51
|
Voisin J, Farina F, Naphade S, Fontaine M, Tshilenge K, Galicia Aguirre C, Lopez‐Ramirez A, Dancourt J, Ginisty A, Sasidharan Nair S, Lakshika Madushani K, Zhang N, Lejeune F, Verny M, Campisi J, Ellerby LM, Neri C. FOXO3 targets are reprogrammed as Huntington's disease neural cells and striatal neurons face senescence with p16 INK4a increase. Aging Cell 2020; 19:e13226. [PMID: 33156570 PMCID: PMC7681055 DOI: 10.1111/acel.13226] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 06/26/2020] [Accepted: 07/26/2020] [Indexed: 12/21/2022] Open
Abstract
Neurodegenerative diseases (ND) have been linked to the critical process in aging—cellular senescence. However, the temporal dynamics of cellular senescence in ND conditions is unresolved. Here, we show senescence features develop in human Huntington's disease (HD) neural stem cells (NSCs) and medium spiny neurons (MSNs), including the increase of p16INK4a, a key inducer of cellular senescence. We found that HD NSCs reprogram the transcriptional targets of FOXO3, a major cell survival factor able to repress cell senescence, antagonizing p16INK4a expression via the FOXO3 repression of the transcriptional modulator ETS2. Additionally, p16INK4a promotes cellular senescence features in human HD NSCs and MSNs. These findings suggest that cellular senescence may develop during neuronal differentiation in HD and that the FOXO3‐ETS2‐p16INK4a axis may be part of molecular responses aimed at mitigating this phenomenon. Our studies identify neuronal differentiation with accelerated aging of neural progenitors and neurons as an alteration that could be linked to NDs.
Collapse
Affiliation(s)
- Jessica Voisin
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | - Francesca Farina
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | | | - Morgane Fontaine
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | | | | | | | - Julia Dancourt
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | - Aurélie Ginisty
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | - Satish Sasidharan Nair
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | | | | | - François‐Xavier Lejeune
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | - Marc Verny
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| | - Judith Campisi
- Buck Institute for Research on Aging Novato CA USA
- Lawrence Berkeley National Laboratory Berkeley CA USA
| | | | - Christian Neri
- Centre National de la Recherche Scientifique UMR 8256 Institut National de la Santé et de la Recherche Médicale ERL U1164 Assistance Publique‐Hôpitaux de Paris Brain‐C Lab Sorbonne Université Paris France
| |
Collapse
|
52
|
Abruzzo PM, Canaider S, Pizzuti V, Pampanella L, Casadei R, Facchin F, Ventura C. Herb-Derived Products: Natural Tools to Delay and Counteract Stem Cell Senescence. Stem Cells Int 2020; 2020:8827038. [PMID: 33101419 PMCID: PMC7568162 DOI: 10.1155/2020/8827038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Cellular senescence plays a very important role in organismal aging increasing with age and in age-related diseases (ARDs). This process involves physiological, structural, biochemical, and molecular changes of cells, leading to a characteristic trait referred to "senescence-associated secretory phenotype (SASP)." In particular, with aging, stem cells (SCs) in situ exhibit a diminished capacity of self-renewal and show a decline in their functionality. The identification of interventions able to prevent the accumulation of senescent SCs in the organism or to pretreat cultured multipotent mesenchymal stromal cells (MSCs) prior to employing them for cell therapy is a main purpose of medical research. Many approaches have been investigated and resulted effective to prevent or counteract SC senescence in humans, as well as other animal models. In this work, we have reviewed the chance of using a number of herb-derived products as novel tools in the treatment of cell senescence, highlighting the efficacy of these agents, often still far from being clearly understood.
Collapse
Affiliation(s)
- Provvidenza M. Abruzzo
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Silvia Canaider
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering-Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Valeria Pizzuti
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Luca Pampanella
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Raffaella Casadei
- Department for Life Quality Studies (QuVi), University of Bologna, Corso D'Augusto 237, 47921 Rimini, Italy
| | - Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering-Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Carlo Ventura
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering-Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| |
Collapse
|
53
|
Luan Y, Zhang W, Xie J, Mao J. CDKN2A inhibits cell proliferation and invasion in cervical cancer through LDHA-mediated AKT/mTOR pathway. Clin Transl Oncol 2020; 23:222-228. [DOI: 10.1007/s12094-020-02409-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/23/2020] [Indexed: 12/19/2022]
|
54
|
Liu C, Gao J, Su G, Xiang Y, Wan L. MicroRNA-1202 plays a vital role in osteoarthritis via KCNQ1OT1 has-miR-1202-ETS1 regulatory pathway. J Orthop Surg Res 2020; 15:130. [PMID: 32252801 PMCID: PMC7137223 DOI: 10.1186/s13018-020-01655-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/27/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND This study aimed to explore the molecular mechanism of osteoarthritis (OA) and provide information about new genes as potential targets for OA treatment. METHODS Gene expression profile of GSE105027, including 12 OA serum samples (OA group) and 12 healthy serum samples (ctrl group), was downloaded. The differentially expressed miRNAs (DEMs) as well as miRNA-mRNAs interactions were investigated, followed by function and pathway investigation. Then the protein-protein interaction (PPI) network was performed. Furthermore, the long non-coding RNA (lncRNA)-miRNA-mRNA interactions (competing endogenous RNAs, ceRNAs) were investigated. RESULTS A total of 17 downregulated miRNAs were revealed between OA and ctrl groups. These DEMs such as has-miR-1202 were mainly enriched in GO functions like histone acetyltransferase binding and KEGG pathways like cellular senescence. The integrated PPI network analysis showed that has-miR-1202, has-miR-33b-3p, has-miR-940, has-miR-4284, and has-miR-4281 were 5 downregulated miRNAs in this network. Furthermore, the lncRNA-miRNA-mRNA interactions such as KCNQ1OT1-has-miR-1202-ETS1 were revealed in the present ceRNA network. CONCLUSION Key DEMs such as miR-33b-3p, miR-940, and miR-1202 may be involved in OA. miR-1202 may regulate OA development via histone acetyltransferase pathway binding function and cellular senescence pathway. Furthermore, KCNQ1OT1-has-miR-1202-ETS1 might be vital for the process of OA.
Collapse
Affiliation(s)
- Changzeng Liu
- Department of Orthopaedics, No. 904th Hospital of the Joint Logistics Support Force of PLA, Liangxi District, Wuxi, 214044, Jiangsu, China.
| | - Jianming Gao
- Department of Orthopaedics, No. 904th Hospital of the Joint Logistics Support Force of PLA, Liangxi District, Wuxi, 214044, Jiangsu, China
| | - Guangyan Su
- Department of Orthopaedics, No. 904th Hospital of the Joint Logistics Support Force of PLA, Liangxi District, Wuxi, 214044, Jiangsu, China
| | - Yang Xiang
- Department of Orthopaedics, No. 904th Hospital of the Joint Logistics Support Force of PLA, Liangxi District, Wuxi, 214044, Jiangsu, China
| | - Le Wan
- Department of Orthopaedics, No. 904th Hospital of the Joint Logistics Support Force of PLA, Liangxi District, Wuxi, 214044, Jiangsu, China
| |
Collapse
|
55
|
Anerillas C, Abdelmohsen K, Gorospe M. Regulation of senescence traits by MAPKs. GeroScience 2020; 42:397-408. [PMID: 32300964 PMCID: PMC7205942 DOI: 10.1007/s11357-020-00183-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 03/10/2020] [Indexed: 01/10/2023] Open
Abstract
A phenotype of indefinite growth arrest acquired in response to sublethal damage, cellular senescence affects normal aging and age-related disease. Mitogen-activated protein kinases (MAPKs) are capable of sensing changes in cellular conditions, and in turn elicit adaptive responses including cell senescence. MAPKs modulate the levels and function of many proteins, including proinflammatory factors and factors in the p21/p53 and p16/RB pathways, the main senescence-regulatory axes. Through these actions, MAPKs implement key traits of senescence-growth arrest, cell survival, and the senescence-associated secretory phenotype (SASP). In this review, we summarize and discuss our current knowledge of the impact of MAPKs in senescence. In addition, given that eliminating or suppressing senescent cells can improve health span, we discuss the function and possible exploitation of MAPKs in the elimination (senolysis) or suppression (senostasis) of senescent cells.
Collapse
Affiliation(s)
- Carlos Anerillas
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA.
| |
Collapse
|
56
|
Wu C, Yang P, Liu B, Tang Y. Is there a CDKN2A-centric network in pancreatic ductal adenocarcinoma? Onco Targets Ther 2020; 13:2551-2562. [PMID: 32273725 PMCID: PMC7108878 DOI: 10.2147/ott.s232464] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/19/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer has a high mortality rate and its incidence has risen rapidly in recent years. Meanwhile, the diagnosis and treatment of this cancer remain challenging. Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, but, currently, no sufficiently effective modalities for its treatment exist. The early diagnosis rate of pancreatic cancer is low and most patients have reached an advanced stage at the time of diagnosis. PDAC evolves from precancerous lesions and is highly aggressive and metastatic. It is essential to understand how the disease progresses and metastasizes. CDKN2A mutations are very common in PDAC. Therefore, here, we have performed a literature review and discuss the role of CDKN2A and some related genes in the development of PDAC, as well as the basis of gene targeting with a correlation coefficient of CDKN2A above 0.9 on the STRING website. It is noteworthy that the interaction of CDKN2A with each gene has been reported in the literature. The role of these genes and CDKN2A in PDAC may provide new directions that will advance the current knowledge base and treatment options since cancer progression is realized through interactions among cells. Our findings provide new insights into the treatment of PADC that can, to some extent, improve the diagnosis rate and quality of life of patients.
