1
|
Murugan AK, Kannan S, Alzahrani AS. TERT promoter mutations in gliomas: Molecular roles in tumorigenesis, metastasis, diagnosis, prognosis, therapeutic targeting, and drug resistance. Biochim Biophys Acta Rev Cancer 2025; 1880:189243. [PMID: 39674418 DOI: 10.1016/j.bbcan.2024.189243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Telomerase reverse transcriptase (TERT), a critical player in cellular immortalization, has emerged as a focal point of investigation due to its frequent promoter mutations in various human malignancies. TERT promoter mutations exhibit a significant role in tumorigenesis, fostering unbridled cellular proliferation and survival. This comprehensive review delves into the landscape of TERT promoter mutations and their profound implications in cancer, particularly within the context of gliomas. This article meticulously examines the intricate interplay between TERT promoter mutations and the metastatic cascade, shedding light on their capacity to orchestrate invasive behavior in gliomas. Moreover, this review describes the recent trends in therapeutic targeting of the TERT and dissects the evolving landscape of drug resistance associated with TERT mutations, providing insights into potential therapeutic challenges. In addition, the diagnostic and prognostic implications of TERT promoter mutations in gliomas are scrutinized, unraveling their potential as robust biomarkers. It also discusses the recent advancements in molecular diagnostics, illustrating the promise of TERT mutations as diagnostic tools and prognostic indicators. This review collectively aims to contribute to a deeper understanding of TERT promoter mutations in gliomas, offering a foundation for future research endeavors and paving the way for innovative strategies in glioma management.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia.
| | - Siddarth Kannan
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| | - Ali S Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
2
|
Chen J, Si J, Li Q, Zhang W, He J. Unlocking the potential of senescence-related gene signature as a diagnostic and prognostic biomarker in sepsis: insights from meta-analyses, single-cell RNA sequencing, and in vitro experiments. Aging (Albany NY) 2024; 16:3989-4013. [PMID: 38412321 PMCID: PMC10929830 DOI: 10.18632/aging.205574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/08/2024] [Indexed: 02/29/2024]
Abstract
Cellular senescence is closely associated with the pathogenesis of sepsis. However, the diagnostic and prognostic value of senescence-related genes remain unclear. In this study, 866 senescence-related genes were collected from CellAge. The training cohort, GSE65682, which included 42 control and 760 sepsis samples, was obtained from the Gene Expression Omnibus (GEO). Feature selection was performed using gene expression difference detection, LASSO analysis, random forest, and Cox regression. TGFBI and MAD1L1 were ultimately selected for inclusion in the multivariate Cox regression model. Clustering based on the expressions of TGFBI and MAD1L1 was significantly associated with sepsis characteristics and prognoses (all P < 0.05). The risk signature served as a reliable prognostic predictor across the GSE65682, GSE95233, and GSE4607 cohorts (pooled hazard ratio = 4.27; 95% confidence interval [CI] = 1.63-11.17). Furthermore, it also served as a robust classifier to distinguish sepsis samples from control cases across 14 cohorts (pooled odds ratio = 5.88; 95% CI = 3.54-9.77). Single-cell RNA sequencing analyses from five healthy controls and four sepsis subjects indicated that the risk signature could reflect the senescence statuses of monocytes and B cells; this finding was then experimentally validated in THP-1 and IM-9 cells in vitro (both P < 0.05). In all, a senescence-related gene signature was developed as a prognostic and diagnostic biomarker for sepsis, providing cut-in points to uncover underlying mechanisms and a promising clinical tool to support precision medicine.
Collapse
Affiliation(s)
- Jia Chen
- Department of Emergency, Panyu Maternal and Child Care Service Centre of Guangzhou, Hexian Memorial Affiliated Hospital of Southern Medical University, Panyu, Guangzhou 511400, Guangdong Province, China
| | - Jinhong Si
- Department of Respiratory Medicine, Panyu Maternal and Child Care Service Centre of Guangzhou, Hexian Memorial Affiliated Hospital of Southern Medical University, Panyu, Guangzhou 511400, Guangdong Province, China
| | - Qiankun Li
- Department of Emergency, Panyu Maternal and Child Care Service Centre of Guangzhou, Hexian Memorial Affiliated Hospital of Southern Medical University, Panyu, Guangzhou 511400, Guangdong Province, China
| | - Weihong Zhang
- Department of Emergency, Panyu Maternal and Child Care Service Centre of Guangzhou, Hexian Memorial Affiliated Hospital of Southern Medical University, Panyu, Guangzhou 511400, Guangdong Province, China
| | - Jiahao He
- Department of Emergency, Panyu Maternal and Child Care Service Centre of Guangzhou, Hexian Memorial Affiliated Hospital of Southern Medical University, Panyu, Guangzhou 511400, Guangdong Province, China
| |
Collapse
|
3
|
Ali JH, Walter M. Combining old and new concepts in targeting telomerase for cancer therapy: transient, immediate, complete and combinatory attack (TICCA). Cancer Cell Int 2023; 23:197. [PMID: 37679807 PMCID: PMC10483736 DOI: 10.1186/s12935-023-03041-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Telomerase can overcome replicative senescence by elongation of telomeres but is also a specific element in most cancer cells. It is expressed more vastly than any other tumor marker. Telomerase as a tumor target inducing replicative immortality can be overcome by only one other mechanism: alternative lengthening of telomeres (ALT). This limits the probability to develop resistance to treatments. Moreover, telomerase inhibition offers some degree of specificity with a low risk of toxicity in normal cells. Nevertheless, only one telomerase antagonist reached late preclinical studies. The underlying causes, the pitfalls of telomerase-based therapies, and future chances based on recent technical advancements are summarized in this review. Based on new findings and approaches, we propose a concept how long-term survival in telomerase-based cancer therapies can be significantly improved: the TICCA (Transient Immediate Complete and Combinatory Attack) strategy.
