1
|
Liu Y, Luo D, Lu Y, Tan L. E2F transcription factor 1 as a potential prognostic biomarker and promotes tumor proliferation in skin cutaneous melanoma. Pathol Res Pract 2025; 269:155875. [PMID: 40023140 DOI: 10.1016/j.prp.2025.155875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/08/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Cutaneous melanoma (SKCM) is a highly aggressive malignancy with an overall poor prognosis. The expression of E2F transcription factor 1 in SKCM tissues was analyzed using the data acquired from the Cancer Genome Atlas (TCGA)and Gene Expression Omnibus (GEO)databases. The expression of E2F1 was also analyzed in human biopsies using immunohistology, and its expression was detected in SKCM cell lines using qPCR and WB technology. The prognostic value of E2F transcription factor 1 in SKCM was investigated using Kaplan-Meier survival and Cox analysis. The Tumor Immunization Estimation Resource (TIMER) was used to analyze the invasion of immune cells and associated markers of immune cells. The prognostic value of E2F transcription factor 1 DNA methylation levels for each CpG was analyzed by means of the MethSurv. The results were verified in the Human Protein Atlas (HPA) database. Individuals with elevated E2F transcription factor 1 expression had a worse prognosis (HR=1.43, p = 0.01). Using the expression level of E2F transcription factor 1, a reliable distinction between tumor and normal tissues can be made (Area Under the Curve =0.949). Moreover, immune infiltrations analysis showed that the mRNA expression levels and somatic copy number alterations (SCNA) in E2F transcription factor 1 were significantly correlated with recruitment of several immune cells. Our study showed that overexpression of E2F transcription factor 1 was significantly associated with poor prognosis and malignant phenotype of melanoma cells in SKCM patients.
Collapse
Affiliation(s)
- Ying Liu
- Plastic surgery Department, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing 100038, China
| | - Dan Luo
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yuangang Lu
- Department of Plastic and Cosmetic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China. https://twitter.com/
| | - Liuchang Tan
- Department of Plastic and Cosmetic Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China.
| |
Collapse
|
2
|
Chembukavu SN, Lindsay AJ. Therapy-induced senescence in breast cancer: an overview. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:902-920. [PMID: 39280248 PMCID: PMC11390292 DOI: 10.37349/etat.2024.00254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/06/2024] [Indexed: 09/18/2024] Open
Abstract
Outcomes for women with breast cancer have improved dramatically in recent decades. However, many patients present with intrinsic drug resistance and others are initially sensitive to anti-cancer drugs but acquire resistance during the course of their treatment, leading to recurrence and/or metastasis. Drug therapy-induced senescence (TIS) is a form of drug resistance characterised by the induction of cell cycle arrest and the emergence of a senescence-associated secretory phenotype (SASP) that can develop in response to chemo- and targeted- therapies. A wide range of anticancer interventions can lead to cell cycle arrest and SASP induction, by inducing genotoxic stress, hyperactivation of signalling pathways or oxidative stress. TIS can be anti-tumorigenic in the short-term, but pro-tumorigenic in the long-term by creating a pro-inflammatory and immunosuppressive microenvironment. Moreover, the SASP can promote angiogenesis and epithelial-mesenchymal transition in neighbouring cells. In this review, we will describe the characteristics of TIS in breast cancer and detail the changes in phenotype that accompany its induction. We also discuss strategies for targeting senescent cancer cells in order to prevent or delay tumour recurrence.
Collapse
Affiliation(s)
- Suraj Narayanan Chembukavu
- Membrane Trafficking and Disease Laboratory, School of Biochemistry & Cell Biology, Biosciences Institute, University College Cork, Cork, T12 YT20, Ireland
| | - Andrew J Lindsay
- Membrane Trafficking and Disease Laboratory, School of Biochemistry & Cell Biology, Biosciences Institute, University College Cork, Cork, T12 YT20, Ireland
| |
Collapse
|
3
|
Inhibitor of Growth Factors Regulate Cellular Senescence. Cancers (Basel) 2022; 14:cancers14133107. [PMID: 35804879 PMCID: PMC9264871 DOI: 10.3390/cancers14133107] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Five members of the Inhibitor of Growth (ING) family share a highly conserved plant homeodomian with affinity to the specific histone modification H3K4me3. Since some ING family members are preferentially associated with histone acetyltransferaseactivity while other members with histone deacetlyse activity, the ING family membres are epigenetic regulators. Interestingly, ING members can regulate the induction cellular senescence in both primray untransformed human cells as well as human cancer cells. We discuss here the up-to-date knowledge about their regulatory activity within the cellular senescent program. Abstract The Inhibitor of Growth (ING) proteins are a group of tumor suppressors with five conserved genes. A common motif of ING factors is the conserved plant homeodomain (PHD), with which they bind to chromatin as readers of the histone mark trimethylated histone H3 (H3K4me3). These genes often produce several protein products through alternative splicing events. Interestingly, ING1 and ING2 participate in the establishment of the repressive mSIN3a-HDAC complexes, whereas ING3, ING4, and ING5 are associated with the activating HAT protein complexes. In addition to the modulation of chromatin’s structure, they regulate cell cycle transition, cellular senescence, repair of DNA damage, apoptosis, and angiogenic pathways. They also have fundamental effects on regulating cellular senescence in cancer cells. In the current review, we explain their role in cellular senescence based on the evidence obtained from cell line and animal studies, particularly in the context of cancer.
