1
|
UVB Inhibits Proliferation, Cell Cycle and Induces Apoptosis via p53, E2F1 and Microtubules System in Cervical Cancer Cell Lines. Int J Mol Sci 2021; 22:ijms22105197. [PMID: 34068980 PMCID: PMC8157236 DOI: 10.3390/ijms22105197] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
Ultraviolet (UV) exposure has been linked to skin damage and carcinogenesis, but recently UVB has been proposed as a therapeutic approach for cancer. Herein, we investigated the cellular and molecular effects of UVB in immortal and tumorigenic HPV positive and negative cells. Cells were irradiated with 220.5 to 1102.5 J/m2 of UVB and cell proliferation was evaluated by crystal violet, while cell cycle arrest and apoptosis analysis were performed through flow cytometry. UVB effect on cells was recorded at 661.5 J/m2 and it was exacerbated at 1102.5 J/m2. All cell lines were affected by proliferation inhibition, cell cycle ablation and apoptosis induction, with different degrees depending on tumorigenesis level or HPV type. Analysis of the well-known UV-responsive p53, E2F1 and microtubules system proteins was performed in SiHa cells in response to UVB through Western-blotting assays. E2F1 and the Microtubule-associated protein 2 (MAP2) expression decrease correlated with cellular processes alteration while p53 and Microtubule-associated Protein 1S (MAP1S) expression switch was observed since 882 J/m2, suggesting they were required under more severe cellular damage. However, expression transition of α-Tubulin3C and β-Tubulin was abruptly noticed until 1102.5 J/m2 and particularly, γ-Tubulin protein expression remained without alteration. This study provides insights into the effect of UVB in cervical cancer cell lines.
Collapse
|
2
|
Xia Y, Lv J, Jiang T, Li B, Li Y, He Z, Xuan Z, Sun G, Wang S, Li Z, Wang W, Wang L, Xu Z. CircFAM73A promotes the cancer stem cell-like properties of gastric cancer through the miR-490-3p/HMGA2 positive feedback loop and HNRNPK-mediated β-catenin stabilization. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:103. [PMID: 33731207 PMCID: PMC7972245 DOI: 10.1186/s13046-021-01896-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/26/2021] [Indexed: 12/31/2022]
Abstract
Background Circular RNAs (circRNAs) have emerged as a new subclass of regulatory RNAs that play critical roles in various cancers. Cancer stem cells (CSCs), a small subset of cancer cells, are believed to possess the capacities to initiate tumorigenesis and promote progression. Although accumulating evidence has suggested that cells with CSC-like properties are crucial for the malignancy of gastric cancer (GC), it remains unclear whether circRNAs are related to the acquisition of CSC-like properties in GC. Methods CircFAM73A expression was analyzed by GEO datasets and verified in GC samples. The roles of circFAM73A in GC cell proliferation, migration, cisplatin resistance, and CSC-like properties were determined by a series of functional experiments both in vitro and in vivo. RNA pulldown was used to explore the miRNAs and proteins binding to circFAM73A. Bioinformatic analysis and experimental verification confirmed the downstream targets of circFAM73A. The regulation of circFAM73A by HMGA2 was verified by ChIP and RIP assays. Results Elevated circFAM73A expression was confirmed in GC tissues, and higher circFAM73A predicted poor prognosis in GC patients. The upregulation of circFAM73A enhanced CSC-like properties in GC, thus facilitating cell proliferation, migration, and cisplatin resistance. Mechanistically, circFAM73A promoted GC malignancy by regulating miR-490-3p/HMGA2 in a positive feedback loop and recruiting HNRNPK to facilitate β-catenin stabilization. Moreover, HMGA2 further enhanced E2F1 and HNRNPL activity, which in turn promoted circFAM73A expression. Conclusions Our work demonstrates the crucial role of circFAM73A in the CSC-like properties of GC and uncovers a positive feedback loop in circFAM73A regulation that leads to the progression of gastric cancer, which may provide new insights into circRNA-based diagnostic and therapeutic strategies. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01896-9.
Collapse
Affiliation(s)
- Yiwen Xia
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Jialun Lv
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Tianlu Jiang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Ying Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Zhongyuan He
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Zhe Xuan
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Guangli Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Sen Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Weizhi Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Linjun Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China.
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Nanjing, Jiangsu Province, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
3
|
Brash DE. Rethinking Causation for Data-intensive Biology: Constraints, Cancellations, and Quantized Organisms: Causality in complex organisms is sculpted by constraints rather than instigators, with outcomes perhaps better described by quantized patterns than rectilinear pathways. Bioessays 2020; 42:e1900135. [PMID: 32484248 PMCID: PMC7518294 DOI: 10.1002/bies.201900135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 03/19/2020] [Indexed: 12/22/2022]
Abstract
Complex organisms thwart the simple rectilinear causality paradigm of "necessary and sufficient," with its experimental strategy of "knock down and overexpress." This Essay organizes the eccentricities of biology into four categories that call for new mathematical approaches; recaps for the biologist the philosopher's recent refinements to the causation concept and the mathematician's computational tools that handle some but not all of the biological eccentricities; and describes overlooked insights that make causal properties of physical hierarchies such as emergence and downward causation straightforward. Reviewing and extrapolating from similar situations in physics, it is suggested that new mathematical tools for causation analysis incorporating feedback, signal cancellation, nonlinear dependencies, physical hierarchies, and fixed constraints rather than instigative changes will reveal unconventional biological behaviors. These include "eigenisms," organisms that are limited to quantized states; trajectories that steer a system such as an evolving species toward optimal states; and medical control via distributed "sheets" rather than single control points.
Collapse
Affiliation(s)
- Douglas E Brash
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, 06520-8040, USA
| |
Collapse
|
4
|
Tam HW, Hall JR, Messenger ZJ, Jima DD, House JS, Linder K, Smart RC. C/EBPβ suppresses keratinocyte autonomous type 1 IFN response and p53 to increase cell survival and susceptibility to UVB-induced skin cancer. Carcinogenesis 2019; 40:1099-1109. [PMID: 30698678 PMCID: PMC10893916 DOI: 10.1093/carcin/bgz012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/04/2019] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
p53 is activated by DNA damage and oncogenic stimuli to regulate senescence, apoptosis and cell-cycle arrest, which are essential to prevent cancer. Here, we utilized UVB radiation, a potent inducer of DNA damage, p53, apoptosis and skin cancer to investigate the mechanism of CCAAT/enhancer binding protein-β (C/EBPβ) in regulating p53-mediated apoptosis in keratinocytes and to test whether the deletion of C/EBPβ in epidermis can protect mice from UVB-induced skin cancer. UVB-treatment of C/EBPβ skin conditional knockout (CKOβ) mice increased p53 protein levels in epidermis and enhanced p53-dependent apoptotic activity 3-fold compared with UVB-treated control mice. UVB increased C/EBPβ levels through a p53-dependent pathway and stimulated the formation of a C/EBPβ-p53 protein complex; knockdown of C/EBPβ increased p53 protein stability in keratinocytes. These results suggest a p53-C/EBPβ feedback loop, whereby C/EBPβ, a transcriptional target of a p53 pathway, functions as a survival factor by negatively regulating p53 apoptotic activity in response to DNA damage. RNAseq analysis of UVB-treated CKOβ epidermis unexpectedly revealed that type 1 interferon (IFN) pathway was the most highly enriched pathway. Numerous pro-apoptotic interferon stimulated genes were upregulated including some known to enhance p53 apoptosis. Our results indicate that p53 and IFN pathways function together in response to DNA damage to result in the activation of extrinsic apoptosis pathways and caspase 8 cleavage. Last, we observed CKOβ mice were resistant to UVB-induced skin cancer. Our results suggest that C/EBPβ represses apoptosis through keratinocyte autonomous suppression of the type 1 IFN response and p53 to increase cell survival and susceptibility to UVB-induced skin cancer.
Collapse
Affiliation(s)
- Hann W Tam
- Toxicology Program, North Carolina State University, Raleigh, NC, USA
| | - Jonathan R Hall
- Toxicology Program, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | | | - Dereje D Jima
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| | - John S House
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| | - Keith Linder
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, USA
| | - Robert C Smart
- Toxicology Program, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
5
|
Saenz-Ponce N, Pillay R, de Long LM, Kashyap T, Argueta C, Landesman Y, Hazar-Rethinam M, Boros S, Panizza B, Jacquemyn M, Daelemans D, Gannon OM, Saunders NA. Targeting the XPO1-dependent nuclear export of E2F7 reverses anthracycline resistance in head and neck squamous cell carcinomas. Sci Transl Med 2018; 10:eaar7223. [PMID: 29950445 DOI: 10.1126/scitranslmed.aar7223] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 05/18/2018] [Indexed: 12/17/2022]
Abstract
Patient mortality rates have remained stubbornly high (40%) for the past 35 years in head and neck squamous cell carcinoma (HNSCC) due to inherent or acquired drug resistance. Thus, a critical issue in advanced SCC is to identify and target the mechanisms that contribute to therapy resistance. We report that the transcriptional inhibitor, E2F7, is mislocalized to the cytoplasm in >80% of human HNSCCs, whereas the transcriptional activator, E2F1, retains localization to the nucleus in SCC. This results in an imbalance in the control of E2F-dependent targets such as SPHK1, which is derepressed and drives resistance to anthracyclines in HNSCC. Specifically, we show that (i) E2F7 is subject to exportin 1 (XPO1)-dependent nuclear export, (ii) E2F7 is selectively mislocalized in most of SCC and multiple other tumor types, (iii) mislocalization of E2F7 in HNSCC causes derepression of Sphk1 and drives anthracycline resistance, and (iv) anthracycline resistance can be reversed with a clinically available inhibitor of XPO1, selinexor, in xenotransplant models of HNSCC. Thus, we have identified a strategy to repurpose anthracyclines for use in SCC. More generally, we provide a strategy to restore the balance of E2F1 (activator) and E2F7 (inhibitor) activity in cancer.
