents, such as camptothecin (CPT), induced SOD1 acetylation by disrupting its binding with SIRT1. CPT-induced SOD1 acetylation was stimulated by its provoked ROS, suggesting a positive feedback loop, in which ROS per se impairs the antioxidative defence of cancer cells and reinforces oxidative stress stimulated by anticancer agents. The intrinsic abundance of SOD1 acetylation varied among cancer cells, and high level of SOD1 acetylation was correlated with elevated sensitivity to CPT. Together, our findings gained mechanistic insights into how cytotoxic agents fine tune the intracellular ROS homeostasis to strengthen their anticancer effects, and suggested SOD1 acetylation as a candidate biomarker for predicting response to CPT-based chemotherapy.
Collapse
|
37
|
Magni M, Ruscica V, Restelli M, Fontanella E, Buscemi G, Zannini L. CCAR2/DBC1 is required for Chk2-dependent KAP1 phosphorylation and repair of DNA damage. Oncotarget 2016; 6:17817-31. [PMID: 26158765 PMCID: PMC4627348 DOI: 10.18632/oncotarget.4417] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 05/29/2015] [Indexed: 01/26/2023] Open
Abstract
Cell cycle and apoptosis regulator 2 (CCAR2, formerly known as DBC1) is a nuclear protein largely involved in DNA damage response, apoptosis, metabolism, chromatin structure and transcription regulation. Upon DNA lesions, CCAR2 is phosphorylated by the apical kinases ATM/ATR and this phosphorylation enhances CCAR2 binding to SIRT1, leading to SIRT1 inhibition, p53 acetylation and p53-dependent apoptosis. Recently, we found that also the checkpoint kinase Chk2 and the proteasome activator REGγ are required for efficient CCAR2-mediated inhibition of SIRT1 and induction of p53-dependent apoptosis. Here, we report that CCAR2 is required for the repair of heterochromatic DNA lesions, as cells knock-out for CCAR2 retain, at late time-points after genotoxic treatment, abnormal levels of DNA damage-associated nuclear foci, whose timely resolution is reinstated by HP1β depletion. Conversely, repair of DNA damages in euchromatin are not affected by CCAR2 absence. We also report that the impairment in heterochromatic DNA repair is caused by defective Chk2 activation, detectable in CCAR2 ablated cells, which finally impacts on the phosphorylation of the Chk2 substrate KAP1 that is required for the induction of heterochromatin relaxation and DNA repair. These studies further extend and confirm the role of CCAR2 in the DNA damage response and DNA repair and illustrate a new mechanism of Chk2 activity regulation. Moreover, the involvement of CCAR2 in the repair of heterochromatic DNA breaks suggests a new role for this protein in the maintenance of chromosomal stability, which is necessary to prevent cancer formation.
Collapse
Affiliation(s)
- Martina Magni
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Vincenzo Ruscica
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Current address: Max Planck Institute for Developmental Biology, Tubingen, Germany
| | - Michela Restelli
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Enrico Fontanella
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giacomo Buscemi
- Department of Biosciences, University of Milan, Milan, Italy
| | - Laura Zannini
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
38
|
San Jose-Eneriz E, Agirre X, Rodríguez-Otero P, Prosper F. Epigenetic regulation of cell signaling pathways in acute lymphoblastic leukemia. Epigenomics 2016; 5:525-38. [PMID: 24059799 DOI: 10.2217/epi.13.56] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a heterogeneous cancer that is characterized by rapid and uncontrolled proliferation of immature B- or T-lymphoid precursors. Although ALL has been regarded as a genetic disease for many years, the crucial importance of epigenetic alterations in leukemogenesis has become increasingly evident. Epigenetic mechanisms, which include DNA methylation and histone modifications, are critical for gene regulation during many key biological processes. Here, we review the cell signaling pathways that are regulated by DNA methylation or histone modifications in ALL. Recent studies have highlighted the fundamental role of these modifications in ALL development, and suggested that future investigation into the specific genes and pathways that are altered by epigenetic mechanisms can contribute to the development of novel drug-based therapies for ALL.