Collapse
Affiliation(s)
- Jaber Haj Ali
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Universitätsmedizin Rostock, Ernst-Heydemann-Straße 6, 18057, Rostock, Germany
| | - Michael Walter
- Institute of Clinical Chemistry and Laboratory Medicine, Universitätsmedizin Rostock, Ernst-Heydemann-Straße 6, 18057, Rostock, Germany.
| |
Collapse
|
4
|
The MYCL and MXD1 transcription factors regulate the fitness of murine dendritic cells. Proc Natl Acad Sci U S A 2020; 117:4885-4893. [PMID: 32071205 PMCID: PMC7060746 DOI: 10.1073/pnas.1915060117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We previously found that MYCL is required by a Batf3-dependent classical dendritic cell subset (cDC1) for optimal CD8 T cell priming, but the underlying mechanism has remained unclear. The MAX-binding proteins encompass a family of transcription factors with overlapping DNA-binding specificities, conferred by a C-terminal basic helix-loop-helix domain, which mediates heterodimerization. Thus, regulation of transcription by these factors is dependent on divergent N-terminal domains. The MYC family, including MYCL, has actions that are reciprocal to the MXD family, which is mediated through the recruitment of higher-order activator and repressor complexes, respectively. As potent proto-oncogenes, models of MYC family function have been largely derived from their activity at supraphysiological levels in tumor cell lines. MYC and MYCN have been studied extensively, but empirical analysis of MYCL function had been limited due to highly restricted, lineage-specific expression in vivo. Here we observed that Mycl is expressed in immature cDC1s but repressed on maturation, concomitant with Mxd1 induction in mature cDC1s. We hypothesized that MYCL and MXD1 regulate a shared, but reciprocal, transcriptional program during cDC1 maturation. In agreement, immature cDC1s in Mycl -/- -deficient mice exhibited reduced expression of genes that regulate core biosynthetic processes. Mature cDC1s from Mxd1 -/- mice exhibited impaired ability to inhibit the transcriptional signature otherwise supported by MYCL. The present study reveals LMYC and MXD1 as regulators of a transcriptional program that is modulated during the maturation of Batf3-dependent cDC1s.
Collapse
|
5
|
Downregulation of endometrial mesenchymal marker SUSD2 causes cell senescence and cell death in endometrial carcinoma cells. PLoS One 2017; 12:e0183681. [PMID: 28841682 PMCID: PMC5571916 DOI: 10.1371/journal.pone.0183681] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/09/2017] [Indexed: 12/13/2022] Open
Abstract
The cause of death among the majority of endometrial cancer patients involves migration of cancer cells within the peritoneal cavity and subsequent implantation of cancer spheroids into neighbouring organs. It is, thereby, important to identify factors that mediate metastasis. Cell adhesion and migration are modified by the mesenchymal stem cell (MSC) marker Sushi domain containing 2 (SUSD2), a type I transmembrane protein that participates in the orchestration of cell adhesion and migration through interaction with its partner Galactosidase-binding soluble-1 (LGALS1). MSCs have emerged as attractive targets in cancer therapy. Human endometrial adenocarcinoma (Ishikawa) cells were treated with TGFβ (10 ng/ml) for 72h. SUSD2, LGALS1 and MKI67 transcript levels were quantified using qRT-PCR. The proportion of SUSD2 positive (SUSD2+) cells and SMAD2/3 abundance were quantified by FACS and Western blotting, respectively. Senescent cells were identified with β-galactosidase staining; cell cycle and cell death were quantified using Propidium Iodide staining. Treatment of endometrial cancer cells (Ishikawa cells) with TGFβ (10 ng/ml) significantly decreased SUSD2 transcript levels and the proportion of SUSD2 positive cells. Silencing of SUSD2 using siRNA resulted in senescence and cell death of Ishikawa cells via activation of SMAD2/3. These findings suggest that SUSD2 counteracts senescence and cell death and is thus a potential chemotherapeutic target in human endometrial cancer.
Collapse
|
6
|
Concomitant underexpression of TGFBR2 and overexpression of hTERT are associated with poor prognosis in cervical cancer. Sci Rep 2017; 7:41670. [PMID: 28195144 PMCID: PMC5307321 DOI: 10.1038/srep41670] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/28/2016] [Indexed: 12/26/2022] Open
Abstract
The human telomerase reverse transcriptase (hTERT) is highly expressed in a variety of tumors. The transforming growth factor beta receptor type II (TGFBR2) is a downstream protein of transforming growth factor beta (TGF-β) which suppresses telomerase activity. However, the relevance of survival to the expression of TGFBR2, hTERT or TGFBR2/hTERT has not been previously investigated in cervical cancer tissues. Our study showed that patients with low level of TGFBR2 were associated with poor prognosis (HR = 1.704, P = 0.021), but no significant relevance between hTERT expression and survival (HR = 1.390, P = 0.181). However, a combination of low level of TGFBR2 and high level of hTERT was associated with a worse survival (HR = 1.892, P = 0.020), which had higher impact of hazard ratio (HR) on the overall survival (OS) than the low TGFBR2 expression alone. Knockdown of TGFBR2 expression by shRNA in Hela cells increased cell proliferation, cell invasion, G1/S transition and telomere homeostasis but decreased cell apoptosis. Overexpressing TGFBR2 and inhibiting hTERT suppressed Hela cell growth. These results would lead us to further explore whether a phenotype of TGFBR2low/hTERThigh could be considered as a predictor of poor prognosis, and whether simultaneous use of TGFBR2 agonist and hTERT inhibitor could be developed as a therapeutic strategy.
