1
|
The Yun/Prohibitin complex regulates adult Drosophila intestinal stem cell proliferation through the transcription factor E2F1. Proc Natl Acad Sci U S A 2022; 119:2111711119. [PMID: 35115400 PMCID: PMC8832997 DOI: 10.1073/pnas.2111711119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2021] [Indexed: 01/02/2023] Open
Abstract
Stem cells maintain tissue homeostasis. We identified a factor, Yun, required for proliferation of normal and transformed intestinal stem cells in adult Drosophila. Yun acts as a scaffold to stabilize the Prohibitin (PHB) complex previously implicated in various cellular and developmental processes and diseases. The Yun/PHB complex acts downstream of EGFR/MAPK signaling and affects the levels of E2F1 to regulate intestinal stem cell proliferation. The role of the PHB complex in cell proliferation is evolutionarily conserved. Our results provide insight into the underlying mechanisms of how stem cell proliferation is properly controlled during tissue homeostasis and tumorigenesis. Stem cells constantly divide and differentiate to maintain adult tissue homeostasis, and uncontrolled stem cell proliferation leads to severe diseases such as cancer. How stem cell proliferation is precisely controlled remains poorly understood. Here, from an RNA interference (RNAi) screen in adult Drosophila intestinal stem cells (ISCs), we identify a factor, Yun, required for proliferation of normal and transformed ISCs. Yun is mainly expressed in progenitors; our genetic and biochemical evidence suggest that it acts as a scaffold to stabilize the Prohibitin (PHB) complex previously implicated in various cellular and developmental processes and diseases. We demonstrate that the Yun/PHB complex is regulated by and acts downstream of EGFR/MAPK signaling. Importantly, the Yun/PHB complex interacts with and positively affects the levels of the transcription factor E2F1 to regulate ISC proliferation. In addition, we find that the role of the PHB complex in cell proliferation is evolutionarily conserved. Thus, our study uncovers a Yun/PHB-E2F1 regulatory axis in stem cell proliferation.
Collapse
|
2
|
Concepts in Oncolytic Adenovirus Therapy. Int J Mol Sci 2021; 22:ijms221910522. [PMID: 34638863 PMCID: PMC8508870 DOI: 10.3390/ijms221910522] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 02/07/2023] Open
Abstract
Oncolytic adenovirus therapy is gaining importance as a novel treatment option for the management of various cancers. Different concepts of modification within the adenovirus vector have been identified that define the mode of action against and the interaction with the tumour. Adenoviral vectors allow for genetic manipulations that restrict tumour specificity and also the expression of specific transgenes in order to support the anti-tumour effect. Additionally, replication of the virus and reinfection of neighbouring tumour cells amplify the therapeutic effect. Another important aspect in oncolytic adenovirus therapy is the virus induced cell death which is a process that activates the immune system against the tumour. This review describes which elements in adenovirus vectors have been identified for modification not only to utilize oncolytic adenovirus vectors into conditionally replicating adenoviruses (CRAds) that allow replication specifically in tumour cells but also to confer specific characteristics to these viruses. These advances in development resulted in clinical trials that are summarized based on the conceptual design.
Collapse
|
3
|
Multipronged activity of combinatorial miR-143 and miR-506 inhibits Lung Cancer cell cycle progression and angiogenesis in vitro. Sci Rep 2018; 8:10495. [PMID: 30002440 PMCID: PMC6043488 DOI: 10.1038/s41598-018-28872-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/02/2018] [Indexed: 01/04/2023] Open
Abstract
Lung cancer (LC) is the leading cause of cancer-related deaths. Downregulation of CDK1, 4 and 6, key regulators of cell cycle progression, correlates with decreased LC cell proliferation. Enforced expression of miRNAs (miRs) is a promising approach to regulate genes. Here, we study the combinatorial treatment of miR-143 and miR-506 to target the CDK1, 4/6 genes, respectively. We analyzed the differential expression of CDK genes by qPCR, and western blot, and evaluated changes in the cell cycle distribution upon combinatorial treatment. We used an antibody microarray analysis to evaluate protein expression, focusing on the cell cycle pathway, and performed RNA-sequencing for pathway analysis. The combinatorial miR treatment significantly downregulated CDK1, 4 and 6 expression, and induced a shift of the cell cycle populations, indicating a G1 and G2 cell cycle block. The two miRs induces strong cytotoxic activity, with potential synergism, and a significant Caspase 3/7 activation. We identified a strong inhibition of tube formation in the presence or absence VEGF in an in vitro angiogenesis model. Together with the pathways analysis of the RNA-sequencing data, our findings establish the combinatorial miR transfection as a viable strategy for lung cancer treatment that merits further investigation.