Collapse
Affiliation(s)
- Chu Wu
- Cancer Research Institute, Key Laboratory of Tumor Cellular & Molecular Pathology, Medical College of Hengyang, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Ping Yang
- Cancer Research Institute, Key Laboratory of Tumor Cellular & Molecular Pathology, Medical College of Hengyang, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Bingxue Liu
- Cancer Research Institute, Key Laboratory of Tumor Cellular & Molecular Pathology, Medical College of Hengyang, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yunlian Tang
- Cancer Research Institute, Key Laboratory of Tumor Cellular & Molecular Pathology, Medical College of Hengyang, University of South China, Hengyang, Hunan 421001, People's Republic of China
| |
Collapse
|
57
|
Sharma S, Plotkin M. Id1 expression in kidney endothelial cells protects against diabetes-induced microvascular injury. FEBS Open Bio 2020; 10:1447-1462. [PMID: 31957231 PMCID: PMC7396439 DOI: 10.1002/2211-5463.12793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/25/2019] [Accepted: 01/16/2020] [Indexed: 11/08/2022] Open
Abstract
The inhibitor of differentiation (Id) transcription regulators, which are induced in response to oxidative stress, promote cell proliferation and inhibit senescence. Inhibitor of differentiation 1 (Id1) expression is limited to endothelial cells (EC) in the normal mouse kidney and is required for a normal response to injury. Endothelial dysfunction leads to the development of diabetic nephropathy, and so, we hypothesized that endothelial Id1 may help protect against hyperglycemia-induced microvascular injury and nephropathy. Here, we tested this hypothesis by using streptozotocin to induce diabetes in Id1 knockout (KO) mice and WT B6;129 littermates and examining the mice at 3 months. Expression of Id1 was observed to be increased 15-fold in WT kidney EC, and Id1 KO mice exhibited increased mesangial and myofibroblast proliferation, matrix deposition, and albuminuria compared with WT mice. Electron microscopy demonstrated peritubular capillary EC injury and lumen narrowing, and fluorescence microangiography showed a 45% reduction in capillary perfusion area with no reduction in CD31-stained areas in Id1 KO mice. Microarray analysis of EC isolated from WT and KO control and diabetic mice demonstrated activation of senescence pathways in KO cells. Kidneys from KO diabetic mice showed increased histological expression of senescence markers. In addition, premature senescence in cultured KO EC was also seen in response to oxidative stress. In conclusion, endothelial Id1 upregulation with hyperglycemia protects against microvascular injury and senescence and subsequent nephropathy.
Collapse
Affiliation(s)
| | - Matthew Plotkin
- Department of Nephrology, John L. McClellan VA Hospital, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
58
|
Loo TM, Miyata K, Tanaka Y, Takahashi A. Cellular senescence and senescence-associated secretory phenotype via the cGAS-STING signaling pathway in cancer. Cancer Sci 2019; 111:304-311. [PMID: 31799772 PMCID: PMC7004529 DOI: 10.1111/cas.14266] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is historically regarded as a tumor suppression mechanism to prevent damaged cells from aberrant proliferation in benign and premalignant tumors. However, recent findings have suggested that senescent cells contribute to tumorigenesis and age‐associated pathologies through the senescence‐associated secretory phenotype (SASP). Therefore, to control age‐associated cancer, it is important to understand the molecular mechanisms of the SASP in the cancer microenvironment. New findings have suggested that the cyclic GMP‐AMP synthase (cGAS)‐stimulator of interferon genes (STING) signaling pathway, a critical indicator of innate immune response, triggers the SASP in response to accumulation of cytoplasmic DNA (cytoplasmic chromatin fragments, mtDNA and cDNA) in senescent cells. Notably, the cGAS‐STING signaling pathway promotes or inhibits tumorigenesis depending on the biological context in vivo, indicating that it may be a potential therapeutic target for cancer. Herein, we review the regulatory machinery and biological function of the SASP via the cGAS‐STING signaling pathway in cancer.
Collapse
Affiliation(s)
- Tze Mun Loo
- Project for Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kenichi Miyata
- Project for Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoko Tanaka
- Project for Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akiko Takahashi
- Project for Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan.,PRESTO, JST, Saitama, Japan.,PRIME, AMED, Tokyo, Japan
| |
Collapse
|
59
|
KOBASHIGAWA SHINKO, M. SAKAGUCHI YOSHIHIKO, MASUNAGA SHINICHIRO, MORI EIICHIRO. Stress-induced Cellular Senescence Contributes to Chronic Inflammation and Cancer Progression. ACTA ACUST UNITED AC 2019. [DOI: 10.3191/thermalmed.35.41] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- SHINKO KOBASHIGAWA
- Department of Future Basic Medicine, Nara Medical University
- Kyoto University, Institute of Integrated Radiation and Nuclear Science
| | | | | | - EIICHIRO MORI
- Department of Future Basic Medicine, Nara Medical University
| |
Collapse
|
60
|
Inhibitor of DNA-Binding Protein 4 Suppresses Cancer Metastasis through the Regulation of Epithelial Mesenchymal Transition in Lung Adenocarcinoma. Cancers (Basel) 2019; 11:cancers11122021. [PMID: 31847356 PMCID: PMC6966672 DOI: 10.3390/cancers11122021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Metastasis is a predominant cause of cancer death and the major challenge in treating lung adenocarcinoma (LADC). Therefore, exploring new metastasis-related genes and their action mechanisms may provide new insights for developing a new combative approach to treat lung cancer. Previously, our research team discovered that the expression of the inhibitor of DNA binding 4 (Id4) was inversely related to cell invasiveness in LADC cells by cDNA microarray screening. However, the functional role of Id4 and its mechanism of action in lung cancer metastasis remain unclear. In this study, we report that the expression of Id4 could attenuate cell migration and invasion in vitro and cancer metastasis in vivo. Detailed analyses indicated that Id4 could promote E-cadherin expression through the binding of Slug, cause the occurrence of mesenchymal-epithelial transition (MET), and inhibit cancer metastasis. Moreover, the examination of the gene expression database (GSE31210) also revealed that high-level expression of Id4/E-cadherin and low-level expression of Slug were associated with a better clinical outcome in LADC patients. In summary, Id4 may act as a metastatic suppressor, which could not only be used as an independent predictor but also serve as a potential therapeutic for LADC treatment.
Collapse
|
61
|
O'Hara SP, Splinter PL, Trussoni CE, Guicciardi ME, Splinter NP, Al Suraih MS, Nasser-Ghodsi N, Stollenwerk D, Gores GJ, LaRusso NF. The transcription factor ETS1 promotes apoptosis resistance of senescent cholangiocytes by epigenetically up-regulating the apoptosis suppressor BCL2L1. J Biol Chem 2019; 294:18698-18713. [PMID: 31659122 PMCID: PMC6901313 DOI: 10.1074/jbc.ra119.010176] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/15/2019] [Indexed: 12/21/2022] Open
Abstract
Primary sclerosing cholangitis (PSC) is an idiopathic, progressive cholangiopathy. Cholangiocyte senescence is important in PSC pathogenesis, and we have previously reported that senescence is regulated by the transcription factor ETS proto-oncogene 1 (ETS1) and associated with overexpression of BCL2 like 1 (BCL2L1 or BCL-xL), an anti-apoptotic BCL2-family member. Here, we further explored the mechanisms regulating BCL-xL-mediated, apoptosis resistance in senescent cholangiocytes and uncovered that ETS1 and the histone acetyltransferase E1A-binding protein P300 (EP300 or p300) both promote BCL-xL transcription. Using immunofluorescence, we found that BCL-xL protein expression is increased both in cholangiocytes of livers from individuals with PSC and a mouse model of PSC. Using an in vitro model of lipopolysaccharide-induced senescence in normal human cholangiocytes (NHCs), we found increased BCL-xL mRNA and protein levels, and ChIP-PCRs indicated increased occupancy of ETS1, p300, and histone 3 Lys-27 acetylation (H3K27Ac) at the BCL-xL promoter. Using co-immunoprecipitation and proximity ligation assays, we further demonstrate that ETS1 and p300 physically interact in senescent but not control NHCs. Additionally, mutagenesis of predicted ETS1-binding sites within the BCL-xL promoter blocked luciferase reporter activity, and CRISPR/Cas9-mediated genetic deletion of ETS1 reduced senescence-associated BCL-xL expression. In senescent NHCs, TRAIL-mediated apoptosis was reduced ∼70%, and ETS1 deletion or RNAi-mediated BCL-xL suppression increased apoptosis. Overall, our results suggest that ETS1 and p300 promote senescent cholangiocyte resistance to apoptosis by modifying chromatin and inducing BCL-xL expression. These findings reveal ETS1 as a central regulator of both cholangiocyte senescence and the associated apoptosis-resistant phenotype.
Collapse
Affiliation(s)
- Steven P O'Hara
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905.
| | - Patrick L Splinter
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Christy E Trussoni
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Maria Eugenia Guicciardi
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Noah P Splinter
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Mohammed S Al Suraih
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Navine Nasser-Ghodsi
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Deborah Stollenwerk
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Nicholas F LaRusso
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
62
|
Ohtani N. Deciphering the mechanism for induction of senescence-associated secretory phenotype (SASP) and its role in ageing and cancer development. J Biochem 2019; 166:289-295. [PMID: 31297533 DOI: 10.1093/jb/mvz055] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/10/2019] [Indexed: 12/17/2023] Open
Abstract
Cellular senescence is an irreversible form of cell cycle arrest that can be induced by persistent DNA damage, and is well known to function as an important tumour suppression mechanism. Cellular senescence is detected in aged organisms; thus, it is also recognized as a hallmark of organismal ageing. Unlike apoptotic cells, senescent cells can survive for long periods of time. Recently, it has been shown that the late stage of senescent cells are capable of expressing a variety of secreted proteins such as cytokines, chemokines and proteases, and this condition is now known as senescence-associated secretory phenotype (SASP). These secreted factors are involved in myriad of physiological functions including tissue repair and clearance of damaged cells. Alternatively, these factors may promote detrimental effects, such as chronic inflammation or cancer progression, should the SASP persist. Recent scientific advances have indicated that innate immune responses, particularly involving the cGAS-STING pathway, trigger SASP induction. Therefore, developing a strategy to regulate SASP may provide scientific insights for the management of age-associated diseases and the implementation of healthy ageing in the future.
Collapse
Affiliation(s)
- Naoko Ohtani
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, 1-4-3, Asahi-machi, Abeno-ku, Osaka, Japan
| |
Collapse
|
63
|
Henry AP, Probert K, Stewart CE, Thakker D, Bhaker S, Azimi S, Hall IP, Sayers I. Defining a role for lung function associated gene GSTCD in cell homeostasis. Respir Res 2019; 20:172. [PMID: 31370853 PMCID: PMC6676530 DOI: 10.1186/s12931-019-1146-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022] Open
Abstract
Genome wide association (GWA) studies have reproducibly identified signals on chromosome 4q24 associated with lung function and COPD. GSTCD (Glutathione S-transferase C-terminal domain containing) represents a candidate causal gene in this locus, however little is currently known about the function of this protein. We set out to further our understanding of the role of GSTCD in cell functions and homeostasis using multiple molecular and cellular approaches in airway relevant cells. Recombinant expression of human GSTCD in conjunction with a GST activity assay did not identify any enzymatic activity for two GSTCD isoforms questioning the assignment of this protein to this family of enzymes. Protein structure analyses identified a potential methyltransferase domain contained within GSTCD, with these enzymes linked to cell viability and apoptosis. Targeted knockdown (siRNA) of GSTCD in bronchial epithelial cells identified a role for GSTCD in cell viability as proliferation rates were not altered. To provide greater insight we completed transcriptomic analyses on cells with GSTCD expression knocked down and identified several differentially expressed genes including those implicated in airway biology; fibrosis e.g. TGFBR1 and inflammation e.g. IL6R. Pathway based transcriptomic analyses identified an over-representation of genes related to adipogenesis which may suggest additional functions for GSTCD. These findings identify potential additional functions for GSTCD in the context of airway biology beyond the hypothesised GST activity and warrant further investigation.