Collapse
|
7
|
Bai H, Gao Y, Hoyle DL, Cheng T, Wang ZZ. Suppression of Transforming Growth Factor-β Signaling Delays Cellular Senescence and Preserves the Function of Endothelial Cells Derived from Human Pluripotent Stem Cells. Stem Cells Transl Med 2016; 6:589-600. [PMID: 28191769 PMCID: PMC5442820 DOI: 10.5966/sctm.2016-0089] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/09/2016] [Indexed: 12/15/2022] Open
Abstract
Transplantation of vascular cells derived from human pluripotent stem cells (hPSCs) offers an attractive noninvasive method for repairing the ischemic tissues and for preventing the progression of vascular diseases. Here, we found that in a serum‐free condition, the proliferation rate of hPSC‐derived endothelial cells is quickly decreased, accompanied with an increased cellular senescence, resulting in impaired gene expression of endothelial nitric oxide synthase (eNOS) and impaired vessel forming capability in vitro and in vivo. To overcome the limited expansion of hPSC‐derived endothelial cells, we screened small molecules for specific signaling pathways and found that inhibition of transforming growth factor‐β (TGF‐β) signaling significantly retarded cellular senescence and increased a proliferative index of hPSC‐derived endothelial cells. Inhibition of TGF‐β signaling extended the life span of hPSC‐derived endothelial and improved endothelial functions, including vascular network formation on Matrigel, acetylated low‐density lipoprotein uptake, and eNOS expression. Exogenous transforming growth factor‐β1 increased the gene expression of cyclin‐dependent kinase inhibitors, p15Ink4b, p16Ink4a, and p21CIP1, in endothelial cells. Conversely, inhibition of TGF‐β reduced the gene expression of p15Ink4b, p16Ink4a, and p21CIP1. Our findings demonstrate that the senescence of newly generated endothelial cells from hPSCs is mediated by TGF‐β signaling, and manipulation of TGF‐β signaling offers a potential target to prevent vascular aging. Stem Cells Translational Medicine2017;6:589–600
Collapse
Affiliation(s)
- Hao Bai
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yongxing Gao
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dixie L. Hoyle
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Blood Cell Therapy and Technology, Tianjin, People's Republic of China
| | - Zack Z. Wang
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Blood Cell Therapy and Technology, Tianjin, People's Republic of China
| |
Collapse
|
8
|
Yaswen P, MacKenzie KL, Keith WN, Hentosh P, Rodier F, Zhu J, Firestone GL, Matheu A, Carnero A, Bilsland A, Sundin T, Honoki K, Fujii H, Georgakilas AG, Amedei A, Amin A, Helferich B, Boosani CS, Guha G, Ciriolo MR, Chen S, Mohammed SI, Azmi AS, Bhakta D, Halicka D, Niccolai E, Aquilano K, Ashraf SS, Nowsheen S, Yang X. Therapeutic targeting of replicative immortality. Semin Cancer Biol 2015; 35 Suppl:S104-S128. [PMID: 25869441 PMCID: PMC4600408 DOI: 10.1016/j.semcancer.2015.03.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 03/06/2015] [Accepted: 03/13/2015] [Indexed: 12/15/2022]
Abstract
One of the hallmarks of malignant cell populations is the ability to undergo continuous proliferation. This property allows clonal lineages to acquire sequential aberrations that can fuel increasingly autonomous growth, invasiveness, and therapeutic resistance. Innate cellular mechanisms have evolved to regulate replicative potential as a hedge against malignant progression. When activated in the absence of normal terminal differentiation cues, these mechanisms can result in a state of persistent cytostasis. This state, termed “senescence,” can be triggered by intrinsic cellular processes such as telomere dysfunction and oncogene expression, and by exogenous factors such as DNA damaging agents or oxidative environments. Despite differences in upstream signaling, senescence often involves convergent interdependent activation of tumor suppressors p53 and p16/pRB, but can be induced, albeit with reduced sensitivity, when these suppressors are compromised. Doses of conventional genotoxic drugs required to achieve cancer cell senescence are often much lower than doses required to achieve outright cell death. Additional therapies, such as those targeting cyclin dependent kinases or components of the PI3K signaling pathway, may induce senescence specifically in cancer cells by circumventing defects in tumor suppressor pathways or exploiting cancer cells’ heightened requirements for telomerase. Such treatments sufficient to induce cancer cell senescence could provide increased patient survival with fewer and less severe side effects than conventional cytotoxic regimens. This positive aspect is countered by important caveats regarding senescence reversibility, genomic instability, and paracrine effects that may increase heterogeneity and adaptive resistance of surviving cancer cells. Nevertheless, agents that effectively disrupt replicative immortality will likely be valuable components of new combinatorial approaches to cancer therapy.