Collapse
Affiliation(s)
- Natalia Saenz-Ponce
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Rachael Pillay
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Lilia Merida de Long
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | | | | | | | | | - Samuel Boros
- Department of Pathology, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Benedict Panizza
- Department of Ear Nose and Throat, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
- School of Medicine, University of Queensland, Princess Alexandra Hospital, Woolloongabba, Queensland 4102, Australia
| | - Maarten Jacquemyn
- Katholieke Universiteit Leuven, Department of Microbiology and Immunology, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Herestraat 49, 3000 Leuven, Belgium
| | - Dirk Daelemans
- Katholieke Universiteit Leuven, Department of Microbiology and Immunology, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Herestraat 49, 3000 Leuven, Belgium
| | - Orla M Gannon
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Nicholas A Saunders
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland 4102, Australia.
| |
Collapse
|
6
|
Ertosun MG, Hapil FZ, Osman Nidai O. E2F1 transcription factor and its impact on growth factor and cytokine signaling. Cytokine Growth Factor Rev 2016; 31:17-25. [PMID: 26947516 DOI: 10.1016/j.cytogfr.2016.02.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 02/27/2016] [Indexed: 12/13/2022]
Abstract
E2F1 is a transcription factor involved in cell cycle regulation and apoptosis. The transactivation capacity of E2F1 is regulated by pRb. In its hypophosphorylated form, pRb binds and inactivates DNA binding and transactivating functions of E2F1. The growth factor stimulation of cells leads to activation of CDKs (cyclin dependent kinases), which in turn phosphorylate Rb and hyperphosphorylated Rb is released from E2F1 or E2F1/DP complex, and free E2F1 can induce transcription of several genes involved in cell cycle entry, induction or inhibition of apoptosis. Thus, growth factors and cytokines generally utilize E2F1 to direct cells to either fate. Furthermore, E2F1 regulates expressions of various cytokines and growth factor receptors, establishing positive or negative feedback mechanisms. This review focuses on the relationship between E2F1 transcription factor and cytokines (IL-1, IL-2, IL-3, IL-6, TGF-beta, G-CSF, LIF), growth factors (EGF, KGF, VEGF, IGF, FGF, PDGF, HGF, NGF), and interferons (IFN-α, IFN-β and IFN-γ).
Collapse
Affiliation(s)
- Mustafa Gokhan Ertosun
- Akdeniz University, Faculty of Medicine, Department of Medical Biology and Genetic, Kampus, Antalya 07070, Turkey
| | - Fatma Zehra Hapil
- Akdeniz University, Faculty of Medicine, Department of Medical Biology and Genetic, Kampus, Antalya 07070, Turkey
| | - Ozes Osman Nidai
- Akdeniz University, Faculty of Medicine, Department of Medical Biology and Genetic, Kampus, Antalya 07070, Turkey.
| |
Collapse
|
7
|
Castillo DS, Campalans A, Belluscio LM, Carcagno AL, Radicella JP, Cánepa ET, Pregi N. E2F1 and E2F2 induction in response to DNA damage preserves genomic stability in neuronal cells. Cell Cycle 2016; 14:1300-14. [PMID: 25892555 DOI: 10.4161/15384101.2014.985031] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
E2F transcription factors regulate a wide range of biological processes, including the cellular response to DNA damage. In the present study, we examined whether E2F family members are transcriptionally induced following treatment with several genotoxic agents, and have a role on the cell DNA damage response. We show a novel mechanism, conserved among diverse species, in which E2F1 and E2F2, the latter specifically in neuronal cells, are transcriptionally induced after DNA damage. This upregulation leads to increased E2F1 and E2F2 protein levels as a consequence of de novo protein synthesis. Ectopic expression of these E2Fs in neuronal cells reduces the level of DNA damage following genotoxic treatment, while ablation of E2F1 and E2F2 leads to the accumulation of DNA lesions and increased apoptotic response. Cell viability and DNA repair capability in response to DNA damage induction are also reduced by the E2F1 and E2F2 deficiencies. Finally, E2F1 and E2F2 accumulate at sites of oxidative and UV-induced DNA damage, and interact with γH2AX DNA repair factor. As previously reported for E2F1, E2F2 promotes Rad51 foci formation, interacts with GCN5 acetyltransferase and induces histone acetylation following genotoxic insult. The results presented here unveil a new mechanism involving E2F1 and E2F2 in the maintenance of genomic stability in response to DNA damage in neuronal cells.
Collapse
Affiliation(s)
- Daniela S Castillo
- a Laboratorio de Biología Molecular; Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales ; Universidad de Buenos Aires ; Ciudad de Buenos Aires , Argentina
| | | | | | | | | | | | | |
Collapse
|
8
|
Zhou Y, Huang WF, Feng Q, Shi SD, Li EL, Li KH, Wu RS, Wu LQ. Toll-like receptor 4 regulates hepatitis B virus related hepatic carcinoma cell proliferation by controlling cyclin-dependent kinase 4/6 expression. Shijie Huaren Xiaohua Zazhi 2015; 23:3029-3037. [DOI: 10.11569/wcjd.v23.i19.3029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the effect of Toll-like receptor 4 (TLR4) on the proliferation of hepatitis B virus (HBV) related hepatic carcinoma cells and the underlying mechanism.
METHODS: The expression of TLR4, cyclin-dependent kinase (CDK) 4 and CDK6 protein in 36 HBV-related hepatic carcinoma tissues and matched adjacent tissues were detected by Western blot, and their correlations in carcinoma tissues were analyzed. Three TLR4 specific siRNAs and a negative control were synthesized and transfected into hepatoma cells Hep-3B using liposomes. The siRNA sequence with the best performance was selected for Western blot analysis. The expression of TLR4, CDK4 and CDK6 proteins was measured by Western blot assay before and after transfection. The proliferation of hepatoma cells was observed by MTT assay and colony formation assay.
RESULTS: The overall expression levels of TLR4, CDK4 and CDK6 proteins in HBV-related hepatic carcinoma were significantly higher than those in the matched adjacent tissues (P < 0.05). There was a positive correlation between TLR4 and CDK4 expression (r = 0.66, P < 0.05), and between TLR4 and CDK6 expression (r = 0.57, P < 0.05). Using the best interference sequence (TLR4-siRNA-03), it was found that the protein levels of CDK4 and CDK6 were significantly decreased in the TLR4-siRNA group (P < 0.05), compared to the control group. In addition, the proliferation of Hep-3B cells and the colony formation rate were both decreased (P < 0.05) in the TLR4-siRNA group.
CONCLUSION: TLR4 may regulate the proliferation of HBV-related hepatic carcinoma cells by controlling the expression of CDK4 and CDK6.
Collapse
|
9
|
Boros G, Miko E, Muramatsu H, Weissman D, Emri E, van der Horst GTJ, Szegedi A, Horkay I, Emri G, Karikó K, Remenyik É. Identification of Cyclobutane Pyrimidine Dimer-Responsive Genes Using UVB-Irradiated Human Keratinocytes Transfected with In Vitro-Synthesized Photolyase mRNA. PLoS One 2015; 10:e0131141. [PMID: 26121660 PMCID: PMC4488231 DOI: 10.1371/journal.pone.0131141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/27/2015] [Indexed: 12/16/2022] Open
Abstract
Major biological effects of UVB are attributed to cyclobutane pyrimidine dimers (CPDs), the most common photolesions formed on DNA. To investigate the contribution of CPDs to UVB-induced changes of gene expression, a model system was established by transfecting keratinocytes with pseudouridine-modified mRNA (Ψ-mRNA) encoding CPD-photolyase. Microarray analyses of this model system demonstrated that more than 50% of the gene expression altered by UVB was mediated by CPD photolesions. Functional classification of the gene targets revealed strong effects of CPDs on the regulation of the cell cycle and transcriptional machineries. To confirm the microarray data, cell cycle-regulatory genes, CCNE1 and CDKN2B that were induced exclusively by CPDs were selected for further investigation. Following UVB irradiation, expression of these genes increased significantly at both mRNA and protein levels, but not in cells transfected with CPD-photolyase Ψ-mRNA and exposed to photoreactivating light. Treatment of cells with inhibitors of c-Jun N-terminal kinase (JNK) blocked the UVB-dependent upregulation of both genes suggesting a role for JNK in relaying the signal of UVB-induced CPDs into transcriptional responses. Thus, photolyase mRNA-based experimental platform demonstrates CPD-dependent and -independent events of UVB-induced cellular responses, and, as such, has the potential to identify novel molecular targets for treatment of UVB-mediated skin diseases.
Collapse
Affiliation(s)
- Gábor Boros
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Edit Miko
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Hiromi Muramatsu
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Eszter Emri
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Andrea Szegedi
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Dermatological Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Irén Horkay
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabriella Emri
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- * E-mail:
| | - Katalin Karikó
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Éva Remenyik
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
10
|
Abstract
p53 plays a key role in regulating DNA damage response by suppressing cell cycle progression or inducing apoptosis depending on extent of DNA damage. However, it is not clear why mild genotoxic stress favors growth arrest, whereas excessive lesions signal cells to die. Here we showed that TAp73, a p53 homologue thought to have a similar function as p53, restrains the transcriptional activity of p53 and prevents excessive activation of its downstream targets upon low levels of DNA damage, which results in cell cycle arrest. Extensive DNA damage triggers TAp73 depletion through ubiquitin/proteasome-mediated degradation of E2F1, leading to enhanced transcriptional activation by p53 and subsequent induction of apoptosis. These findings provide novel insights into the regulation of p53 function and suggest that TAp73 keeps p53 activity in check in regulating cell fate decisions upon genotoxic stress.
Collapse
|
11
|
Hazar-Rethinam M, de Long LM, Gannon OM, Boros S, Vargas AC, Dzienis M, Mukhopadhyay P, Saenz-Ponce N, Dantzic DDE, Simpson F, Saunders NA. RacGAP1 Is a Novel Downstream Effector of E2F7-Dependent Resistance to Doxorubicin and Is Prognostic for Overall Survival in Squamous Cell Carcinoma. Mol Cancer Ther 2015; 14:1939-50. [PMID: 26018753 DOI: 10.1158/1535-7163.mct-15-0076] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/15/2015] [Indexed: 11/16/2022]
Abstract
We have previously shown that E2F7 contributes to drug resistance in head and neck squamous cell carcinoma (HNSCC) cells. Considering that dysregulation of responses to chemotherapy-induced cytotoxicity is one of the major reasons for treatment failure in HNSCC, identifying the downstream effectors that regulate E2F7-dependent sensitivity to chemotherapeutic agents may have direct clinical impact. We used transcriptomic profiling to identify candidate pathways that contribute to E2F7-dependent resistance to doxorubicin. We then manipulated the expression of the candidate pathway using overexpression and knockdown in in vitro and in vivo models of SCC to demonstrate causality. In addition, we examined the expression of E2F7 and RacGAP1 in a custom tissue microarray (TMA) generated from HNSCC patient samples. Transcriptomic profiling identified RacGAP1 as a potential mediator of E2F7-dependent drug resistance. We validated E2F7-dependent upregulation of RacGAP1 in doxorubicin-insensitive SCC25 cells. Extending this, we found that selective upregulation of RacGAP1 induced doxorubicin resistance in previously sensitive KJDSV40. Similarly, stable knockdown of RacGAP1 in insensitive SCC25 cells induced sensitivity to doxorubicin in vitro and in vivo. RacGAP1 expression was validated in a TMA, and we showed that HNSCCs that overexpress RacGAP1 are associated with a poorer patient overall survival. Furthermore, E2F7-induced doxorubicin resistance was mediated via RacGAP1-dependent activation of AKT. Finally, we show that SCC cells deficient in RacGAP1 grow slower and are sensitized to the cytotoxic actions of doxorubicin in vivo. These findings identify RacGAP1 overexpression as a novel prognostic marker of survival and a potential target to sensitize SCC to doxorubicin.