Collapse
Affiliation(s)
- Amanda P Henry
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK.
| | - Kelly Probert
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Ceri E Stewart
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Dhruma Thakker
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Sangita Bhaker
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Sheyda Azimi
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Ian P Hall
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Ian Sayers
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
64
|
PRMT7 methylates and suppresses GLI2 binding to SUFU thereby promoting its activation. Cell Death Differ 2019; 27:15-28. [PMID: 31000813 DOI: 10.1038/s41418-019-0334-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 03/25/2019] [Accepted: 04/08/2019] [Indexed: 01/20/2023] Open
Abstract
Cellular senescence is implicated in aging or age-related diseases. Sonic hedgehog (Shh) signaling, an inducer of embryonic development, has recently been demonstrated to inhibit cellular senescence. However, the detailed mechanisms to activate Shh signaling to prevent senescence is not well understood. Here, we demonstrate that Protein arginine methyltransferase 7 (PRMT7) promotes Shh signaling via GLI2 methylation which is critical for suppression of cellular senescence. PRMT7-deficient mouse embryonic fibroblasts (MEFs) exhibited a premature cellular senescence with accompanied increase in the cell cycle inhibitors p16 and p21. PRMT7 depletion results in reduced Shh signaling activity in MEFs while PRMT7 overexpression enhances GLI2-reporter activities that are sensitive to methylation inhibition. PRMT7 interacts with and methylates GLI2 on arginine residues 225 and 227 nearby a binding region of SUFU, a negative regulator of GLI2. This methylation interferes with GLI2-SUFU binding, leading to facilitation of GLI2 nuclear accumulation and Shh signaling. Taken together, these data suggest that PRMT7 induces GLI2 methylation, reducing its binding to SUFU and increasing Shh signaling, ultimately leading to prevention of cellular senescence.
Collapse
|
65
|
Liu X, Wan M. A tale of the good and bad: Cell senescence in bone homeostasis and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 346:97-128. [PMID: 31122396 DOI: 10.1016/bs.ircmb.2019.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Historically, cellular senescence has been viewed as an irreversible cell-cycle arrest process with distinctive phenotypic alterations that were implicated primarily in aging and tumor suppression. Recent discoveries suggest that cellular senescence represents a series of diverse, dynamic, and heterogeneous cellular states with the senescence-associated secretory phenotype (SASP). Although senescent cells typically contribute to aging and age-related diseases, accumulating evidence has shown that they also have important physiological functions during embryonic development, late pubertal bone growth cessation, and adulthood tissue remodeling. Here, we review the recent research on cellular senescence and SASP, highlighting the key pathways that mediate senescence cell-cycle arrest and initiate SASP. We also summarize recent literature on the role of cellular senescence in maintaining bone homeostasis and mediating age-associated osteoporosis, discussing both the beneficial and adverse roles of cellular senescence in bone during different physiological stages, including bone development, childhood bone growth, adulthood bone remodeling, and bone aging.
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mei Wan
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
66
|
Wu XL, Wang LK, Xue J, Dai YJ, Yang DD, Qu M, Guo F, Han L, Sun GY. A study of the impact of inhibitors of DNA binding-1 on proliferation and migration in human colon carcinoma cells. Kaohsiung J Med Sci 2019; 35:209-213. [PMID: 30887652 DOI: 10.1002/kjm2.12037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/09/2019] [Indexed: 11/09/2022] Open
Abstract
This study aims to explore the effect of an inhibitor of DNA binding-1 (Id-1) on the proliferation and migration of human colon carcinoma cell line SW480 and HT-29. SW480 and HT-29 cells transfected with Id-1-interference sequence were assigned to the experimental groups (inhibition groups 1 and 2), and SW480 and HT-29 cells with blank interference sequence (blank groups) and blank load transfection (blank load groups) were assigned as the control groups. The expression of Id-1 in six groups was detected by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot. Cell proliferation in vitro was assessed by MTT assay. RT-PCR and Western blot results demonstrated that the mRNA and protein expressions of Id-1 in the inhibition group 1 were lower than those in the blank group 1 and blank load group 1. RT-PCR and Western blot results revealed that the mRNA and protein expressions of Id-1 were lower in the inhibition group 2 than in the blank group 2 and blank load group 2. The results of the growth curve revealed that proliferation ability was significantly weaker from the third day in the inhibition groups 1 and 2 than in the blank group and blank load group. Transwell chamber experiment and Matrigel invasion assay revealed that the number of Transwell cells significantly decreased in the inhibition groups 1 and 2 than in the blank groups and blank load groups (P < 0.01). Id-1 significantly promotes the proliferation and migration of human colon carcinoma cell lines SW480 and HT-29.
Collapse
Affiliation(s)
- Xue-Liang Wu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Li-Kun Wang
- Department of Ultrasound, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Jun Xue
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yong-Jun Dai
- Department of Gastrointestinal Surgery, The first Affiliated Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Dong-Dong Yang
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Ming Qu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Fei Guo
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Lei Han
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Guang-Yuan Sun
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| |
Collapse
|
67
|
Tabasso AFS, Jones DJL, Jones GDD, Macip S. Radiotherapy-Induced Senescence and its Effects on Responses to Treatment. Clin Oncol (R Coll Radiol) 2019; 31:283-289. [PMID: 30826201 DOI: 10.1016/j.clon.2019.02.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 12/24/2022]
Abstract
Radiotherapy is still a treatment of choice for many malignancies, often in combination with other strategies. However, its efficacy is limited by the dose that can be safely administered without eliciting serious side-effects, as well as the fact that recurrence is common, particularly in large tumours. Combining radiotherapy with drugs that could sensitise cells to radiation and/or reduce the factors that promote the recovery of the surviving cancer cells is a promising approach. Ionising radiation has been shown to induce senescence and the accumulation of senescent cells creates a microenvironment that facilitates neoplastic growth. This provides a rationale to test the addition of anti-senescent drugs, some of which are already available in the clinic, to radiotherapy protocols. Here, we discuss the relevance of radiotherapy-induced senescent cell accumulation and the potential interventions to minimise its negative effects.
Collapse
Affiliation(s)
- A F S Tabasso
- Leicester Cancer Research Centre, Leicester Royal Infirmary, University of Leicester, Leicester, UK; Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - D J L Jones
- Leicester Cancer Research Centre, Leicester Royal Infirmary, University of Leicester, Leicester, UK
| | - G D D Jones
- Leicester Cancer Research Centre, Leicester Royal Infirmary, University of Leicester, Leicester, UK
| | - S Macip
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK.
| |
Collapse
|
68
|
Zhao W, Zhang S, Wang X, Ma X, Huang B, Chen H, Chen D. ETS1 targets RYBP transcription to inhibit tumor cell proliferation. Biochem Biophys Res Commun 2019; 509:810-816. [DOI: 10.1016/j.bbrc.2019.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 01/02/2019] [Indexed: 12/27/2022]
|
69
|
Calcinotto A, Kohli J, Zagato E, Pellegrini L, Demaria M, Alimonti A. Cellular Senescence: Aging, Cancer, and Injury. Physiol Rev 2019; 99:1047-1078. [PMID: 30648461 DOI: 10.1152/physrev.00020.2018] [Citation(s) in RCA: 706] [Impact Index Per Article: 117.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a permanent state of cell cycle arrest that occurs in proliferating cells subjected to different stresses. Senescence is, therefore, a cellular defense mechanism that prevents the cells to acquire an unnecessary damage. The senescent state is accompanied by a failure to re-enter the cell cycle in response to mitogenic stimuli, an enhanced secretory phenotype and resistance to cell death. Senescence takes place in several tissues during different physiological and pathological processes such as tissue remodeling, injury, cancer, and aging. Although senescence is one of the causative processes of aging and it is responsible of aging-related disorders, senescent cells can also play a positive role. In embryogenesis and tissue remodeling, senescent cells are required for the proper development of the embryo and tissue repair. In cancer, senescence works as a potent barrier to prevent tumorigenesis. Therefore, the identification and characterization of key features of senescence, the induction of senescence in cancer cells, or the elimination of senescent cells by pharmacological interventions in aging tissues is gaining consideration in several fields of research. Here, we describe the known key features of senescence, the cell-autonomous, and noncell-autonomous regulators of senescence, and we attempt to discuss the functional role of this fundamental process in different contexts in light of the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Arianna Calcinotto
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Jaskaren Kohli
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Elena Zagato
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Laura Pellegrini
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Marco Demaria
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| |
Collapse
|
70
|
The dynamic nature of senescence in cancer. Nat Cell Biol 2019; 21:94-101. [PMID: 30602768 DOI: 10.1038/s41556-018-0249-2] [Citation(s) in RCA: 384] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022]
Abstract
Cellular senescence is implicated in physiological and pathological processes spanning development, wound healing, age-related decline in organ functions and cancer. Here, we discuss cell-autonomous and non-cell-autonomous properties of senescence in the context of tumour formation and anticancer therapy, and characterize these properties, such as reprogramming into stemness, tissue remodelling and immune crosstalk, as far more dynamic than suggested by the common view of senescence as an irreversible, static condition.
Collapse
|
71
|
Jiao Y, Feng Y, Wang X. Regulation of Tumor Suppressor Gene CDKN2A and Encoded p16-INK4a Protein by Covalent Modifications. BIOCHEMISTRY (MOSCOW) 2018; 83:1289-1298. [PMID: 30482142 DOI: 10.1134/s0006297918110019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
CDKN2A is one of the most studied tumor suppressor genes. It encodes the p16-INK4a protein that plays a critical role in the cell cycle progression, differentiation, senescence, and apoptosis. Mutations in CDKN2A or dysregulation of its functional activity are frequently associated with various types of human cancer. As a cyclin-dependent kinase inhibitor, p16-INK4a forms a complex with cyclin-dependent kinases 4/6 (CDK4/6) thereby competing with cyclin D. It is believed that the helix-turn-helix structures in the content of tandem ankyrin repeats in p16-INK4a are required for the protein interaction with CDK4. Until recently, the mechanisms considered to be involved in the regulation of p16-INK4a functions and cancer development have been mutations in DNA, homozygous or heterozygous gene loss, and methylation of CDKN2A promoter region. In this review, we discuss recent findings on the regulation of p16-INK4a by covalent modifications at both transcriptional and post-translational levels.
