1
|
Xiao R, Baptista RP, Agyabeng-Dadzie F, Li Y, Dong Y, Schmitz RJ, Glenn TC, Kissinger JC. Deciphering Transcription in Cryptosporidium parvum: Polycistronic Gene Expression and Chromatin Accessibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633476. [PMID: 39868316 PMCID: PMC11761812 DOI: 10.1101/2025.01.17.633476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Once considered rare in eukaryotes, polycistronic mRNA expression has been identified in kinetoplastids and, more recently, green algae, red algae, and certain fungi. This study provides comprehensive evidence supporting the existence of polycistronic mRNA expression in the apicomplexan parasite Cryptosporidium parvum. Leveraging long-read RNA-seq data from different parasite strains and using multiple long-read technologies, we demonstrate the existence of defined polycistronic transcripts containing 2-4 protein encoding genes, several validated with RT-PCR. Some polycistrons exhibit differential expression profiles, usually involving the generation of internal monocistronic transcripts at different times during development. ATAC-seq in sporozoites reveals that polycistronic transcripts usually have a single open chromatin peak at their 5-prime ends, which contains a single E2F binding site motif. Polycistronic genes do not appear enriched for either male or female exclusive genes. This study elucidates a potentially complex layer of gene regulation with distinct chromatin accessibility akin to monocistronic transcripts. This is the first report of polycistronic transcription in an apicomplexan and expands our understanding of gene expression strategies in this medically important organism.
Collapse
Affiliation(s)
- Rui Xiao
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rodrigo P. Baptista
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
- Houston Methodist Research Hospital, Houston, TX, 77030, USA
| | | | - Yiran Li
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA
| | - Yinxin Dong
- Department of Genetics, University of Georgia, Athens, GA, 30602, USA
| | - Robert J. Schmitz
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Department of Genetics, University of Georgia, Athens, GA, 30602, USA
| | - Travis C. Glenn
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Department of Genetics, University of Georgia, Athens, GA, 30602, USA
- Department of Environmental Health Science, University of Georgia, Athens, GA, 30602, USA
| | - Jessica C. Kissinger
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
- Department of Genetics, University of Georgia, Athens, GA, 30602, USA
| |
Collapse
|
2
|
Liu P, Zhang X, Li Z, Wei L, Peng Q, Liu C, Wu Y, Yan Q, Ma J. A significant role of transcription factors E2F in inflammation and tumorigenesis of nasopharyngeal carcinoma. Biochem Biophys Res Commun 2020; 524:816-824. [PMID: 32044038 DOI: 10.1016/j.bbrc.2020.01.158] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 12/11/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor from head and neck with characteristics in remarkable geographic and racial distribution worldwide, which has the important features of vigorous proliferation and inflammatory cells infiltration. By analyzing the expression profile data of NPC, we found that the E2F-related gene sets were highly enriched in NPC tissues. E2F transcription factor family is an important cycle regulator, which can promote the malignant proliferation and tumorigenesis. Here, we showed that E2Fs accelerated malignant phenotypes of NPC cells. RNA sequencing revealed that E2Fs can significantly up-regulate the inflammatory pathways in NPC cells. E2F1, as a transcription factor, can active the transcription activity of IL-6 promoter, and modulate macrophage function through a microenvironment manner. Thus, this study characterized a significant role of E2Fs in inflammation and tumorigenesis of NPC, which provided a promising anti-tumor target in NPC, since E2Fs are highly expressed and activated in NPC.
Collapse
Affiliation(s)
- Peishan Liu
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China; Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, China
| | - Xiaoyue Zhang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China; Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China; NHC Key Laboratory of Carcinogenesis (Central South University), Changsha, China
| | - Zhengshuo Li
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Lingyu Wei
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Qiu Peng
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Can Liu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Yangge Wu
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China
| | - Qun Yan
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, China.
| | - Jian Ma
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China; Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, China; NHC Key Laboratory of Carcinogenesis (Central South University), Changsha, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, China.
| |
Collapse
|
3
|
Li X. Epigenetics and cell cycle regulation in cystogenesis. Cell Signal 2019; 68:109509. [PMID: 31874209 DOI: 10.1016/j.cellsig.2019.109509] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/16/2022]
Abstract
The role of genetic mutations in the development of polycystic kidney disease (PKD), such as alterations in PKD1 and PKD2 genes in autosomal dominant PKD (ADPKD), is well understood. However, the significance of epigenetic mechanisms in the progression of PKD remains unclear and is increasingly being investigated. The term of epigenetics describes a range of mechanisms in genome function that do not solely result from the DNA sequence itself. Epigenetic information can be inherited during mammalian cell division to sustain phenotype specifically and physiologically responsive gene expression in the progeny cells. A multitude of functional studies of epigenetic modifiers and systematic genome-wide mapping of epigenetic marks reveal the importance of epigenomic mechanisms, including DNA methylation, histone/chromatin modifications and non-coding RNAs, in PKD pathologies. Deregulated proliferation is a characteristic feature of cystic renal epithelial cells. Moreover, defects in many of the molecules that regulate the cell cycle have been implicated in cyst formation and progression. Recent evidence suggests that alterations of DNA methylation and histone modifications on specific genes and the whole genome involved in cell cycle regulation and contribute to the pathogenesis of PKD. This review summarizes the recent advances of epigenetic mechanisms in PKD, which helps us to define the term of "PKD epigenetics" and group PKD epigenetic changes in three categories. In particularly, this review focuses on the interplay of epigenetic mechanisms with cell cycle regulation during normal cell cycle progression and cystic cell proliferation, and discusses the potential to detect and quantify DNA methylation from body fluids as diagnostic/prognostic biomarkers. Collectively, this review provides concepts and examples of epigenetics in cell cycle regulation to reveal a broad view of different aspects of epigenetics in biology and PKD, which may facilitate to identify possible novel therapeutic intervention points and to explore epigenetic biomarkers in PKD.
Collapse
Affiliation(s)
- Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, United States of America; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, United States of America.
| |
Collapse
|
4
|
Expression and purification of the recombinant full-length retinoblastoma protein and characterisation of its interaction with the oncoprotein HDM2. Protein Expr Purif 2019; 162:62-66. [DOI: 10.1016/j.pep.2019.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/29/2019] [Indexed: 01/11/2023]
|
5
|
Wang SN, Wang LT, Sun DP, Chai CY, Hsi E, Kuo HT, Yokoyama KK, Hsu SH. Intestine-specific homeobox (ISX) upregulates E2F1 expression and related oncogenic activities in HCC. Oncotarget 2018; 7:36924-36939. [PMID: 27175585 PMCID: PMC5095049 DOI: 10.18632/oncotarget.9228] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/16/2016] [Indexed: 01/29/2023] Open
Abstract
Intestine-specific homeobox (ISX), a newly identified proto-oncogene, is involved in cell proliferation and progression of hepatocellular carcinoma (HCC). However, the underlying mechanisms linking gene expression and tumor formation remain unclear. In this study, we found that ISX transcriptionally activated E2F transcription factor 1 (E2F1) and associated oncogenic activity by directly binding to the E2 site of its promoter. Forced expression of ISX increased the expression of and phosphorylated the serine residue at position 332 of E2F1, which may be translocated into the nucleus to form the E2F1–DP-1 complex, suggesting that the promotion of oncogenic activities of the ISX–E2F1 axis plays a critical role in hepatoma cells. Coexpression of ISX and E2F1 significantly promoted p53 and RB-mediated cell proliferation and anti-apoptosis, and repressed apoptosis and autophagy. In contrast, short hairpin RNAi-mediated attenuation of ISX and E2F1 decreased cell proliferation and malignant transformation, respectively, in hepatoma cells in vitro and in vivo. The mRNA expression of E2F1 and ISX in 238 paired specimens from human HCC patients, and the adjacent, normal tissues exhibited a tumor-specific expression pattern which was highly correlated with disease pathogenesis, patient survival time, progression stage, and poor prognosis. Therefore, our results indicate that E2F1 is an important downstream gene of ISX in hepatoma progression.