Collapse
Affiliation(s)
- Edurne San Jose-Eneriz
- Oncology Division, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | | | | | |
Collapse
|
39
|
Abstract
The nutrient demands of cancer cannot be met by normal cell metabolism. Cancer cells undergo dramatic alteration of metabolic pathways in a process called reprogramming, characterized by increased nutrient uptake and re-purposing of these fuels for biosynthetic, bioenergetic or signaling pathways. Partitioning carbon sources toward growth and away from ATP production necessitates other means of generating energy for biosynthetic reactions. Additionally, cancer cell adaptations frequently lead to increased production of reactive oxygen species and lactic acid, which can be beneficial to cancer growth but also are potentially toxic and must be appropriately cleared. Sirtuins are a family of deacylases and ADP-ribosyltransferases with clear links to regulation of cancer metabolism. Through their unique ability to integrate cellular stress and nutrient status with coordination of metabolic outputs, sirtuins are well poised to play pivotal roles in tumor progression and survival. Here, we review the multi-faceted duties of sirtuins in tackling the metabolic hurdles in cancer. We focus on both beneficial and adverse effects of sirtuins in the regulation of energetic, biosynthetic and toxicity barriers faced by cancer cells.
Collapse
Affiliation(s)
- Natalie J German
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| | - Marcia C Haigis
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
40
|
SENP1-Mediated Desumoylation of DBC1 Inhibits Apoptosis Induced by High Glucose in Bovine Retinal Pericytes. J Ophthalmol 2016; 2016:6392658. [PMID: 27110392 PMCID: PMC4826714 DOI: 10.1155/2016/6392658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/23/2016] [Accepted: 03/02/2016] [Indexed: 12/17/2022] Open
Abstract
Pericyte loss is an early characteristic change in diabetic retinopathy, but its precise molecular mechanisms have not been elucidated. This study investigated the role of SENP1 in pericyte loss in diabetic retinopathy. We demonstrated that a high concentration of glucose inhibited the expression of the Sentrin/SUMO-specific protease 1 (SENP1), which resulted in an increase in DBC1 sumoylation in bovine retinal pericytes (BRPCs). Furthermore, SENP1 overexpression attenuated hyperemia-induced apoptosis of BPRCs, and SENP1 knockdown aggravated this effect. We also provide evidence that DBC1 sumoylation/desumoylation is involved in the SENP1-regulated apoptosis of BRPCs under high glucose conditions. Understanding the role of SENP1 in the pathogenesis of high glucose induced pericyte loss could help elucidate important targets for future pharmacological interventions.
Collapse
|
41
|
YU DENGFENG, JIANG SUJUAN, PAN ZHIPENG, CHENG WEIDONG, ZHANG WENJUN, YAO XIAOKUN, LI YUCHENG, LUN YONGZHI. Expression and clinical significance of Sirt1 in colorectal cancer. Oncol Lett 2016; 11:1167-1172. [PMID: 26893713 PMCID: PMC4738140 DOI: 10.3892/ol.2015.3982] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 10/06/2015] [Indexed: 01/16/2023] Open
Abstract
The objective of the present study was to examine the expression of Silent information regulator 1 (Sirt1) in colorectal cancer and peritumoral normal mucosa tissue, and therefore analyze the role and molecular mechanism of Sirt1 in the pathogenesis of colorectal cancer. Colorectal cancer tissue specimens were employed as the experimental group, and adjacent normal mucosa tissues >5 cm from tumor lesions were used as the control group. The expression of Sirt1 was detected by the immunohistochemical streptavidin peroxidase detection method in paraffin-embedded sections, whilst Sirt1 protein expression was examined by western blot analysis in the fresh tissues. Sirt1 protein was primarily expressed in the nuclei of the tumor cells, and positive staining was brownish-yellow in color. The relative expression quantities of Sirt1 in the peritumoral normal rectal mucosa and rectal carcinoma were 1.15 and 2.62, and the differences between the two groups were statistically significant (P<0.05). The expression level of Sirt1 in colorectal carcinoma was significantly associated with the depth of tumor invasion, differentiation and tumor size (P<0.05). Sirt1 expression was also found to be associated with tumor tissue type, lymph node metastasis, Duke's stage and patient age. These characteristics combined may therefore be used as markers for the early diagnosis of colorectal cancer pathogenesis.