Collapse
Affiliation(s)
- Yang Jiao
- School of Physical Education, Northeast Normal University, Changchun, Jilin, 130024, P. R. China
| | - Yunpeng Feng
- Key Laboratory of Molecular Epigenetics, Ministry of Education, Northeast Normal University, Changchun, Jilin, 130024, P. R. China
| | - Xiuli Wang
- Central Laboratory of General Biology, School of Life Sciences, Northeast Normal University, Changchun, Jilin, 130024, P. R. China.
| |
Collapse
|
72
|
Milanovic M, Yu Y, Schmitt CA. The Senescence-Stemness Alliance - A Cancer-Hijacked Regeneration Principle. Trends Cell Biol 2018; 28:1049-1061. [PMID: 30253901 DOI: 10.1016/j.tcb.2018.09.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/27/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022]
Abstract
Activated oncogenes or anticancer therapies evoke senescent cell-cycle arrest in (pre-)malignant cells, thereby interrupting tumor formation or progression. Physiologically, cellular senescence contributes to embryonic development and tissue regeneration. These observations and the overlap of numerous gene products in senescence and stem cell signaling prompted investigations into whether epigenetic establishment of the senescent state may concomitantly reprogram the cell into a latent stem-like condition, whose functional impact becomes evident when arrested cells resume proliferation. We review here recent discoveries underscoring the unexpected senescence-stemness alliance, elucidate underlying molecular mechanisms, and discuss its fundamentally different implications in normal tissue repair - to replenish the exhausted repopulation capacity - as compared to cancer biology, where usurpation of this natural principle accounts for particularly aggressive tumor behavior.
Collapse
Affiliation(s)
- Maja Milanovic
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Medical Department of Hematology, Oncology and Tumor Immunology, and Molekulares Krebsforschungszentrum (MKFZ), Virchow Campus, 13353 Berlin, Germany
| | - Yong Yu
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Clemens A Schmitt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Medical Department of Hematology, Oncology and Tumor Immunology, and Molekulares Krebsforschungszentrum (MKFZ), Virchow Campus, 13353 Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125 Berlin, Germany; Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium), Partner Site Berlin, Germany; Berlin Institute of Health, Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany.
| |
Collapse
|
73
|
Zhang L, Zhang S, Li A, Zhang A, Zhang S, Chen L. DPY30 is required for the enhanced proliferation, motility and epithelial-mesenchymal transition of epithelial ovarian cancer cells. Int J Mol Med 2018; 42:3065-3072. [PMID: 30221689 PMCID: PMC6202113 DOI: 10.3892/ijmm.2018.3869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/28/2018] [Indexed: 12/20/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is one of the most lethal gynecological malignancies and is known to be associated with the accumulation of various genetic and epigenetic alterations. As a member of the human histone-lysine N-methyltransferase SETD1A (SET1)/histone-lysine N-methyltransferase 2A (MLL) complexes that are required for full SET1/MLL methyltransferase activity, protein dpy-30 homolog (DPY30) catalyzes histone H3K4 methylation, and its dysfunction has been associated with the occurrence of cancer. Therefore, the present study investigated the role of DPY30 in EOC and the potential association between DPY30 expression and the clinicopathological characteristics of EOC. The expression of DPY30 was examined in EOC tissues and cell lines to identify any correlations between the clinico-pathological characteristics of EOC and DPY30 expression, and to determine the effects of DPY30 on EOC cell proliferation, migration and invasion. DPY30 was highly expressed in EOC tissues and cell lines, and high DPY30 expression was significantly associated with notable clinicopathological variables in EOC patients, including International Federation of Gynecology and Obstetrics stage, pathological grade and lymph node metastasis. Functional studies on EOC cell lines demonstrated that DPY30 significantly promoted cell proliferation, migration, and invasion, accelerated cell cycle progression, and promoted epithelial-mesenchymal transition. Chromatin immunoprecipitation assay results revealed that DPY30 regulates histone H3K4 modification via interaction with the vimentin gene promoter, suggesting that DPY30 promotes the transcription of vimentin. Finally, high expression of DPY30 was significantly associated with reduced survival in patients with EOC. The results indicated that DPY30 may act as an oncogene in EOC and thus represents a potential therapeutic target and prognostic marker in EOC.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shuguang Zhang
- Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Aihua Li
- Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Anqi Zhang
- Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Shiqian Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Liang Chen
- Department of Gynecological Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
74
|
Kalimutho M, Sinha D, Jeffery J, Nones K, Srihari S, Fernando WC, Duijf PH, Vennin C, Raninga P, Nanayakkara D, Mittal D, Saunus JM, Lakhani SR, López JA, Spring KJ, Timpson P, Gabrielli B, Waddell N, Khanna KK. CEP55 is a determinant of cell fate during perturbed mitosis in breast cancer. EMBO Mol Med 2018; 10:e8566. [PMID: 30108112 PMCID: PMC6127888 DOI: 10.15252/emmm.201708566] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 07/15/2018] [Accepted: 07/18/2018] [Indexed: 12/28/2022] Open
Abstract
The centrosomal protein, CEP55, is a key regulator of cytokinesis, and its overexpression is linked to genomic instability, a hallmark of cancer. However, the mechanism by which it mediates genomic instability remains elusive. Here, we showed that CEP55 overexpression/knockdown impacts survival of aneuploid cells. Loss of CEP55 sensitizes breast cancer cells to anti-mitotic agents through premature CDK1/cyclin B activation and CDK1 caspase-dependent mitotic cell death. Further, we showed that CEP55 is a downstream effector of the MEK1/2-MYC axis. Blocking MEK1/2-PLK1 signaling therefore reduced outgrowth of basal-like syngeneic and human breast tumors in in vivo models. In conclusion, high CEP55 levels dictate cell fate during perturbed mitosis. Forced mitotic cell death by blocking MEK1/2-PLK1 represents a potential therapeutic strategy for MYC-CEP55-dependent basal-like, triple-negative breast cancers.
Collapse
Affiliation(s)
- Murugan Kalimutho
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- School of Natural Sciences, Griffith University, Nathan, Qld, Australia
| | - Debottam Sinha
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- School of Natural Sciences, Griffith University, Nathan, Qld, Australia
| | - Jessie Jeffery
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Qld, Australia
| | - Sriganesh Srihari
- Computational Systems Biology Laboratory, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Qld, Australia
| | | | - Pascal Hg Duijf
- University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Claire Vennin
- Cancer Division, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Prahlad Raninga
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | | | - Deepak Mittal
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Jodi M Saunus
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- Centre for Clinical Research, The University of Queensland, Herston, Qld, Australia
| | - Sunil R Lakhani
- Centre for Clinical Research, The University of Queensland, Herston, Qld, Australia
- School of Medicine, The University of Queensland, Herston, Qld, Australia
- Pathology Queensland, The Royal Brisbane and Women's Hospital, Herston, Qld, Australia
| | - J Alejandro López
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- School of Natural Sciences, Griffith University, Nathan, Qld, Australia
| | - Kevin J Spring
- Liverpool Clinical School, University of Western Sydney, Liverpool, NSW, Australia
- Ingham Institute, Liverpool Hospital, Liverpool, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW, Australia
| | - Paul Timpson
- Cancer Division, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Brian Gabrielli
- University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| |
Collapse
|
75
|
Niu LL, Cheng CL, Li MY, Yang SL, Hu BG, Chong CCN, Chan SL, Ren J, Chen GG, Lai PBS. ID1-induced p16/IL6 axis activation contributes to the resistant of hepatocellular carcinoma cells to sorafenib. Cell Death Dis 2018; 9:852. [PMID: 30154433 PMCID: PMC6113298 DOI: 10.1038/s41419-018-0926-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
Sorafenib is the only approved drug for the treatment of advanced hepatocellular carcinoma (HCC). However, its efficacy is limited by the emergence of primary and/or acquired resistance. Senescence-associated secretory phenotype (SASP)-mediated chemo-resistance, which depends on the secreted bioactive molecules, has attracted increasing attention but never revealed in HCC. In this study, we investigated the effect of SASP-related p16/IL6 axis on sorafenib resistance in HCC. Initially, we noticed that HCC cells with a high level of p16/IL6 axis exhibited a low sensitivity to sorafenib. Further in vivo and in vitro studies demonstrated that such a primary resistance resulted from ID1-mediated activation of p16/IL6 axis. Overexpression of ID1 or IL6 blocking in sorafenib-resistant HCC cells could increase the cytotoxicity of sorafenib. Moreover, SASP-related p16/IL6 axis contributed to the formation of acquired resistance in cells received long-term exposure to sorafenib. In acquired sorafenib-resistant cells, ID1 low expression, p16/IL6 axis up-regulation, and AKT phosphorylation activation were observed. A reduced cytotoxicity of sorafenib was detected when sorafenib-sensitive cells incubated with conditioned media from the resistant cells, accompanied by the stimulation of AKT phosphorylation. The reversal of sorafenib resistance could be achieved through ID1 overexpression, IL6 blocking, and AKT pathway inhibition. Our study reveals that SASP-related p16/IL6 axis activation is responsible for sorafenib resistance, which will be a novel strategy to prevent the drug resistance.
Collapse
Affiliation(s)
- Lei-Lei Niu
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.,Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Chuan-le Cheng
- Department of Thoracic Surgery, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, China
| | - Ming-Yue Li
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Sheng-Li Yang
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.,Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bao-Guang Hu
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.,Department of Gastrointestinal Surgery, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Charing C N Chong
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Stephen L Chan
- Department of Clinical Oncology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - Jianwei Ren
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - George G Chen
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| | - Paul B S Lai
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China.
| |
Collapse
|
76
|
Senescence-associated ribosome biogenesis defects contributes to cell cycle arrest through the Rb pathway. Nat Cell Biol 2018; 20:789-799. [PMID: 29941930 DOI: 10.1038/s41556-018-0127-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/21/2018] [Indexed: 01/05/2023]
Abstract
Cellular senescence is a tumour suppressor programme characterized by a stable cell cycle arrest. Here we report that cellular senescence triggered by a variety of stimuli leads to diminished ribosome biogenesis and the accumulation of both rRNA precursors and ribosomal proteins. These defects were associated with reduced expression of several ribosome biogenesis factors, the knockdown of which was also sufficient to induce senescence. Genetic analysis revealed that Rb but not p53 was required for the senescence response to altered ribosome biogenesis. Mechanistically, the ribosomal protein S14 (RPS14 or uS11) accumulates in the soluble non-ribosomal fraction of senescent cells, where it binds and inhibits CDK4 (cyclin-dependent kinase 4). Overexpression of RPS14 is sufficient to inhibit Rb phosphorylation, inducing cell cycle arrest and senescence. Here we describe a mechanism for maintaining the senescent cell cycle arrest that may be relevant for cancer therapy, as well as biomarkers to identify senescent cells.