Collapse
|
4
|
Shin EY, Soung NK, Schwartz MA, Kim EG. Altered endocytosis in cellular senescence. Ageing Res Rev 2021; 68:101332. [PMID: 33753287 PMCID: PMC8131247 DOI: 10.1016/j.arr.2021.101332] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/02/2021] [Accepted: 03/15/2021] [Indexed: 01/10/2023]
Abstract
Cellular senescence occurs in response to diverse stresses (e.g., telomere shortening, DNA damage, oxidative stress, oncogene activation). A growing body of evidence indicates that alterations in multiple components of endocytic pathways contribute to cellular senescence. Clathrin-mediated endocytosis (CME) and caveolae-mediated endocytosis (CavME) represent major types of endocytosis that are implicated in senescence. More recent research has also identified a chromatin modifier and tumor suppressor that contributes to the induction of senescence via altered endocytosis. Here, molecular regulators of aberrant endocytosis-induced senescence are reviewed and discussed in the context of their capacity to serve as senescence-inducing stressors or modifiers.
Collapse
Affiliation(s)
- Eun-Young Shin
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju, 28644, South Korea
| | - Nak-Kyun Soung
- World Class Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang-eup, Cheongju, 28116, South Korea
| | - Martin Alexander Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, And Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06511, USA; Wellcome Trust Centre for Cell-matrix Research, University of Manchester, Manchester, UK.
| | - Eung-Gook Kim
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju, 28644, South Korea.
| |
Collapse
|
5
|
Wanner E, Thoppil H, Riabowol K. Senescence and Apoptosis: Architects of Mammalian Development. Front Cell Dev Biol 2021; 8:620089. [PMID: 33537310 PMCID: PMC7848110 DOI: 10.3389/fcell.2020.620089] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
Mammalian development involves an exquisite choreography of cell division, differentiation, locomotion, programmed cell death, and senescence that directs the transformation of a single cell zygote to a mature organism containing on the order of 40 trillion cells in humans. How a single totipotent zygote undergoes the rapid stages of embryonic development to form over 200 different cell types is complex in the extreme and remains the focus of active research. Processes such as programmed cell death or apoptosis has long been known to occur during development to help sculpt organs and tissue systems. Other processes such as cellular senescence, long thought to only occur in pathologic states such as aging and tumorigenesis have been recently reported to play a vital role in development. In this review, we focus on apoptosis and senescence; the former as an integral mechanism that plays a critical role not only in mature organisms, but that is also essential in shaping mammalian development. The latter as a well-defined feature of aging for which some reports indicate a function in development. We will dissect the dual roles of major gene families, pathways such as Hox, Rb, p53, and epigenetic regulators such as the ING proteins in both early and the late stages and how they play antagonistic roles by increasing fitness and decreasing mortality early in life but contribute to deleterious effects and pathologies later in life.
Collapse
Affiliation(s)
- Emma Wanner
- Department of Biology, Faculty of Science, University of Calgary, Calgary, AB, Canada
| | - Harikrishnan Thoppil
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Karl Riabowol
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
6
|
Kang D, Jung SH, Lee GH, Lee S, Park HJ, Ko YG, Kim YN, Lee JS. Sulfated syndecan 1 is critical to preventing cellular senescence by modulating fibroblast growth factor receptor endocytosis. FASEB J 2020; 34:10316-10328. [PMID: 32530114 DOI: 10.1096/fj.201902714r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 05/06/2020] [Accepted: 05/20/2020] [Indexed: 01/10/2023]
Abstract
Cellular senescence can be triggered by various intrinsic and extrinsic stimuli. We previously reported that silencing of 3'-phosphoadenosine 5'-phosphosulfate synthetase 2 (PAPSS2) induces cellular senescence through augmented fibroblast growth factor receptor 1 (FGFR1) signaling. However, the exact molecular mechanism connecting heparan sulfation and cellular senescence remains unclear. Here, we investigated the potential involvement of heparan sulfate proteoglycans (HSPGs) in augmented FGFR1 signaling and cellular senescence. Depletion of several types of HSPGs revealed that cells depleted of syndecan 1 (SDC1) exhibited typical senescence phenotypes, and those depleted of PAPSS2-, SDC1-, or heparan sulfate 2-O sulfotransferase 1 (HS2ST1) showed decreased FGFR1 internalization along with hyperresponsiveness to and prolonged activation of fibroblast growth factor 2 (FGF2)-stimulated FGFR1- v-akt murine thymoma viral oncogene homolog (AKT) signaling. Clathrin- and caveolin-mediated FGFR1 endocytosis contributed to cellular senescence through the FGFR1-AKT-p53-p21 signaling pathway. Dynasore treatment triggered senescence phenotypes, augmented FGFR1-AKT-p53-p21 signaling, and decreased SDC1 expression. Finally, the replicatively and prematurely senescent cells were characterized by decreases of SDC1 expression and FGFR1 internalization, and an increase in FGFR1-AKT-p53-p21 signaling. Together, our results demonstrate that properly sulfated SDC1 plays a critical role in preventing cellular senescence through the regulation of FGFR1 endocytosis.