Collapse
Affiliation(s)
- Mehlika Hazar-Rethinam
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Lilia Merida de Long
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Orla M Gannon
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Samuel Boros
- Department of Pathology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Ana Cristina Vargas
- Department of Pathology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Marcin Dzienis
- Department of Medical Oncology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Pamela Mukhopadhyay
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Natalia Saenz-Ponce
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Daniel D E Dantzic
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nicholas A Saunders
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
12
|
Long noncoding RNA lincRNA-p21 is the major mediator of UVB-induced and p53-dependent apoptosis in keratinocytes. Cell Death Dis 2015; 6:e1700. [PMID: 25789975 PMCID: PMC4385943 DOI: 10.1038/cddis.2015.67] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 02/09/2015] [Accepted: 02/17/2015] [Indexed: 12/16/2022]
Abstract
LincRNA-p21 is a long noncoding RNA and a transcriptional target of p53 and HIF-1α. LincRNA-p21 regulates gene expression in cis and trans, mRNA translation, protein stability, the Warburg effect, and p53-dependent apoptosis and cell cycle arrest in doxorubicin-treated mouse embryo fibroblasts. p53 plays a key role in the response of skin keratinocytes to UVB-induced DNA damage by inducing cell cycle arrest and apoptosis. In skin cancer development, UVB-induced mutation of p53 allows keratinocytes upon successive UVB exposures to evade apoptosis and cell cycle arrest. We hypothesized that lincRNA-p21 has a key functional role in UVB-induced apoptosis and/or cell cycle arrest in keratinocytes and loss of lincRNA-p21 function results in the evasion of apoptosis and/or cell cycle arrest. We observed that lincRNA-p21 transcripts are highly inducible by UVB in mouse and human keratinocytes in culture and in mouse skin in vivo. LincRNA-p21 is regulated at the transcriptional level in response to UVB, and the UVB induction of lincRNA-p21 in keratinocytes and in vivo in mouse epidermis is primarily through a p53-dependent pathway. Knockdown of lincRNA-p21 blocked UVB-induced apoptosis in mouse and human keratinocytes, and lincRNA-p21 was responsible for the majority of UVB-induced and p53-mediated apoptosis in keratinocytes. Knockdown of lincRNA-p21 had no effect on cell proliferation in untreated or UVB-treated keratinocytes. An early event in skin cancer is the mutation of a single p53 allele. We observed that a mutant p53+/R172H allele expressed in mouse epidermis (K5Cre+/tg;LSLp53+/R172H) showed a significant dominant-negative inhibitory effect on UVB-induced lincRNA-p21 transcription and apoptosis in epidermis. We conclude lincRNA-p21 is highly inducible by UVB and has a key role in triggering UVB-induced apoptotic death. We propose that the mutation of a single p53 allele provides a pro-oncogenic function early in skin cancer development through a dominant inhibitory effect on UVB-induced lincRNA-p21 expression and the subsequent evasion of UVB-induced apoptosis.
Collapse
|
13
|
Hazar-Rethinam M, de Long LM, Gannon OM, Topkas E, Boros S, Vargas AC, Dzienis M, Mukhopadhyay P, Simpson F, Endo-Munoz L, Saunders NA. A novel E2F/sphingosine kinase 1 axis regulates anthracycline response in squamous cell carcinoma. Clin Cancer Res 2014; 21:417-27. [PMID: 25411162 DOI: 10.1158/1078-0432.ccr-14-1962] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Head and neck squamous cell carcinomas (HNSCC) are frequently drug resistant and have a mortality rate of 45%. We have previously shown that E2F7 may contribute to drug resistance in SCC cells. However, the mechanism and pathways involved remain unknown. EXPERIMENTAL DESIGN We used transcriptomic profiling to identify candidate pathways that may contribute to E2F7-dependent resistance to anthracyclines. We then manipulated the activity/expression of the candidate pathway using overexpression, knockdown, and pharmacological inhibitors in in vitro and in vivo models of SCC to demonstrate causality. In addition, we examined the expression of E2F7 and a downstream effector in a tissue microarray (TMA) generated from HNSCC patient samples. RESULTS E2F7-deficient keratinocytes were selectively sensitive to doxorubicin and this was reversed by overexpressing E2F7. Transcriptomic profiling identified Sphingosine kinase 1 (Sphk1) as a potential mediator of E2F7-dependent drug resistance. Knockdown and overexpression studies revealed that Sphk1 was a downstream target of E2F7. TMA studies showed that E2F7 overexpression correlated with Sphk1 overexpression in human HNSCC. Moreover, inhibition of Sphk1 by shRNA or the Sphk1-specific inhibitor, SK1-I (BML-EI411), enhanced the sensitivity of SCC cells to doxorubicin in vitro and in vivo. Furthermore, E2F7-induced doxorubicin resistance was mediated via Sphk1-dependent activation of AKT in vitro and in vivo. CONCLUSION We identify a novel drugable pathway in which E2F7 directly increases the transcription and activity of the Sphk1/S1P axis resulting in activation of AKT and subsequent drug resistance. Collectively, this novel combinatorial therapy can potentially be trialed in humans using existing agents.
Collapse
Affiliation(s)
- Mehlika Hazar-Rethinam
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Lilia Merida de Long
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Orla M Gannon
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Eleni Topkas
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Samuel Boros
- Department of Pathology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Ana Cristina Vargas
- Department of Pathology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Marcin Dzienis
- Department of Medical Oncology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Pamela Mukhopadhyay
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Liliana Endo-Munoz
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nicholas A Saunders
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
14
|
Deregulation of Rb-E2F1 axis causes chromosomal instability by engaging the transactivation function of Cdc20-anaphase-promoting complex/cyclosome. Mol Cell Biol 2014; 35:356-69. [PMID: 25368385 DOI: 10.1128/mcb.00868-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The E2F family of transcription factors regulates genes involved in various aspects of the cell cycle. Beyond the well-documented role in G1/S transition, mitotic regulation by E2F has also been reported. Proper mitotic progression is monitored by the spindle assembly checkpoint (SAC). The SAC ensures bipolar separation of chromosomes and thus prevents aneuploidy. There are limited reports on the regulation of the SAC by E2F. Our previous work identified the SAC protein Cdc20 as a novel transcriptional regulator of the mitotic ubiquitin carrier protein UbcH10. However, none of the Cdc20 transcription complex proteins have any known DNA binding domain. Here we show that an E2F1-DP1 heterodimer is involved in recruitment of the Cdc20 transcription complex to the UBCH10 promoter and in transactivation of the gene. We further show that inactivation of Rb can facilitate this transactivation process. Moreover, this E2F1-mediated regulation of UbcH10 influences mitotic progression. Deregulation of this pathway results in premature anaphase, chromosomal abnormalities, and aneuploidy. We conclude that excess E2F1 due to Rb inactivation recruits the complex of Cdc20 and the anaphase-promoting complex/cyclosome (Cdc20-APC/C) to deregulate the expression of UBCH10, leading to chromosomal instability in cancer cells.
Collapse
|
15
|
Liu Z, Long X, Chao C, Yan C, Wu Q, Hua S, Zhang Y, Wu A, Fang W. Knocking down CDK4 mediates the elevation of let-7c suppressing cell growth in nasopharyngeal carcinoma. BMC Cancer 2014; 14:274. [PMID: 24751144 PMCID: PMC4014407 DOI: 10.1186/1471-2407-14-274] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 04/08/2014] [Indexed: 11/17/2022] Open
Abstract
Background CDK4 is a protein kinase in the CDK family important for G1/S phase cell cycle progression. However, the roles and molecular mechanisms of CDK4 triggering nasopharynx carcinogenesis are still unclear. Methods Lentiviral-vector mediated shRNA was used to suppress CDK4 expression and examine its molecular mechanisms. Using immunohistochemistry, we analyzed CDK4 protein expression in clinicopathologically characterized nasopharyngeal carcinoma (NPC) cases and nasopharyngeal tissues (NPs). Survival curves were plotted by the Kaplan-Meier method and compared using the log-rank test. Results In this investigation, we knocked down CDK4 expression and observed that NPC cell growth and cell cycle progression were significantly blocked by suppressing expression of CCND1, CDK6, and E2F1 as well as elevated p21 expression. Further, we found that reduced CDK4 expression elevated the expression of let-7c, a tumor-suppressive miRNA modulated by E2F1. We found that let-7c was markedly downregulated in NPC tissues compared to NPs and suppressed cell growth and cell cycle progression by modulating p15/p16/CDK4/E2F1 pathway. Finally, CDK4 protein was observed to be overexpressed in NPC tissues and could be considered an unfavorable prognosis factor for NPC patients although its independent prognostic value did not reach statistical significance (p = 0.087). Conclusions Our results demonstrated that overexpressed CDK4 is an unfavorable prognostic factor which suppresses the expression of tumor suppressive-factor let-7c through p21/CCND1/CDK6/E2F1 signaling, and inhibits cell proliferation by p15/p16/CDK4/E2F1 feedback signaling in NPC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yajie Zhang
- Department of Pathology, Guangzhou Medical University, Guangzhou 510182, China.
| | | | | |
Collapse
|
16
|
Biswas AK, Mitchell DL, Johnson DG. E2F1 responds to ultraviolet radiation by directly stimulating DNA repair and suppressing carcinogenesis. Cancer Res 2014; 74:3369-77. [PMID: 24741006 DOI: 10.1158/0008-5472.can-13-3216] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In response to DNA damage, the E2F1 transcription factor is phosphorylated at serine 31 (serine 29 in mouse) by the ATM or ATR kinases, which promotes E2F1 protein stabilization. Phosphorylation of E2F1 also leads to the recruitment of E2F1 to sites of DNA damage, where it functions to enhance DNA repair. To study the role of this E2F1 phosphorylation event in vivo, a knock-in mouse model was generated, in which serine 29 was mutated to alanine. The S29A mutation impairs E2F1 stabilization in response to ultraviolet (UV) radiation and doxorubicin treatment, but has little effect on the expression of E2F target genes. The apoptotic and proliferative responses to acute UV radiation exposure are also similar between wild-type and E2f1(S29A/) (S29A) mice. As expected, the S29A mutation prevents E2F1 association with damaged DNA and reduces DNA repair efficiency. Moreover, E2f1(S29A/) (S29A) mice display increased sensitivity to UV-induced skin carcinogenesis. This knock-in mouse model thus links the ability of E2F1 to directly promote DNA repair with the suppression of tumor development.