Collapse
|
4
|
Hoare J, Campbell N, Carapuça E. Oncolytic virus immunotherapies in ovarian cancer: moving beyond adenoviruses. Porto Biomed J 2018; 3:e7. [PMID: 31595233 PMCID: PMC6726300 DOI: 10.1016/j.pbj.0000000000000007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/04/2018] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer is the 5th most common cancer in UK women with a high relapse rate. The overall survival for ovarian cancer has remained low for decades prompting a real need for new therapies. Recurrent ovarian cancer remains confined in the peritoneal cavity in >80% of the patients, providing an opportunity for locoregional administration of novel therapeutics, including gene and viral therapy approaches. Immunotherapy is an expanding field, and includes oncolytic viruses as well as monoclonal antibodies, immune checkpoint inhibitors, and therapeutic vaccines. Oncolytic viruses cause direct cancer cell cytolysis and immunogenic cell death and subsequent release of tumor antigens that will prime for a potent tumor-specific immunity. This effect may be further enhanced when the viruses are engineered to express, or coadministered with, immunostimulatory molecules. Currently, the most commonly used and well-characterized vectors utilized for virotherapy purposes are adenoviruses. They have been shown to work synergistically with traditional chemotherapy and radiotherapy and have met with success in clinical trials. However, pre-existing immunity and poor in vivo models limit our ability to fully investigate the potential of oncolytic adenovirus as effective immunotherapies which in turn fosters the need to develop alternative viral vectors. In this review we cover recent advances in adenovirus-based oncolytic therapies targeting ovarian cancer and recent advances in mapping immune responses to oncolytic virus therapies in ovarian cancer.
Collapse
Affiliation(s)
- Joseph Hoare
- Centre for Molecular Oncology, Barts Cancer Institute - a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| | - Nicola Campbell
- Centre for Molecular Oncology, Barts Cancer Institute - a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| | - Elisabete Carapuça
- Centre for Molecular Oncology, Barts Cancer Institute - a CRUK Centre of Excellence, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
5
|
Efficient Transformation of Primary Human Mesenchymal Stromal Cells by Adenovirus Early Region 1 Oncogenes. J Virol 2016; 91:JVI.01782-16. [PMID: 27795433 DOI: 10.1128/jvi.01782-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/15/2016] [Indexed: 11/20/2022] Open
Abstract
Previous observations that human amniotic fluid cells (AFC) can be transformed by human adenovirus type 5 (HAdV-5) E1A/E1B oncogenes prompted us to identify the target cells in the AFC population that are susceptible to transformation. Our results demonstrate that one cell type corresponding to mesenchymal stem/stroma cells (hMSCs) can be reproducibly transformed by HAdV-5 E1A/E1B oncogenes as efficiently as primary rodent cultures. HAdV-5 E1-transformed hMSCs exhibit all properties commonly associated with a high grade of oncogenic transformation, including enhanced cell proliferation, anchorage-independent growth, increased growth rate, and high telomerase activity as well as numerical and structural chromosomal aberrations. These data confirm previous work showing that HAdV preferentially transforms cells of mesenchymal origin in rodents. More importantly, they demonstrate for the first time that human cells with stem cell characteristics can be completely transformed by HAdV oncogenes in tissue culture with high efficiency. Our findings strongly support the hypothesis that undifferentiated progenitor cells or cells with stem cell-like properties are highly susceptible targets for HAdV-mediated cell transformation and suggest that virus-associated tumors in humans may originate, at least in part, from infections of these cell types. We expect that primary hMSCs will replace the primary rodent cultures in HAdV viral transformation studies and are confident that these investigations will continue to uncover general principles of viral oncogenesis that can be extended to human DNA tumor viruses as well. IMPORTANCE It is generally believed that transformation of primary human cells with HAdV-5 E1 oncogenes is very inefficient. However, a few cell lines have been successfully transformed with HAdV-5 E1A and E1B, indicating that there is a certain cell type which is susceptible to HAdV-mediated transformation. Interestingly, all those cell lines have been derived from human embryonic tissue, albeit the exact cell type is not known yet. We show for the first time the successful transformation of primary human mesenchymal stromal cells (hMSCs) by HAdV-5 E1A and E1B. Further, we show upon HAdV-5 E1A and E1B expression that these primary progenitor cells exhibit features of tumor cells and can no longer be differentiated into the adipogenic, chondrogenic, or osteogenic lineage. Hence, primary hMSCs represent a robust and novel model system to elucidate the underlying molecular mechanisms of adenovirus-mediated transformation of multipotent human progenitor cells.