Collapse
Affiliation(s)
- DENG-FENG YU
- Liaoning Provincial University Key Laboratory of Biophysics, College of Medicine, Dalian University, Dalian, Liaoning 116622, P.R. China
- Department of Anorectal Surgery, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning 116021, P.R. China
| | - SU-JUAN JIANG
- Liaoning Provincial University Key Laboratory of Biophysics, College of Medicine, Dalian University, Dalian, Liaoning 116622, P.R. China
- Department of Gynecology and Obstetrics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - ZHI-PENG PAN
- Liaoning Provincial University Key Laboratory of Biophysics, College of Medicine, Dalian University, Dalian, Liaoning 116622, P.R. China
| | - WEI-DONG CHENG
- Department of Anorectal Surgery, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning 116021, P.R. China
| | - WEN-JUN ZHANG
- Department of Anorectal Surgery, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning 116021, P.R. China
| | - XIAO-KUN YAO
- Liaoning Provincial University Key Laboratory of Biophysics, College of Medicine, Dalian University, Dalian, Liaoning 116622, P.R. China
| | - YU-CHENG LI
- Department of Dermatology, Yuzhou People's Hospital, Xuchang, Henan 461670, P.R. China
| | - YONG-ZHI LUN
- Liaoning Provincial University Key Laboratory of Biophysics, College of Medicine, Dalian University, Dalian, Liaoning 116622, P.R. China
| |
Collapse
|
42
|
Buscemi G, Ricci C, Zannini L, Fontanella E, Plevani P, Delia D. Bimodal regulation of p21(waf1) protein as function of DNA damage levels. Cell Cycle 2015; 13:2901-12. [PMID: 25486478 PMCID: PMC4615108 DOI: 10.4161/15384101.2014.946852] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Human p21Waf1 protein is well known for being transcriptionally induced by p53 and activating the cell cycle checkpoint arrest in response to DNA breaks. Here we report that p21Waf1 protein undergoes a bimodal regulation, being upregulated in response to low doses of DNA damage but rapidly and transiently degraded in response to high doses of DNA lesions. Responsible for this degradation is the checkpoint kinase Chk1, which phosphorylates p21Waf1 on T145 and S146 residues and induces its proteasome-dependent proteolysis. The initial p21Waf1 degradation is then counteracted by the ATM-Chk2 pathway, which promotes the p53-dependent accumulation of p21Waf1 at any dose of damage. We also found that p21Waf1 ablation favors the activation of an apoptotic program to eliminate otherwise irreparable cells. These findings support a model in which in human cells a balance between ATM-Chk2-p53 and the ATR-Chk1 pathways modulates p21Waf1 protein levels in relation to cytostatic and cytotoxic doses of DNA damage.
Collapse
Affiliation(s)
- G Buscemi
- a Department of Experimental Oncology; Fondazione IRCCS Istituto Nazionale dei Tumori ; Milan , Italy
| | | | | | | | | | | |
Collapse
|
43
|
DBC1/CCAR2 is involved in the stabilization of androgen receptor and the progression of osteosarcoma. Sci Rep 2015; 5:13144. [PMID: 26249023 PMCID: PMC4642542 DOI: 10.1038/srep13144] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/21/2015] [Indexed: 02/07/2023] Open
Abstract
Deleted in breast cancer 1 (DBC1/CCAR2) is a protein of interest because of its diverse roles in tumorigenesis and its possible role as an androgen receptor (AR) co-activator. However, there are limited studies on the role of DBC1 in osteosarcoma. Therefore, we investigated the role of DBC1 and AR and their relationship in osteosarcoma. Immunohistochemical expression of DBC1 and AR was significantly associated with higher clinical stage and higher histologic grade, and predicted shorter survival. Especially, DBC1 expression was an independent prognostic indicator of overall survival (p = 0.005) and relapse-free survival (p = 0.004) by multivariate analysis. In osteosarcoma cell lines, U2OS and SaOS2, the knock down of DBC1 and AR with siRNA significantly reduced cellular proliferation and inhibited proliferation-related signaling. In addition, the knock down of DBC1 and AR decreased the invasion activity and inhibited invasion-related signaling of osteosarcoma cells. Interestingly, DBC1 affects the stabilization of AR protein via a mechanism involving the ubiquitination of AR. Proteosome-mediated degradation and poly-ubiquitination of AR were increased with the knock-down of DBC1. In conclusion, this study has shown that DBC1 is involved in the stabilization of AR protein and DBC1-AR pathways might be involved in the progression of osteosarcoma.