Collapse
Affiliation(s)
- Anup Kumar Biswas
- Authors' Affiliations: Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Science Park; and The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - David L Mitchell
- Authors' Affiliations: Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Science Park; and The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TexasAuthors' Affiliations: Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Science Park; and The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - David G Johnson
- Authors' Affiliations: Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Science Park; and The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TexasAuthors' Affiliations: Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Science Park; and The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
17
|
Yamaguchi K, Yamaguchi R, Takahashi N, Ikenoue T, Fujii T, Shinozaki M, Tsurita G, Hata K, Niida A, Imoto S, Miyano S, Nakamura Y, Furukawa Y. Overexpression of cohesion establishment factor DSCC1 through E2F in colorectal cancer. PLoS One 2014; 9:e85750. [PMID: 24465681 PMCID: PMC3894995 DOI: 10.1371/journal.pone.0085750] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 11/30/2013] [Indexed: 02/06/2023] Open
Abstract
Ctf18-replication factor C complex including Dscc1 (DNA replication and sister chromatid cohesion 1) is implicated in sister chromatid cohesion, DNA replication, and genome stability in S. cerevisiae and C. elegans. We previously performed gene expression profiling in primary colorectal cancer cells in order to identify novel molecular targets for the treatment of colorectal cancer. A feature of the cancer-associated transcriptional signature revealed from this effort is the elevated expression of the proto-oncogene DSCC1. Here, we have interrogated the molecular basis for deviant expression of human DSCC1 in colorectal cancer and its ability to promote survival of cancer cells. Quantitative PCR and immunohistochemical analyses corroborated that the expression level of DSCC1 is elevated in 60-70% of colorectal tumors compared to their matched noncancerous colonic mucosa. An in silico evaluation of the presumptive DSCC1 promoter region for consensus DNA transcriptional regulatory elements revealed a potential role for the E2F family of DNA-binding proteins in controlling DSCC1 expression. RNAi-mediated reduction of E2F1 reduced expression of DSCC1 in colorectal cancer cells. Gain- and loss-of-function experiments demonstrated that DSCC1 is involved in the viability of cancer cells in response to genotoxic stimuli. We reveal that E2F-dependent expression of DSCC1 confers anti-apoptotic properties in colorectal cancer cells, and that its suppression may be a useful option for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- * E-mail:
| | - Rui Yamaguchi
- Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Norihiko Takahashi
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tsuneo Ikenoue
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tomoaki Fujii
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masaru Shinozaki
- Department of Surgery, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Giichiro Tsurita
- Department of Surgery, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Keisuke Hata
- Department of Surgery, Research Hospital, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsushi Niida
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiya Imoto
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yusuke Nakamura
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Costa C, Paramio JM, Santos M. Skin Tumors Rb(eing) Uncovered. Front Oncol 2013; 3:307. [PMID: 24381932 PMCID: PMC3865458 DOI: 10.3389/fonc.2013.00307] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/04/2013] [Indexed: 11/23/2022] Open
Abstract
The Rb1 gene was the first bona fide tumor suppressor identified and cloned more than 25 years ago. Since then, a plethora of studies have revealed the functions of pRb and the existence of a sophisticated and strictly regulated pathway that modulates such functional roles. An emerging paradox affecting Rb1 in cancer connects the relatively low number of mutations affecting Rb1 gene in specific human tumors, compared with the widely functional inactivation of pRb in most, if not in all, human cancers. The existence of a retinoblastoma family of proteins pRb, p107, and p130 and their potential unique and overlapping functions as master regulators of cell cycle progression and transcriptional modulation by similar processes, may provide potential clues to explain such conundrum. Here, we will review the development of different genetically engineered mouse models, in particular those affecting stratified epithelia, and how they have offered new avenues to understand the roles of the Rb family members and their targets in the context of tumor development and progression.
Collapse
Affiliation(s)
- Clotilde Costa
- Molecular Oncology Unit, Department of Basic Research, Centro de Investigaciones Energéticas Medioambientales y Teconológicas (ed70A) , Madrid , Spain
| | - Jesús M Paramio
- Molecular Oncology Unit, Department of Basic Research, Centro de Investigaciones Energéticas Medioambientales y Teconológicas (ed70A) , Madrid , Spain
| | - Mirentxu Santos
- Molecular Oncology Unit, Department of Basic Research, Centro de Investigaciones Energéticas Medioambientales y Teconológicas (ed70A) , Madrid , Spain
| |
Collapse
|
19
|
Liu JL, Zeng GZ, Liu XL, Liu YQ, Hu ZG, Liu Y, Tan NH, Zhou GB. Small compound bigelovin exerts inhibitory effects and triggers proteolysis of E2F1 in multiple myeloma cells. Cancer Sci 2013; 104:1697-704. [PMID: 24118350 DOI: 10.1111/cas.12295] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 09/16/2013] [Accepted: 09/22/2013] [Indexed: 12/30/2022] Open
Abstract
Multiple myeloma (MM) is a currently incurable blood cancer. Here we tested the effects of a small compound bigelovin on MM cells, and reported that it caused cell cycle arrest and subsequently induced apoptosis. Bigelovin triggered proteolysis of E2F1, which could be inhibited by caspase inhibitor. To investigate the clinical relevance, the expression of E2F1 in MM specimens was tested, and the results showed that E2F1 was overexpressed in 25-57% of MM patients and was associated with higher International Staging System (ISS) stage. These results suggest that E2F1 may be important for MM pathogenesis, and bigelovin could serve as a lead compound for the development of E2F1 inhibitor.
Collapse
Affiliation(s)
- Jing-Lei Liu
- Division of Molecular Carcinogenesis and Targeted Therapy for Cancer, Guangzhou Institutes of Biomedicine and Health & State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Science and Technology of China, Hefei, China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Requirement for phosphorylation of P53 at Ser312 in suppression of chemical carcinogenesis. Sci Rep 2013; 3:3105. [PMID: 24173284 PMCID: PMC3813944 DOI: 10.1038/srep03105] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/11/2013] [Indexed: 12/23/2022] Open
Abstract
The p53 tumour suppressor is activated in response to a wide variety of genotoxic stresses, frequently via post-translational modification. Using a knock in mouse model with a Ser312 to Ala mutation, we show here that phosphorylation of p53 on Ser312 helps to prevent tumour induction by the alkylating agent MNU, which predominantly caused T cell lymphomas. This is consistent with our previous observation that p53312A/A mice are more susceptible to X-ray induced tumourigenesis. Phosphorylation on Ser312 aids p53's interaction with E2F1, and enhances p53-mediated apoptosis. Loss of E2F1 alone does not affect tumour susceptibility to MNU, but its absence partially rescues tumour formation in p53312A/A mice, thus reflecting the oncogenic properties of E2F1. Our data confirms the participation of Ser312 phosphorylation in tumour suppression by p53.
Collapse
|
21
|
Frauenstein K, Sydlik U, Tigges J, Majora M, Wiek C, Hanenberg H, Abel J, Esser C, Fritsche E, Krutmann J, Haarmann-Stemmann T. Evidence for a novel anti-apoptotic pathway in human keratinocytes involving the aryl hydrocarbon receptor, E2F1, and checkpoint kinase 1. Cell Death Differ 2013; 20:1425-34. [PMID: 23912710 DOI: 10.1038/cdd.2013.102] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 06/14/2013] [Accepted: 07/04/2013] [Indexed: 01/22/2023] Open
Abstract
Exposure of keratinocytes (KC) to ultraviolet (UV) radiation results in the initiation of apoptosis, a protective mechanism that eliminates cells harboring irreparable DNA damage. Hence, a manipulation of UV-induced apoptosis may significantly influence photocarcinogenesis. We have discovered that the aryl hydrocarbon receptor (AHR), a key regulator of drug metabolism and an UVB-sensitive transcription factor, serves an anti-apoptotic function in UVB-irradiated human KC. Chemical and shRNA-mediated inhibition of AHR signaling sensitized KC to UVB-induced apoptosis by decreasing the expression of E2F1 and its target gene checkpoint kinase 1 (CHK1). The decreased expression of these cell-cycle regulators was due to an enhanced expression of p27(KIP1) and an associated decrease in phosphorylation of both cyclin-dependent kinase 2 and its substrate molecule retinoblastoma protein. The subsequent inhibition of E2F1 autoregulation and downstream CHK1 expression resulted in an enhanced susceptibility of damaged cells to undergo apoptosis. Accordingly, ectopic overexpression of either E2F1 or CHK1 in AHR-knockdown KC attenuated the observed sensitization to UVB-induced apoptosis. Using an AHR-knockout SKH-1 hairless mouse model, we next demonstrated the physiological relevance of the anti-apoptotic function of AHR. In contrast to their AHR-proficient littermates, the constitutive expression of E2F1 and CHK1 was significantly reduced in the skin of AHR-knockout mice. Accordingly, a single exposure of the animals to UVB resulted in an enhanced cleavage of caspase-3 in the skin of AHR-knockout mice. These results identify for the first time the AHR-E2F1-CHK1 axis as a novel anti-apoptotic pathway in KC, which may represent a suitable target for chemoprevention of non-melanoma skin cancer.