Collapse
|
6
|
Rosewell Shaw A, Suzuki M. Recent advances in oncolytic adenovirus therapies for cancer. Curr Opin Virol 2016; 21:9-15. [PMID: 27379906 DOI: 10.1016/j.coviro.2016.06.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 06/14/2016] [Accepted: 06/17/2016] [Indexed: 12/11/2022]
Abstract
Oncolytic adenoviruses (Onc.Ads) selectively replicate in and lyse cancer cells and are therefore commonly used vectors in clinical trials for cancer gene therapy. Building upon the well-characterized adenoviral natural tropism, genetic modification of Onc.Ad can enhance/regulate their transduction and replication within specific cancer cell types. However, Onc.Ad-mediated tumor cell lysis cannot fully eliminate tumors. The hostile tumor microenvironment provides many barriers to efficient oncolytic virotherapy, as tumors develop structure and immune-evasion mechanisms in order to grow and ultimately spread. For these reasons, Onc.Ads modified to deliver structural or immune modulatory molecules (Armed Onc.Ads) have been developed to overcome the physical and immunological barriers of solid tumors. The combination of oncolysis with tumor microenvironment modulation/destruction may provide a promising platform for Ad-based cancer gene therapy.
Collapse
Affiliation(s)
- Amanda Rosewell Shaw
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| | - Masataka Suzuki
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
7
|
Spratford CM, Kumar JP. Inhibition of Daughterless by Extramacrochaetae mediates Notch-induced cell proliferation. Development 2015; 142:2058-68. [PMID: 25977368 DOI: 10.1242/dev.121855] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/16/2015] [Indexed: 12/30/2022]
Abstract
During development, the rate of cell proliferation must be constantly monitored so that an individual tissue achieves its correct size. Mutations in genes that normally promote tissue growth often result in undersized, disorganized and non-functional organs. However, mutations in genes that encode growth inhibitors can trigger the onset of tumorigenesis and cancer. The developing eye of the fruit fly, Drosophila melanogaster, has become a premier model system for studies that are focused on identifying the molecular mechanisms that underpin growth control. Here, we examine the mechanism by which the Notch pathway, a major contributor to growth, promotes cell proliferation in the developing eye. Current models propose that the Notch pathway directly influences cell proliferation by regulating growth-promoting genes such as four-jointed, cyclin D1 and E2f1. Here, we show that, in addition to these mechanisms, some Notch signaling is devoted to blocking the growth-suppressing activity of the bHLH DNA-binding protein Daughterless (Da). We demonstrate that Notch signaling activates the expression of extramacrochaetae (emc), which encodes a helix-loop-helix (HLH) transcription factor. Emc, in turn, then forms a biochemical complex with Da. As Emc lacks a basic DNA-binding domain, the Emc-Da heterodimer cannot bind to and regulate genomic targets. One effect of Da sequestration is to relieve the repression on growth. Here, we present data supporting our model that Notch-induced cell proliferation in the developing eye is mediated in part by the activity of Emc.