Collapse
Affiliation(s)
- Donghee Kang
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Medical Research Center, Inha University College of Medicine, Incheon, Korea
| | - Seung Hee Jung
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Medical Research Center, Inha University College of Medicine, Incheon, Korea
| | - Gun-Hee Lee
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Medical Research Center, Inha University College of Medicine, Incheon, Korea
| | - Seongju Lee
- Medical Research Center, Inha University College of Medicine, Incheon, Korea.,Department of Anatomy, Inha University College of Medicine, Incheon, Korea
| | - Heon Joo Park
- Medical Research Center, Inha University College of Medicine, Incheon, Korea.,Department of Microbiology, Inha University College of Medicine, Incheon, Korea
| | - Young-Gyu Ko
- Division of Life Sciences, Korea University, Seoul, Korea
| | - Yong-Nyun Kim
- Division of Translational Science, National Cancer Center, Goyang, Korea
| | - Jae-Seon Lee
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Medical Research Center, Inha University College of Medicine, Incheon, Korea
| |
Collapse
|
7
|
Shin EY, Park JH, You ST, Lee CS, Won SY, Park JJ, Kim HB, Shim J, Soung NK, Lee OJ, Schwartz MA, Kim EG. Integrin-mediated adhesions in regulation of cellular senescence. SCIENCE ADVANCES 2020; 6:eaay3909. [PMID: 32494696 PMCID: PMC7202880 DOI: 10.1126/sciadv.aay3909] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 02/21/2020] [Indexed: 05/10/2023]
Abstract
Bioinformatic and functional data link integrin-mediated cell adhesion to cellular senescence; however, the significance of and molecular mechanisms behind these connections are unknown. We now report that the focal adhesion-localized βPAK-interacting exchange factor (βPIX)-G protein-coupled receptor kinase interacting protein (GIT) complex controls cellular senescence in vitro and in vivo. βPIX and GIT levels decline with age. βPIX knockdown induces cellular senescence, which was prevented by reexpression. Loss of βPIX induced calpain cleavage of the endocytic adapter amphiphysin 1 to suppress clathrin-mediated endocytosis (CME); direct competition of GIT1/2 for the calpain-binding site on paxillin mediates this effect. Decreased CME and thus integrin endocytosis induced abnormal integrin signaling, with elevated reactive oxygen species production. Blocking integrin signaling inhibited senescence in human fibroblasts and mouse lungs in vivo. These results reveal a central role for integrin signaling in cellular senescence, potentially identifying a new therapeutic direction.
Collapse
Affiliation(s)
- Eun-Young Shin
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Jin-Hee Park
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Soon-Tae You
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Chan-Soo Lee
- Food Standard Division Scientific Office, Ministry of Food and Drug Safety (KFDA), Osong-eup, Cheongju 28159, Korea
| | - So-Yoon Won
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Jung-Jin Park
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Han-Byeol Kim
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Jaegal Shim
- Comparative Biomedicine Research Branch, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Nak-Kyun Soung
- World Class Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang-eup, Cheongju 28116, Korea
| | - Ok-Jun Lee
- Department of Pathology, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Martin Alexander Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
- Corresponding author. (E.-G.K.); (M.A.S.)
| | - Eung-Gook Kim
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju 28644, Korea
- Corresponding author. (E.-G.K.); (M.A.S.)
| |
Collapse
|
8
|
He M, Shen L, Jiang C, Gao G, Wang K, Jiao Y, Sun L, Cui Y, Ke Z, Yang Z. Rab22a is a novel prognostic marker for cell progression in breast cancer. Int J Mol Med 2020; 45:1037-1046. [PMID: 32124943 PMCID: PMC7053859 DOI: 10.3892/ijmm.2020.4486] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 01/07/2020] [Indexed: 12/30/2022] Open
Abstract
Breast cancer (BC) is the most common female malignant tumor worldwide. The mechanism of tumorigenesis is still unclear. Ras‑related proteins in brain (Rab)22a belongs to the Ras superfamily, which may act as an oncogene and participate in carcinogenesis. The present study aims to identify whether Rab22a could be a novel biomarker of prognosis and determine the effects of Rab22a on BC cell progression. A total 258 BC and 56 para‑tumor or non‑tumor formalin fixed paraffin embedded tissues were stained through immunohistochemistry. The association between Rab22a expression and clinicopathological features, as well as overall survival status were analyzed. The expression level of Rab22a in breast cell lines were detected using reverse transcription‑quantitative PCR and western blotting. SK‑BR‑3 cells were infected with Rab22a short hairpin RNA lenti‑virus and the ability of cell proliferation, migration and invasion were measured. Gene Set Enrichment Analysis (GSEA) was employed to analyze the pathways involved in the Rab22a mRNA high level group. Rab22a was found to be overexpressed in BC tissues and upregulated in BC cells. High expression of Rab22a was related to a poor prognosis of patients with BC. Knockdown of Rab22a decreased the proliferation, migration and invasion ability of BC cells. GSEA indicated that certain pathways, including mammalian target of rapamycin complex 1 and protein secretion were upregulated, while pathways, such as hypoxia and KRas were downregulated in the Rab22a high level group. Rab22a is of prognostic value for BC and necessary for BC cell proliferation.
Collapse
Affiliation(s)
- Miao He
- Department of Anesthesia, The Second Hospital of Jilin University, Changchun, Jilin 130022
| | - Leihua Shen
- Department of General Surgery, Xi'an Central Hospital, Xi'an, Shanxi 710000
- Department of Breast Surgery
| | - Chengwei Jiang
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033
| | - Ge Gao
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033
| | | | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021
| | | | | | - Zirui Ke
- Department of Breast Surgery
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | | |
Collapse
|
9
|
Wang L, Jin S, Dai P, Zhang T, Shi Y, Ai G, Shao X, Xie Y, Xu J, Chen Z, Gao Z. p57 Kip2 is a master regulator of human adipose derived stem cell quiescence and senescence. Stem Cell Res 2020; 44:101759. [PMID: 32224418 DOI: 10.1016/j.scr.2020.101759] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 03/01/2020] [Accepted: 03/05/2020] [Indexed: 12/30/2022] Open
Abstract
Although human adipose derived stem cells (hADSCs) hold great promises for regenerative medicine, their key biological properties remain poorly understood. In particular, proliferation defects resulted from deep quiescence (dormancy) and senescence represent a major hurdle in hADSC production and clinical application. We have developed a model system for mechanistic dissection of hADSC quiescence and senescence. p57Kip2, a major CDK inhibitor, was highly expressed in quiescent and senescent hADSCs but its level quickly declined upon stem cell activation. p57Kip2 overexpression induced quiescence in spite of proliferative signals and its knockdown promoted cell cycle reentry even with induction of quiescence presumably through modulating the CDK2-CyclinE1 complex. Given its key role in quiescence and senescence, p57Kip2 may be exploited for innovative strategies to amplify hADSCs of high quality for clinics.