Collapse
Affiliation(s)
- Shen-Nien Wang
- Division of Hepatobiliary Surgery, Department of Surgery, Faculty of Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Ting Wang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ding-Ping Sun
- Division of General Surgery, Department of Surgery, Chi-Mei Medical Center, Tainan, Taiwan.,Department of Food Science and Technology, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung, Taiwan
| | - Edward Hsi
- Department of Genome Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsing-Tao Kuo
- Department of Internal Medicine, Division of Hepatogastroenterology, Chi-Mei Medical Center, Tainan, Taiwan.,Department of Senior Citizen Service Management, Chia Nan University of Pharmacy & Science, Tainan, Taiwan
| | - Kazunari K Yokoyama
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Research Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Faculty of Science and Engineering, Tokushima Bunri University, Sanuki, Japan.,Center of Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Hsien Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center of Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
6
|
Seoane M, Costoya JA, Arce VM. Uncoupling Oncogene-Induced Senescence (OIS) and DNA Damage Response (DDR) triggered by DNA hyper-replication: lessons from primary mouse embryo astrocytes (MEA). Sci Rep 2017; 7:12991. [PMID: 29021613 PMCID: PMC5636792 DOI: 10.1038/s41598-017-13408-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/22/2017] [Indexed: 11/15/2022] Open
Abstract
Oncogene-induced senescence (OIS) is a complex process, in which activation of oncogenic signals during early tumorigenesis results in a high degree of DNA replication stress. The ensuing response to the DNA damage produces a permanent G1 arrest that prevents unlimited cell proliferation and lessens the development of tumours. However, despite the role of OIS in the proliferative arrest resulting from an activating oncogenic-lesion has obtained wide support, there is also evidence indicating that cells may overcome oncogene-induced senescence under some circumstances. In this study, we have investigated the possibility that some of the assumptions on the role of DNA damage response (DDR) in triggering OIS may depend on the fact that most of the available data were obtained in mouse embryo fibroblast. By comparing the degree of OIS observed in mouse embryo fibroblasts (MEF) and mouse embryo astrocytes (MEA) obtained from the same individuals we have demonstrated that, despite truthful activation of DDR in both cell types, significant levels of OIS were only detected in MEF. Therefore, this uncoupling between OIS and DDR observed in astrocytes supports the intriguingly possibility that OIS is not a widespread response mechanism to DDR.
Collapse
Affiliation(s)
- Marcos Seoane
- Molecular Oncology Laboratory MOL. Departamento de Fisioloxia, Facultade de Medicina and Centro de Investigación en Medicina Molecular e Enfermidades Crónicas (CiMUS). Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - José A Costoya
- Molecular Oncology Laboratory MOL. Departamento de Fisioloxia, Facultade de Medicina and Centro de Investigación en Medicina Molecular e Enfermidades Crónicas (CiMUS). Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS). Universidade de Santiago de Compostela, Santiago de Compostela, Spain.
| | - Víctor M Arce
- Molecular Oncology Laboratory MOL. Departamento de Fisioloxia, Facultade de Medicina and Centro de Investigación en Medicina Molecular e Enfermidades Crónicas (CiMUS). Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
7
|
Neault M, Mallette F, Richard S. miR-137 Modulates a Tumor Suppressor Network-Inducing Senescence in Pancreatic Cancer Cells. Cell Rep 2016; 14:1966-78. [DOI: 10.1016/j.celrep.2016.01.068] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 12/10/2015] [Accepted: 01/22/2016] [Indexed: 12/18/2022] Open
|
8
|
Law ME, Corsino PE, Narayan S, Law BK. Cyclin-Dependent Kinase Inhibitors as Anticancer Therapeutics. Mol Pharmacol 2015; 88:846-52. [PMID: 26018905 PMCID: PMC4613943 DOI: 10.1124/mol.115.099325] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/27/2015] [Indexed: 12/20/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) have been considered promising drug targets for a number of years, but most CDK inhibitors have failed rigorous clinical testing. Recent studies demonstrating clear anticancer efficacy and reduced toxicity of CDK4/6 inhibitors such as palbociclib and multi-CDK inhibitors such as dinaciclib have rejuvenated the field. Favorable results with palbociclib and its recent U.S. Food and Drug Administration approval demonstrate that CDK inhibitors with narrow selectivity profiles can have clinical utility for therapy based on individual tumor genetics. A brief overview of results obtained with ATP-competitive inhibitors such as palbociclib and dinaciclib is presented, followed by a compilation of new avenues that have been pursued toward the development of novel, non-ATP-competitive CDK inhibitors. These creative ways to develop CDK inhibitors are presented along with crystal structures of these agents complexed with CDK2 to highlight differences in their binding sites and mechanisms of action. The recent successes of CDK inhibitors in the clinic, combined with the potential for structure-based routes to the development of non-ATP-competitive CDK inhibitors, and evidence that CDK inhibitors may have use in suppressing chromosomal instability and in synthetic lethal drug combinations inspire optimism that CDK inhibitors will become important weapons in the fight against cancer.
Collapse
Affiliation(s)
- Mary E Law
- Departments of Pharmacology and Therapeutics (M.E.L., P.E.C., B.K.L.), Anatomy and Cell Biology (S.N.), and the University of Florida Health Cancer Center (M.E.L., P.E.C., S.N., B.K.L.), University of Florida, Gainesville, Florida
| | - Patrick E Corsino
- Departments of Pharmacology and Therapeutics (M.E.L., P.E.C., B.K.L.), Anatomy and Cell Biology (S.N.), and the University of Florida Health Cancer Center (M.E.L., P.E.C., S.N., B.K.L.), University of Florida, Gainesville, Florida
| | - Satya Narayan
- Departments of Pharmacology and Therapeutics (M.E.L., P.E.C., B.K.L.), Anatomy and Cell Biology (S.N.), and the University of Florida Health Cancer Center (M.E.L., P.E.C., S.N., B.K.L.), University of Florida, Gainesville, Florida
| | - Brian K Law
- Departments of Pharmacology and Therapeutics (M.E.L., P.E.C., B.K.L.), Anatomy and Cell Biology (S.N.), and the University of Florida Health Cancer Center (M.E.L., P.E.C., S.N., B.K.L.), University of Florida, Gainesville, Florida
| |
Collapse
|
9
|
Ngo DC, Ververis K, Tortorella SM, Karagiannis TC. Introduction to the molecular basis of cancer metabolism and the Warburg effect. Mol Biol Rep 2015; 42:819-23. [PMID: 25672512 DOI: 10.1007/s11033-015-3857-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In differentiated normal cells, the conventional route of glucose metabolism involves glycolysis, followed by the citric acid cycle and electron transport chain to generate usable energy in the form of adenosine triphosphate (ATP). This occurs in the presence of oxygen. In hypoxic conditions, normal cells undergo anaerobic glycolysis to yield significantly less energy producing lactate as a product. As first highlighted in the 1920s by Otto Warburg, the metabolism exhibited by tumor cells involves an increased rate of aerobic glycolysis, known as the Warburg effect. In aerobic glycolysis, pyruvate molecules yielded from glycolysis are converted into fewer molecules of ATP even in the presence of oxygen. Evidence indicates that the reasons as to why tumor cells undergo aerobic glycolysis include: (1) the shift in priority to accumulate biomass rather than energy production, (2) the evasion of apoptosis as fewer reactive oxygen species are released by the mitochondria and (3) the production of lactate to further fuel growth of tumors. In this mini-review we discuss emerging molecular aspects of cancer metabolism and the Warburg effect. Aspects of the Warburg effect are analyzed in the context of the established hallmarks of cancer including the role of oncogenes and tumor suppressor genes.