Collapse
|
44
|
Lee J, Adelmant G, Marto JA, Lee DH. Dephosphorylation of DBC1 by Protein Phosphatase 4 Is Important for p53-Mediated Cellular Functions. Mol Cells 2015; 38:697-704. [PMID: 26194823 PMCID: PMC4546941 DOI: 10.14348/molcells.2015.0066] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/30/2015] [Accepted: 06/08/2015] [Indexed: 12/17/2022] Open
Abstract
Deleted in breast cancer-1 (DBC1) contributes to the regulation of cell survival and apoptosis. Recent studies demonstrated that DBC is phosphorylated at Thr454 by ATM/ATR kinases in response to DNA damage, which is a critical event for p53 activation and apoptosis. However, how DBC1 phosphorylation is regulated has not been studied. Here we show that protein phosphatase 4 (PP4) dephosphorylates DBC1, regulating its role in DNA damage response. PP4R2, a regulatory subunit of PP4, mediates the interaction between DBC1 and PP4C, a catalytic subunit. PP4C efficiently dephosphorylates pThr454 on DBC1 in vitro, and the depletion of PP4C/PP4R2 in cells alters the kinetics of DBC1 phosphorylation and p53 activation, and increases apoptosis in response to DNA damage, which are compatible with the expression of the phosphomimetic DBC-1 mutant (T454E). These suggest that the PP4-mediated dephosphorylation of DBC1 is necessary for efficient damage responses in cells.
Collapse
Affiliation(s)
- Jihye Lee
- Department of Biological Sciences, College of Science, Chonnam National University, Gwangju 500-757,
Korea
| | | | | | - Dong-Hyun Lee
- Department of Biological Sciences, College of Science, Chonnam National University, Gwangju 500-757,
Korea
| |
Collapse
|
45
|
HIPK2 restricts SIRT1 activity upon severe DNA damage by a phosphorylation-controlled mechanism. Cell Death Differ 2015; 23:110-22. [PMID: 26113041 PMCID: PMC4815982 DOI: 10.1038/cdd.2015.75] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 01/23/2023] Open
Abstract
Upon severe DNA damage a cellular signalling network initiates a cell death response through activating tumour suppressor p53 in association with promyelocytic leukaemia (PML) nuclear bodies. The deacetylase Sirtuin 1 (SIRT1) suppresses cell death after DNA damage by antagonizing p53 acetylation. To facilitate efficient p53 acetylation, SIRT1 function needs to be restricted. How SIRT1 activity is regulated under these conditions remains largely unclear. Here we provide evidence that SIRT1 activity is limited upon severe DNA damage through phosphorylation by the DNA damage-responsive kinase HIPK2. We found that DNA damage provokes interaction of SIRT1 and HIPK2, which phosphorylates SIRT1 at Serine 682 upon lethal damage. Furthermore, upon DNA damage SIRT1 and HIPK2 colocalize at PML nuclear bodies, and PML depletion abrogates DNA damage-induced SIRT1 Ser682 phosphorylation. We show that Ser682 phosphorylation inhibits SIRT1 activity and impacts on p53 acetylation, apoptotic p53 target gene expression and cell death. Mechanistically, we found that DNA damage-induced SIRT1 Ser682 phosphorylation provokes disruption of the complex between SIRT1 and its activator AROS. Our findings indicate that phosphorylation-dependent restriction of SIRT1 activity by HIPK2 shapes the p53 response.