Collapse
Affiliation(s)
- Lian Wang
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; The Lifeng institute of Regenerative Medicine, Tongji University, Shanghai 200092, China
| | - Shengkai Jin
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; The Lifeng institute of Regenerative Medicine, Tongji University, Shanghai 200092, China; Advanced Institute of Translational Medicine, Tongji University School of Medicine, Shanghai 200092 China
| | - Peibin Dai
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; The Lifeng institute of Regenerative Medicine, Tongji University, Shanghai 200092, China; Advanced Institute of Translational Medicine, Tongji University School of Medicine, Shanghai 200092 China
| | - Tianran Zhang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; The Lifeng institute of Regenerative Medicine, Tongji University, Shanghai 200092, China; Advanced Institute of Translational Medicine, Tongji University School of Medicine, Shanghai 200092 China
| | - Yanghua Shi
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; The Lifeng institute of Regenerative Medicine, Tongji University, Shanghai 200092, China; Advanced Institute of Translational Medicine, Tongji University School of Medicine, Shanghai 200092 China
| | - Guihai Ai
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaowen Shao
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; The Lifeng institute of Regenerative Medicine, Tongji University, Shanghai 200092, China
| | - Yutong Xie
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jun Xu
- East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Zhongping Chen
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Zhengliang Gao
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; The Lifeng institute of Regenerative Medicine, Tongji University, Shanghai 200092, China; Advanced Institute of Translational Medicine, Tongji University School of Medicine, Shanghai 200092 China.
| |
Collapse
|
10
|
Thoppil H, Riabowol K. Senolytics: A Translational Bridge Between Cellular Senescence and Organismal Aging. Front Cell Dev Biol 2020; 7:367. [PMID: 32039197 PMCID: PMC6987374 DOI: 10.3389/fcell.2019.00367] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/16/2019] [Indexed: 11/22/2022] Open
Abstract
Aging is defined as a progressive decrease in physiological function accompanied by a steady increase in mortality. The antagonistic pleiotropy theory proposes that aging is largely due to the natural selection of genes and pathways that increase fitness and decrease mortality early in life but contribute to deleterious effects and pathologies later in life. Cellular senescence is one such mechanism, which results in a permanent cell cycle arrest that has been described as a mechanism to limit cancer cell growth. However, recent studies have also suggested a dark side of senescence in which a build-up of senescent cells with age leads to increased inflammation due to a senescence-associated secretory phenotype (SASP). This phenotype that includes many cytokines promotes tumorigenesis and can exhaust the pool of immune cells in the body. Studies clearing senescent cells from mice using the p16-based transgene INK-ATTAC have shown that senescent cells can impact both organismal aging and lifespan. Here we discuss these advances that have resulted in the development of a whole new class of compounds known as senolytics, some of which are currently undergoing clinical trials in humans for treating a variety of age-related pathologies such as osteoarthritis.
Collapse
Affiliation(s)
- Harikrishnan Thoppil
- Arnie Charbonneau Cancer Institute, Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Karl Riabowol
- Arnie Charbonneau Cancer Institute, Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, Department of Oncology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
11
|
Archambeau J, Blondel A, Pedeux R. Focus-ING on DNA Integrity: Implication of ING Proteins in Cell Cycle Regulation and DNA Repair Modulation. Cancers (Basel) 2019; 12:cancers12010058. [PMID: 31878273 PMCID: PMC7017203 DOI: 10.3390/cancers12010058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/16/2022] Open
Abstract
The ING family of tumor suppressor genes is composed of five members (ING1-5) involved in cell cycle regulation, DNA damage response, apoptosis and senescence. All ING proteins belong to various HAT or HDAC complexes and participate in chromatin remodeling that is essential for genomic stability and signaling pathways. The gatekeeper functions of the INGs are well described by their role in the negative regulation of the cell cycle, notably by modulating the stability of p53 or the p300 HAT activity. However, the caretaker functions are described only for ING1, ING2 and ING3. This is due to their involvement in DNA repair such as ING1 that participates not only in NERs after UV-induced damage, but also in DSB repair in which ING2 and ING3 are required for accumulation of ATM, 53BP1 and BRCA1 near the lesion and for the subsequent repair. This review summarizes evidence of the critical roles of ING proteins in cell cycle regulation and DNA repair to maintain genomic stability.
Collapse
|
12
|
miR-135b-5p enhances doxorubicin-sensitivity of breast cancer cells through targeting anterior gradient 2. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:26. [PMID: 30665445 PMCID: PMC6341729 DOI: 10.1186/s13046-019-1024-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/06/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND The pro-oncogenic anterior gradient 2 (AGR2) is involved in tumor growth and drug resistance of breast cancer. Mechanisms that regulate expression of AGR2 still need to be elucidated. METHODS In this study, expression levels of AGR2 and miR-135b-5p were analyzed in different breast cancer cell lines as well as in clinical breast cancer tissues. The in vitro and in vivo functional effect of AGR2 and miR-135b-5p were also investigated. A luciferase reporter assay was applied to confirm the interaction between miR-135b-5p and AGR2 mRNA. RESULTS We identified AGR2 as a target of miR-135b-5p. Expression of AGR2 was up-regulated in doxorubicin-resistant breast cancer cells. AGR2 mediated doxorubicin-sensitivity of breast cancer cells both in vitro and in vivo. miR-135b-5p negatively regulated AGR2-expression of breast cancer cells increasing doxorubicin-sensitivity. However, miR-135b-5p was down-regulated in doxorubicin-resistant breast cancer cells as well as during treatment with doxorubicin, which might be a probable reason for over-expression of AGR2. Up-regulation of miR-135b-5p increased doxorubicin-sensitivity of breast cancer cells in vivo. In addition, levels of AGR2 negatively correlated with levels of miR-135b-5p in clinical breast cancer tissue samples. CONCLUSION Our results highlight the potential of miR-135b-5p as a target for treating AGR2-expressing breast cancer with doxorubicin-resistance.