Collapse
Affiliation(s)
- Paul Yaswen
- Life Sciences Division, Lawrence Berkeley National Lab, Berkeley, CA, United States.
| | - Karen L MacKenzie
- Children's Cancer Institute Australia, Kensington, New South Wales, Australia.
| | | | | | | | - Jiyue Zhu
- Washington State University College of Pharmacy, Pullman, WA, United States.
| | | | | | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, HUVR, Consejo Superior de Investigaciones Cientificas, Universdad de Sevilla, Seville, Spain.
| | | | | | | | | | | | | | - Amr Amin
- United Arab Emirates University, Al Ain, United Arab Emirates; Cairo University, Cairo, Egypt
| | - Bill Helferich
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| | | | - Gunjan Guha
- SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust, Guildford, Surrey, United Kingdom
| | | | - Asfar S Azmi
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | | | | | | | | | - S Salman Ashraf
- United Arab Emirates University, Al Ain, United Arab Emirates; Cairo University, Cairo, Egypt
| | | | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| |
Collapse
|
9
|
Wu J, Niu J, Li X, Wang X, Guo Z, Zhang F. TGF-β1 induces senescence of bone marrow mesenchymal stem cells via increase of mitochondrial ROS production. BMC DEVELOPMENTAL BIOLOGY 2014; 14:21. [PMID: 24886313 PMCID: PMC4031602 DOI: 10.1186/1471-213x-14-21] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 05/06/2014] [Indexed: 11/17/2022]
Abstract
Background Bone marrow derived mesenchymal stem cells (bmMSCs) are multipotent cells that can differentiate into diverse cell types, including cardiomyocytes. BmMSC-based transplantation is capable of repairing acute and chronic myocardial infarction. Prior to the transplantation, MSCs are usually induced in vitro by biological reagents and chemicals for directional differentiation. Transforming growth factor beta (TGF-β) is one of the most commonly used biological reagents for induction of cardiomyocyte differentiation of bmMSCs. Previous studies have shown that TGF-β induces senescence in several cell types. However, whether TGF-β affects senescence of bmMSCs has not been elucidated. The goal of this study was to investigate the effect of TGF-β1 on senescence of bmMSCs and the underlying mechanisms. Results We found that TGF-β1 increased activity of senescence-associated-galactosidase (SA-Gal) and production of mitochondrial reactive oxygen species (mtROS) in bmMSCs in a dose-dependent manner. TGF-β1 also significantly decreased expression of superoxide dismutase 2 (SOD2) and Id1, and increased expression of 4-Hydroxynonenal (4-HNE) subunits and p16 in bmMSCs in a dose-dependent manner. Pre-treatment with mtROS inhibitor acetyl-L-carnitine (ALCAR, 0.1 mM) significantly inhibited TGF-β1-induced mtROS production and SA-Gal activity. Conclusion TGF-β1 can induce senescence of bmMSCs, which at least partially depends on mtROS production.
Collapse
Affiliation(s)
| | | | | | | | | | - Fenxi Zhang
- Department of Anatomy, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
10
|
Comegna M, Succoio M, Napolitano M, Vitale M, D'Ambrosio C, Scaloni A, Passaro F, Zambrano N, Cimino F, Faraonio R. Identification of miR-494 direct targets involved in senescence of human diploid fibroblasts. FASEB J 2014; 28:3720-33. [PMID: 24823364 DOI: 10.1096/fj.13-239129] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cellular senescence is a permanent cell cycle arrest triggered by different stimuli. We recently identified up-regulation of microRNA (miR)-494 as a component of the genetic program leading to senescence of human diploid IMR90 fibroblasts. Here, we used 2-dimensional differential gel electrophoresis (2D-DIGE) coupled to mass spectrometry to profile protein expression changes induced by adoptive overexpression of miR-494 in IMR90 cells. miR-494 induced robust perturbation of the IMR90 proteome by significantly (P≤0.05) down-regulating a number of proteins. Combination of mass spectrometry-based identification of down-regulated proteins and bioinformatic prediction of the miR-494 binding sites on the relevant mRNAs identified 26 potential targets of miR-494. Among them, computational analysis identified 7 potential evolution-conserved miR-494 targets. Functional miR-494 binding sites were confirmed in 3'-untranslated regions (UTRs) of 4 of them [heterogeneous nuclear ribonucleoprotein A3 (hnRNPA3), protein disulfide isomerase A3 (PDIA3), UV excision repair protein RAD23 homolog B (RAD23B), and synaptotagmin-binding cytoplasmic RNA-interacting protein (SYNCRIP)/heterogeneous nuclear ribonucleoprotein Q (hnRNPQ)]. Their reduced expression correlated with miR-494 up-regulation in senescent cells. RNA interference-mediated knockdown of hnRNPA3 and, to a lesser extent, RAD23B mirrored the senescent phenotype induced by miR-494 overexpression, blunting cell proliferation and causing up-regulation of SA-β-galactosidase and DNA damage. Ectopic expression of hnRNPA3 or RAD23B slowed the appearance of the senescent phenotype induced by miR-494. Overall, these findings identify novel miR-494 direct targets that are involved in cellular senescence.