Collapse
|
46
|
DBC1 promotes anoikis resistance of gastric cancer cells by regulating NF-κB activity. Oncol Rep 2015; 34:843-9. [PMID: 26035299 DOI: 10.3892/or.2015.4007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/06/2015] [Indexed: 11/05/2022] Open
Abstract
Deleted in breast cancer 1 (DBC1) has been known to be overexpressed and serves as a poor prognostic indicator of several human cancers. In this study, we examined DBC1 expression in a total of 142 gastric cancer tissues by immunohistochemistry and revealed that DBC1 was overexpressed in gastric cancer and significantly associated with TNM stage and lymph node metastasis. The in vitro experiments showed that DBC1 expression correlated with the ability of anoikis resistance in gastric cancer cells, which has been defined as critical to metastasis. Furthermore, the results showed that the IKK-β/NF-κB signaling pathway was involved in the regulation of anoikis resistance by DBC1 in gastric cancer cells. Taken together, the results indicated that DBC1 promotes anoikis resistance in gastric cancer cells by regulating NF-κB activity and may thus be a new therapeutic target for preventing potential metastasis.
Collapse
|
47
|
Han B, Wang TD, Shen SM, Yu Y, Mao C, Yao ZJ, Wang LS. Annonaceous acetogenin mimic AA005 induces cancer cell death via apoptosis inducing factor through a caspase-3-independent mechanism. BMC Cancer 2015; 15:139. [PMID: 25885900 PMCID: PMC4379763 DOI: 10.1186/s12885-015-1133-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/24/2015] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Annonaceous acetogenins are a family of natural products with antitumor activities. Annonaceous acetogenin mimic AA005 reportedly inhibits mammalian mitochondrial NADH-ubiquinone reductase (Complex I) and induces gastric cancer cell death. However, the mechanisms underlying its cell-death-inducing activity are unclear. METHODS We used SW620 colorectal adenocarcinoma cells to study AA005 cytotoxic activity. Cell deaths were determined by Trypan blue assay and flow cytometry, and related proteins were characterized by western blot. Immunofluorescence and subcellular fractionation were used to evaluate AIF nuclear translocation. Reactive oxygen species were assessed by using redox-sensitive dye DCFDA. RESULTS AA005 induces a unique type of cell death in colorectal adenocarcinoma cells, characterized by lack of caspase-3 activation or apoptotic body formation, sensitivity to poly (ADP-ribose) polymerase inhibitor Olaparib (AZD2281) but not pan-caspase inhibitor Z-VAD.fmk, and dependence on apoptosis-inducing factor (AIF). AA005 treatment also reduced expression of mitochondrial Complex I components, and leads to accumulation of intracellular reactive oxygen species (ROS) at the early stage. Blocking ROS formation significantly suppresses AA005-induced cell death in SW620 cells. Moreover, blocking activation of RIP-1 by necroptosis inhibitor necrotatin-1 inhibits AIF translocation and partially suppresses AA005-induced cell death in SW620 cells demonstrating that RIP-1 protein may be essential for cell death. CONCLUSIONS AA005 may trigger the cell death via mediated by AIF through caspase-3 independent pathway. Our work provided new mechanisms for AA005-induced cancer cell death and novel clues for cancer treatment via AIF dependent cell death.
Collapse
Affiliation(s)
- Bing Han
- Center for Molecular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Tong-Dan Wang
- Center for Molecular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Shao-Ming Shen
- Center for Molecular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yun Yu
- Center for Molecular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chan Mao
- State Key Laboratory of Coordination Chemistry, Institute of Chemical Biology and Drug Innovation, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, P. R. China.
| | - Zhu-Jun Yao
- State Key Laboratory of Coordination Chemistry, Institute of Chemical Biology and Drug Innovation, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, P. R. China. .,Shanghai Universities E-Institute for Chemical Biology, Shanghai, 200025, China.
| | - Li-Shun Wang
- Center for Molecular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Universities E-Institute for Chemical Biology, Shanghai, 200025, China.