Collapse
|
13
|
Personalized prediction of genes with tumor-causing somatic mutations based on multi-modal deep Boltzmann machine. Neurocomputing 2019. [DOI: 10.1016/j.neucom.2018.02.096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
14
|
Bertschmann J, Thalappilly S, Riabowol K. The ING1a model of rapid cell senescence. Mech Ageing Dev 2019; 177:109-117. [DOI: 10.1016/j.mad.2018.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/21/2018] [Accepted: 06/16/2018] [Indexed: 12/17/2022]
|
15
|
Hinze C, Boucrot E. Endocytosis in proliferating, quiescent and terminally differentiated cells. J Cell Sci 2018; 131:131/23/jcs216804. [PMID: 30504135 DOI: 10.1242/jcs.216804] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Endocytosis mediates nutrient uptake, receptor internalization and the regulation of cell signaling. It is also hijacked by many bacteria, viruses and toxins to mediate their cellular entry. Several endocytic routes exist in parallel, fulfilling different functions. Most studies on endocytosis have used transformed cells in culture. However, as the majority of cells in an adult body have exited the cell cycle, our understanding is biased towards proliferating cells. Here, we review the evidence for the different pathways of endocytosis not only in dividing, but also in quiescent, senescent and terminally differentiated cells. During mitosis, residual endocytosis is dedicated to the internalization of caveolae and specific receptors. In non-dividing cells, clathrin-mediated endocytosis (CME) functions, but the activity of alternative processes, such as caveolae, macropinocytosis and clathrin-independent routes, vary widely depending on cell types and functions. Endocytosis supports the quiescent state by either upregulating cell cycle arrest pathways or downregulating mitogen-induced signaling, thereby inhibiting cell proliferation. Endocytosis in terminally differentiated cells, such as skeletal muscles, adipocytes, kidney podocytes and neurons, supports tissue-specific functions. Finally, uptake is downregulated in senescent cells, making them insensitive to proliferative stimuli by growth factors. Future studies should reveal the molecular basis for the differences in activities between the different cell states.
Collapse
Affiliation(s)
- Claudia Hinze
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Emmanuel Boucrot
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK .,Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London WC1E 7HX, UK
| |
Collapse
|
16
|
Rajarajacholan UK, Thalappilly S, Riabowol K. ING1 regulates rRNA levels by altering nucleolar chromatin structure and mTOR localization. Nucleic Acids Res 2017; 45:1776-1792. [PMID: 27903908 PMCID: PMC5389678 DOI: 10.1093/nar/gkw1161] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 11/10/2016] [Indexed: 01/31/2023] Open
Abstract
Epigenetic, transcriptional and signaling processes in the nucleolus regulate rRNA transcription and cell growth. We report here that the tumor suppressor ING1b binds rDNA, regulates rDNA chromatin modifications and affects nucleolar localization of mTOR to modulate rRNA levels. ING1 represses rDNA transcription by recruiting HDAC1 to rDNA loci, increasing its association with the NoRC complex and deacetylating the histone H3K9 and H3K27 marks of active transcription. Loss of ING1 enhances nucleolar localization of phospho-mTOR and its association with Raptor and GβL, even during rapamycin treatment. ING1 inhibits rDNA transcription by inhibiting UBF activity and its interaction with mTOR. Regulation of rDNA heterochromatin and rRNA synthesis by ING1 is also apparent during normal cell growth and during cell stress. Moreover, this function was also important during PMA induced differentiation of THP1 cells, since knocking down ING1 affected the process by inhibiting rRNA transcriptional repression. These observations show that ING1 regulates the nucleolar epigenome and rDNA transcription suggesting that regulation of protein synthesis might serve as the basis for ING1 function as a type II tumor suppressor.
Collapse
Affiliation(s)
- Uma Karthika Rajarajacholan
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Subhash Thalappilly
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Karl Riabowol
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Oncology, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
17
|
Wang F, Wang AY, Chesnelong C, Yang Y, Nabbi A, Thalappilly S, Alekseev V, Riabowol K. ING5 activity in self-renewal of glioblastoma stem cells via calcium and follicle stimulating hormone pathways. Oncogene 2017; 37:286-301. [PMID: 28925404 PMCID: PMC5799773 DOI: 10.1038/onc.2017.324] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/10/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022]
Abstract
Stem cell-like brain tumor initiating cells (BTICs) cause recurrence of glioblastomas, with BTIC 'stemness' affected by epigenetic mechanisms. The ING family of epigenetic regulators (ING1-5) function by targeting histone acetyltransferase (HAT) or histone deacetylase complexes to the H3K4me3 mark to alter histone acetylation and subsequently, gene expression. Here we find that ectopic expression of ING5, the targeting subunit of HBO1, MOZ and MORF HAT complexes increases expression of the Oct4, Olig2 and Nestin stem cell markers, promotes self-renewal, prevents lineage differentiation and increases stem cell pools in BTIC populations. This activity requires the plant homeodomain region of ING5 that interacts specifically with the H3K4me3 mark. ING5 also enhances PI3K/AKT and MEK/ERK activity to sustain self-renewal of BTICs over serial passage of stem cell-like spheres. ING5 exerts these effects by activating transcription of calcium channel and follicle stimulating hormone pathway genes. In silico analyses of The Cancer Genome Atlas data suggest that ING5 is a positive regulator of BTIC stemness, whose expression negatively correlates with patient prognosis, especially in the Proneural and Classical subtypes, and in tumors with low SOX2 expression. These data suggest that altering histone acetylation status and signaling pathways induced by ING5 may provide useful clinical strategies to target tumor resistance and recurrence in glioblastoma.