Collapse
Affiliation(s)
- Darleen C Ngo
- Epigenomic Medicine, The Alfred Medical Research and Education Precinct, Baker IDI Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
| | | | | | | |
Collapse
|
10
|
Choi J, Yang ES, Cha K, Whang J, Choi WJ, Avraham S, Kim TA. The Nuclear Matrix Protein, NRP/B, Acts as a Transcriptional Repressor of E2F-mediated Transcriptional Activity. J Cancer Prev 2014; 19:187-98. [PMID: 25337588 PMCID: PMC4189505 DOI: 10.15430/jcp.2014.19.3.187] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 08/29/2014] [Accepted: 08/29/2014] [Indexed: 12/12/2022] Open
Abstract
Background: NRP/B, a family member of the BTB/Kelch repeat proteins, is implicated in neuronal and cancer development, as well as the regulation of oxidative stress responses in breast and brain cancer. Our previous studies indicate that the NRP/B-BTB/POZ domain is involved in the dimerization of NRP/B and in a complex formation with the tumor suppressor, retinoblastoma protein. Although much evidence supports the potential role of NRP/B as a tumor suppressor, the molecular mechanisms of NRP/B action on E2F transcription factors have not been elucidated. Methods: Three-dimensional modeling of NRP/B was used to generate point mutations in the BTB/Kelch domains. Tet-on inducible NRP/B expression was established. The NRP/B deficient breast cancer cell line, MDA-MB-231, was generated using lentiviral shNRP/B to evaluate the effect of NRP/B on cell proliferation, invasion and migration. Immunoprecipitation was performed to verify the interaction of NRP/B with E2F and histone deacetylase (HDAC-1), and the expression level of NRP/B protein was analyzed by Western blot analysis. Changes in cell cycle were determined by flow cytometry. Transcriptional activities of E2F transcription factors were measured by chloramphenicol acetyltransferase (CAT) activity. Results: Ectopic overexpression of NRP/B demonstrated that the NRP/B-BTB/POZ domain plays a critical role in E2F-mediated transcriptional activity. Point mutations within the BTB/POZ domain restored E2-promoter activity inhibited by NRP/B. Loss of NRP/B enhanced the proliferation and migration of breast cancer cells. Endogenous NRP/B interacted with E2F and HDAC1. Treatement with an HDAC inhibitor, trichostatin A (TSA), abolished the NRP/B-mediated suppression of E2-promoter activity. Gain or loss of NRP/B in HeLa cells confirmed the transcriptional repressive capability of NRP/B on the E2F target genes, Cyclin E and HsORC (Homo sapiens Origin Recognition Complex). Conclusions: The present study shows that NRP/B acts as a transcriptional repressor by interacting with the co-repressors, HDAC1, providing new insight into the molecular mechanisms of NRP/B on tumor suppression.
Collapse
Affiliation(s)
- Jina Choi
- CHA Cancer Institute, CHA University, Seoul, Korea
| | - Eun Sung Yang
- Cancer Cell Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kiweon Cha
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - John Whang
- Cancer Cell Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Shalom Avraham
- Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Tae-Aug Kim
- CHA Cancer Institute, CHA University, Seoul, Korea ; Cancer Cell Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
11
|
KMTase Set7/9 is a critical regulator of E2F1 activity upon genotoxic stress. Cell Death Differ 2014; 21:1889-99. [PMID: 25124555 DOI: 10.1038/cdd.2014.108] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 05/01/2014] [Accepted: 06/09/2014] [Indexed: 12/20/2022] Open
Abstract
During the recent years lysine methyltransferase Set7/9 ((Su(var)-3-9, Enhancer-of-Zeste, Trithorax) domain containing protein 7/9) has emerged as an important regulator of different transcription factors. In this study, we report a novel function for Set7/9 as a critical co-activator of E2 promoter-binding factor 1 (E2F1)-dependent transcription in response to DNA damage. By means of various biochemical, cell biology, and bioinformatics approaches, we uncovered that cell-cycle progression through the G1/S checkpoint of tumour cells upon DNA damage is defined by the threshold of expression of both E2F1 and Set7/9. The latter affects the activity of E2F1 by indirectly modulating histone modifications in the promoters of E2F1-dependent genes. Moreover, Set7/9 differentially affects E2F1 transcription targets: it promotes cell proliferation via expression of the CCNE1 gene and represses apoptosis by inhibiting the TP73 gene. Our biochemical screening of the panel of lung tumour cell lines suggests that these two factors are critically important for transcriptional upregulation of the CCNE1 gene product and hence successful progression through cell cycle. These findings identify Set7/9 as a potential biomarker in tumour cells with overexpressed E2F1 activity.
Collapse
|
12
|
Chakrabarti L, Wang BD, Lee NH, Sandler AD. A mechanism linking Id2-TGFβ crosstalk to reversible adaptive plasticity in neuroblastoma. PLoS One 2013; 8:e83521. [PMID: 24376712 PMCID: PMC3871549 DOI: 10.1371/journal.pone.0083521] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/05/2013] [Indexed: 12/19/2022] Open
Abstract
The ability of high-risk neuroblastoma to survive unfavorable growth conditions and multimodal therapy has produced an elusive childhood cancer with remarkably poor prognosis. A novel phenomenon enabling neuroblastoma to survive selection pressure is its capacity for reversible adaptive plasticity. This plasticity allows cells to transition between highly proliferative anchorage dependent (AD) and slow growing, anoikis-resistant anchorage independent (AI) phenotypes. Both phenotypes are present in established mouse and human tumors. The differential gene expression profile of the two cellular phenotypes in the mouse Neuro2a cell line delineated pathways of proliferation in AD cells or tyrosine kinase activation/ apoptosis inhibition in AI cells. A 20 fold overexpression of inhibitor of differentiation 2 (Id2) was identified in AD cells while up-regulation of genes involved in anoikis resistance like PI3K/Akt, Erk, Bcl2 and integrins was observed in AI cells. Similarly, differential expression of Id2 and other genes of interest were also observed in the AD and AI phenotypes of human neuroblastoma cell lines, SK-N-SH and IMR-32; as well as in primary human tumor specimens. Forced down-regulation of Id2 in AD cells or overexpression in AI cells induced the cells to gain characteristics of the other phenotype. Id2 binds both TGFβ and Smad2/3 and appears critical for maintaining the proliferative phenotype at least partially through negative regulation of the TGFβ/Smad pathway. Simultaneously targeting the differential molecular pathways governing reversible adaptive plasticity resulted in 50% cure of microscopic disease and delayed tumor growth in established mouse neuroblastoma tumors. We present a mechanism that accounts for reversible adaptive plasticity and a molecular basis for combined targeted therapies in neuroblastoma.
Collapse
Affiliation(s)
- Lina Chakrabarti
- The Joseph E. Robert Center for Surgical Care, Children’s National Medical Center, Washington, D.C., United States of America
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Medical Center, Washington, D.C., United States of America
| | - Bi-Dar Wang
- Department of Pharmacology and Physiology, George Washington University Medical Center, Washington, D.C., United States of America
| | - Norman H. Lee
- Department of Pharmacology and Physiology, George Washington University Medical Center, Washington, D.C., United States of America
| | - Anthony D. Sandler
- The Joseph E. Robert Center for Surgical Care, Children’s National Medical Center, Washington, D.C., United States of America
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Medical Center, Washington, D.C., United States of America
- * E-mail:
| |
Collapse
|
13
|
Garcia-Jove Navarro M, Basset C, Arcondéguy T, Touriol C, Perez G, Prats H, Lacazette E. Api5 contributes to E2F1 control of the G1/S cell cycle phase transition. PLoS One 2013; 8:e71443. [PMID: 23940755 PMCID: PMC3737092 DOI: 10.1371/journal.pone.0071443] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 07/05/2013] [Indexed: 02/02/2023] Open
Abstract
Background The E2f transcription factor family has a pivotal role in controlling the cell fate in general, and in particular cancer development, by regulating the expression of several genes required for S phase entry and progression through the cell cycle. It has become clear that the transcriptional activation of at least one member of the family, E2F1, can also induce apoptosis. An appropriate balance of positive and negative regulators appears to be necessary to modulate E2F1 transcriptional activity, and thus cell fate. Methodology/Principal Findings In this report, we show that Api5, already known as a regulator of E2F1 induced-apoptosis, is required for the E2F1 transcriptional activation of G1/S transition genes, and consequently, for cell cycle progression and cell proliferation. Api5 appears to be a cell cycle regulated protein. Removal of Api5 reduces cyclin E, cyclin A, cyclin D1 and Cdk2 levels, causing G1 cell cycle arrest and cell cycle delay. Luciferase assays established that Api5 directly regulates the expression of several G1/S genes under E2F1 control. Using protein/protein and protein/DNA immunoprecipitation studies, we demonstrate that Api5, even if not physically interacting with E2F1, contributes positively to E2F1 transcriptional activity by increasing E2F1 binding to its target promoters, through an indirect mechanism. Conclusion/Significance The results described here support the pivotal role of cell cycle related proteins, that like E2F1, may act as tumor suppressors or as proto-oncogenes during cancer development, depending on the behavior of their positive and negative regulators. According to our findings, Api5 contributes to E2F1 transcriptional activation of cell cycle-associated genes by facilitating E2F1 recruitment onto its target promoters and thus E2F1 target gene transcription.