Collapse
Affiliation(s)
- Carrie M Spratford
- Department of Biology, Indiana University, Bloomington, IN 47405, USA Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | - Justin P Kumar
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
8
|
Popov B, Petrov N. pRb-E2F signaling in life of mesenchymal stem cells: Cell cycle, cell fate, and cell differentiation. Genes Dis 2014; 1:174-187. [PMID: 30258863 PMCID: PMC6150080 DOI: 10.1016/j.gendis.2014.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/14/2014] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into various mesodermal lines forming fat, muscle, bone, and other lineages of connective tissue. MSCs possess plasticity and under special metabolic conditions may transform into cells of unusual phenotypes originating from ecto- and endoderm. After transplantation, MSCs release the humoral factors promoting regeneration of the damaged tissue. During last five years, the numbers of registered clinical trials of MSCs have increased about 10 folds. This gives evidence that MSCs present a new promising resource for cell therapy of the most dangerous diseases. The efficacy of the MSCs therapy is limited by low possibilities to regulate their conversion into cells of damaged tissues that is implemented by the pRb-E2F signaling. The widely accepted viewpoint addresses pRb as ubiquitous regulator of cell cycle and tumor suppressor. However, current publications suggest that basic function of the pRb-E2F signaling in development is to regulate cell fate and differentiation. Through facultative and constitutive chromatin modifications, pRb-E2F signaling promotes transient and stable cells quiescence, cell fate choice to differentiate, to senesce, or to die. Loss of pRb is associated with cancer cell fate. pRb regulates cell fate by retaining quiescence of one cell population in favor of commitment of another or by suppression of genes of different cell phenotype. pRb is the founder member of the "pocket protein" family possessing functional redundancy. Critical increase in the efficacy of the MSCs based cell therapy will depend on precise understanding of various aspects of the pRb-E2F signaling.
Collapse
Affiliation(s)
- Boris Popov
- Institute of Cytology, Russian Academy of Sciences, St.Petersburg, 4, Tikhoretsky Av., 194064, Russia
| | | |
Collapse
|
9
|
Munro S, Oppermann U, La Thangue NB. Pleiotropic effect of somatic mutations in the E2F subunit DP-1 gene in human cancer. Oncogene 2013; 33:3594-603. [DOI: 10.1038/onc.2013.316] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 05/31/2013] [Accepted: 07/09/2013] [Indexed: 11/09/2022]
|
10
|
Evidence for autoregulation and cell signaling pathway regulation from genome-wide binding of the Drosophila retinoblastoma protein. G3-GENES GENOMES GENETICS 2012; 2:1459-72. [PMID: 23173097 PMCID: PMC3484676 DOI: 10.1534/g3.112.004424] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 09/20/2012] [Indexed: 12/24/2022]
Abstract
The retinoblastoma (RB) tumor suppressor protein is a transcriptional cofactor with essential roles in cell cycle and development. Physical and functional targets of RB and its paralogs p107/p130 have been studied largely in cultured cells, but the full biological context of this family of proteins' activities will likely be revealed only in whole organismal studies. To identify direct targets of the major Drosophila RB counterpart in a developmental context, we carried out ChIP-Seq analysis of Rbf1 in the embryo. The association of the protein with promoters is developmentally controlled; early promoter access is globally inhibited, whereas later in development Rbf1 is found to associate with promoter-proximal regions of approximately 2000 genes. In addition to conserved cell-cycle-related genes, a wholly unexpected finding was that Rbf1 targets many components of the insulin, Hippo, JAK/STAT, Notch, and other conserved signaling pathways. Rbf1 may thus directly affect output of these essential growth-control and differentiation pathways by regulation of expression of receptors, kinases and downstream effectors. Rbf1 was also found to target multiple levels of its own regulatory hierarchy. Bioinformatic analysis indicates that different classes of genes exhibit distinct constellations of motifs associated with the Rbf1-bound regions, suggesting that the context of Rbf1 recruitment may vary within the Rbf1 regulon. Many of these targeted genes are bound by Rbf1 homologs in human cells, indicating that a conserved role of RB proteins may be to adjust the set point of interlinked signaling networks essential for growth and development.