Collapse
Affiliation(s)
- F Wang
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - A Y Wang
- Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - C Chesnelong
- Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Y Yang
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - A Nabbi
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - S Thalappilly
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - V Alekseev
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - K Riabowol
- Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
18
|
How and why do toxic conformers of aberrant proteins accumulate during ageing? Essays Biochem 2017; 61:317-324. [DOI: 10.1042/ebc20160085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/04/2017] [Accepted: 04/21/2017] [Indexed: 12/18/2022]
Abstract
Ageing can be defined as a gradual decline in cellular and physical functions accompanied by an increased sensitivity to the environment and risk of death. The increased risk of mortality is causally connected to a gradual, intracellular accumulation of so-called ageing factors, of which damaged and aggregated proteins are believed to be one. Such aggregated proteins also contribute to several age-related neurodegenerative disorders e.g. Alzheimer’s, Parkinson’s, and Huntington’s diseases, highlighting the importance of protein quality control (PQC) in ageing and its associated diseases. PQC consists of two interrelated systems: the temporal control system aimed at refolding, repairing, and/or removing aberrant proteins and their aggregates and the spatial control system aimed at harnessing the potential toxicity of aberrant proteins by sequestering them at specific cellular locations. The accumulation of toxic conformers of aberrant proteins during ageing is often declared to be a consequence of an incapacitated temporal PQC system—i.e. a gradual decline in the activity of chaperones and proteases. Here, we review the current knowledge on PQC in relation to ageing and highlight that the breakdown of both temporal and spatial PQC may contribute to ageing and thus comprise potential targets for therapeutic interventions of the ageing process.
Collapse
|
19
|
Zhang R, Jin J, Shi J, Hou Y. INGs are potential drug targets for cancer. J Cancer Res Clin Oncol 2017; 143:189-197. [PMID: 27544390 DOI: 10.1007/s00432-016-2219-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
PURPOSE The inhibitor of growth (ING) family consists of ING1, ING2, ING3, ING4 and ING5, which function as the type II tumor suppressors. INGs regulate cell proliferation, senescence, apoptosis, differentiation, angiogenesis, DNA repair, metastasis, and invasion by multiple pathways. In addition, INGs increase cancer cell sensitivity for chemotherapy and radiotherapy, while clinical observations show that INGs are frequently lost in some types of cancers. The aim of the study was to summarize the recent progress regarding INGs regulating tumor progression. METHODS The literatures of INGs regulating tumor progression were searched and assayed. RESULTS The regulating signaling pathways of ING1, ING2, ING3 or ING4 on tumor progression were shown. The mechanisms of INGs on tumor suppression were also assayed. CONCLUSIONS This review better summarized the signaling mechanism of INGs on tumor suppression, which provides a candidate therapy strategy for cancers.
Collapse
Affiliation(s)
- Runyun Zhang
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Jianhua Jin
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China
| | - Juanjuan Shi
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| | - Yongzhong Hou
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China.
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| |
Collapse
|
20
|
Nagano T, Nakano M, Nakashima A, Onishi K, Yamao S, Enari M, Kikkawa U, Kamada S. Identification of cellular senescence-specific genes by comparative transcriptomics. Sci Rep 2016; 6:31758. [PMID: 27545311 PMCID: PMC4992837 DOI: 10.1038/srep31758] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/27/2016] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence is defined as permanent cell cycle arrest induced by various stresses. Although the p53 transcriptional activity is essential for senescence induction, the downstream genes that are crucial for senescence remain unsolved. Here, by using a developed experimental system in which cellular senescence or apoptosis is induced preferentially by altering concentration of etoposide, a DNA-damaging drug, we compared gene expression profiles of senescent and apoptotic cells by microarray analysis. Subtraction of the expression profile of apoptotic cells identified 20 genes upregulated specifically in senescent cells. Furthermore, 6 out of 20 genes showed p53-dependent upregulation by comparing gene expression between p53-proficient and -deficient cells. These 6 genes were also upregulated during replicative senescence of normal human diploid fibroblasts, suggesting that upregulation of these genes is a general phenomenon in senescence. Among these genes, 2 genes (PRODH and DAO) were found to be directly regulated by p53, and ectopic expression of 4 genes (PRODH, DAO, EPN3, and GPR172B) affected senescence phenotypes induced by etoposide treatment. Collectively, our results identified several proteins as novel downstream effectors of p53-mediated senescence and provided new clues for further research on the complex signalling networks underlying the induction and maintenance of senescence.
Collapse
Affiliation(s)
- Taiki Nagano
- Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan.,Department of Biology, Graduate School of Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Masayuki Nakano
- Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan.,Department of Biology, Graduate School of Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Akio Nakashima
- Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan.,Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Kengo Onishi
- Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan.,Department of Biology, Graduate School of Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Shunsuke Yamao
- Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan.,Department of Biology, Graduate School of Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Masato Enari
- Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Ushio Kikkawa
- Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan.,Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Shinji Kamada
- Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan.,Department of Biology, Graduate School of Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| |
Collapse
|
21
|
Rajarajacholan UK, Riabowol K. Aging with ING: a comparative study of different forms of stress induced premature senescence. Oncotarget 2016; 6:34118-27. [PMID: 26439691 PMCID: PMC4741440 DOI: 10.18632/oncotarget.5947] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 09/09/2015] [Indexed: 12/12/2022] Open
Abstract
Cell senescence contributes to organismal aging and is induced by telomere erosion and an ensuing DNA damage signal as cells reach the end of their replicative lifespan in vitro or in vivo. Stresses induced by oncogene or tumor suppressor hyperactivation, oxidative stress, ionizing radiation and other DNA damaging agents result in forms of stress induced premature senescence (SIPS) that show similarities to replicative senescence. Since replicative senescence and SIPS occur over many days and many population doublings of the mass cultures of primary cells used to study senescence, the sequence of events that occur downstream of senescence signaling can be challenging to define. Here we compare a new model of ING1a-induced senescence with several other forms of senescence. The ING1a epigenetic regulator synchronously induces senescence in mass cultures several-fold faster than all other agents, taking 24 and 36 hours to activate the Rb/ p16INK4a, but not the p53 tumor suppressor axis to efficiently induce senescence. ING1a induces expression of intersectin 2, a scaffold protein necessary for endocytosis, altering the stoichiometry of endocytosis proteins, subsequently blocking growth factor uptake leading to activation of Rb signaling to block cell growth. ING1a acts as a novel link in the activation of the Rb pathway that can impose senescence in the absence of activating p53-mediated DNA damage signaling, and should prove useful in defining the molecular events contributing to Rb-induced senescence.