Collapse
Affiliation(s)
| | - Céline Basset
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Cancer Department, Toulouse, France
| | - Tania Arcondéguy
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Cancer Department, Toulouse, France
| | - Christian Touriol
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Cancer Department, Toulouse, France
| | - Guillaume Perez
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Cancer Department, Toulouse, France
| | - Hervé Prats
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Cancer Department, Toulouse, France
| | - Eric Lacazette
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Cancer Department, Toulouse, France
- * E-mail:
| |
Collapse
|
14
|
Raj N, Zhang L, Wei Y, Arnosti DN, Henry RW. Rbf1 degron dysfunction enhances cellular DNA replication. Cell Cycle 2012; 11:3731-8. [PMID: 22895052 DOI: 10.4161/cc.21665] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The E2F family of transcription factors contributes to oncogenesis through activation of multiple genes involved in cellular proliferation, a process that is opposed by the Retinoblastoma tumor suppressor protein (RB). RB also increases E2F1 stability by inhibiting its proteasome-mediated degradation, but the consequences of this post-translational regulation of E2F1 remain unknown. To better understand the mechanism of E2F stabilization and its physiological relevance, we examined the streamlined Rbf1-dE2F1 network in Drosophila. During embryonic development, Rbf1 is insulated from ubiquitin-mediated turnover by the COP9 signalosome, a multi-protein complex that modulates E3 ubiquitin ligase activity. Here, we report that the COP9 signalosome also protects the Cullin4-E3 ligase that is responsible for dE2F1 proteasome-mediated destruction. This dual role of the COP9 signalosome may serve to buffer E2F levels, enhancing its turnover via Cul4 protection and its stabilization through protection of Rbf1. We further show that Rbf1-mediated stabilization of dE2F1 and repression of dE2F1 cell cycle-target genes are distinct properties. Removal of an evolutionarily conserved Rbf1 C terminal degron disabled Rbf1 repression without affecting dE2F1 stabilization. This mutant form of Rbf1 also enhanced G(1)-to-S phase progression when expressed in Rbf1-containing S2 embryonic cells, suggesting that such mutations may generate gain-of-function properties relevant to cellular transformation. Consistent with this idea, several studies have identified mutations in the homologous C terminal domains of RB and p130 in human cancer.
Collapse
Affiliation(s)
- Nitin Raj
- Program in Genetics, Michigan State University, East Lansing, MI, USA
| | | | | | | | | |
Collapse
|
15
|
Naemi FMA, Ali S, Kirby JA. Antibody-mediated allograft rejection: the emerging role of endothelial cell signalling and transcription factors. Transpl Immunol 2011; 25:96-103. [PMID: 21782944 DOI: 10.1016/j.trim.2011.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 07/08/2011] [Accepted: 07/08/2011] [Indexed: 10/18/2022]
Abstract
The presence of antibodies against class I human leukocyte antigens (HLA) can cause the development of chronic allograft rejection. Although endothelial cell activation has been identified as a main effector, the mechanisms mediating this process are not fully understood. Exposure of endothelium to antibodies against HLA antigens induces cell activation which promotes rejection. This activation process can be divided into two phases: an early response in which intracellular signalling proteins and transcription factors are activated, and a later change in protein expression and cell function. In this review, antibody-mediated endothelial signalling and the role of transcription factors in organ transplantation will be described with a particular focus on their potential to mediate some of the graft-damaging effects of anti-HLA class I antibodies.
Collapse
Affiliation(s)
- Fatmah M A Naemi
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | | | | |
Collapse
|
16
|
Boulay PL, Schlienger S, Lewis-Saravalli S, Vitale N, Ferbeyre G, Claing A. ARF1 controls proliferation of breast cancer cells by regulating the retinoblastoma protein. Oncogene 2011; 30:3846-61. [PMID: 21478909 DOI: 10.1038/onc.2011.100] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ADP-ribosylation factors (ARFs) 1 and 6 are small GTP-binding proteins, highly expressed and activated in several breast cancer cell lines and are associated with enhanced migration and invasiveness. In this study, we report that ARF1 has a critical role in cell proliferation. Depletion of this GTPase or expression of a dominant negative form, which both resulted in diminished ARF1 activity, led to sustained cell-growth arrest. This cellular response was associated with the induction of senescent markers in highly invasive breast cancer cells as well as in control mammary epithelial cells by a mechanism regulating retinoblastoma protein (pRB) function. When examining the role of ARF1, we found that this GTPase was highly activated in normal proliferative conditions, and that a limited amount could be found in the nucleus, associated with the chromatin of MDA-MB-231 cells. However, when cells were arrested in the G(0)/G(1) phase or transfected with a dominant negative form of ARF1, the total level of activated ARF1 was markedly reduced and the GTPase significantly enriched in the chromatin. Using biochemical approaches, we demonstrated that the GDP-bound form of ARF1 directly interacted with pRB, but not other members of this family of proteins. In addition, depletion of ARF1 or expression of ARF1T(31)N resulted in the constitutive association of pRB and E2F1, thereby stabilizing the interaction of E2F1 as well as pRB at endogenous sites of target gene promoters, preventing expression of E2F target genes, such as cyclin D1, Mcm6 and E2F1, important for cell-cycle progression. These novel findings provide direct physiological and molecular evidence for the role of ARF1 in controlling cell proliferation, dependent on its ability to regulate pRB/E2F1 activity and gene expression for enhanced proliferation and breast cancer progression.
Collapse
Affiliation(s)
- P-L Boulay
- Department of Pharmacology, University of Montreal, Québec, Canada
| | | | | | | | | | | |
Collapse
|
17
|
Xie W, Jin L, Mei Y, Wu M. E2F1 represses beta-catenin/TCF activity by direct up-regulation of Siah1. J Cell Mol Med 2010; 13:1719-1727. [PMID: 20187294 DOI: 10.1111/j.1582-4934.2008.00423.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Transcription factor E2F1 is a key regulator of cell proliferation and apoptosis. Its activity is strictly controlled by the pRB/E2F pathway. In the majority of cancer cells, however, this pathway is frequently found deregulated, and the underlying mechanism involving transcriptional control by E2F1 has not yet been fully elucidated. Here we report the identification of two putative E2F1-binding sites located upstream from Siah1 transcription start site (+1). Chromatin immunoprecipitation assay reveals that transcription factor E2F1 is capable of binding to the putative sites, and luciferase reporter assay shows that E2F1 can activate transcription from the Siah1 promoter. Ectopic expression of E2F1 elevates the Siah1 level, hence suppressing the beta-catenin/TCF activity. Consistently, knock-down of endogenous E2F1 by a shRNA strategy results in reduced expression of Siah1. Moreover, repression of beta-catenin/TCF activity by E2F1 can be attenuated by shRNA-based repression of endogenous Siah1, implying that Siah1 is a bona fide E2F1 target gene, which at least partly, mediates the suppression of beta-catenin/TCF signalling pathway.
Collapse
Affiliation(s)
- Wei Xie
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Lei Jin
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Yide Mei
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Mian Wu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
18
|
Brown KC, Witte TR, Hardman WE, Luo H, Chen YC, Carpenter AB, Lau JK, Dasgupta P. Capsaicin displays anti-proliferative activity against human small cell lung cancer in cell culture and nude mice models via the E2F pathway. PLoS One 2010; 5:e10243. [PMID: 20421925 PMCID: PMC2857654 DOI: 10.1371/journal.pone.0010243] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Accepted: 03/24/2010] [Indexed: 11/18/2022] Open
Abstract
Background Small cell lung cancer (SCLC) is characterized by rapid progression and low survival rates. Therefore, novel therapeutic agents are urgently needed for this disease. Capsaicin, the active ingredient of chilli peppers, displays anti-proliferative activity in prostate and epidermoid cancer in vitro. However, the anti-proliferative activity of capsaicin has not been studied in human SCLCs. The present manuscript fills this void of knowledge and explores the anti-proliferative effect of capsaicin in SCLC in vitro and in vivo. Methodology/Principal Findings BrdU assays and PCNA ELISAs showed that capsaicin displays robust anti-proliferative activity in four human SCLC cell lines. Furthermore, capsaicin potently suppressed the growth of H69 human SCLC tumors in vivo as ascertained by CAM assays and nude mice models. The second part of our study attempted to provide insight into molecular mechanisms underlying the anti-proliferative activity of capsaicin. We found that the anti-proliferative activity of capsaicin is correlated with a decrease in the expression of E2F-responsive proliferative genes like cyclin E, thymidylate synthase, cdc25A and cdc6, both at mRNA and protein levels. The transcription factor E2F4 mediated the anti-proliferative activity of capsaicin. Ablation of E2F4 levels by siRNA methodology suppressed capsaicin-induced G1 arrest. ChIP assays demonstrated that capsaicin caused the recruitment of E2F4 and p130 on E2F-responsive proliferative promoters, thereby inhibiting cell proliferation. Conclusions/Significance Our findings suggest that the anti-proliferative effects of capsaicin could be useful in the therapy of human SCLCs.