Collapse
Affiliation(s)
- K Frauenstein
- IUF-Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
E2F1 apoptosis counterattacked: evil strikes back. Trends Mol Med 2013; 19:89-98. [DOI: 10.1016/j.molmed.2012.10.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/23/2012] [Accepted: 10/23/2012] [Indexed: 12/15/2022]
|
23
|
Abstract
The specific ablation of Rb1 gene in epidermis (Rb(F/F);K14cre) promotes proliferation and altered differentiation but does not produce spontaneous tumour development. These phenotypic changes are associated with increased expression of E2F members and E2F-dependent transcriptional activity. Here, we have focused on the possible dependence on E2F1 gene function. We have generated mice that lack Rb1 in epidermis in an inducible manner (Rb(F/F);K14creER(TM)). These mice are indistinguishable from those lacking pRb in this tissue in a constitutive manner (Rb(F/F);K14cre). In an E2F1-null background (Rb(F/F);K14creER(TM); and E2F1(-/-) mice), the phenotype due to acute Rb1 loss is not ameliorated by E2F1 loss, but rather exacerbated, indicating that pRb functions in epidermis do not rely solely on E2F1. On the other hand, Rb(F/F);K14creER(TM);E2F1(-/-) mice develop spontaneous epidermal tumours of hair follicle origin with high incidence. These tumours, which retain a functional p19(arf)/p53 axis, also show aberrant activation of β-catenin/Wnt pathway. Gene expression studies revealed that these tumours display relevant similarities with specific human tumours. These data demonstrate that the Rb/E2F1 axis exerts essential functions not only in maintaining epidermal homoeostasis, but also in suppressing tumour development in epidermis, and that the disruption of this pathway may induce tumour progression through specific alteration of developmental programs.
Collapse
|
24
|
Hazar-Rethinam M, Endo-Munoz L, Gannon O, Saunders N. The role of the E2F transcription factor family in UV-induced apoptosis. Int J Mol Sci 2011; 12:8947-60. [PMID: 22272113 PMCID: PMC3257110 DOI: 10.3390/ijms12128947] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 11/15/2011] [Accepted: 11/30/2011] [Indexed: 11/16/2022] Open
Abstract
The E2F transcription factor family is traditionally associated with cell cycle control. However, recent data has shown that activating E2Fs (E2F1-3a) are potent activators of apoptosis. In contrast, the recently cloned inhibitory E2Fs (E2F7 and 8) appear to antagonize E2F-induced cell death. In this review we will discuss (i) the potential role of E2Fs in UV-induced cell death and (ii) the implications of this to the development of UV-induced cutaneous malignancies.
Collapse
Affiliation(s)
- Mehlika Hazar-Rethinam
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Queensland 4102, Australia; E-Mails: (M.H.-R.); (L.E.-M.); (O.G.)
| | - Liliana Endo-Munoz
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Queensland 4102, Australia; E-Mails: (M.H.-R.); (L.E.-M.); (O.G.)
| | - Orla Gannon
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Queensland 4102, Australia; E-Mails: (M.H.-R.); (L.E.-M.); (O.G.)
| | - Nicholas Saunders
- Epithelial Pathobiology Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, Queensland 4102, Australia; E-Mails: (M.H.-R.); (L.E.-M.); (O.G.)
- School of Biomedical Sciences, University of Queensland, Queensland 4072, Australia
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +61-7-3176-5894; Fax: +61-7-3176-5946
| |
Collapse
|
25
|
Chen J, Zhu F, Weaks RL, Biswas AK, Guo R, Li Y, Johnson DG. E2F1 promotes the recruitment of DNA repair factors to sites of DNA double-strand breaks. Cell Cycle 2011; 10:1287-94. [PMID: 21512314 DOI: 10.4161/cc.10.8.15341] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The E2F1 transcription factor is post-translationally modified and stabilized in response to various forms of DNA damage to regulate the expression of cell cycle and pro-apoptotic genes. E2F1 also forms foci at DNA double-strand breaks (DSBs) but the function of E2F1 at sites of damage is unknown. Here we demonstrate that the absence of E2F1 leads to spontaneous DNA breaks and impaired recovery following exposure to ionizing radiation. E2F1 deficiency results in defective NBS1 phosphorylation and foci formation in response to DSBs but does not affect NBS1 expression levels. Moreover, an increased association between NBS1 and E2F1 is observed in response to DNA damage, suggesting that E2F1 may promote NBS1 foci formation through a direct or indirect interaction at sites of DNA breaks. E2F1 deficiency also impairs RPA and Rad51 foci formation indicating that E2F1 is important for DNA end resection and the formation of single-stranded DNA at DSBs. These findings establish new roles for E2F1 in the DNA damage response, which may directly contribute to DNA repair and genome maintenance.
Collapse
Affiliation(s)
- Jie Chen
- The University of Texas M.D. Anderson Cancer Center; Smithville, TX, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Guo R, Chen J, Mitchell DL, Johnson DG. GCN5 and E2F1 stimulate nucleotide excision repair by promoting H3K9 acetylation at sites of damage. Nucleic Acids Res 2010; 39:1390-7. [PMID: 20972224 PMCID: PMC3045616 DOI: 10.1093/nar/gkq983] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Chromatin structure is known to be a barrier to DNA repair and a large number of studies have now identified various factors that modify histones and remodel nucleosomes to facilitate repair. In response to ultraviolet (UV) radiation several histones are acetylated and this enhances the repair of DNA photoproducts by the nucleotide excision repair (NER) pathway. However, the molecular mechanism by which UV radiation induces histone acetylation to allow for efficient NER is not completely understood. We recently discovered that the E2F1 transcription factor accumulates at sites of UV-induced DNA damage and directly stimulates NER through a non-transcriptional mechanism. Here we demonstrate that E2F1 associates with the GCN5 acetyltransferase in response to UV radiation and recruits GCN5 to sites of damage. UV radiation induces the acetylation of histone H3 lysine 9 (H3K9) and this requires both GCN5 and E2F1. Moreover, as previously observed for E2F1, knock down of GCN5 results in impaired recruitment of NER factors to sites of damage and inefficient DNA repair. These findings demonstrate a direct role for GCN5 and E2F1 in NER involving H3K9 acetylation and increased accessibility to the NER machinery.
Collapse
Affiliation(s)
- Ruifeng Guo
- Department of Molecular Carcinogenesis, UT MD Anderson Cancer Center, Science Park-Research Division, 1808 Park Road 1C, PO Box 389, Smithville, TX 78957, USA
| | | | | | | |
Collapse
|
27
|
Tavana O, Benjamin CL, Puebla-Osorio N, Sang M, Ullrich SE, Ananthaswamy HN, Zhu C. Absence of p53-dependent apoptosis leads to UV radiation hypersensitivity, enhanced immunosuppression and cellular senescence. Cell Cycle 2010; 9:3328-36. [PMID: 20703098 DOI: 10.4161/cc.9.16.12688] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Genotoxic stress triggers the p53 tumor suppressor network to activate cellular responses that lead to cell cycle arrest, DNA repair, apoptosis or senescence. This network functions mainly through transactivation of different downstream targets, including cell cycle inhibitor p21, which is required for short-term cell cycle arrest or long-term cellular senescence, or proapoptotic genes such as p53 upregulated modulator of apoptosis (PUMA) and Noxa. However, the mechanism that switches from cell cycle arrest to apoptosis is still unknown. In this study, we found that mice harboring a hypomorphic mutant p53, R172P, a mutation that abrogates p53-mediated apoptosis while keeping cell cycle control mostly intact, are more susceptible to ultraviolet-B (UVB)-induced skin damage, inflammation and immunosuppression than wild-type mice. p53(R172P) embryonic fibroblasts (MEFs) are hypersensitive to UVB and prematurely senesce after UVB exposure, in stark contrast to wild-type MEFs, which undergo apoptosis. However, these mutant cells are able to repair UV-induced DNA lesions, indicating that the UV hypersensitive phenotype results from the subsequent damage response. Mutant MEFs show an induction of p53 and p21 after UVR, while wild-type MEFs additionally induce PUMA and Noxa. Importantly, p53(R172P) MEFs failed to downregulate anti-apoptotic protein Bcl-2, which has been shown to play an important role in p53-dependent apoptosis. Taken together, these data demonstrate that in the absence of p53-mediated apoptosis, cells undergo cellular senescence to prevent genomic instability. Our results also indicate that p53-dependent apoptosis may play an active role in balancing cellular growth.
Collapse
Affiliation(s)
- Omid Tavana
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Hirano G, Izumi H, Kidani A, Yasuniwa Y, Han B, Kusaba H, Akashi K, Kuwano M, Kohno K. Enhanced expression of PCAF endows apoptosis resistance in cisplatin-resistant cells. Mol Cancer Res 2010; 8:864-72. [PMID: 20530585 DOI: 10.1158/1541-7786.mcr-09-0458] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Histone acetyltransferase (HAT) regulates transcription. We have previously shown that two HAT genes, Clock and Tip60, are overexpressed, and upregulate glutathione biosynthesis and the expression of DNA repair genes in cisplatin-resistant cells. To better understand the mechanism of HAT-related drug resistance, we investigated the role of another HAT gene, p300/CBP-associated factor (PCAF), and found that PCAF was also overexpressed in cisplatin-resistant cells and endowed an antiapoptotic phenotype through enhanced E2F1 expression. PCAF-overexpressing cells showed enhanced expression of E2F1 and conferred cell resistance to chemotherapeutic agents. Downregulation of PCAF decreased E2F1 expression and sensitized cells to chemotherapeutic agents. Moreover, knockdown of PCAF induced G(1) arrest and apoptosis. These results suggest that PCAF is one of pleiotropic factors for drug resistance and seems to be critical for cancer cell growth.
Collapse
Affiliation(s)
- Gen Hirano
- Department of Molecular Biology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Guo R, Chen J, Zhu F, Biswas AK, Berton TR, Mitchell DL, Johnson DG. E2F1 localizes to sites of UV-induced DNA damage to enhance nucleotide excision repair. J Biol Chem 2010; 285:19308-15. [PMID: 20413589 DOI: 10.1074/jbc.m110.121939] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The E2F1 transcription factor is a well known regulator of cell proliferation and apoptosis, but its role in the DNA damage response is less clear. Using a local UV irradiation technique and immunofluorescence staining, E2F1 is shown to accumulate at sites of DNA damage. Localization of E2F1 to UV-damaged DNA requires the ATM and Rad3-related (ATR) kinase and serine 31 of E2F1 but not an intact DNA binding domain. E2F1 deficiency does not appear to affect the expression of nucleotide excision repair (NER) factors, such as XPC and XPA. However, E2F1 depletion does impair the recruitment of NER factors to sites of damage and reduces the efficiency of DNA repair. E2F1 mutants unable to bind DNA or activate transcription retain the ability to stimulate NER. These findings demonstrate that E2F1 has a direct, non-transcriptional role in DNA repair involving increased recruitment of NER factors to sites of damage.