Collapse
Affiliation(s)
| | - Karl Riabowol
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada.,Department of Oncology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
22
|
Nabbi A, Almami A, Thakur S, Suzuki K, Boland D, Bismar TA, Riabowol K. ING3 protein expression profiling in normal human tissues suggest its role in cellular growth and self-renewal. Eur J Cell Biol 2015; 94:214-22. [PMID: 25819753 DOI: 10.1016/j.ejcb.2015.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 03/03/2015] [Accepted: 03/03/2015] [Indexed: 12/17/2022] Open
Abstract
Members of the INhibitor of Growth (ING) family of proteins act as readers of the epigenetic code through specific recognition of the trimethylated form of lysine 4 of histone H3 (H3K4Me3) by their plant homeodomains. The founding member of the family, ING1, was initially identified as a tumor suppressor with altered regulation in a variety of cancer types. While alterations in ING1 and ING4 levels have been reported in a variety of cancer types, little is known regarding ING3 protein levels in normal or transformed cells due to a lack of reliable immunological tools. In this study we present the characterization of a new monoclonal antibody we have developed against ING3 that specifically recognizes human and mouse ING3. The antibody works in western blots, immunofluorescence, immunoprecipitation and immunohistochemistry. Using this antibody we show that ING3 is most highly expressed in small intestine, bone marrow and epidermis, tissues in which cells undergo rapid proliferation and renewal. Consistent with this observation, we show that ING3 is expressed at significantly higher levels in proliferating versus quiescent epithelial cells. These data suggest that ING3 levels may serve as a surrogate for growth rate, and suggest possible roles for ING3 in growth and self renewal and related diseases such as cancer.
Collapse
Affiliation(s)
- Arash Nabbi
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Oncology, Southern Alberta Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Amal Almami
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Oncology, Southern Alberta Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Satbir Thakur
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Oncology, Southern Alberta Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Keiko Suzuki
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Oncology, Southern Alberta Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Donna Boland
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Oncology, Southern Alberta Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tarek A Bismar
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Oncology, Southern Alberta Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Pathology & Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Karl Riabowol
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Oncology, Southern Alberta Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
23
|
LincRNA-p21 acts as a mediator of ING1b-induced apoptosis. Cell Death Dis 2015; 6:e1668. [PMID: 25741593 PMCID: PMC4385912 DOI: 10.1038/cddis.2015.15] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/02/2015] [Accepted: 01/07/2015] [Indexed: 02/08/2023]
Abstract
ING1b is a tumor suppressor that affects transcription, cell cycle control and apoptosis. ING1b is deregulated in disease, and its activity is closely linked to that of p53. In addition to regulating protein-coding genes, we found that ING1b also influences the expression of large intergenic non-coding RNAs (lincRNAs). In particular, lincRNA-p21 was significantly induced after DNA-damage stress or by ING1b overexpression. Furthermore, lincRNA-p21 expression in response to DNA damage was significantly attenuated in cells lacking ING1b. LincRNA-p21 is also a target of p53 and can trigger apoptosis in mouse cell models. We found that this function of lincRNA-p21 is conserved in human cell models. Moreover, ING1b and p53 could function independently to influence lincRNA-p21 expression. However, their effects become more additive under conditions of stress. In particular, ING1b regulates lincRNA-p21 levels by binding to its promoter and is required for induction of lincRNA-p21 by p53. The ability of ING1b to cause apoptosis is also impaired in the absence of lincRNA-p21. Surprisingly, deletion of the ING1b plant homeodomain, which allows it to bind histones and regulate chromatin structure, did not alter regulation of lincRNA-p21. Our findings suggest that ING1b induces lincRNA-p21 expression independently of histone 3 lysine 4 trimethylation mark recognition and that lincRNA-p21 functions downstream of ING1b. Thus, regulation at the level of lincRNA-p21 may represent the point at which ING1b and p53 pathways converge to induce apoptosis under specific stress conditions.
Collapse
|
24
|
Satpathy S, Guérillon C, Kim TS, Bigot N, Thakur S, Bonni S, Riabowol K, Pedeux R. SUMOylation of the ING1b tumor suppressor regulates gene transcription. Carcinogenesis 2014; 35:2214-23. [PMID: 24903338 DOI: 10.1093/carcin/bgu126] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The INhibitor of Growth (ING) proteins are encoded as multiple isoforms in five ING genes (ING1 -5) and act as type II tumor suppressors. They are growth inhibitory when overexpressed and are frequently mislocalized or downregulated in several forms of cancer. ING1 and ING2 are stoichiometric members of histone deacetylase complexes, whereas ING3-5 are stoichiometric components of different histone acetyltransferase complexes. The INGs target these complexes to histone marks, thus acting as epigenetic regulators. ING proteins affect angiogenesis, apoptosis, DNA repair, metastasis and senescence, but how the proteins themselves are regulated is not yet clear. Here, we find a small ubiquitin-like modification (SUMOylation) of the ING1b protein and identify lysine 193 (K193) as the preferred ING1b SUMO acceptor site. We also show that PIAS4 is the E3 SUMO ligase responsible for ING1b SUMOylation on K193. Sequence alignment reveals that the SUMO consensus site on ING1b contains a phosphorylation-dependent SUMOylation motif (PDSM) and our data indicate that the SUMOylation on K193 is enhanced by the S199D phosphomimic mutant. Using an ING1b protein mutated at the major SUMOylation site (ING1b E195A), we further demonstrate that ING1b SUMOylation regulates the binding of ING1b to the ISG15 and DGCR8 promoters, consequently regulating ISG15 and DGCR8 transcription. These results suggest a role for ING1b SUMOylation in the regulation of gene transcription.