Collapse
Affiliation(s)
- Marika Comegna
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy; Center of Genetics Engineering (CEINGE) Biotecnologie Avanzate s.c. a r.l, Naples, Italy; and
| | - Mariangela Succoio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy; Center of Genetics Engineering (CEINGE) Biotecnologie Avanzate s.c. a r.l, Naples, Italy; and
| | - Marco Napolitano
- Fondazione SDN, Istituto di Ricerca Diagnostica e Nucleare, Naples, Italy
| | - Monica Vitale
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy; Center of Genetics Engineering (CEINGE) Biotecnologie Avanzate s.c. a r.l, Naples, Italy; and
| | - Chiara D'Ambrosio
- Proteomics and Mass Spectrometry Laboratory, National Research Council, Naples, Italy
| | - Andrea Scaloni
- Proteomics and Mass Spectrometry Laboratory, National Research Council, Naples, Italy
| | - Fabiana Passaro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy; Fondazione SDN, Istituto di Ricerca Diagnostica e Nucleare, Naples, Italy; Center of Genetics Engineering (CEINGE) Biotecnologie Avanzate s.c. a r.l, Naples, Italy; and
| | - Nicola Zambrano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy; Center of Genetics Engineering (CEINGE) Biotecnologie Avanzate s.c. a r.l, Naples, Italy; and
| | - Filiberto Cimino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy; Fondazione SDN, Istituto di Ricerca Diagnostica e Nucleare, Naples, Italy; Center of Genetics Engineering (CEINGE) Biotecnologie Avanzate s.c. a r.l, Naples, Italy; and
| | - Raffaella Faraonio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy; Center of Genetics Engineering (CEINGE) Biotecnologie Avanzate s.c. a r.l, Naples, Italy; and
| |
Collapse
|
11
|
Qian Y, Yang L, Cao S. Telomeres and telomerase in T cells of tumor immunity. Cell Immunol 2014; 289:63-9. [PMID: 24727158 DOI: 10.1016/j.cellimm.2014.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 03/05/2014] [Accepted: 03/24/2014] [Indexed: 02/08/2023]
Abstract
Telomeres are specific nucleoprotein structures at the end of a eukaryotic chromosomes characterized by repeats of the sequence TTAGGG and regulated by the enzyme telomerase which prevents their degradation, loss, rearrangement and end-to-end fusion. During activation, T lymphocytes actively divide, albeit through only a finite number of cell divisions due to shortening of telomeres. However, studies have demonstrated that human telomerase reverse transcriptase (hTERT), thought to be the major component regulating telomerase activity, can enhance the proliferation of T cells when overexpressed. There are many treatments for cancers, most of which are targeting the telomere and telomerase of tumor cells. However, the hTERT-transduced T cells improve their potential for proliferation, making them an appropriate cell resource for tumor adoptive immunotherapy, a procedure whereby T cells are isolated from patients, expanded ex vivo and eventually delivered back into the patients, provides a new approach for tumor therapy through improved overall survival rates in cancer patients. In this review, we will focus on the telomerase activity in T cells, the regulation of telomerase activity, and hTERT-transduced T cells used in adoptive immunotherapy for cancer.
Collapse
Affiliation(s)
- Yaqin Qian
- Department of Immunology, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China; National Clinical Research Center of Cancer, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China; Research Center of Lung Cancer, Tianjin, China
| | - Lili Yang
- Department of Immunology, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China; National Clinical Research Center of Cancer, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China; Research Center of Lung Cancer, Tianjin, China.
| | - Shui Cao
- Department of Immunology, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China; National Clinical Research Center of Cancer, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China; Research Center of Lung Cancer, Tianjin, China.
| |
Collapse
|
12
|
Olivieri F, Rippo MR, Monsurrò V, Salvioli S, Capri M, Procopio AD, Franceschi C. MicroRNAs linking inflamm-aging, cellular senescence and cancer. Ageing Res Rev 2013; 12:1056-68. [PMID: 23688930 DOI: 10.1016/j.arr.2013.05.001] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/30/2013] [Accepted: 05/08/2013] [Indexed: 01/01/2023]
Abstract
Epidemiological and experimental data demonstrate a strong correlation between age-related chronic inflammation (inflamm-aging) and cancer development. However, a comprehensive approach is needed to clarify the underlying molecular mechanisms. Chronic inflammation has mainly been attributed to continuous immune cells activation, but the cellular senescence process, which may involve acquisition of a senescence-associated secretory phenotype (SASP), can be another important contributor, especially in the elderly. MicroRNAs (miRs), a class of molecules involved in gene expression regulation, are emerging as modulators of some pathways, including NF-κB, mTOR, sirtuins, TGF-β and Wnt, that may be related to inflammation, cellular senescence and age-related diseases, cancer included. Interestingly, cancer development is largely avoided or delayed in centenarians, where changes in some miRs are found in plasma and leukocytes. We identified miRs that can be considered as senescence-associated (SA-miRs), inflammation-associated (inflamma-miRs) and cancer-associated (onco-miRs). Here we review recent findings concerning three of them, miR-21, -126 and -146a, which target mRNAs belonging to the NF-κB pathway; we discuss their ability to link cellular senescence, inflamm-aging and cancer and their changes in centenarians, and provide an update on the possibility of using miRs to block accumulation of senescent cells to prevent formation of a microenvironment favoring cancer development and progression.