Collapse
Affiliation(s)
- Kathleen C. Brown
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States of America
| | - Ted R. Witte
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States of America
| | - W. Elaine Hardman
- Department of Biochemistry and Microbiology, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States of America
| | - Haitao Luo
- Department of Biology, Alderson-Broaddus College, Phillipi, West Virginia, United States of America
| | - Yi C. Chen
- Department of Biology, Alderson-Broaddus College, Phillipi, West Virginia, United States of America
| | - A. Betts Carpenter
- Department of Anatomy and Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States of America
| | - Jamie K. Lau
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States of America
| | - Piyali Dasgupta
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States of America
- * E-mail:
| |
Collapse
|
19
|
Rizzo P, Miao H, D'Souza G, Osipo C, Yun J, Zhao H, Mascarenhas J, Wyatt D, Antico G, Hao L, Yao K, Rajan P, Hicks C, Siziopikou K, Selvaggi S, Bashir A, Bhandari D, Marchese A, Lendahl U, Qin JZ, Tonetti DA, Albain K, Nickoloff BJ, Miele L. Cross-talk between notch and the estrogen receptor in breast cancer suggests novel therapeutic approaches. Cancer Res 2008; 68:5226-35. [PMID: 18593923 PMCID: PMC4445363 DOI: 10.1158/0008-5472.can-07-5744] [Citation(s) in RCA: 275] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
High expression of Notch-1 and Jagged-1 mRNA correlates with poor prognosis in breast cancer. Elucidating the cross-talk between Notch and other major breast cancer pathways is necessary to determine which patients may benefit from Notch inhibitors, which agents should be combined with them, and which biomarkers indicate Notch activity in vivo. We explored expression of Notch receptors and ligands in clinical specimens, as well as activity, regulation, and effectors of Notch signaling using cell lines and xenografts. Ductal and lobular carcinomas commonly expressed Notch-1, Notch-4, and Jagged-1 at variable levels. However, in breast cancer cell lines, Notch-induced transcriptional activity did not correlate with Notch receptor levels and was highest in estrogen receptor alpha-negative (ERalpha(-)), Her2/Neu nonoverexpressing cells. In ERalpha(+) cells, estradiol inhibited Notch activity and Notch-1(IC) nuclear levels and affected Notch-1 cellular distribution. Tamoxifen and raloxifene blocked this effect, reactivating Notch. Notch-1 induced Notch-4. Notch-4 expression in clinical specimens correlated with proliferation (Ki67). In MDA-MB231 (ERalpha(-)) cells, Notch-1 knockdown or gamma-secretase inhibition decreased cyclins A and B1, causing G(2) arrest, p53-independent induction of NOXA, and death. In T47D:A18 (ERalpha(+)) cells, the same targets were affected, and Notch inhibition potentiated the effects of tamoxifen. In vivo, gamma-secretase inhibitor treatment arrested the growth of MDA-MB231 tumors and, in combination with tamoxifen, caused regression of T47D:A18 tumors. Our data indicate that combinations of antiestrogens and Notch inhibitors may be effective in ERalpha(+) breast cancers and that Notch signaling is a potential therapeutic target in ERalpha(-) breast cancers.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Proliferation
- Drug Delivery Systems
- Estradiol/administration & dosage
- Estradiol/pharmacology
- Estrogen Receptor alpha/antagonists & inhibitors
- Estrogen Receptor alpha/metabolism
- Estrogen Receptor alpha/physiology
- Female
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Invasiveness
- Neoplasms, Hormone-Dependent/drug therapy
- Proto-Oncogene Proteins/metabolism
- RNA, Small Interfering/administration & dosage
- Receptor Cross-Talk/physiology
- Receptor, Notch1/metabolism
- Receptor, Notch4
- Receptors, Notch/antagonists & inhibitors
- Receptors, Notch/metabolism
- Receptors, Notch/physiology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Paola Rizzo
- Breast Cancer Program, Loyola University Chicago
| | - Haixi Miao
- Breast Cancer Program, Loyola University Chicago
| | - Gwendolyn D'Souza
- Breast Cancer Program, Loyola University Chicago
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Clodia Osipo
- Breast Cancer Program, Loyola University Chicago
| | - Jieun Yun
- Breast Cancer Program, Loyola University Chicago
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Huiping Zhao
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Joaquina Mascarenhas
- Breast Cancer Program, Loyola University Chicago
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Debra Wyatt
- Breast Cancer Program, Loyola University Chicago
| | | | - Lu Hao
- Breast Cancer Program, Loyola University Chicago
| | | | - Prabha Rajan
- Breast Cancer Program, Loyola University Chicago
| | - Chindo Hicks
- Breast Cancer Program, Loyola University Chicago
| | | | - Suzanne Selvaggi
- Department of Pathology, University of Wisconsin, Madison, Wisconsin, Stockholm, Sweden
| | - Amina Bashir
- Breast Cancer Program, Loyola University Chicago
| | - Deepali Bhandari
- Breast Cancer Program, Loyola University Chicago
- Department of Pharmacology, Loyola University Chicago, Maywood, Illinois
| | - Adriano Marchese
- Breast Cancer Program, Loyola University Chicago
- Department of Pharmacology, Loyola University Chicago, Maywood, Illinois
| | | | - Jian-Zhong Qin
- Skin Cancer Program, Cardinal Bernardin Cancer Center, Loyola University Chicago
| | - Debra A. Tonetti
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Kathy Albain
- Breast Cancer Program, Loyola University Chicago
| | - Brian J. Nickoloff
- Skin Cancer Program, Cardinal Bernardin Cancer Center, Loyola University Chicago
| | - Lucio Miele
- Breast Cancer Program, Loyola University Chicago
| |
Collapse
|
20
|
Wiseman DA, Werner SR, Crowell PL. Cell cycle arrest by the isoprenoids perillyl alcohol, geraniol, and farnesol is mediated by p21(Cip1) and p27(Kip1) in human pancreatic adenocarcinoma cells. J Pharmacol Exp Ther 2007; 320:1163-70. [PMID: 17138864 DOI: 10.1124/jpet.106.111666] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pancreatic cancer, the fourth leading cause of cancer-associated mortality in the United States, usually presents in an advanced stage and is generally refractory to chemotherapy. As such, there is a great need for novel therapies for this disease. The naturally derived isoprenoids perillyl alcohol, farnesol, and geraniol have chemotherapeutic potential in pancreatic and other tumor types. However, their mechanisms of action in these systems are not completely defined. In this study, we investigated isoprenoid effects on the cell cycle and observed a similar antiproliferative mechanism of action among the three compounds. First, when given in combination, the isoprenoids exhibited an additive antiproliferative effect against MIA PaCa-2 human pancreatic cancer cells. Furthermore, all three compounds induced a G(0)/G(1) cell cycle arrest that coincided with an increase in the expression of the cyclin kinase inhibitor proteins p21(Cip1) and p27(Kip1) and a reduction in cyclin A, cyclin B1, and cyclin-dependent kinase (Cdk) 2 protein levels. Immunoprecipitation studies demonstrated increased association of both p21(Cip1) and p27(Kip1) with Cdk2 as well as diminished Cdk2 kinase activity after isoprenoid exposure, indicating a cell cycle-inhibitory role for p21(Cip1) and p27(Kip1) in pancreatic adenocarcinoma cells. When siRNA was used to inhibit expression of p21(Cip1) and p27(Kip1) proteins in MIA PaCa-2 cells, conditional resistance to all three isoprenoid compounds was evident. Given similar findings in this cell line and in BxPC-3 human pancreatic adenocarcinoma cells, we conclude that the chemotherapeutic isoprenoid compounds perillyl alcohol, farnesol, and geraniol invoke a p21(Cip1)- and p27(Kip1)-dependent antiproliferative mechanism in human pancreatic adenocarcinoma cells.