Collapse
Affiliation(s)
- Shankha Satpathy
- Department of Biochemistry and Molecular Biology, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada, Present address: The Novo Nordisk Foundation Center for Protein Research, Copenhagen, Denmark
| | - Claire Guérillon
- INSERM U917, Microenvironnement et Cancer, 350043 Rennes, France, Université de Rennes 1, 350043 Rennes, France and
| | - Tae-Sun Kim
- Department of Biochemistry and Molecular Biology, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Nicolas Bigot
- INSERM U917, Microenvironnement et Cancer, 350043 Rennes, France, Université de Rennes 1, 350043 Rennes, France and
| | - Satbir Thakur
- INSERM U917, Microenvironnement et Cancer, 350043 Rennes, France
| | - Shirin Bonni
- INSERM U917, Microenvironnement et Cancer, 350043 Rennes, France
| | - Karl Riabowol
- Department of Biochemistry and Molecular Biology, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada,
| | - Rémy Pedeux
- INSERM U917, Microenvironnement et Cancer, 350043 Rennes, France
| |
Collapse
|
25
|
Keep-ING balance: tumor suppression by epigenetic regulation. FEBS Lett 2014; 588:2728-42. [PMID: 24632289 DOI: 10.1016/j.febslet.2014.03.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/06/2014] [Indexed: 12/26/2022]
Abstract
Cancer cells accumulate genetic and epigenetic changes that alter gene expression to drive tumorigenesis. Epigenetic silencing of tumor suppressor, cell cycle, differentiation and DNA repair genes contributes to neoplastic transformation. The ING (inhibitor of growth) proteins (ING1-ING5) have emerged as a versatile family of growth regulators, phospholipid effectors, histone mark sensors and core components of HDAC1/2 - and several HAT chromatin-modifying complexes. This review will describe the characteristic pathways by which ING family proteins differentially affect the Hallmarks of Cancer and highlight the various epigenetic mechanisms by which they regulate gene expression. Finally, we will discuss their potentials as biomarkers and therapeutic targets in epigenetic treatment strategies.
Collapse
|
26
|
Qi L, Zhang Y. Truncation of inhibitor of growth family protein 5 effectively induces senescence, but not apoptosis in human tongue squamous cell carcinoma cell line. Tumour Biol 2013; 35:3139-44. [PMID: 24254310 DOI: 10.1007/s13277-013-1410-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 11/08/2013] [Indexed: 10/26/2022] Open
Abstract
In these studies, inhibitor of growth protein 5 (ING5) and various fragments of it were overexpressed in the human tongue squamous cell carcinoma cell line, HSC-3. The roles of ING5 in HSC-3 cells were then identified in vitro. Our results indicate that intact ING5 can inhibit proliferation and induce apoptosis in HSC-3 cells. Moreover, two truncated fragments of ING5 (aa 1-184 and aa 107-226) can induce cellular senescence. To analyze the signaling pathway involved, western blotting was performed. In these assays, two truncated fragments of ING5 were found to inhibit the cyclin E and CDK2 expression. These results are consistent with the S phase arrest observed with the overexpression of truncated ING5. However, the mechanisms of ING5-induced cellular senescence remain unclear, and extensive investigations are required in future studies.
Collapse
Affiliation(s)
- Lin Qi
- Department of Orthodontics, School of Stomatology, China Medical University, Shenyang, Liaoning, China
| | | |
Collapse
|
27
|
Wu DS, Shen JZ, Yu AF, Fu HY, Zhou HR, Shen SF. Epigallocatechin-3-gallate and trichostatin A synergistically inhibit human lymphoma cell proliferation through epigenetic modification of p16INK4a. Oncol Rep 2013; 30:2969-75. [PMID: 24064951 DOI: 10.3892/or.2013.2734] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 08/19/2013] [Indexed: 11/06/2022] Open
Abstract
DNA methylation and histone deacetylation play important roles in the occurrence and development of cancers by inactivating the expression of tumor suppressors, including p16(INK4a), a cyclin-dependent kinase inhibitor. The present study investigated the effect of epigallocatechin-3-gallate (EGCG) alone or in combination with trichostatin A (TSA) on p16(INK4a) gene expression and growth in human malignant lymphoma CA46 cells. CA46 cell viability and cell cycle were analyzed; methylation of the p16(INK4a) gene was assessed by nested methylation-specific PCR (n-MSP). p16(INK4a )mRNA and protein expression was determined by real-time quantitative PCR and western blot analyses, respectively. Both EGCG and TSA alone inhibited CA46 cell proliferation; the combined treatment (6 µg/ml EGCG and 15 ng/ml TSA) significantly reduced CA46 cell proliferation from 24 to 96 h (all P<0.001). Cells treated with 24 µg/ml EGCG or the combination treatment (6 µg/ml EGCG and 15 ng/ml TSA) had lower proliferative indices when compared to the other groups. Co-treatment with EGCG and TSA decreased p16(INK4a) gene methylation, which coincided with increased p16(INK4a) mRNA and protein expression. Thus, EGCG and TSA synergistically reactivate p16(INK4a) gene expression in part through reducing promoter methylation, which may decrease CA46 cell proliferation.
Collapse
Affiliation(s)
- Dan-Sen Wu
- Department of Hematology, Union Hospital of Fujian Medical University, Fujian Institute of Hematology, Fuzhou, Fujian 35001, P.R. China
| | | | | | | | | | | |
Collapse
|
28
|
Burgess DJ. Senescence: ConnectING endocytosis. Nat Rev Cancer 2013; 13:294-5. [PMID: 23535845 DOI: 10.1038/nrc3511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|