Collapse
Affiliation(s)
- Ruifeng Guo
- Department of Carcinogenesis, University of Texas MD Anderson Cancer Center, Science Park Research Division, Smithville, Texas 78957, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Chen D, Pacal M, Wenzel P, Knoepfler PS, Leone G, Bremner R. Division and apoptosis of E2f-deficient retinal progenitors. Nature 2010; 462:925-9. [PMID: 20016601 DOI: 10.1038/nature08544] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Accepted: 09/25/2009] [Indexed: 12/16/2022]
Abstract
The activating E2f transcription factors (E2f1, E2f2 and E2f3) induce transcription and are widely viewed as essential positive cell cycle regulators. Indeed, they drive cells out of quiescence, and the 'cancer cell cycle' in Rb1 null cells is E2f-dependent. Absence of activating E2fs in flies or mammalian fibroblasts causes cell cycle arrest, but this block is alleviated by removing repressive E2f or the tumour suppressor p53, respectively. Thus, whether activating E2fs are indispensable for normal division is an area of debate. Activating E2fs are also well known pro-apoptotic factors, providing a defence against oncogenesis, yet E2f1 can limit irradiation-induced apoptosis. In flies this occurs through repression of hid (also called Wrinkled; Smac/Diablo in mammals). However, in mammals the mechanism is unclear because Smac/Diablo is induced, not repressed, by E2f1, and in keratinocytes survival is promoted indirectly through induction of DNA repair targets. Thus, a direct pro-survival function for E2f1-3 and/or its relevance beyond irradiation has not been established. To address E2f1-3 function in normal cells in vivo we focused on the mouse retina, which is a relatively simple central nervous system component that can be manipulated genetically without compromising viability and has provided considerable insight into development and cancer. Here we show that unlike fibroblasts, E2f1-3 null retinal progenitor cells or activated Müller glia can divide. We attribute this effect to functional interchangeability with Mycn. However, loss of activating E2fs caused downregulation of the p53 deacetylase Sirt1, p53 hyperacetylation and elevated apoptosis, establishing a novel E2f-Sirt1-p53 survival axis in vivo. Thus, activating E2fs are not universally required for normal mammalian cell division, but have an unexpected pro-survival role in development.
Collapse
Affiliation(s)
- Danian Chen
- Toronto Western Research Institute, University Health Network, Department of Ophthalmology, and Laboratory Medicine and Pathobiology, University of Toronto, Ontario M5T 2S8, Canada
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
The skin is the largest organ of the body and protects the organism against external physical, chemical and biological insults, such as wounding, ultraviolet radiation and micro-organisms. The epidermis is the upper part of the skin that is continuously renewed. The keratinocytes are the major cell type in the epidermis and undergo a specialized form of programmed cell death, called cornification, which is different from classical apoptosis. In keep with this view, several lines of evidence indicate that NF-kB is an important factor providing protection against keratinocyte apoptosis in homeostatic and inflammatory conditions. In contrast, the hair follicle is an epidermal appendage that shows cyclic apoptosis-driven involution, as part of the normal hair cycle. The different cell death programs need to be well orchestrated to maintain skin homeostasis. One of the major environmental insults to the skin is UVB radiation, causing the occurrence of apoptotic sunburn cells. Deregulation of cell death mechanisms in the skin can lead to diseases such as cancer, necrolysis and graft-versus-host disease. Here we review the apoptotic and the anti-apoptotic mechanisms in skin homeostasis and disease.
Collapse
|
32
|
Zheng C, Ren Z, Wang H, Zhang W, Kalvakolanu DV, Tian Z, Xiao W. E2F1 Induces Tumor Cell Survival via Nuclear Factor-κB–Dependent Induction of EGR1 Transcription in Prostate Cancer Cells. Cancer Res 2009; 69:2324-31. [DOI: 10.1158/0008-5472.can-08-4113] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
33
|
Endo-Munoz L, Dahler A, Teakle N, Rickwood D, Hazar-Rethinam M, Abdul-Jabbar I, Sommerville S, Dickinson I, Kaur P, Paquet-Fifield S, Saunders N. E2F7 can regulate proliferation, differentiation, and apoptotic responses in human keratinocytes: implications for cutaneous squamous cell carcinoma formation. Cancer Res 2009; 69:1800-8. [PMID: 19223542 DOI: 10.1158/0008-5472.can-08-2725] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The E2F family of transcription factors plays a crucial role in the regulation of genes involved in cell proliferation, differentiation, and apoptosis. In keratinocytes, the inhibition of E2F is a key step in the control and initiation of squamous differentiation. Because the product of the recently identified E2F7a/E2F7b gene has been shown to repress E2F-regulated promoters, and to be abundant in skin, we examined its role in the epidermis. Our results indicate that E2F7b mRNA expression is selectively associated with proliferation-competent keratinocytes. Moreover, E2F7 was able to antagonize E2F1-induced proliferation and apoptosis. In contrast, although E2F7 was able to inhibit proliferation and initiate differentiation, it was unable to antagonize the differentiation suppression induced by E2F1. These data indicate that E2F7-mediated suppression of proliferation and apoptosis acts through E2F1-dependent pathways, whereas E2F7-induced differentiation acts through an E2F1-independent pathway. These data also suggest that proliferation, differentiation, and survival of primary human keratinocytes can be controlled by the relative ratio of E2F1 to E2F7. Because deregulated proliferation, differentiation, and apoptosis are hallmarks of cancer, we examined the expression levels of E2F1 and E2F7 in cutaneous squamous cell carcinomas (CSCC). We found that both genes were overexpressed in CSCCs compared with normal epidermis. Furthermore, inhibition of E2F7 in a SCC cell line sensitized the cells to UV-induced apoptosis and doxorubicin-induced apoptosis. Combined, these data suggest that the selected disruption of E2F1 and E2F7 in keratinocytes is likely to contribute to CSCC formation and may prove to be a viable therapeutic target.
Collapse
Affiliation(s)
- Liliana Endo-Munoz
- Translational Research Unit, Cancer Collaborative Group, Princess Alexandra Hospital, Wooloongabba, Queensland 4102, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Polager S, Ginsberg D. E2F - at the crossroads of life and death. Trends Cell Biol 2008; 18:528-35. [PMID: 18805009 DOI: 10.1016/j.tcb.2008.08.003] [Citation(s) in RCA: 234] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Revised: 08/05/2008] [Accepted: 08/05/2008] [Indexed: 12/16/2022]
Abstract
The retinoblastoma tumor suppressor, pRb, restricts cell-cycle progression mainly by regulating members of the E2F-transcription-factor family. The Rb pathway is often inactivated in human tumors, resulting in deregulated-E2F activity that promotes proliferation or cell death, depending on the cellular context. Specifically, the outcome of deregulated-E2F activity is determined by integration of signals coming from the cellular DNA and the external environment. Alterations in cell proliferation and cell-death pathways are key features of transformed cells and, therefore, an understanding of the variables that determine the outcome of E2F activation is pivotal for cancer research and treatment. In this review, we discuss recent studies that have elucidated some of the signals affecting E2F activity and that have revealed additional E2F targets and functions, thereby enriching the understanding of this versatile transcription-factor family.
Collapse
Affiliation(s)
- Shirley Polager
- The Mina and Everard Goodman Faculty of Life Science, Bar Ilan University, Ramat Gan 52900, Israel
| | | |
Collapse
|
35
|
Palacios G, Talos F, Nemajerova A, Moll UM, Petrenko O. E2F1 plays a direct role in Rb stabilization and p53-independent tumor suppression. Cell Cycle 2008; 7:1776-81. [PMID: 18583939 DOI: 10.4161/cc.7.12.6030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
To better understand the role of E2F1 in tumor formation, we analyzed spontaneous tumorigenesis in p53(-/-)E2F1(+/+) and p53(-/-)E2F1(-/-) mice. We show that the combined loss of p53 and E2F1 leads to an increased incidence of sarcomas and carcinomas compared to the loss of p53 alone. E2F1-deficient tumors show wide chromosomal variation, indicative of genomic instability. Consistent with this, p53(-/-)E2F1(-/-) primary fibroblasts have a reduced capacity to maintain genomic stability when exposed to S-phase inhibitors or genotoxic drugs. A major mechanism of E2F1's contribution to genomic integrity lies in mediating stabilization and engagement of the Rb protein.
Collapse
Affiliation(s)
- Gustavo Palacios
- Department of Pathology, State University of New York at Stony Brook, Stony Brook, New York 11794, USA
| | | | | | | | | |
Collapse
|
36
|
Chen D, Yu Z, Zhu Z, Lopez CD. E2F1 regulates the base excision repair gene XRCC1 and promotes DNA repair. J Biol Chem 2008; 283:15381-9. [PMID: 18348985 PMCID: PMC2397471 DOI: 10.1074/jbc.m710296200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 03/12/2008] [Indexed: 11/06/2022] Open
Abstract
The E2F1 transcription factor activates S-phase-promoting genes, mediates apoptosis, and stimulates DNA repair through incompletely understood mechanisms. XRCC1 (x-ray repair cross-complementing group 1) protein is important for efficient single strand break/base excision repair. Although both damage and proliferative signals increase XRCC1 levels, the mechanisms regulating XRCC1 transcription remain unclear. To study these upstream mechanisms, the XRCC1 promoter was cloned into a luciferase reporter. Ectopic expression of wild-type E2F1, but not an inactive mutant E2F1(132E), activated the XRCC1 promoter-luciferase reporter, and deletion of predicted E2F1 binding sites in the promoter attenuated E2F1-induced activation. Endogenous XRCC1 expression increased in cells conditionally expressing wild-type, but not mutant E2F1, and methyl methanesulfonate-induced DNA damage stimulated XRCC1 expression in E2F1(+/+) but not E2F1(-/-) mouse embryo fibroblasts (MEFs). Additionally, E2F1(-/-) MEFs displayed attenuated DNA repair after methyl methanesulfonate-induced damage compared with E2F1(+/+) MEFs. Moreover, Chinese hamster ovary cells with mutant XRCC1 (EM9) were more sensitive to E2F1-induced apoptosis compared with Chinese hamster ovary cells with wild-type XRCC1 (AA8). These results provide new mechanistic insight into the role of the E2F pathway in maintaining genomic stability.