Collapse
|
77
|
Fry EA, Inoue K. Aberrant expression of ETS1 and ETS2 proteins in cancer. CANCER REPORTS AND REVIEWS 2018; 2:10.15761/CRR.1000151. [PMID: 29974077 PMCID: PMC6027756 DOI: 10.15761/crr.1000151] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The ETS transcription factors regulate expression of genes involved in normal cell development, proliferation, differentiation, angiogenesis, and apoptosis, consisting of 28 family members in humans. Dysregulation of these transcription factors facilitates cell proliferation in cancers, and several members participate in invasion and metastasis by activating gene transcription. ETS1 and ETS2 are the founding members of the ETS family and regulate transcription by binding to ETS sequences. They are both involved in oncogenesis and tumor suppression depending on the biological situations used. The essential roles of ETS proteins in human telomere maintenance have been suggested, which have been linked to creation of new Ets binding sites. Recently, preferential binding of ETS2 to gain-of-function mutant p53 and ETS1 to wild type p53 (WTp53) has been suggested, raising the tumor promoting role for the former and tumor suppressive role for the latter. The oncogenic and tumor suppressive functions of ETS1 and 2 proteins have been discussed.
Collapse
Affiliation(s)
- Elizabeth A. Fry
- The Dept. of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| | - Kazushi Inoue
- The Dept. of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| |
Collapse
|
78
|
Baker DJ, Petersen RC. Cellular senescence in brain aging and neurodegenerative diseases: evidence and perspectives. J Clin Invest 2018; 128:1208-1216. [PMID: 29457783 PMCID: PMC5873891 DOI: 10.1172/jci95145] [Citation(s) in RCA: 307] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Along with a general decline in overall health, most chronic degenerative human diseases are inherently associated with increasing age. Age-associated cognitive impairments and neurodegenerative diseases, such as Parkinson's and Alzheimer's diseases, are potentially debilitating conditions that lack viable options for treatment, resulting in a tremendous economic and societal cost. Most high-profile clinical trials for neurodegenerative diseases have led to inefficacious results, suggesting that novel approaches to treating these pathologies are needed. Numerous recent studies have demonstrated that senescent cells, which are characterized by sustained cell cycle arrest and production of a distinct senescence-associated secretory phenotype, accumulate with age and at sites of age-related diseases throughout the body, where they actively promote tissue deterioration. Cells with features of senescence have been detected in the context of brain aging and neurodegenerative disease, suggesting that they may also promote dysfunction. Here, we discuss the evidence implicating senescent cells in neurodegenerative diseases, the mechanistic contribution of these cells that may actively drive neurodegeneration, and how these cells or their effects may be targeted therapeutically.
Collapse
Affiliation(s)
- Darren J. Baker
- Department of Biochemistry and Molecular Biology
- Department of Pediatric and Adolescent Medicine, and
| | | |
Collapse
|
79
|
Du WW, Yang W, Chen Y, Wu ZK, Foster FS, Yang Z, Li X, Yang BB. Foxo3 circular RNA promotes cardiac senescence by modulating multiple factors associated with stress and senescence responses. Eur Heart J 2018; 38:1402-1412. [PMID: 26873092 DOI: 10.1093/eurheartj/ehw001] [Citation(s) in RCA: 348] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 12/31/2015] [Indexed: 12/20/2022] Open
Abstract
Aims Circular RNAs are a subclass of non-coding RNAs detected within mammalian cells. This study was designed to test the roles of a circular RNA circ-Foxo3 in senescence using in vitro and in vivo approaches. Methods and results Using the approaches of molecular and cellular biology, we show that a circular RNA generated from a member of the forkhead family of transcription factors, Foxo3, namely circ-Foxo3, was highly expressed in heart samples of aged patients and mice, which was correlated with markers of cellular senescence. Doxorubicin-induced cardiomyopathy was aggravated by ectopic expression of circ-Foxo3 but was relieved by silencing endogenous circ-Foxo3. We also found that silencing circ-Foxo3 inhibited senescence of mouse embryonic fibroblasts and that ectopic expression of circ-Foxo3 induced senescence. We found that circ-Foxo3 was mainly distributed in the cytoplasm, where it interacted with the anti-senescent protein ID-1 and the transcription factor E2F1, as well as the anti-stress proteins FAK and HIF1α. Conclusion We conclude that ID-1, E2F1, FAK, and HIF1α interact with circ-Foxo3 and are retained in the cytoplasm and could no longer exert their anti-senescent and anti-stress roles, resulting in increased cellular senescence.
Collapse
Affiliation(s)
- William W Du
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, S-Wing Research Building, 2075 Bayview Ave, Toronto M4N 3M5, Canada
| | - Weining Yang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Yu Chen
- 2nd Department of Cardiac Surgery, The First Hospital, Sun Yet-Sen University, Guangzhou, China
| | - Zhong-Kai Wu
- 2nd Department of Cardiac Surgery, The First Hospital, Sun Yet-Sen University, Guangzhou, China
| | | | - Zhenguo Yang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, S-Wing Research Building, 2075 Bayview Ave, Toronto M4N 3M5, Canada
| | - Xiangmin Li
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, S-Wing Research Building, 2075 Bayview Ave, Toronto M4N 3M5, Canada.,State Key Laboratory of Applied Microbiology Southern China (The Ministry-Province Joint Development), Guangdong Institute of Microbiology, Guangzhou 510070, PR China
| | - Burton B Yang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, S-Wing Research Building, 2075 Bayview Ave, Toronto M4N 3M5, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| |
Collapse
|
80
|
Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of Cellular Senescence. Trends Cell Biol 2018; 28:436-453. [PMID: 29477613 DOI: 10.1016/j.tcb.2018.02.001] [Citation(s) in RCA: 1500] [Impact Index Per Article: 214.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a permanent state of cell cycle arrest that promotes tissue remodeling during development and after injury, but can also contribute to the decline of the regenerative potential and function of tissues, to inflammation, and to tumorigenesis in aged organisms. Therefore, the identification, characterization, and pharmacological elimination of senescent cells have gained attention in the field of aging research. However, the nonspecificity of current senescence markers and the existence of different senescence programs strongly limit these tasks. Here, we describe the molecular regulators of senescence phenotypes and how they are used for identifying senescent cells in vitro and in vivo. We also highlight the importance that these levels of regulations have in the development of therapeutic targets.
Collapse
Affiliation(s)
- Alejandra Hernandez-Segura
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jamil Nehme
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
81
|
Abstract
The ARF and INK4a genes are located in the same CDKN2a locus, both showing its tumor suppressive activity. ARF has been shown to detect potentially harmful oncogenic signals, making incipient cancer cells undergo senescence or apoptosis. INK4a, on the other hand, responds to signals from aging in a variety of tissues including islets of Langerhans, neuronal cells, and cancer stem cells in general. It also detects oncogenic signals from incipient cancer cells to induce them senescent to prevent neoplastic transformation. Both of these genes are inactivated by gene deletion, promoter methylation, frame shift, and aberrant splicing although mutations changing the amino acid sequences affect only the latter. Recent studies indicated that polycomb gene products EZH2 and BMI1 repressed p16INK4a expression in primary cells, but not in cells deficient for pRB protein function. It was also reported that that p14ARF inhibits the stability of the p16INK4a protein in human cancer cell lines and mouse embryonic fibroblasts through its interaction with regenerating islet-derived protein 3γ. Overexpression of INK4a is associated with better prognosis of cancer when it is associated with human papilloma virus infection. However, it has a worse prognostic value in other tumors since it is an indicator of pRB loss. The p16INK4a tumor suppressive protein can thus be used as a biomarker to detect early stage cancer cells as well as advanced tumor cells with pRB inactivation since it is not expressed in normal cells.
Collapse
Affiliation(s)
- Kazushi Inoue
- The Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, NC 27157
| | - Elizabeth A Fry
- The Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, NC 27157
| |
Collapse
|
82
|
Kohli JS, Mir H, Wasif A, Chong H, Akhras V, Kumar R, Nagore E, Bennett DC. ETS1, nucleolar and non-nucleolar TERT expression in nevus to melanoma progression. Oncotarget 2017; 8:104408-104417. [PMID: 29262649 PMCID: PMC5732815 DOI: 10.18632/oncotarget.22254] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/03/2017] [Indexed: 11/25/2022] Open
Abstract
TERT (telomerase reverse transcriptase) is the catalytic component of telomerase. TERT shows little expression in normal somatic cells but is commonly re-expressed in cancers, facilitating immortalization. Recently-discovered TERT promoter mutations create binding sites for ETS-family transcription factors to upregulate TERT. ETS1 is reported to be important for TERT upregulation in melanoma. However it is unclear when in melanoma progression TERT and ETS1 proteins are expressed. To elucidate this question, ETS1 and TERT immunohistochemistry were performed on a panel of benign (n=27) and dysplastic nevi (n=34), radial growth phase (n=29), vertical growth phase (n=25) and metastatic melanomas (n=27). Lesions were scored by percentage of positive cells. ETS1 was readily detectable in all lesions, but not in normal melanocytes. TERT was located in either the nucleolus, the nucleoplasm (non-nucleolar) or both. Non-nucleolar TERT increased in prevalence with progression, from 19% of benign nevi to 78% of metastases. It did not however correlate with cell proliferation (Ki-67 immunostaining), nor differ significantly in prevalence between primary melanomas with or without a TERT promoter mutation. These results demonstrate that ETS1 is expressed very early in melanoma progression, and interestingly only non-nucleolar TERT correlates clearly in prevalence with melanoma progression. It can be acquired at various stages and by mechanisms other than promoter mutations.