| |
Collapse
|
48
|
Magni M, Ruscica V, Buscemi G, Kim JE, Nachimuthu BT, Fontanella E, Delia D, Zannini L. Chk2 and REGγ-dependent DBC1 regulation in DNA damage induced apoptosis. Nucleic Acids Res 2014; 42:13150-60. [PMID: 25361978 PMCID: PMC4245943 DOI: 10.1093/nar/gku1065] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 10/15/2014] [Indexed: 01/08/2023] Open
Abstract
Human DBC1 (Deleted in Breast Cancer 1; KIAA1967; CCAR2) is a protein implicated in the regulation of apoptosis, transcription and histone modifications. Upon DNA damage, DBC1 is phosphorylated by ATM/ATR on Thr454 and this modification increases its inhibitory interaction with SIRT1, leading to p53 acetylation and p53-dependent apoptosis. Here, we report that the inhibition of SIRT1 by DBC1 in the DNA damage response (DDR) also depends on Chk2, the transducer kinase that is activated by ATM upon DNA lesions and contributes to the spreading of DNA damage signal. Indeed we found that inactivation of Chk2 reduces DBC1-SIRT1 binding, thus preventing p53 acetylation and DBC1-induced apoptosis. These events are mediated by Chk2 phosphorylation of the 11S proteasome activator REGγ on Ser247, which increases REGγ-DBC1 interaction and SIRT1 inhibition. Overall our results clarify the mechanisms underlying the DBC1-dependent SIRT1 inhibition and link, for the first time, Chk2 and REGγ to the ATM-DBC1-SIRT1 axis.
Collapse
Affiliation(s)
- Martina Magni
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Vincenzo Ruscica
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Giacomo Buscemi
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | - Ja-Eun Kim
- Department of Pharmacology, School of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | - Enrico Fontanella
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Domenico Delia
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Laura Zannini
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| |
Collapse
|
49
|
Park JH, Lee SW, Yang SW, Yoo HM, Park JM, Seong MW, Ka SH, Oh KH, Jeon YJ, Chung CH. Modification of DBC1 by SUMO2/3 is crucial for p53-mediated apoptosis in response to DNA damage. Nat Commun 2014; 5:5483. [DOI: 10.1038/ncomms6483] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 10/06/2014] [Indexed: 12/14/2022] Open
|
50
|
Liu T, Lin YH, Leng W, Jung SY, Zhang H, Deng M, Evans D, Li Y, Luo K, Qin B, Qin J, Yuan J, Lou Z. A divergent role of the SIRT1-TopBP1 axis in regulating metabolic checkpoint and DNA damage checkpoint. Mol Cell 2014; 56:681-95. [PMID: 25454945 DOI: 10.1016/j.molcel.2014.10.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 07/02/2014] [Accepted: 10/06/2014] [Indexed: 11/16/2022]
Abstract
DNA replication is executed only when cells have sufficient metabolic resources and undamaged DNA. Nutrient limitation and DNA damage cause a metabolic checkpoint and DNA damage checkpoint, respectively. Although SIRT1 activity is regulated by metabolic stress and DNA damage, its function in these stress-mediated checkpoints remains elusive. Here we report that the SIRT1-TopBP1 axis functions as a switch for both checkpoints. With glucose deprivation, SIRT1 is activated and deacetylates TopBP1, resulting in TopBP1-Treslin disassociation and DNA replication inhibition. Conversely, SIRT1 activity is inhibited under genotoxic stress, resulting in increased TopBP1 acetylation that is important for the TopBP1-Rad9 interaction and activation of the ATR-Chk1 pathway. Mechanistically, we showed that acetylation of TopBP1 changes the conformation of TopBP1, thereby facilitating its interaction with distinct partners in DNA replication and checkpoint activation. Taken together, our studies identify the SIRT1-TopBP1 axis as a key signaling mode in the regulation of the metabolic checkpoint and the DNA damage checkpoint.
Collapse
Affiliation(s)
- Tongzheng Liu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yi-Hui Lin
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Wenchuan Leng
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sung Yun Jung
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haoxing Zhang
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Min Deng
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Debra Evans
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yunhui Li
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Kuntian Luo
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Bo Qin
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jun Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jian Yuan
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Zhenkun Lou
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|