Collapse
|
13
|
Hubackova S, Krejcikova K, Bartek J, Hodny Z. IL1- and TGFβ-Nox4 signaling, oxidative stress and DNA damage response are shared features of replicative, oncogene-induced, and drug-induced paracrine 'bystander senescence'. Aging (Albany NY) 2013; 4:932-51. [PMID: 23385065 PMCID: PMC3615160 DOI: 10.18632/aging.100520] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Many cancers arise at sites of infection and inflammation. Cellular senescence, a permanent state of cell cycle arrest that provides a barrier against tumorigenesis, is accompanied by elevated proinflammatory cytokines such as IL1, IL6, IL8 and TNFα. Here we demonstrate that media conditioned by cells undergoing any of the three main forms of senescence, i.e. replicative, oncogene- and drug-induced, contain high levels of IL1, IL6, and TGFb capable of inducing reactive oxygen species (ROS)-mediated DNA damage response (DDR). Persistent cytokine signaling and activated DDR evoke senescence in normal bystander cells, accompanied by activation of the JAK/STAT, TGFβ/SMAD and IL1/NFκB signaling pathways. Whereas inhibition of IL6/STAT signaling had no effect on DDR induction in bystander cells, inhibition of either TGFβ/SMAD or IL1/NFκB pathway resulted in decreased ROS production and reduced DDR in bystander cells. Simultaneous inhibition of both TGFβ/SMAD and IL1/NFκB pathways completely suppressed DDR indicating that IL1 and TGFβ cooperate to induce and/or maintain bystander senescence. Furthermore, the observed IL1- and TGFβ-induced expression of NAPDH oxidase Nox4 indicates a mechanistic link between the senescence-associated secretory phenotype (SASP) and DNA damage signaling as a feature shared by development of all major forms of paracrine bystander senescence.
Collapse
Affiliation(s)
- Sona Hubackova
- Department of Genome Integrity, Institute of Molecular Genetics, v.v.i., Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | |
Collapse
|
14
|
Chen TH, Kuro-O M, Chen CH, Sue YM, Chen YC, Wu HH, Cheng CY. The secreted Klotho protein restores phosphate retention and suppresses accelerated aging in Klotho mutant mice. Eur J Pharmacol 2012; 698:67-73. [PMID: 23041151 DOI: 10.1016/j.ejphar.2012.09.032] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 09/14/2012] [Accepted: 09/22/2012] [Indexed: 01/03/2023]
Abstract
Klotho was identified as the responsible gene in a mutant mouse line whose disruption results in a variety of premature aging-related phenotypes. Nonetheless, the related mechanisms were still unknown. Many studies report that dietary phosphate restriction and genetic ablation of vitamin D pathways indirectly reverse premature aging processes in these mice. Furthermore, transgenic overexpression of klotho in mice extends their life span through inhibition of insulin and IGF1 signaling. We found that intraperitoneal injection of recombinant soluble Klotho protein at dose of 0.02 mg/kg every other day effectively extends the life span of kl/kl mice by 17.4%. Soluble Klotho administration also ameliorated premature aging-related phenotype, such as growth retardation, premature thymus involution and vascular calcification, and effectively enhanced urinary phosphate excretion in kl/kl mice. Klotho treatment attenuated renal fibrosis through down-regulation of transforming growth factor-β signaling as well as reduced cellular senescence through down-regulation of p21-cip1 mRNA levels. In addition, soluble Klotho treatment significantly reduced both renal and aorta calcium deposits. In conclusion, our study shows the therapeutic potential of soluble Klotho protein to treat age-related disorders in mice.
Collapse
Affiliation(s)
- Tso-Hsiao Chen
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, No 111, Section 3 Hsin-Long Road, Wen-Shang District, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
15
|
Regulation of the human catalytic subunit of telomerase (hTERT). Gene 2012; 498:135-46. [PMID: 22381618 DOI: 10.1016/j.gene.2012.01.095] [Citation(s) in RCA: 205] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 01/29/2012] [Accepted: 01/30/2012] [Indexed: 12/12/2022]
Abstract
Over the past decade, there has been much interest in the regulation of telomerase, the enzyme responsible for maintaining the integrity of chromosomal ends, and its crucial role in cellular immortalization, tumorigenesis, and the progression of cancer. Telomerase activity is characterized by the expression of the telomerase reverse transcriptase (TERT) gene, suggesting that TERT serves as the major limiting agent for telomerase activity. Recent discoveries have led to characterization of various interactants that aid in the regulation of human TERT (hTERT), including numerous transcription factors; further supporting the pivotal role that transcription plays in both the expression and repression of telomerase. Several studies have suggested that epigenetic modulation of the hTERT core promoter region may provide an additional level of regulation. Although these studies have provided essential information on the regulation of hTERT, there has been ambiguity of the role of methylation within the core promoter region and the subsequent binding of various activating and repressive agents. As a result, we found it necessary to consolidate and summarize these recent developments and elucidate these discrepancies. In this review, we focus on the co-regulation of hTERT via transcriptional regulation, the presence or absence of various activators and repressors, as well as the epigenetic pathways of DNA methylation and histone modifications.
Collapse
|
16
|
Lüscher B. MAD1 and its life as a MYC antagonist: an update. Eur J Cell Biol 2011; 91:506-14. [PMID: 21917351 DOI: 10.1016/j.ejcb.2011.07.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Revised: 07/21/2011] [Accepted: 07/25/2011] [Indexed: 12/16/2022] Open
Abstract
The MYC/MAX/MAD network is of central importance for controlling cell physiology. The network is compiled of transcriptional regulators that form different heterodimers, which can either activate or repress the expression of target genes. Thus these proteins function as a molecular switch to control gene expression. MAD1, a member of this network, acts as a transcriptional repressor. It interacts with MAX to form the OFF position of the switch, antagonizing MYC/MAX complexes that define the ON position. MAD1 regulates cell proliferation and apoptosis through a number of target genes. In addition recent evidence indicates that the expression and activity of MAD1 are regulated at multiple levels. Here the recent developments are summarized, in comparison to MYC, of our understanding how the expression of the MAD1 gene and protein are controlled and what the functional consequences and downstream effectors of MAD1 are, which relay its activity as a transcriptional regulator.