Collapse
Affiliation(s)
- Jaskaren S. Kohli
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, UK
- Current/Present address: European Research Institute for The Biology of Aging, University Medical Center Groningen, Groningen, The Netherlands
| | - Hira Mir
- Department of Cellular Pathology, St George’s University Hospitals NHS Foundation Trust, London, UK
- Current/Present address: King’s College Hospital Foundation Trust, London, UK
| | - Afsheen Wasif
- Department of Cellular Pathology, St George’s University Hospitals NHS Foundation Trust, London, UK
- Current/Present address: King’s College Hospital Foundation Trust, London, UK
| | - Heung Chong
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, UK
- Department of Cellular Pathology, St George’s University Hospitals NHS Foundation Trust, London, UK
| | - Victoria Akhras
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, UK
- Department of Dermatology, St George’s University Hospitals NHS Foundation Trust, London, UK
| | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Eduardo Nagore
- Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Dorothy C. Bennett
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, UK
| |
Collapse
|
83
|
Nakade K, Lin CS, Chen XY, Tsai MH, Wuputra K, Zhu ZW, Pan JZ, Yokoyama KK. Jun dimerization protein 2 controls hypoxia-induced replicative senescence via both the p16 Ink4a-pRb and Arf-p53 pathways. FEBS Open Bio 2017; 7:1793-1804. [PMID: 29123987 PMCID: PMC5666393 DOI: 10.1002/2211-5463.12325] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 09/13/2017] [Accepted: 09/22/2017] [Indexed: 11/08/2022] Open
Abstract
The main regulators of replicative senescence in mice are p16Ink4a and Arf, inhibitors of cell cycle progression. Jun dimerization protein 2 (JDP2)-deficient mouse embryonic fibroblasts are resistant to replicative senescence through recruitment of the Polycomb repressive complexes 1 and 2 to the promoter of the gene that encodes p16Ink4a and inhibits the methylation of lysine 27 of the histone H3 locus. However, whether or not JDP2 is able to regulate the chromatin signaling of either p16Ink4a-pRb or Arf-p53, or both, in response to oxidative stress remains elusive. Thus, this study sought to clarify this point. We demonstrated that the introduction of JDP2 leads to upregulation of p16Ink4a and Arf and decreases cell proliferation in the presence of environmental (20% O2), but not in low (3% O2) oxygen. JDP2-mediated growth suppression was inhibited by the downregulation of both p16Ink4a and Arf. Conversely, the forced expression of p16Ink4a or Arf inhibited cell growth even in the absence of JDP2. The downregulation of both the p53 and pRb pathways, but not each individually, was sufficient to block JDP2-dependent growth inhibition. These data suggest that JDP2 induces p16Ink4a and Arf by mediating signals from oxidative stress, resulting in cell cycle arrest via both the p16Ink4a-pRb and Arf-p53 pathways.
Collapse
Affiliation(s)
- Koji Nakade
- Gene Engineering Division RIKEN BioResource Center Tsukuba Japan
| | - Chang-Shen Lin
- Graduate Institute of Medicine Kaohsiung Medical University Taiwan.,Department of Biological Sciences National Sun Yat-sen University Kaohsiung Taiwan
| | - Xiao-Yu Chen
- Institute of Animal Husbandry and Veterinary Zhe Jiang Academy of Agricultural Sciences China
| | - Ming-Ho Tsai
- Graduate Institute of Medicine Kaohsiung Medical University Taiwan.,Center for Stem Cell Research Kaohsiung Medical University Taiwan.,Center of Infectious Disease and Cancer Research Kaohsiung Medical University Taiwan.,Center for Environmental Medicine Kaohsiung Medical University Taiwan
| | - Kenly Wuputra
- Graduate Institute of Medicine Kaohsiung Medical University Taiwan.,Center for Stem Cell Research Kaohsiung Medical University Taiwan.,Center of Infectious Disease and Cancer Research Kaohsiung Medical University Taiwan.,Center for Environmental Medicine Kaohsiung Medical University Taiwan
| | - Zhi-Wei Zhu
- Institute of Animal Husbandry and Veterinary Zhe Jiang Academy of Agricultural Sciences China
| | - Jian-Zhi Pan
- Gene Engineering Division RIKEN BioResource Center Tsukuba Japan.,Institute of Animal Husbandry and Veterinary Zhe Jiang Academy of Agricultural Sciences China
| | - Kazunari K Yokoyama
- Graduate Institute of Medicine Kaohsiung Medical University Taiwan.,Center for Stem Cell Research Kaohsiung Medical University Taiwan.,Center of Infectious Disease and Cancer Research Kaohsiung Medical University Taiwan.,Center for Environmental Medicine Kaohsiung Medical University Taiwan.,Faculty of Molecular Preventive Medicine Graduate School of Medicine The University of Tokyo Japan.,Faculty of Science and Engineering Tokushima Bunri University Sanuki Japan
| |
Collapse
|
84
|
Childs BG, Gluscevic M, Baker DJ, Laberge RM, Marquess D, Dananberg J, van Deursen JM. Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov 2017; 16:718-735. [PMID: 28729727 PMCID: PMC5942225 DOI: 10.1038/nrd.2017.116] [Citation(s) in RCA: 792] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronological age represents the single greatest risk factor for human disease. One plausible explanation for this correlation is that mechanisms that drive ageing might also promote age-related diseases. Cellular senescence, which is a permanent state of cell cycle arrest induced by cellular stress, has recently emerged as a fundamental ageing mechanism that also contributes to diseases of late life, including cancer, atherosclerosis and osteoarthritis. Therapeutic strategies that safely interfere with the detrimental effects of cellular senescence, such as the selective elimination of senescent cells (SNCs) or the disruption of the SNC secretome, are gaining significant attention, with several programmes now nearing human clinical studies.
Collapse
Affiliation(s)
| | | | - Darren J Baker
- Departments of Biochemistry and Molecular Biology, Mayo Clinic
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 1st St. SW, Rochester, Minnesota 55905, USA
| | - Remi-Martin Laberge
- Unity Biotechnology, 3280 Bayshore Boulevard Suite 100, Brisbane, California 94005, USA
| | - Dan Marquess
- Unity Biotechnology, 3280 Bayshore Boulevard Suite 100, Brisbane, California 94005, USA
| | - Jamie Dananberg
- Unity Biotechnology, 3280 Bayshore Boulevard Suite 100, Brisbane, California 94005, USA
| | - Jan M van Deursen
- Departments of Biochemistry and Molecular Biology, Mayo Clinic
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 1st St. SW, Rochester, Minnesota 55905, USA
| |
Collapse
|
85
|
Yang L, Xing S, Wang K, Yi H, Du B. Paeonol attenuates aging MRC-5 cells and inhibits epithelial–mesenchymal transition of premalignant HaCaT cells induced by aging MRC-5 cell-conditioned medium. Mol Cell Biochem 2017; 439:117-129. [DOI: 10.1007/s11010-017-3141-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 08/02/2017] [Indexed: 12/15/2022]
|
86
|
Monchusi B, Ntwasa M. Methyl pyruvate protects a normal lung fibroblast cell line from irinotecan-induced cell death: Potential use as adjunctive to chemotherapy. PLoS One 2017; 12:e0182789. [PMID: 28797070 PMCID: PMC5552298 DOI: 10.1371/journal.pone.0182789] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/24/2017] [Indexed: 12/02/2022] Open
Abstract
The Warburg Effect, characterized by increased rate of glycolysis even under normoxic conditions, is one of the hallmarks of cancer. Relatively lower oxidative phosphorylation (OXPHOS) is also a characteristic feature in cancer cells. We hypothesized that interference with this phenomenon, by introducing exogenous pyruvate, would upset this cancer phenotype and boost the energy requirements of normal cells. We find that methyl pyruvate protects irinotecan-treated normal lung fibroblast cell line (MRC-5) probably by turning off the p53/p21 axis of the apoptotic pathways. When the MRC-5 fibroblasts recover in drug-free medium, the intrinsic apoptotic pathway is also turned off and the cells survive with no discernible exponential growth during the observation period. In contrast, the mere introduction of exogenous pyruvate kills the lung cancer cell line (A549). Although, functional p53 is important in the drug-induced cancer cell death, it is probably not essential because cancer cell lines with mutated p53 also die albeit less efficiently. We conclude that methyl pyruvate may preferentially kill cancer cells and protect normal cells during chemotherapy.
Collapse
Affiliation(s)
- Bernice Monchusi
- School of Molecular & Cell Biology, Gatehouse 514, University of the Witwatersrand, Wits, Johannesburg, Republic of South Africa
| | - Monde Ntwasa
- Department of Life & Consumer Sciences, 211 Calabash Building, University of South Africa, Florida, Johannesburg, Republic of South Africa
- * E-mail:
| |
Collapse
|
87
|
D'Arcangelo D, Tinaburri L, Dellambra E. The Role of p16 INK4a Pathway in Human Epidermal Stem Cell Self-Renewal, Aging and Cancer. Int J Mol Sci 2017; 18:ijms18071591. [PMID: 28737694 PMCID: PMC5536078 DOI: 10.3390/ijms18071591] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 07/13/2017] [Accepted: 07/19/2017] [Indexed: 12/31/2022] Open
Abstract
The epidermis is a self-renewing tissue. The balance between proliferation and differentiation processes is tightly regulated to ensure the maintenance of the stem cell (SC) population in the epidermis during life. Aging and cancer may be considered related endpoints of accumulating damages within epidermal self-renewing compartment. p16INK4a is a potent inhibitor of the G1/S-phase transition of the cell cycle. p16INK4a governs the processes of SC self-renewal in several tissues and its deregulation may result in aging or tumor development. Keratinocytes are equipped with several epigenetic enzymes and transcription factors that shape the gene expression signatures of different epidermal layers and allow dynamic and coordinated expression changes to finely balance keratinocyte self-renewal and differentiation. These factors converge their activity in the basal layer to repress p16INK4a expression, protecting cells from senescence, and preserving epidermal homeostasis and regeneration. Several stress stimuli may activate p16INK4a expression that orchestrates cell cycle exit and senescence response. In the present review, we discuss the role of p16INK4a regulators in human epidermal SC self-renewal, aging and cancer.