Collapse
Affiliation(s)
- Dean A Wiseman
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | | | | |
Collapse
|
21
|
White MK, Khalili K. Interaction of retinoblastoma protein family members with large T-antigen of primate polyomaviruses. Oncogene 2006; 25:5286-93. [PMID: 16936749 DOI: 10.1038/sj.onc.1209618] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The retinoblastoma gene product pRb and other members of the Rb family of pocket proteins have a central role in the regulation of cell cycle progression. Soon after its discovery, pRb was found to interact with the transforming oncoproteins of DNA tumor viruses and this led to rapid advances in our understanding of the mechanisms of viral transformation and cell cycle progression. DNA viruses of the polyomavirus family have small, circular, double-stranded DNA genomes contained within non-enveloped icosahedral capsids and are highly tumorigenic in experimental animals. At least three types of polyomavirus infect humans: JC virus (JCV), BK virus (BKV) and Simian Vacuolating virus-40. The early region of these viruses encodes the transforming proteins large T-antigen and small t-antigen, which are involved in viral replication and also promote transformation of cells in culture and oncogenesis in vivo. Binding of T-antigen to pRb promotes the activation of the E2F family of transcription factors, which induce the expression of cellular genes required for S phase. In the context of lytic infection, this cell cycle progression is necessary for viral replication because polyomaviruses rely on S phase-specific host factors for their DNA synthesis. In the context of cellular transformation and tumorigenesis, T-antigen/pRB interaction is an indispensable event.
Collapse
Affiliation(s)
- M K White
- Center for Neurovirology, Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA 19122, USA
| | | |
Collapse
|
22
|
Morris KE, Schang LM, Brindley DN. Lipid phosphate phosphatase-2 activity regulates S-phase entry of the cell cycle in Rat2 fibroblasts. J Biol Chem 2006; 281:9297-306. [PMID: 16467304 DOI: 10.1074/jbc.m511710200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Lipid phosphates are potent mediators of cell signaling and control processes including development, cell migration and division, blood vessel formation, wound repair, and tumor progression. Lipid phosphate phosphatases (LPPs) regulate the dephosphorylation of lipid phosphates, thus modulating their signals and producing new bioactive compounds both at the cell surface and in intracellular compartments. Knock-down of endogenous LPP2 in fibroblasts delayed cyclin A accumulation and entry into S-phase of the cell cycle. Conversely, overexpression of LPP2, but not a catalytically inactive mutant, caused premature S-phase entry, accompanied by premature cyclin A accumulation. At high passage, many LPP2 overexpressing cells arrested in G(2)/M and the rate of proliferation declined severely. This was accompanied by changes in proteins and lipids characteristic of senescence. Additionally, arrested LPP2 cells contained decreased lysophosphatidate concentrations and increased ceramide. These effects of LPP2 activity were not reproduced by overexpression or knock-down of LPP1 or LPP3. This work identifies a novel and specific role for LPP2 activity and bioactive lipids in regulating cell cycle progression.
Collapse
Affiliation(s)
- Katherine E Morris
- Signal Transduction Research Group and Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | | | | |
Collapse
|
23
|
Abstract
Tumor suppressor gene silencing by DNA hypermethylation contributes to tumorigenesis in many tumor types. This aberrant methylation may be due to increased expression and activity of DNA methyltransferases, which catalyze the transfer of methyl groups from S-adenosylmethionine to cytosines in CpG dinucleotides. Elevated expression of the maintenance DNA methyltransferase, DNA methyltransferase 1 (DNMT-1), has been shown in carcinomas of the colon, lung, liver, and prostate. Based on the nearly ubiquitous alterations of both DNA methylation and the retinoblastoma protein (pRb) pathway found in human cancer, we investigated a potential regulatory pathway linking the two alterations in murine and human prostate epithelial cells. Analysis of DNA methyltransferase levels in Rb-/- murine prostate epithelial cell lines revealed elevated Dnmt-1 levels. Genomic DNA sequence analysis identified conserved E2F consensus binding sites in proximity to the transcription initiation points of murine and human Dnmt-1. Furthermore, the Dnmt-1 promoter was shown to be regulated by the pRb/E2F pathway in murine and human cell lines of epithelial and fibroblast origin. In the absence of pRb, Dnmt-1 transcripts exhibited aberrant cell cycle regulation and Rb-/- cells showed aberrant methylation of the paternally expressed gene 3 (Peg3) tumor suppressor gene. These findings show a link between inactivation of the pRb pathway and induction of DNA hypermethylation of CpG island-containing genes in tumorigenesis.
Collapse
Affiliation(s)
- Michael T McCabe
- Department of Urology, Michigan Urology Center, University of Michigan, Ann Arbor, Michigan 48109-0944, USA
| | | | | |
Collapse
|
24
|
Hirsch HA, Jawdekar GW, Lee KA, Gu L, Henry RW. Distinct mechanisms for repression of RNA polymerase III transcription by the retinoblastoma tumor suppressor protein. Mol Cell Biol 2004; 24:5989-99. [PMID: 15199152 PMCID: PMC480882 DOI: 10.1128/mcb.24.13.5989-5999.2004] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The retinoblastoma (RB) protein represses global RNA polymerase III transcription of genes that encode nontranslated RNAs, potentially to control cell growth. However, RNA polymerase III-transcribed genes exhibit diverse promoter structures and factor requirements for transcription, and a universal mechanism explaining global repression is uncertain. We show that RB represses different classes of RNA polymerase III-transcribed genes via distinct mechanisms. Repression of human U6 snRNA (class 3) gene transcription occurs through stable promoter occupancy by RB, whereas repression of adenovirus VAI (class 2) gene transcription occurs in the absence of detectable RB-promoter association. Endogenous RB binds to a human U6 snRNA gene in both normal and cancer cells that maintain functional RB but not in HeLa cells whose RB function is disrupted by the papillomavirus E7 protein. Both U6 promoter association and transcriptional repression require the A/B pocket domain and C region of RB. These regions of RB contribute to U6 promoter targeting through numerous interactions with components of the U6 general transcription machinery, including SNAP(C) and TFIIIB. Importantly, RB also concurrently occupies a U6 promoter with RNA polymerase III during repression. These observations suggest a novel mechanism for RB function wherein RB can repress U6 transcription at critical steps subsequent to RNA polymerase III recruitment.
Collapse
Affiliation(s)
- Heather A Hirsch
- Program in Cell and Molecular Biology, Michigan State University, East Lansing 48824, USA
| | | | | | | | | |
Collapse
|
25
|
Miliani de Marval PL, Macias E, Conti CJ, Rodriguez-Puebla ML. Enhanced malignant tumorigenesis in Cdk4 transgenic mice. Oncogene 2004; 23:1863-73. [PMID: 14647432 PMCID: PMC2859671 DOI: 10.1038/sj.onc.1207309] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In a previous study, we reported that overexpression of cyclin-dependent kinase-4 (CDK4) in mouse epidermis results in epidermal hyperplasia, hypertrophy and severe dermal fibrosis. In this study, we have investigated the susceptibility to skin tumor formation by forced expression of CDK4. Skin tumors from transgenic mice showed a dramatic increase in the rate of malignant progression to squamous cell carcinomas (SCC) in an initiation-promotion protocol. Histopathological analysis of papillomas from transgenic mice showed an elevated number of premalignant lesions characterized by dysplasia and marked atypia. Interestingly, transgenic mice also developed tumors in initiated but not promoted skin, demonstrating that CDK4 replaced the action of tumor promoters. These results suggest that expression of cyclin D1 upon ras activation synergizes with CDK4 overexpression. However, cyclin D1 transgenic mice and double transgenic mice for cyclin D1 and CDK4 did not show increased malignant progression in comparison to CDK4 transgenic mice. Biochemical analysis of tumors showed that CDK4 sequesters the CDK2 inhibitors p27Kip1 and p21Cip1, suggesting that indirect activation of CDK2 plays an important role in tumor development. These results indicate that, contrary to the general assumption, the catalytic subunit, CDK4, has higher oncogenic activity than cyclin D1, revealing a potential use of CDK4 as therapeutic target.