Collapse
Affiliation(s)
| | | | | | - Charles D. Lopez
- Department of Medicine, Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, Oregon 97239
| |
Collapse
|
37
|
Lee J, Park CK, Park JO, Lim T, Park YS, Lim HY, Lee I, Sohn TS, Noh JH, Heo JS, Kim S, Lim DH, Kim KM, Kang WK. Impact of E2F-1 expression on clinical outcome of gastric adenocarcinoma patients with adjuvant chemoradiation therapy. Clin Cancer Res 2008; 14:82-8. [PMID: 18172256 DOI: 10.1158/1078-0432.ccr-07-0612] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE There are no reliable prognostic markers that identify gastric cancer patients who may benefit from adjuvant chemoradiation therapy. E2F-1 was shown to be associated with radiosensitivity and chemosensitivity in certain tumor types. Therefore, we analyzed expression and prognostic significance of E2F-1 along with thymidylate synthase (TS) in R(0)-resected gastric adenocarcinoma patients, who underwent adjuvant chemoradiation therapy with 5-fluorouracil (5-FU) and leucovorin. EXPERIMENTAL DESIGN The chemosensitivity to 5-FU and radiosensitivity were tested in three E2F-1-overexpressed gastric cancer cell lines in vitro. The expressions of TS and E2F-1 were analyzed in 467 R(0)-resected primary gastric cancer patients, who received adjuvant chemoradiation therapy with 5-FU and leucovorin using tissue microarray. RESULTS The E2F-1 immunopositivity rate was 22.2% (103 of 465 samples) with a cutoff value of 5% immunoreactivity, whereas the TS-positive expression occurred in 19.0% of the 463 tumors tested. Using stepwise Cox proportional hazards regression modeling, multivariate analyses showed that the E2F-1 immunopositivity predicted more favorable survival as compared with the E2F-1 immunonegativity with borderline statistical significance [P = 0.050, hazard ratio (HR) = 0.702, 95% confidence interval, 0.487, 1.013]. However, the E2F-1 immunopositivity did not retain its statistical significance at multivariate analysis for predicting disease-free survival (data not shown, P = 0.270), but stage was the only influential factor for disease-free survival in stages IB to IV (M(0)) patients (P < 0.001). TS immunopositivity did not influence survival (P = 0.459) or disease-free survival (P = 0.447). CONCLUSION E2F-1 is a potentially novel independent prognostic factor that may identify gastric cancer patients who will likely benefit from adjuvant chemoradiation therapy following curative resection.
Collapse
Affiliation(s)
- Jeeyun Lee
- Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Disruption of pRB-E2F interactions by E1A is a key event in the adenoviral life cycle that drives expression of early viral transcription and induces cell cycle progression. This function of E1A is complicated by E2F1, an E2F family member that controls multiple processes besides proliferation, including apoptosis and DNA repair. Recently, a second interaction site in pRB that only contacts E2F1 has been discovered, allowing pRB to control proliferation separately from other E2F1-dependent activities. Based on this new insight into pRB-E2F1 regulation, we investigated how E1A affects control of E2F1 by pRB. Our data reveal that pRB-E2F1 interactions are resistant to E1A-mediated disruption. Using mutant forms of pRB that selectively force E2F1 to bind through only one of the two binding sites on pRB, we determined that E1A is unable to disrupt E2F1's unique interaction with pRB. Furthermore, analysis of pRB-E2F complexes during adenoviral infection reveals the selective maintenance of pRB-E2F1 interactions despite the presence of E1A. Our experiments also demonstrate that E2F1 functions to maintain cell viability in response to E1A expression. This suggests that adenovirus E1A's seemingly complex mechanism of disrupting pRB-E2F interactions provides selectivity in promoting viral transcription and cell cycle advancement, while maintaining cell viability.
Collapse
|
39
|
Iaquinta PJ, Lees JA. Life and death decisions by the E2F transcription factors. Curr Opin Cell Biol 2007; 19:649-57. [PMID: 18032011 DOI: 10.1016/j.ceb.2007.10.006] [Citation(s) in RCA: 241] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Accepted: 10/06/2007] [Indexed: 11/28/2022]
Abstract
The E2F transcription factors are critical regulators of genes required for appropriate progression through the cell cycle, and in special circumstances they can also promote the expression of another class of genes that function in the apoptotic program. Since E2Fs can initiate both cell proliferation and cell death, it is not surprising that the pro-apoptotic capacity of these proteins is subject to complex regulation. Recent study has expanded our knowledge of the factors influencing E2F-induced apoptosis as well as downstream targets of E2F in this process.
Collapse
Affiliation(s)
- Phillip J Iaquinta
- Center for Cancer Research, Massachusetts Institute of Technology, E17-517B, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | | |
Collapse
|
40
|
Opavsky R, Tsai SY, Guimond M, Arora A, Opavska J, Becknell B, Kaufmann M, Walton NA, Stephens JA, Fernandez SA, Muthusamy N, Felsher DW, Porcu P, Caligiuri MA, Leone G. Specific tumor suppressor function for E2F2 in Myc-induced T cell lymphomagenesis. Proc Natl Acad Sci U S A 2007; 104:15400-5. [PMID: 17881568 PMCID: PMC2000495 DOI: 10.1073/pnas.0706307104] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Deregulation of the Myc pathway and deregulation of the Rb pathway are two of the most common abnormalities in human malignancies. Recent in vitro experiments suggest a complex cross-regulatory relationship between Myc and Rb that is mediated through the control of E2F. To evaluate the functional connection between Myc and E2Fs in vivo, we used a bitransgenic mouse model of Myc-induced T cell lymphomagenesis and analyzed tumor progression in mice deficient for E2f1, E2f2, or E2f3. Whereas the targeted inactivation of E2f1 or E2f3 had no significant effect on tumor progression, loss of E2f2 accelerated lymphomagenesis. Interestingly, loss of a single copy of E2f2 also accelerated tumorigenesis, albeit to a lesser extent, suggesting a haploinsufficient function for this locus. The combined ablation of E2f1 or E2f3, along with E2f2, did not further accelerate tumorigenesis. Myc-overexpressing T cells were more resistant to apoptosis in the absence of E2f2, and the reintroduction of E2F2 into these tumor cells resulted in an increase of apoptosis and inhibition of tumorigenesis. These results identify the E2f2 locus as a tumor suppressor through its ability to modulate apoptosis.
Collapse
Affiliation(s)
- Rene Opavsky
- *Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine and Public Health and
- Department of Molecular Genetics, College of Biological Sciences
| | - Shih-Yin Tsai
- *Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine and Public Health and
- Department of Molecular Genetics, College of Biological Sciences
| | - Martin Guimond
- *Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine and Public Health and
- Division of Hematology and Oncology, Department of Internal Medicine, and
| | - Anjulie Arora
- *Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine and Public Health and
- Department of Molecular Genetics, College of Biological Sciences
| | - Jana Opavska
- *Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine and Public Health and
- Department of Molecular Genetics, College of Biological Sciences
| | - Brian Becknell
- *Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine and Public Health and
- Division of Hematology and Oncology, Department of Internal Medicine, and
| | - Michael Kaufmann
- *Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine and Public Health and
- Department of Molecular Genetics, College of Biological Sciences
| | - Nathaniel A. Walton
- *Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine and Public Health and
- Department of Molecular Genetics, College of Biological Sciences
| | | | | | | | - Dean W. Felsher
- Division of Oncology, Department of Medicine, Stanford University, CCSR 1105B, 269 Campus Drive, Stanford, CA 94305-5151
| | - Pierluigi Porcu
- Division of Hematology and Oncology, Department of Internal Medicine, and
| | - Michael A. Caligiuri
- *Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine and Public Health and
- Division of Hematology and Oncology, Department of Internal Medicine, and
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210; and
| | - Gustavo Leone
- *Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, College of Medicine and Public Health and
- Department of Molecular Genetics, College of Biological Sciences
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210; and
- **To whom correspondence should be addressed. E-mail:
| |
Collapse
|
41
|
Lim CA, Yao F, Wong JJY, George J, Xu H, Chiu KP, Sung WK, Lipovich L, Vega VB, Chen J, Shahab A, Zhao XD, Hibberd M, Wei CL, Lim B, Ng HH, Ruan Y, Chin KC. Genome-wide mapping of RELA(p65) binding identifies E2F1 as a transcriptional activator recruited by NF-kappaB upon TLR4 activation. Mol Cell 2007; 27:622-35. [PMID: 17707233 DOI: 10.1016/j.molcel.2007.06.038] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Revised: 05/04/2007] [Accepted: 06/25/2007] [Indexed: 12/18/2022]
Abstract
NF-kappaB is a key mediator of inflammation. Here, we mapped the genome-wide loci bound by the RELA subunit of NF-kappaB in lipopolysaccharide (LPS)-stimulated human monocytic cells, and together with global gene expression profiling, found an overrepresentation of the E2F1-binding motif among RELA-bound loci associated with NF-kappaB target genes. Knockdown of endogenous E2F1 impaired the LPS inducibility of the proinflammatory cytokines CCL3(MIP-1alpha), IL23A(p19), TNF-alpha, and IL1-beta. Upon LPS stimulation, E2F1 is rapidly recruited to the promoters of these genes along with p50/RELA heterodimer via a mechanism that is dependent on NF-kappaB activation. Together with the observation that E2F1 physically interacts with p50/RELA in LPS-stimulated cells, our findings suggest that NF-kappaB recruits E2F1 to fully activate the transcription of NF-kappaB target genes. Global gene expression profiling subsequently revealed a spectrum of NF-kappaB target genes that are positively regulated by E2F1, further demonstrating the critical role of E2F1 in the Toll-like receptor 4 pathway.
Collapse
Affiliation(s)
- Ching-Aeng Lim
- Laboratory of Immunology and Virology, Genome Institute of Singapore, 138672 Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Knezevic D, Zhang W, Rochette PJ, Brash DE. Bcl-2 is the target of a UV-inducible apoptosis switch and a node for UV signaling. Proc Natl Acad Sci U S A 2007; 104:11286-91. [PMID: 17586682 PMCID: PMC2040891 DOI: 10.1073/pnas.0701318104] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sunlight's UVB radiation triggers cell signaling at multiple sites to induce apoptosis. The integration of these signal entry sites is not understood. Here we show that P53 and E2f1 constitute a UV-inducible apoptosis switch. At low-UV doses, wild-type cells resemble the OFF state of an siP53-treated cell, whereas at high-UV doses, the apoptosis frequency transitions to the fully ON behavior of an siE2f1-treated cell. The switch's target is Bcl-2: Rapid Bcl-2 down-regulation in response to UVB-induced DNA photoproducts is lost in P53-deficient cells, but, as for apoptosis, is restored when both P53 and its inhibited target E2f1 are absent. P53's down-regulation of Bcl-2 is mediated entirely through E2f1. Bcl-2 is also down-regulated by a separate pathway triggered by DNA photoproducts in the absence of P53 and E2f1. Four UV pathways terminating on Bcl-2 contribute to apoptosis after UVB irradiation. The apoptosis lost in p53(-/-) is completely restored by siBcl-2, implying that Bcl-2 is a rate-limiting member of this network. These results identify Bcl-2 as an integrator of several UV-induced proapoptotic signals and show that it, in turn, suppresses a direct UV-apoptosis pathway. UV-induced apoptosis requires both UV activation of the direct pathway and a separate UV disinhibition of this pathway through P53-E2f1-Bcl-2.