Collapse
Affiliation(s)
- Daniela D'Arcangelo
- Laboratory of Vascular Pathology, Istituto Dermopatico dell'Immacolata, Istituto di Ricovero e Cura a Carattere Scientifico (IDI-IRCCS), Fondazione Luigi Maria Monti (FLMM), via Monti di Creta 104, 00167 Rome, Italy.
| | - Lavinia Tinaburri
- Molecular and Cell Biology Laboratory, Istituto Dermopatico dell'Immacolata, Istituto di Ricovero e Cura a Carattere Scientifico (IDI-IRCCS), Fondazione Luigi Maria Monti (FLMM), via Monti di Creta 104, 00167 Rome, Italy.
| | - Elena Dellambra
- Molecular and Cell Biology Laboratory, Istituto Dermopatico dell'Immacolata, Istituto di Ricovero e Cura a Carattere Scientifico (IDI-IRCCS), Fondazione Luigi Maria Monti (FLMM), via Monti di Creta 104, 00167 Rome, Italy.
| |
Collapse
|
88
|
Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat Commun 2017; 8:15287. [PMID: 28508895 PMCID: PMC5440838 DOI: 10.1038/ncomms15287] [Citation(s) in RCA: 553] [Impact Index Per Article: 69.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 03/13/2017] [Indexed: 12/20/2022] Open
Abstract
Emerging evidence is revealing that exosomes contribute to many aspects of physiology and disease through intercellular communication. However, the biological roles of exosome secretion in exosome-secreting cells have remained largely unexplored. Here we show that exosome secretion plays a crucial role in maintaining cellular homeostasis in exosome-secreting cells. The inhibition of exosome secretion results in the accumulation of nuclear DNA in the cytoplasm, thereby causing the activation of cytoplasmic DNA sensing machinery. This event provokes the innate immune response, leading to reactive oxygen species (ROS)-dependent DNA damage response and thus induce senescence-like cell-cycle arrest or apoptosis in normal human cells. These results, in conjunction with observations that exosomes contain various lengths of chromosomal DNA fragments, indicate that exosome secretion maintains cellular homeostasis by removing harmful cytoplasmic DNA from cells. Together, these findings enhance our understanding of exosome biology, and provide valuable new insights into the control of cellular homeostasis.
Collapse
|
89
|
Song XQ, Su LN, Wei HP, Liu YH, Yin HF, Li JH, Zhu DX, Zhang AL. The effect of Id1gene silencing on the neural differentiation of MSCs. BIOTECHNOL BIOTEC EQ 2017. [DOI: 10.1080/13102818.2017.1286234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Affiliation(s)
- Xiao-qing Song
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Li-ning Su
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Hui-ping Wei
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Ying-hui Liu
- Department of Agriculture Science, Agriculture and Forestry College of Hebei North University, Zhangjiakou, China
| | - Hai-feng Yin
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Ji-hong Li
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Deng-xiang Zhu
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Ai-lan Zhang
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| |
Collapse
|
90
|
Zhao S, Cao M, Wu H, Hu Y, Xue X. 5-aza-2'-deoxycytidine Inhibits the Proliferation of Lung Fibroblasts in Neonatal Rats Exposed to Hyperoxia. Pediatr Neonatol 2017; 58:122-127. [PMID: 27663361 DOI: 10.1016/j.pedneo.2015.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/03/2015] [Accepted: 11/30/2015] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND A persistent increase in the number of lung fibroblasts (LFs) is found in the interstitium of the lungs of infants with bronchopulmonary dysplasia (BPD), which leads to lung fibrosis. P16 methylation plays an important role in the pathogenesis of BPD. 5-aza-2'-deoxycytidine (5-aza-CdR) is a major methyltransferase-specific inhibitor. This study investigated the effects of 5-aza-CdR on LFs in vitro from a hyperoxia-induced lung fibrosis model in newborn rats. METHODS Methylation-specific polymerase chain reaction (PCR) and Western blotting were performed to determine P16 gene methylation status and protein expression after LFs were treated with 0 μmol/L, 0.5 μmol/L, 1.0 μmol/L, and 5.0 μmol/L 5-aza-CdR for 120 hours. Proliferation was assessed by an MTT assay after LFs were treated with 0 μmol/L, 0.5 μmol/L, 1.0 μmol/L, and 5.0 μmol/L 5-aza-CdR for 24 hours, 48 hours, 72 hours, 96 hours, and 120 hours. At the final time point, cells were also analyzed by flow cytometry to identify any change in their cell cycle profiles. RESULTS A methylated P16 gene promoter was detected in hyperoxia LFs. Following treatment with 5-aza-CdR, partial methylation and demethylation was detected. The expression protein's level of the P16 gene was significantly higher in the 5.0 μmol/L 5-aza-CdR-treated group compared with that in the control group (p < 0.01). The cell growth rate at each tested time point was lower in the 5-aza-CdR-treated group compared with that in the control group after 72 hours (p < 0.01). Flow cytometry revealed that the cells in the 1.0 μmol/L and 5.0 μmol/L 5-aza-CdR-treated groups were apparently arrested in the G0/G1 phase and that the number of cells in the S phase was significantly lower than the control group (p < 0.01). CONCLUSION 5-aza-CdR inhibits the growth of the LFs in hyperoxia-induced neonatal BPD rats in vitro by demethylating the P16 gene.
Collapse
Affiliation(s)
- Shimeng Zhao
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Meiling Cao
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hongmin Wu
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yu Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xindong Xue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
91
|
Kalimutho M, Bain AL, Mukherjee B, Nag P, Nanayakkara DM, Harten SK, Harris JL, Subramanian GN, Sinha D, Shirasawa S, Srihari S, Burma S, Khanna KK. Enhanced dependency of KRAS-mutant colorectal cancer cells on RAD51-dependent homologous recombination repair identified from genetic interactions in Saccharomyces cerevisiae. Mol Oncol 2017; 11:470-490. [PMID: 28173629 PMCID: PMC5527460 DOI: 10.1002/1878-0261.12040] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 01/10/2017] [Accepted: 01/27/2017] [Indexed: 01/08/2023] Open
Abstract
Activating KRAS mutations drive colorectal cancer tumorigenesis and influence response to anti‐EGFR‐targeted therapy. Despite recent advances in understanding Ras signaling biology and the revolution in therapies for melanoma using BRAF inhibitors, no targeted agents have been effective in KRAS‐mutant cancers, mainly due to activation of compensatory pathways. Here, by leveraging the largest synthetic lethal genetic interactome in yeast, we identify that KRAS‐mutated colorectal cancer cells have augmented homologous recombination repair (HRR) signaling. We found that KRAS mutation resulted in slowing and stalling of the replication fork and accumulation of DNA damage. Moreover, we found that KRAS‐mutant HCT116 cells have an increase in MYC‐mediated RAD51 expression with a corresponding increase in RAD51 recruitment to irradiation‐induced DNA double‐strand breaks (DSBs) compared to genetically complemented isogenic cells. MYC depletion using RNA interference significantly reduced IR‐induced RAD51 foci formation and HRR. On the contrary, overexpression of either HA‐tagged wild‐type (WT) MYC or phospho‐mutant S62A increased RAD51 protein levels and hence IR‐induced RAD51 foci. Likewise, depletion of RAD51 selectively induced apoptosis in HCT116‐mutant cells by increasing DSBs. Pharmacological inhibition targeting HRR signaling combined with PARP inhibition selectivity killed KRAS‐mutant cells. Interestingly, these differences were not seen in a second isogenic pair of KRAS WT and mutant cells (DLD‐1), likely due to their nondependency on the KRAS mutation for survival. Our data thus highlight a possible mechanism by which KRAS‐mutant‐dependent cells drive HRR in vitro by upregulating MYC‐RAD51 expression. These data may offer a promising therapeutic vulnerability in colorectal cancer cells harboring otherwise nondruggable KRAS mutations, which warrants further investigation in vivo.
Collapse
Affiliation(s)
- Murugan Kalimutho
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,School of Natural Sciences, Griffith University, Nathan, Australia
| | - Amanda L Bain
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Bipasha Mukherjee
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Purba Nag
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,School of Natural Sciences, Griffith University, Nathan, Australia
| | - Devathri M Nanayakkara
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Sarah K Harten
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Janelle L Harris
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Goutham N Subramanian
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Debottam Sinha
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,School of Natural Sciences, Griffith University, Nathan, Australia
| | - Senji Shirasawa
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Japan
| | - Sriganesh Srihari
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | - Sandeep Burma
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kum Kum Khanna
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
92
|
O'Hara SP, Splinter PL, Trussoni CE, Pisarello MJL, Loarca L, Splinter NS, Schutte BF, LaRusso NF. ETS Proto-oncogene 1 Transcriptionally Up-regulates the Cholangiocyte Senescence-associated Protein Cyclin-dependent Kinase Inhibitor 2A. J Biol Chem 2017; 292:4833-4846. [PMID: 28184004 PMCID: PMC5377799 DOI: 10.1074/jbc.m117.777409] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/06/2017] [Indexed: 12/13/2022] Open
Abstract
Primary sclerosing cholangitis (PSC) is a chronic, fibroinflammatory cholangiopathy (disease of the bile ducts) of unknown pathogenesis. We reported that cholangiocyte senescence features prominently in PSC and that neuroblastoma RAS viral oncogene homolog (NRAS) is activated in PSC cholangiocytes. Additionally, persistent microbial insult (e.g. LPSs) induces cyclin-dependent kinase inhibitor 2A (CDKN2A/p16INK4a) expression and senescence in cultured cholangiocytes in an NRAS-dependent manner. However, the molecular mechanisms involved in LPS-induced cholangiocyte senescence and NRAS-dependent regulation of CDKN2A remain unclear. Using our in vitro senescence model, we found that LPS-induced CDKN2A expression coincided with a 4.5-fold increase in ETS1 (ETS proto-oncogene 1) mRNA, suggesting that ETS1 is involved in regulating CDKN2A This idea was confirmed by RNAi-mediated suppression or genetic deletion of ETS1, which blocked CDKN2A expression and reduced cholangiocyte senescence. Furthermore, site-directed mutagenesis of a predicted ETS-binding site within the CDKN2A promoter abolished luciferase reporter activity. Pharmacological inhibition of RAS/MAPK reduced ETS1 and CDKN2A protein expression and CDKN2A promoter-driven luciferase activity by ∼50%. In contrast, constitutively active NRAS expression induced ETS1 and CDKN2A protein expression, whereas ETS1 RNAi blocked this increase. Chromatin immunoprecipitation-PCR detected increased ETS1 and histone 3 lysine 4 trimethylation (H3K4Me3) at the CDKN2A promoter following LPS-induced senescence. Additionally, phospho-ETS1 expression was increased in cholangiocytes of human PSC livers and in the Abcb4 (Mdr2)-/- mouse model of PSC. These data pinpoint ETS1 and H3K4Me3 as key transcriptional regulators in NRAS-induced expression of CDKN2A, and this regulatory axis may therefore represent a potential therapeutic target for PSC treatment.