Collapse
Affiliation(s)
- Paula L. Miliani de Marval
- North Carolina State University, College of Veterinary Medicine, Department of Molecular Biomedical Sciences - Raleigh, North Carolina 27606
- The University of Texas M. D. Anderson Cancer Center, Department of Carcinogenesis, Smithville, Texas 78957
| | - Everardo Macias
- North Carolina State University, College of Veterinary Medicine, Department of Molecular Biomedical Sciences - Raleigh, North Carolina 27606
| | - Claudio J. Conti
- The University of Texas M. D. Anderson Cancer Center, Department of Carcinogenesis, Smithville, Texas 78957
| | - Marcelo L. Rodriguez-Puebla
- North Carolina State University, College of Veterinary Medicine, Department of Molecular Biomedical Sciences - Raleigh, North Carolina 27606
| |
Collapse
|
26
|
Abriss B, Hollweg G, Gressner AM, Weiskirchen R. Adenoviral-mediated transfer of p53 or retinoblastoma protein blocks cell proliferation and induces apoptosis in culture-activated hepatic stellate cells. J Hepatol 2003; 38:169-78. [PMID: 12547405 DOI: 10.1016/s0168-8278(02)00361-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND/AIMS The principal cellular effectors of fibrosis in liver are hepatic stellate cells (HSC). In response to liver injury these quiescent cells undergo a phenotypic change to a myofibroblastic cell type, proliferate and secrete matrix components. Thus, removal of activated HSC should be beneficial for the prognosis of hepatic fibrogenesis and preserve organ function. METHODS The purpose of this study was to investigate whether administration of adenoviruses constitutively expressing the p53 tumor suppressor or the retinoblastoma protein (pRb) is sufficient to induce cell arrest or apoptosis in culture-activated HSC. The expression of the transgenes was confirmed by Western blot analysis and immunohistochemistry. RESULTS Both proteins were expressed mainly in the nucleus and their expression was associated with a marked inhibition of cell proliferation and induction of apoptosis as determined by measurement of phosphatidylserine exposed at the surface, proliferation assay, induction of the p21 cyclin-dependent kinase inhibitor, and an increase of caspase-3 activity. Additionally, electron microscopic analysis confirmed that activation of the p53-mediated pathway in HSC results in chromatin and cytoplasmic condensation, typical features of ongoing apoptosis. CONCLUSIONS Our results indicate that transduction of p53 or pRb offers a feasible approach to induce apoptosis in activated HSC. Thus, targeted transfer of these proteins into HSC may be potentially useful for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Bärbel Abriss
- Institute of Clinical Chemistry and Pathobiochemistry, RWTH-University Hospital, D-52074, Aachen, Germany
| | | | | | | |
Collapse
|
27
|
Ferreira R, Naguibneva I, Mathieu M, Ait-Si-Ali S, Robin P, Pritchard LL, Harel-Bellan A. Cell cycle-dependent recruitment of HDAC-1 correlates with deacetylation of histone H4 on an Rb-E2F target promoter. EMBO Rep 2001; 2:794-9. [PMID: 11520855 PMCID: PMC1084028 DOI: 10.1093/embo-reports/kve173] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The transcription factor E2F, which is a key element in the control of cell proliferation, is repressed by Rb and other pocket proteins in growth-arrested differentiating cells, as well as in proliferating cells when they progress through early G1. It is not known whether similar mechanisms are operative in the two situations. A body of data suggests that E2F repression by pocket proteins involves class I histone deacetylases (HDACs). It has been hypothesized that these enzymes are recruited to E2F target promoters where they deacetylate histones. Here we have tested this hypothesis directly by using formaldehyde cross-linked chromatin immunoprecipitation (XChIP) assays to evaluate HDAC association in living cells. Our data show that a histone deacetylase, HDAC-1, is stably bound to an E2F target promoter during early G1 in proliferating cells and released at the G1-S transition. In addition, our results reveal an inverse correlation between HDAC-1 recruitment and histone H4 acetylation on specific lysines.
Collapse
Affiliation(s)
- R Ferreira
- Laboratoire 'Oncogenèse, Différenciation et Transduction du Signal', CNRS UPR 9079, Institut Fédératif André Lwoff, 7 rue Guy Moquet, 94801 Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
28
|
Petkova V, Romanowski MJ, Sulijoadikusumo I, Rohne D, Kang P, Shenk T, Usheva A. Interaction between YY1 and the retinoblastoma protein. Regulation of cell cycle progression in differentiated cells. J Biol Chem 2001; 276:7932-6. [PMID: 11118439 DOI: 10.1074/jbc.m007411200] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Overexpression of the transcription factor YY1 activates DNA synthesis in differentiated primary human coronary artery smooth muscle cells. Overexpression of the retinoblastoma protein together with YY1 blocked this effect. In growth-arrested cells, YY1 resides in a complex with the retinoblastoma protein, but the complex is not detected in serum-stimulated S phase cultures, indicating that the interaction of the retinoblastoma protein and YY1 is cell cycle-regulated. Recombinant retinoblastoma protein directly interacts with YY1, destabilizing the interaction of YY1 with DNA and inhibiting its transcription initiator function in vitro. We conclude that in differentiated cells elevation of the nuclear level of YY1 protein favors progression into the S phase, and we propose that this activity is regulated by its interaction with the retinoblastoma protein.
Collapse
Affiliation(s)
- V Petkova
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachuttes 02215, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Rodriguez-Puebla ML, LaCava M, Miliani De Marval PL, Jorcano JL, Richie ER, Conti CJ. Cyclin D2 overexpression in transgenic mice induces thymic and epidermal hyperplasia whereas cyclin D3 expression results only in epidermal hyperplasia. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 157:1039-50. [PMID: 10980142 PMCID: PMC1885715 DOI: 10.1016/s0002-9440(10)64616-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In a previous report, we described the effects of cyclin D1 expression in epithelial tissues of transgenic mice. To study the involvement of D-type cyclins (D1, D2, and D3) in epithelial growth and differentiation and their putative role as oncogenes in skin, transgenic mice were developed which carry cyclin D2 or D3 genes driven by a keratin 5 promoter. As expected, both transgenic lines showed expression of these proteins in most of the squamous tissues analyzed. Epidermal proliferation increased in transgenic animals and basal cell hyperplasia was observed. All of the animals also had a minor thickening of the epidermis. The pattern of expression of keratin 1 and keratin 5 indicated that epidermal differentiation was not affected. Transgenic K5D2 mice developed mild thymic hyperplasia that reversed at 4 months of age. On the other hand, high expression of cyclin D3 in the thymus did not produce hyperplasia. This model provides in vivo evidence of the action of cyclin D2 and cyclin D3 as mediators of proliferation in squamous epithelial cells. A direct comparison among the three D-type cyclin transgenic mice suggests that cyclin D1 and cyclin D2 have similar roles in epithelial thymus cells. However, overexpression of each D-type cyclin produces a distinct phenotype in thymic epithelial cells.
Collapse
Affiliation(s)
- M L Rodriguez-Puebla
- University of Texas M. D. Anderson Cancer Center, Science Park-Research Division, Smithville, Texas 78957, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
In any multi-cellular organism, the balance between cell division and cell death maintains a constant cell number. Both cell division cycle and cell death are highly regulated events. Whether the cell will proceed through the cycle or not, depends upon whether the conditions required at the checkpoints during the cycle are fulfilled. In higher eucaryotic cells, such as mammalian cells, signals that arrest the cycle usually act at a G1 checkpoint. Cells that pass this restriction point are committed to complete the cycle. Regulation of the G1 phase of the cell cycle is extremely complex and involves many different families of proteins such as retinoblastoma family, cyclin dependent kinases, cyclins, and cyclin kinase inhibitors.
Collapse
Affiliation(s)
- D Donjerkovic
- Department of Immunology, Holland Laboratory for the Biomedical Sciences, American Red Cross, Rockville, MD, USA
| | | |
Collapse
|
31
|
Lee J, Choi YH, Nguyen P, Kim JS, Lee SJ, Trepel JB. Cyclic AMP induces inhibition of cyclin A expression and growth arrest in human hepatoma cells. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1449:261-8. [PMID: 10209305 DOI: 10.1016/s0167-4889(99)00019-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Classical cytotoxic therapy has been minimally useful in the treatment of hepatocellular carcinoma. In an effort to develop a new approach to the treatment of this neoplasm, we have investigated the signal transduction pathways regulating the growth of human hepatoma cells. In the data reported here, cyclic AMP (cAMP), a negative growth regulator for many cells of epithelial origin, induced G1 synchronization and apoptosis in the HepG2 human hepatoma cell line. The effects of cAMP on the components of the G1/S transition were analyzed. There was no detectable effect of two different cAMP analogs, 8-bromo cAMP or dibutyryl cAMP on the level of the D-type cyclins, cyclin E, cyclin-dependent kinase 2, cyclin-dependent kinase 4, p53, or the cyclin-dependent kinase inhibitors p21 or p27. In contrast, the cAMP analogs induced a dramatic downregulation of cyclin A protein, cyclin A messenger RNA, and cyclin A-dependent kinase activity. Cyclin A-dependent kinase has been shown to be required for the G1-S transition. Furthermore, cyclin A deregulation has been implicated in the pathogenesis of hepatocellular carcinoma. The data reported here suggest a novel signal transduction-based approach to hepatoma therapy.