Collapse
Affiliation(s)
- Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52057 Aachen, Germany.
| |
Collapse
|
17
|
Wang ML, Walsh R, Robinson KL, Burchard J, Bartz SR, Cleary M, Galloway DA, Grandori C. Gene expression signature of c-MYC-immortalized human fibroblasts reveals loss of growth inhibitory response to TGFβ. Cell Cycle 2011; 10:2540-8. [PMID: 21720214 DOI: 10.4161/cc.10.15.16309] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cancer cells exhibit the ability to proliferate indefinitely, but paradoxically, overexpression of cellular oncogenes in primary cells can result in a rapid and irreversible cell cycle arrest known as oncogene-induced senescence (OIS). However, we have shown that constitutive overexpression of the oncogene c-MYC in primary human foreskin fibroblasts results in a population of cells with unlimited lifespan; these immortalized cells are henceforth referred to as iMYC. Here, in order to further elucidate the mechanisms underlying the immortalization process, a gene expression signature of three independently established iMYC cell lines compared to matched early passage c-MYC overexpressing cells was derived. Network analysis of this "iMYC signature" indicated that a large fraction of the down-regulated genes were functionally connected and major nodes centered around the TGFβ, IL-6 and IGF-1 signaling pathways. Here, we focused on the functional validation of the alteration of TGFβ response during c-MYC-mediated immortalization. The results demonstrate loss of sensitivity of iMYC cells to activation of TGFβ signaling upon ligand addition. Furthermore, we show that aberrant regulation of the p27 tumor suppressor protein in iMYC cells is a key event that contributes to loss of response to TGFβ. These findings highlight the potential to reveal key pathways contributing to the self-renewal of cancer cells through functional mining of the unique gene expression signature of cells immortalized by c-MYC.
Collapse
Affiliation(s)
- Myra L Wang
- Program in Cancer Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Hein N, Jiang K, Cornelissen C, Lüscher B. TGFβ1 enhances MAD1 expression and stimulates promoter-bound Pol II phosphorylation: basic functions of C/EBP, SP and SMAD3 transcription factors. BMC Mol Biol 2011; 12:9. [PMID: 21345218 PMCID: PMC3056803 DOI: 10.1186/1471-2199-12-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 02/23/2011] [Indexed: 12/28/2022] Open
Abstract
Background The MAD1 protein, a member of the MYC/MAX/MAD network of transcriptional regulators, controls cell proliferation, differentiation and apoptosis. MAD1 functions as a transcriptional repressor, one direct target gene being the tumor suppressor PTEN. Repression of this gene is critical to mediate the anti-apoptotic function of MAD1. Under certain conditions it also antagonizes the functions of the oncoprotein MYC. Previous studies have demonstrated that MAD1 expression is controlled by different cytokines and growth factors. Moreover we have recently demonstrated that the MAD1 promoter is controlled by the cytokine granulocyte colony-stimulating factor (G-CSF) through the activation of STAT3, MAP kinases and C/EBP transcription factors. Results We observed that in addition to G-CSF, the cytokine transforming growth factor β (TGFβ1) rapidly induced the expression of MAD1 mRNA and protein in promyelocytic tumor cells. Moreover we found that C/EBP and SP transcription factors cooperated in regulating the expression of MAD1. This cooperativity was dependent on the respective binding sites in the proximal promoter, with the CCAAT boxes being bound by C/EBPα/β heterodimers. Both C/EBP and SP transcription factors bound constitutively to DNA without obvious changes in response to TGFβ1. In addition SMAD3 stimulated the MAD1 reporter, cooperated with C/EBPα and was bound to the core promoter region. Thus SMAD3 appears to be a potential link between TGFβ1 signaling and C/EBP regulated promoter activity. Moreover TGFβ1 stimulated the phosphorylation of polymerase II at serine 2 and its progression into the gene body, consistent with enhanced processivity. Conclusions Our findings suggest that C/EBP and SP factors provide a platform of transcription factors near the core promoter of the MAD1 gene that participate in mediating signal transduction events emanating from different cytokine receptors. SMAD3, a target of TGFβ1 signaling, appears to be functionally relevant. We suggest that a key event induced by TGFβ1 at the MAD1 promoter is the recruitment or activation of cofactors, possibly in complex with C/EBP, SP, and SMAD3 transcriptional regulators, that control polymerase activity.
Collapse
Affiliation(s)
- Nadine Hein
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52057 Aachen, Germany
| | | | | | | |
Collapse
|
19
|
van Riggelen J, Müller J, Otto T, Beuger V, Yetil A, Choi PS, Kosan C, Möröy T, Felsher DW, Eilers M. The interaction between Myc and Miz1 is required to antagonize TGFbeta-dependent autocrine signaling during lymphoma formation and maintenance. Genes Dev 2010; 24:1281-94. [PMID: 20551174 PMCID: PMC2885663 DOI: 10.1101/gad.585710] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The Myc protein suppresses the transcription of several cyclin-dependent kinase inhibitors (CKIs) via binding to Miz1; whether this interaction is important for Myc's ability to induce or maintain tumorigenesis is not known. Here we show that the oncogenic potential of a point mutant of Myc (MycV394D) that is selectively deficient in binding to Miz1 is greatly attenuated. Binding of Myc to Miz1 is continuously required to repress CKI expression and inhibit accumulation of trimethylated histone H3 at Lys 9 (H3K9triMe), a hallmark of cellular senescence, in T-cell lymphomas. Lymphomas that arise express high amounts of transforming growth factor beta-2 (TGFbeta-2) and TGFbeta-3. Upon Myc suppression, TGFbeta signaling is required to induce CKI expression and cellular senescence and suppress tumor recurrence. Binding of Myc to Miz1 is required to antagonize growth suppression and induction of senescence by TGFbeta. We demonstrate that, since lymphomas express high levels of TGFbeta, they are poised to elicit an autocrine program of senescence upon Myc inactivation, demonstrating that TGFbeta is a key factor that establishes oncogene addiction of T-cell lymphomas.