Collapse
Affiliation(s)
- Steven P O'Hara
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Patrick L Splinter
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Christy E Trussoni
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Maria J Lorenzo Pisarello
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Lorena Loarca
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Noah S Splinter
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Bryce F Schutte
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Nicholas F LaRusso
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
93
|
Abstract
Inhibitors of DNA binding and cell differentiation (Id) proteins are members of the large family of the helix-loop-helix (HLH) transcription factors, but they lack any DNA-binding motif. During development, the Id proteins play a key role in the regulation of cell-cycle progression and cell differentiation by modulating different cell-cycle regulators both by direct and indirect mechanisms. Several Id-protein interacting partners have been identified thus far, which belong to structurally and functionally unrelated families, including, among others, the class I and II bHLH transcription factors, the retinoblastoma protein and related pocket proteins, the paired-box transcription factors, and the S5a subunit of the 26 S proteasome. Although the HLH domain of the Id proteins is involved in most of their protein-protein interaction events, additional motifs located in their N-terminal and C-terminal regions are required for the recognition of diverse protein partners. The ability of the Id proteins to interact with structurally different proteins is likely to arise from their conformational flexibility: indeed, these proteins contain intrinsically disordered regions that, in the case of the HLH region, undergo folding upon self- or heteroassociation. Besides their crucial role for cell-fate determination and cell-cycle progression during development, other important cellular events have been related to the Id-protein expression in a number of pathologies. Dysregulated Id-protein expression has been associated with tumor growth, vascularization, invasiveness, metastasis, chemoresistance and stemness, as well as with various developmental defects and diseases. Herein we provide an overview on the structural properties, mode of action, biological function and therapeutic potential of these regulatory proteins.
Collapse
Affiliation(s)
- Cornelia Roschger
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria
| | - Chiara Cabrele
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria.
| |
Collapse
|
94
|
Roschger C, Cabrele C. The Id-protein family in developmental and cancer-associated pathways. Cell Commun Signal 2017; 15:7. [PMID: 28122577 PMCID: PMC5267474 DOI: 10.1186/s12964-016-0161-y] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/29/2016] [Indexed: 01/15/2023] Open
Abstract
Inhibitors of DNA binding and cell differentiation (Id) proteins are members of the large family of the helix-loop-helix (HLH) transcription factors, but they lack any DNA-binding motif. During development, the Id proteins play a key role in the regulation of cell-cycle progression and cell differentiation by modulating different cell-cycle regulators both by direct and indirect mechanisms. Several Id-protein interacting partners have been identified thus far, which belong to structurally and functionally unrelated families, including, among others, the class I and II bHLH transcription factors, the retinoblastoma protein and related pocket proteins, the paired-box transcription factors, and the S5a subunit of the 26 S proteasome. Although the HLH domain of the Id proteins is involved in most of their protein-protein interaction events, additional motifs located in their N-terminal and C-terminal regions are required for the recognition of diverse protein partners. The ability of the Id proteins to interact with structurally different proteins is likely to arise from their conformational flexibility: indeed, these proteins contain intrinsically disordered regions that, in the case of the HLH region, undergo folding upon self- or heteroassociation. Besides their crucial role for cell-fate determination and cell-cycle progression during development, other important cellular events have been related to the Id-protein expression in a number of pathologies. Dysregulated Id-protein expression has been associated with tumor growth, vascularization, invasiveness, metastasis, chemoresistance and stemness, as well as with various developmental defects and diseases. Herein we provide an overview on the structural properties, mode of action, biological function and therapeutic potential of these regulatory proteins.
Collapse
Affiliation(s)
- Cornelia Roschger
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria
| | - Chiara Cabrele
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria.
| |
Collapse
|
95
|
Park H, Kim CH, Jeong JH, Park M, Kim KS. GDF15 contributes to radiation-induced senescence through the ROS-mediated p16 pathway in human endothelial cells. Oncotarget 2016; 7:9634-44. [PMID: 26909594 PMCID: PMC4891072 DOI: 10.18632/oncotarget.7457] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/08/2016] [Indexed: 11/25/2022] Open
Abstract
Growth differentiation factor 15 (GDF15) is an emerging biomarker of cardiovascular risk and disease. Microarray analyses revealed that GDF15 levels were increased during cellular senescence induced by ionizing radiation (IR) in human aortic endothelial cells (HAECs). However, the role of GDF15 in HAEC cellular senescence remains unclear. This study demonstrated that downregulation of GDF15 in HAECs partially prevented cellular senescence triggered by IR, which was confirmed by recovery of cell proliferation and reverse senescence-associated β-galactosidase (SA-β-gal) staining. Conversely, upregulation of GDF15-induced cellular senescence in HAECs, confirmed by G0/G1 cell cycle arrest, decreased during cell proliferation and increased SA-β-gal staining. GDF15-induced cellular senescence was observed in p16-knockdown cells but not in p53-knockdown cells. GDF15 expression in endothelial cells also generated reactive oxygen species (ROS), which led to activation of extracellular signal-regulated kinases (ERKs) and induction of senescence by oxidative stress. These results suggested that GDF15 might play an important role in cellular senescence through a ROS-mediated p16 pathway and contribute to the pathogenesis of atherosclerosis via pro-senescent activity.
Collapse
Affiliation(s)
- Hyejin Park
- Divisions of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Chun-Ho Kim
- Divisions of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jae-Hoon Jeong
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Myungjin Park
- Divisions of Radiation Cancer Science, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Kwang Seok Kim
- Divisions of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| |
Collapse
|
96
|
Yoshizawa H, Senda D, Natori Y, Tanaka R, Mizuno H, Hayashi A. End-to-Side Neurorrhaphy as Schwann Cells Provider to Acellular Nerve Allograft and Its Suitable Application. PLoS One 2016; 11:e0167507. [PMID: 27907118 PMCID: PMC5132318 DOI: 10.1371/journal.pone.0167507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 11/15/2016] [Indexed: 11/23/2022] Open
Abstract
Axonal regeneration relies on support from proliferating host Schwann cells (SCs), and previous studies on acellular nerve allografts (ANGs) suggest that axons can regenerate into ANGs within a limited distance. Numerous studies have demonstrated that the supplementation of ANGs with exogenous factors, such as cultured SCs, stem cells, and growth factors, promote nerve regeneration in ANGs. However, there are several problems associated with their utilization. In this study, we investigated whether end-to-side (ETS) neurorrhaphy, which is an axonal provider, could be useful as an SC provider to support axonal elongation in ANGs. We found that ETS neurorrhaphy effectively promoted SC migration into ANGs when an epineurium window combined with partial neurectomy was performed, and the effectiveness increased when it was applied bilaterally. When we transplanted ANGs containing migrated SCs via ETS neurorrhaphy (hybrid ANGs) to the nerve gap, hybrid ANGs increased the number of regenerated axons and facilitated rapid axonal elongation, particularly when ETS neurorrhaphy was applied to both edges of the graft. This approach may represent a novel application of ETS neurorrhaphy and lead to the development of hybrid ANGs, making ANGs more practical in a clinical setting.
Collapse
Affiliation(s)
- Hidekazu Yoshizawa
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Daiki Senda
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yuhei Natori
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Rica Tanaka
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Ayato Hayashi
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
- * E-mail:
| |
Collapse
|
97
|
BMP-SMAD-ID promotes reprogramming to pluripotency by inhibiting p16/INK4A-dependent senescence. Proc Natl Acad Sci U S A 2016; 113:13057-13062. [PMID: 27794120 DOI: 10.1073/pnas.1603668113] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) patients carry a missense mutation in ACVR1 [617G > A (R206H)] that leads to hyperactivation of BMP-SMAD signaling. Contrary to a previous study, here we show that FOP fibroblasts showed an increased efficiency of induced pluripotent stem cell (iPSC) generation. This positive effect was attenuated by inhibitors of BMP-SMAD signaling (Dorsomorphin or LDN1931890) or transducing inhibitory SMADs (SMAD6 or SMAD7). In normal fibroblasts, the efficiency of iPSC generation was enhanced by transducing mutant ACVR1 (617G > A) or SMAD1 or adding BMP4 protein at early times during the reprogramming. In contrast, adding BMP4 at later times decreased iPSC generation. ID genes, transcriptional targets of BMP-SMAD signaling, were critical for iPSC generation. The BMP-SMAD-ID signaling axis suppressed p16/INK4A-mediated cell senescence, a major barrier to reprogramming. These results using patient cells carrying the ACVR1 R206H mutation reveal how cellular signaling and gene expression change during the reprogramming processes.
Collapse
|
98
|
Leoz ML, Sánchez A, Carballal S, Ruano L, Ocaña T, Pellisé M, Castells A, Balaguer F, Moreira L. Hereditary gastric and pancreatic cancer predisposition syndromes. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.gastre.2016.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
99
|
p16 upregulation is linked to poor prognosis in ERG negative prostate cancer. Tumour Biol 2016; 37:12655-12663. [PMID: 27444279 DOI: 10.1007/s13277-016-5167-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/12/2016] [Indexed: 10/21/2022] Open
Abstract
Altered expression of the p16 tumor suppressor is frequently found in prostate cancer, but its role for tumor development and patient prognosis is disputed. In order to clarify the prognostic role of p16 and to draw conclusions on interactions with key molecular features of prostate cancer, we studied p16 expression in a tissue microarray (TMA) with more than 12,400 prostate cancers and attached clinical, pathological, and molecular data such as ERG status and deletions of 3p13, 5q21, 6q15, and PTEN. p16 immunostaining was absent in non-neoplastic prostate cells but was found in 37 % of 9627 interpretable prostate cancers. Finding p16 expression in 58 % of ERG positive but in only 22 % of ERG negative cancers (p < 0.0001), highlights the known androgen-dependence of both genes. Significant associations between p16 upregulation and tumor phenotype or patient prognosis were strictly limited to the subset of ERG negative cancers. For example, p16 positivity increased from 15 % in Gleason ≤3 + 3 to 38 % in Gleason ≥4 + 4 cancers (p < 0.0001) and was associated with early PSA recurrence (p < 0.0001). p16 upregulation was strongly linked to deletions of PTEN (p < 0.0001), highlighting the interaction of both genes in growth control. In conclusion, p16 upregulation is a strong prognostic factor in ERG negative cancers. The strict limitation of its prognostic impact to a molecularly defined subgroup challenges the concept of molecular prognosis testing without considering molecular subtypes.
Collapse
|
100
|
Abstract
Because the genome stores all genetic information required for growth and development, it is of pivotal importance to maintain DNA integrity, especially during cell division, when the genome is prone to replication errors and damage. Although over the last two decades it has become evident that the basic cell cycle toolbox of plants shares several similarities with those of fungi and mammals, plants appear to have evolved a set of distinct checkpoint regulators in response to different types of DNA stress. This might be a consequence of plants' sessile lifestyle, which exposes them to a set of unique DNA damage-inducing conditions. In this review, we highlight the types of DNA stress that plants typically experience and describe the plant-specific molecular mechanisms that control cell division in response to these stresses.
Collapse
Affiliation(s)
- Zhubing Hu
- Department of Plant Systems Biology, VIB, B-9052 Gent, Belgium
| | - Toon Cools
- Department of Plant Systems Biology, VIB, B-9052 Gent, Belgium
| | | |
Collapse
|