Collapse
Affiliation(s)
- J Lee
- Medicine Branch, Division of Clinical Sciences, National Cancer Institute, National Institutes of Health, Building 10, Room 12N230, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
32
|
Kiuchi N, Nakajima K, Ichiba M, Fukada T, Narimatsu M, Mizuno K, Hibi M, Hirano T. STAT3 is required for the gp130-mediated full activation of the c-myc gene. J Exp Med 1999; 189:63-73. [PMID: 9874564 PMCID: PMC1887683 DOI: 10.1084/jem.189.1.63] [Citation(s) in RCA: 325] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The signal transducers and activators of transcription (STAT) family members have been implicated in regulating the growth, differentiation, and death of normal and transformed cells in response to either extracellular stimuli, including cytokines and growth factors, or intracellular tyrosine kinases. c-myc expression is coordinately regulated by multiple signals in these diverse cellular responses. We show that STAT3 mostly mediates the rapid activation of the c-myc gene upon stimulation of the interleukin (IL)-6 receptor or gp130, a signal transducing subunit of the receptor complexes for the IL-6 cytokine family. STAT3 does so most likely by binding to cis-regulatory region(s) of the c-myc gene. We show that STAT3 binds to a region overlapping with the E2F site in the c-myc promoter and this site is critical for the c-myc gene promoter- driven transcriptional activation by IL-6 or gp130 signals. This is the first identification of the linkage between a member of the STAT family and the c-myc gene activation, and also explains how the IL-6 family of cytokines is capable of inducing the expression of the c-myc gene.
Collapse
Affiliation(s)
- N Kiuchi
- Division of Molecular Oncology, Biomedical Research Center, Osaka University Medical School, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Rodriguez-Puebla ML, Robles AI, Conti CJ. ras activity and cyclin D1 expression: An essential mechanism of mouse skin tumor development. Mol Carcinog 1999. [DOI: 10.1002/(sici)1098-2744(199901)24:1<1::aid-mc1>3.0.co;2-e] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
34
|
Kilbourne EJ, Evans MJ, Karathanasis SK. E1A represses apolipoprotein AI enhancer activity in liver cells through a pRb- and CBP-independent pathway. Nucleic Acids Res 1998; 26:1761-8. [PMID: 9512550 PMCID: PMC147459 DOI: 10.1093/nar/26.7.1761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The apolipoprotein AI (apoAI) promoter/enhancer contains multiple cis -acting elements on which a variety of hepatocyte-enriched and ubiquitous transcription factors function synergistically to regulate liver-specific transcription. Adenovirus E1A proteins repress tissue-specific gene expression and disrupt the differentiated state in a variety of cell types. In this study expression of E1A 12Sor 13S in hepatoblastoma HepG2 cells repressed apoAI enhancer activity 8-fold. Deletion mapping analysis showed that inhibition by E1A was mediated by the apoAI promoter site B. E1A selectively inhibited the ability of HNF3beta and HNF3alpha to transactivate reporter genes controlled by the apoAI site B and the HNF3 binding site from the transthyretin promoter. The E1A-mediated repression of HNF3 activity was not reversed by overexpression of HNF3beta nor did E1A alter nuclear HNF3beta protein levels or inhibit HNF3 binding to DNA in mobility shift assays. Overexpression of two cofactors known to interact with E1A, pRb and CBP failed to overcome inhibition of HNF3 activity. Similarly, mutations in E1A that disrupt its interaction with pRb or CBP did not compromise its ability to repress HNF3beta transcriptional activity. These data suggest that E1A inhibits HNF3 activity by inactivating a limiting cofactor(s) distinct from pRb or CBP.
Collapse
Affiliation(s)
- E J Kilbourne
- Department of Nuclear Receptors, Wyeth-Ayerst Research, 145 King of Prussia Road, Radnor, PA 19087, USA
| | | | | |
Collapse
|
35
|
Dong F, Cress WD, Agrawal D, Pledger WJ. The role of cyclin D3-dependent kinase in the phosphorylation of p130 in mouse BALB/c 3T3 fibroblasts. J Biol Chem 1998; 273:6190-5. [PMID: 9497341 DOI: 10.1074/jbc.273.11.6190] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have observed that cyclin D3-dependent kinase activity is increased in the late G1 phase in BALB/c 3T3 fibroblasts. The profile of cyclin D3-associated activity closely parallels that of cyclin D1, which is also induced after mitogenic stimulation of quiescent cells. These activities correlate with the appearance of hyperphosphorylated p130, an Rb family member important in regulating E2F-4 and E2F-5 activity in fibroblastic cells. We demonstrated, however, that only the cyclin D3 activity efficiently phosphorylated p130 in an in vitro kinase assay. This apparent specificity was further demonstrated by experiments which demonstrated that cyclin D3 was physically associated with p130 at the times when D3-dependent kinase activity and p130 hyperphosphorylation were observed. Examination of E2F by electrophoretic mobility shift assay revealed that E2F-4 DNA binding activity existed in a p130.E2F complex at times before D3-dependent kinase activity was apparent and in a free E2F-4 complex after D3 activity developed. Thus, our data suggest that cyclin D3 preferentially phosphorylates p130 and is thereby specifically targeted to overcoming growth-suppressive control mediated through p130 pathways.
Collapse
Affiliation(s)
- F Dong
- H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, Florida 33612, USA
| | | | | | | |
Collapse
|
36
|
Javier R, Shenk T. Mammary tumors induced by human adenovirus type 9: a role for the viral early region 4 gene. Breast Cancer Res Treat 1996; 39:57-67. [PMID: 8738606 DOI: 10.1007/bf01806078] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Human adenovirus type 9 (Ad9) elicits exclusively estrogen-dependent mammary tumors when injected into female rats. Three different histological types of mammary tumor (benign fibroadenomas, phyllodes tumors, and malignant solid sarcomas) have been described in Ad9-infected animals, with benign fibroadenomas being seen most frequently. Interestingly, in contrast to other adenoviruses, in which oncogenic viral functions are entirely encoded within the E1 region, Ad9 requires an E4 region transforming protein (ORF1) for its unique mammary oncogenicity. Studies of Ad9-induced rat mammary tumors may lead to a detailed molecular understanding for the development of fibroadenoma, a common human breast tumor.
Collapse
MESH Headings
- Adenoviruses, Human/chemistry
- Adenoviruses, Human/genetics
- Animals
- Base Sequence
- DNA, Viral/metabolism
- Estrogens/physiology
- Female
- Fibroadenoma/pathology
- Fibroadenoma/virology
- Gene Expression Regulation, Viral
- Male
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/virology
- Molecular Sequence Data
- Oncogene Proteins, Viral/chemistry
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/physiology
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
Collapse
Affiliation(s)
- R Javier
- Division of Molecular Virology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|
37
|
Wong KK, Zou X, Merrell KT, Patel AJ, Marcu KB, Chellappan S, Calame K. v-Abl activates c-myc transcription through the E2F site. Mol Cell Biol 1995; 15:6535-44. [PMID: 8524218 PMCID: PMC230906 DOI: 10.1128/mcb.15.12.6535] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The v-abl oncogene of Abelson murine leukemia virus encodes a deregulated form of the cellular nonreceptor tyrosine kinase. v-Abl activates c-myc transcription, and c-Myc is an essential downstream component in the v-Abl transformation program. To explore the mechanism by which v-Abl activates c-myc transcription, a cotransfection assay was developed. We show that transactivation of a c-myc promoter by v-Abl requires the SH1 (tyrosine kinase) and SH2 domains of v-Abl; the C-terminal domains are not required for transactivation. The assay also identified the E2F site in the c-myc promoter as a v-Abl-responsive element. In addition, multimerized E2F sites were shown to be sufficient to confer v-Abl-dependent activation on a minimal promoter. This is the first identification of a v-Abl response element for transcriptional activation. v-Abl tyrosine kinase-dependent changes in proteins binding the c-myc E2F site were also demonstrated, including induction of a complex containing DP1, p107, cyclin A, and cdk2. Identification of v-Abl-dependent changes in E2F-binding proteins provides an important link between v-Abl, transcription, cell cycle regulation, and control of cellular growth.
Collapse
Affiliation(s)
- K K Wong
- Integrated Program in Cellular, Molecular and Biophysical Studies, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | | | | | | | |
Collapse
|