Collapse
Affiliation(s)
- Jan van Riggelen
- Department of Medicine, Division of Oncology, Stanford University, School of Medicine, Stanford, California 94304, USA
- Department of Pathology, Division of Oncology, Stanford University, School of Medicine, Stanford, California 94304, USA
| | - Judith Müller
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Tobias Otto
- Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Vincent Beuger
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany
- TaconicArtemis GmbH, 51063 Koeln, Germany
| | - Alper Yetil
- Department of Medicine, Division of Oncology, Stanford University, School of Medicine, Stanford, California 94304, USA
- Department of Pathology, Division of Oncology, Stanford University, School of Medicine, Stanford, California 94304, USA
| | - Peter S. Choi
- Department of Medicine, Division of Oncology, Stanford University, School of Medicine, Stanford, California 94304, USA
- Department of Pathology, Division of Oncology, Stanford University, School of Medicine, Stanford, California 94304, USA
| | - Christian Kosan
- Institut de Recherches Cliniques de Montreal, Université de Montréal, Montreal, Québec H2W 1R7, Canada
| | - Tarik Möröy
- Institut de Recherches Cliniques de Montreal, Université de Montréal, Montreal, Québec H2W 1R7, Canada
| | - Dean W. Felsher
- Department of Medicine, Division of Oncology, Stanford University, School of Medicine, Stanford, California 94304, USA
- Department of Pathology, Division of Oncology, Stanford University, School of Medicine, Stanford, California 94304, USA
- E-MAIL ; FAX (650) 725-1420
| | - Martin Eilers
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany
- Corresponding authors.E-MAIL ; FAX 49-9031-3184113
| |
Collapse
|
20
|
Larsson LG, Henriksson MA. The Yin and Yang functions of the Myc oncoprotein in cancer development and as targets for therapy. Exp Cell Res 2010; 316:1429-37. [PMID: 20382143 DOI: 10.1016/j.yexcr.2010.03.025] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 03/31/2010] [Indexed: 12/21/2022]
Abstract
The Myc proto-oncoprotein coordinates a number of normal physiological processes necessary for growth and expansion of somatic cells by controlling the expression of numerous target genes. Deregulation of MYC as a consequence of carciogenic events enforces cells to undergo a transition to a hyperproliferative state. This increases the risk of additional oncogenic mutations that in turn can result in further tumor progression. However, Myc activation also provokes intrinsic tumor suppressor mechanisms including apoptosis, cellular senescence and DNA damage responses that act as barriers for tumor development and therefore needs to be overcome during tumorigenesis. Myc thus possesses two seemingly contradictory "faces" here referred to as "Yin and Yang". Observations that many tumor suppressor pathways remain intact but are latent in tumor cells opens the possibility that pharmacological inhibition of the Yin or activation of the Yang functions can prevail and offer new attractive approaches for treating diverse types of cancer.
Collapse
Affiliation(s)
- Lars-Gunnar Larsson
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Box 280, 171 77 Stockholm, Sweden.
| | | |
Collapse
|
21
|
Phosphorylation by Cdk2 is required for Myc to repress Ras-induced senescence in cotransformation. Proc Natl Acad Sci U S A 2009; 107:58-63. [PMID: 19966300 DOI: 10.1073/pnas.0900121106] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The MYC and RAS oncogenes are frequently activated in cancer and, together, are sufficient to transform rodent cells. The basis for this cooperativity remains unclear. We found that although Ras interfered with Myc-induced apoptosis, Myc repressed Ras-induced senescence, together abrogating two main barriers of tumorigenesis. Inhibition of cellular senescence required phosphorylation of Myc at Ser-62 by cyclin E/cyclin-dependent kinase (Cdk) 2. Cdk2 interacted with Myc at promoters, where it affected Myc-dependent regulation of genes, including Bmi-1, p16, p21, and hTERT, which encode proteins known to control senescence. Repression of senescence by Myc was abrogated by the Cdk inhibitor p27Kip1, which is induced by antiproliferative signals like IFN-gamma or by pharmacological inhibitors of Cdk2 but not by inhibitors of other Cdks. In contrast, a phospho-mimicking Myc-S62D mutant was resistant to these manipulations. Inhibition of cyclin E/Cdk2 reversed the senescence-associated gene expression pattern imposed by Myc/cyclin E/Cdk2. This indicates a role of Cdk2 as a transcriptional cofactor and activator of the antisenescence function of Myc and provides mechanistic insight into the Myc-p27Kip1 antagonism. Finally, our findings highlight that pharmacological inhibition of Cdk2 activity is a potential therapeutical principle for cancer therapy, in particular for tumors with activated Myc or Ras.
Collapse
|