Collapse
Affiliation(s)
- Dejan Knezevic
- Departments of *Therapeutic Radiology
- Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06525
| | | | | | - Douglas E. Brash
- Departments of *Therapeutic Radiology
- Genetics
- Dermatology, and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
43
|
Gu M, Singh RP, Dhanalakshmi S, Mohan S, Agarwal R. Differential effect of silibinin on E2F transcription factors and associated biological events in chronically UVB-exposed skin versus tumors in SKH-1 hairless mice. Mol Cancer Ther 2006; 5:2121-9. [PMID: 16928834 DOI: 10.1158/1535-7163.mct-06-0052] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UVB radiation-induced DNA damage in skin activates cellular pathways involved in DNA repair, cell cycle regulation, and apoptosis, important events that prevent conversion of damaged skin cells into cancer. We reported recently the efficacy of silibinin against photocarcinogenesis along with altered molecular events in tumors (Cancer Research, 64:6349-56, 2004). The molecular and biological events modulated by silibinin in chronically UVB-irradiated skin leading to cancer prevention, however, are not known. Herein, we describe effect of silibinin on skin 15 and 25 weeks after UVB exposure and compared them with molecular alterations in skin tumors. UVB decreased E2F1 but increased E2F2 and E2F3 protein levels in skin, and these were reversed by silibinin treatment. Silibinin-induced E2F1 was accompanied by an inhibition of apoptosis and decreases in p53 and cyclin-dependent kinase inhibitors. Silibinin-caused decrease in E2F2 and E2F3 was accompanied by reduced levels of cyclin-dependent kinases, cyclins, CDC25C, and mitogen-activated protein kinases and Akt signaling and inhibition of cell proliferation. In tumorigenesis protocols, topical or dietary silibinin significantly inhibited tumor appearance and growth. As opposed to UVB-exposed skin, UVB-induced tumors showed elevated levels of E2F1, but these were reduced in silibinin-treated tumors without any effect on E2F2 and E2F3. Contrary to the inhibition of apoptosis and p53 expression in UVB-exposed skin cells, silibinin increased these variables in tumors. These differential effects of silibinin on E2F1 versus E2F2 and E2F3 and their associated molecular alterations and biological effects in chronic UVB-exposed skin suggest their role in silibinin interference with photocarcinogenesis.
Collapse
Affiliation(s)
- Mallikarjuna Gu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Health Sciences Center, 4200 East Ninth Avenue, Box C238, Denver, 80262, USA
| | | | | | | | | |
Collapse
|
44
|
Yamasaki L. Modeling cell cycle control and cancer with pRB tumor suppressor. Results Probl Cell Differ 2006; 42:227-56. [PMID: 16903213 DOI: 10.1007/b136682] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cancer is a complex syndrome of diseases characterized by the increased abundance of cells that disrupts the normal tissue architecture within an organism. Defining one universal mechanism underlying cancer with the hope of designing a magic bullet against cancer is impossible, largely because there is so much variation between various types of cancer and different individuals. However, we have learned much in past decades about different journeys that a normal cell takes to become cancerous, and that the delicate balance between oncogenes and tumor suppressor is upset, favoring growth and survival of the tumor cell. One of the most important cellular barriers to cancer development is the retinoblastoma tumor suppressor (pRB) pathway, which is inactivated in a wide range of human tumors and controls cell cycle progression via repression of the E2F/DP transcription factor family. Much of the clarity with which we view tumor suppression via pRB is due to our belief in the universality of the cell cycle and our attempts to model tumor pathways in vivo, nowhere so evident as in the multitude of data emerging from mutant mouse models that have been engineered to understand how cell cycle regulators limit growth in vivo and how deregulation of these regulators facilitates cancer development. In spite of this clarity, we have witnessed with incredulity several stunning results in the last 2 years that have challenged the very foundations of the cell cycle paradigm and made us question seriously how important these cell cycle regulators actually are.
Collapse
Affiliation(s)
- Lili Yamasaki
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| |
Collapse
|
45
|
Abstract
Keratinocyte (KC) apoptosis plays a critical role in regulating epidermal development and restraining carcinogenesis. Apoptosis balances proliferation to maintain epidermal thickness, contributes to stratum corneum formation and may eliminate pre-malignant cells. Apart from the normal developmental program, KC apoptosis can be triggered by UV light and other stimuli. Dysfunctional apoptosis occurs in some skin diseases, such as psoriasis and skin cancer. Here we review the current state of knowledge of KC apoptosis, with particular focus on apoptotic signaling pathways and molecular mechanisms of apoptosis control, and discuss new insights into the complex role of apoptosis in skin carcinogenesis that are emerging from mouse models.
Collapse
Affiliation(s)
- Deepak Raj
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - Douglas E. Brash
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Douglas Grossman
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Department of Dermatology, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| |
Collapse
|
46
|
Zhang Z, Wang H, Li M, Rayburn ER, Agrawal S, Zhang R. Stabilization of E2F1 protein by MDM2 through the E2F1 ubiquitination pathway. Oncogene 2005; 24:7238-47. [PMID: 16170383 DOI: 10.1038/sj.onc.1208814] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although previous studies suggested that the tumorigenicity of mouse double minute 2 (MDM2) was due to its negative regulation of p53, the p53-independent interactions may be equally as important. During recent studies utilizing MDM2 inhibitors, we noted that E2F transcription factor 1 (E2F1) was down regulated upon inhibition of MDM2, regardless of the p53 status of the cancer. The present study investigated the mechanisms responsible for the MDM2-mediated increase in E2F1 expression. MDM2 prolongs the half-life of the E2F1 protein by inhibiting its ubiquitination. MDM2 displaces SCF(SKP2), the E2F1 E3 ligase. Direct binding between MDM2 and E2F1 is necessary for the negative effects of MDM2 on E2F1 ubiquitination, and deletion of the MDM2 nuclear localization signal does not result in loss of the ability to increase the E2F1 protein level. The downregulation of E2F1 upon MDM2 inhibition was not due to either pRB or p14(Arf). In addition, E2F1 was responsible for at least part of the inhibition of cell proliferation induced by MDM2 knockdown. In conclusion, the present study provides evidence that stabilization of the E2F1 protein is likely another p53-independent component of MDM2-mediated tumorigenesis. More knowledge about the MDM2-E2F1 interaction may be helpful in developing novel anticancer therapies.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Pharmacology and Toxicology and Division of Clinical Pharmacology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
47
|
Zhang Z, Li M, Rayburn ER, Hill DL, Zhang R, Wang H. Oncogenes as novel targets for cancer therapy (part III): transcription factors. ACTA ACUST UNITED AC 2005; 5:327-38. [PMID: 16196502 DOI: 10.2165/00129785-200505050-00005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This is the third paper in a four-part serial review on potential therapeutic targeting of oncogenes. The previous parts described the involvement of oncogenes in different aspects of cancer growth and development, and considered the new technologies responsible for the advancement of oncogene identification, target validation, and drug design. Because of such advances, new specific and more efficient therapeutic agents can be developed for cancer. This part of the review continues the exploration of various oncogenes that we have grouped within seven categories: growth factors, tyrosine kinases, intermediate signaling molecules, transcription factors, cell cycle regulators, DNA damage repair genes, and genes involved in apoptosis. Part one discussed growth factors and tyrosine kinases and part two discussed intermediate signaling molecules. This portion of the review covers transcription factors and the various strategies being used to inhibit their expression or decrease their activities.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Pharmacology and Toxicology and Division of Clinical Pharmacology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019, USA
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Our understanding of roles for the E2F transcription factor in regulating apoptosis has progressed from asking what E2F can do based on overexpression studies to what E2F actually does based on analyses of loss-of-function mutants. A paper in this issue of Developmental Cell implicates Drosophila E2F1 in context-dependent pro- as well as anti-apoptotic roles in the same tissue following genotoxic stress.
Collapse
Affiliation(s)
- James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Coloradao, Denver, Colorado 80045, USA
| |
Collapse
|
49
|
Abstract
The E2F family of transcription factors is a central modulator of important cellular events, including cell cycle progression, apoptosis and DNA damage response. The role of E2F family members in various human malignancies is yet unclear and may provide vital clues to the diagnosis, prognosis and therapy of cancer patients. In this review we provide a brief but concise overview of E2F function and its putative role in the most common human tumour types.
Collapse
Affiliation(s)
- P K Tsantoulis
- Department of Histology and Embryology, Molecular Carcinogenesis Group, School of Medicine, University of Athens, Antaiou 53 Str, Lamprini, Ano Patissia, GR-11146, Athens, Greece
| | | |
Collapse
|
50
|
Ivanova IA, D'Souza SJA, Dagnino L. Signalling in the epidermis: the E2F cell cycle regulatory pathway in epidermal morphogenesis, regeneration and transformation. Int J Biol Sci 2005; 1:87-95. [PMID: 15951853 PMCID: PMC1142216 DOI: 10.7150/ijbs.1.87] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2004] [Accepted: 02/01/2005] [Indexed: 02/06/2023] Open
Abstract
The epidermis is the outermost layer in the skin, and it is the first line of defence against the environment. The epidermis also provides a barrier against loss of fluids and electrolytes, which is crucial for life. Essential in the maintenance of this tissue is its ability to continually self-renew and regenerate after injury. These two characteristics are critically dependent on the ability of the principal epidermal cell type, the keratinocyte, to proliferate and to respond to differentiation cues. Indeed, the epidermis is a multilayered tissue composed of keratinocyte stem cells and their differentiated progeny. Central for the control of cell proliferation is the E2F transcription factor regulatory network. This signaling network also includes cyclins, cdk, cdk inhibitors and the retinoblastoma (pRb) family of proteins. The biological importance of the E2F/pRb pathway is emphasized by the fact that a majority of human tumours exhibit alterations that disrupt the ability of pRb proteins to inhibit E2F, leading to permanent activation of the latter. Further, E2F is essential for normal epidermal regeneration after injury. Other member of the E2F signaling pathway are also involved in epidermal development and pathophysiology. Thus, whereas the pRb family of proteins is essential for epidermal morphogenesis, abnormal regulation of cyclins and E2F proteins results in tumorgenesis in this tissue. In this review, we discuss the role of each member of this important growth regulatory network in epidermal formation, homeostasis and carcinogenesis.
Collapse
Affiliation(s)
- Iordanka A Ivanova
- Dept. of Physiology & Pharmacology, University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|