Collapse
|
11
|
ncRNA- and Pc2 methylation-dependent gene relocation between nuclear structures mediates gene activation programs. Cell 2012; 147:773-88. [PMID: 22078878 DOI: 10.1016/j.cell.2011.08.054] [Citation(s) in RCA: 506] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 05/05/2011] [Accepted: 08/26/2011] [Indexed: 12/14/2022]
Abstract
Although eukaryotic nuclei contain distinct architectural structures associated with noncoding RNAs (ncRNAs), their potential relationship to regulated transcriptional programs remains poorly understood. Here, we report that methylation/demethylation of Polycomb 2 protein (Pc2) controls relocation of growth-control genes between Polycomb bodies (PcGs) and interchromatin granules (ICGs) in response to growth signals. This movement is the consequence of binding of methylated and unmethylated Pc2 to the ncRNAs TUG1 and MALAT1/NEAT2, located in PcGs and ICGs, respectively. These ncRNAs mediate assembly of multiple corepressors/coactivators and can serve to switch mark recognition by "readers" of the histone code. Additionally, binding of NEAT2 to unmethylated Pc2 promotes E2F1 SUMOylation, leading to activation of the growth-control gene program. These observations delineate a molecular pathway linking the actions of subnuclear structure-specific ncRNAs and nonhistone protein methylation to relocation of transcription units in the three-dimensional space of the nucleus, thus achieving coordinated gene expression programs.
Collapse
|
12
|
Next-generation sequencing reveals regional differences of the α-synuclein methylation state independent of Lewy body disease. Neuromolecular Med 2011; 13:310-20. [PMID: 22042430 DOI: 10.1007/s12017-011-8163-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 10/07/2011] [Indexed: 12/17/2022]
Abstract
The α-synuclein gene (SNCA) plays a major role in the aetiology of Lewy body disease (LBD) including Parkinson's disease (PD). Point mutations and genetic alterations causing elevated gene expression are causally linked to familial PD. To what extent epigenetic changes play a role in the regulation of α-synuclein expression and may contribute to the aetiology of sporadic LBD is a matter of debate. We analysed the methylation state of the promoter region and a CpG-rich region of intron 1 of α-synuclein in several brain regions in sporadic LBD and controls using 454 GS-FLX-based high-resolution bisulphite sequencing. Our results indicate that there are significant differences in the level of methylation between different brain areas. The overall methylation levels in the promoter and intron 1 of α-synuclein are rather low in controls and-in contrast to previously reported findings-are not significantly different from LBD. However, single CpG analysis revealed significant hyper- and hypomethylation at different positions in various brain regions and LBD stages. A slight overall increase in methylation related to LBD patients' age was detected.
Collapse
|
13
|
Sáenz Robles MT, Case A, Chong JL, Leone G, Pipas JM. The retinoblastoma tumor suppressor regulates a xenobiotic detoxification pathway. PLoS One 2011; 6:e26019. [PMID: 22022495 PMCID: PMC3192141 DOI: 10.1371/journal.pone.0026019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 09/15/2011] [Indexed: 11/22/2022] Open
Abstract
The retinoblastoma tumor suppressor (pRb) regulates cell cycle entry, progression and exit by controlling the activity of the E2F-family of transcription factors. During cell cycle exit pRb acts as a transcriptional repressor by associating with E2F proteins and thereby inhibiting their ability to stimulate the expression of genes required for S phase. Indeed, many tumors harbor mutations in the RB gene and the pRb-E2F pathway is compromised in nearly all types of cancers. In this report we show that both pRb and its interacting partners, the transcriptional factors E2F1-2-3, act as positive modulators of detoxification pathways important for metabolizing and clearing xenobiotics—such as toxins and drugs—from the body. Using a combination of conventional molecular biology techniques and microarray analysis of specific cell populations, we have analyzed the detoxification pathway in murine samples in the presence or absence of pRb and/or E2F1-2-3. In this report, we show that both pRb and E2F1-2-3 act as positive modulators of detoxification pathways in mice, challenging the conventional view of E2F1-2-3 as transcriptional repressors negatively regulated by pRb. These results suggest that mutations altering the pRb-E2F axis may have consequences beyond loss of cell cycle control by altering the ability of tissues to remove toxins and to properly metabolize anticancer drugs, and might help to understand the formation and progression rates of different types of cancer, as well as to better design appropriate therapies based on the particular genetic composition of the tumors.
Collapse
Affiliation(s)
- Maria Teresa Sáenz Robles
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ashley Case
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jean-Leon Chong
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Department of Molecular Genetics, College of Biological Sciences and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Gustavo Leone
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Department of Molecular Genetics, College of Biological Sciences and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - James M. Pipas
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
14
|
Mohácsik P, Zeöld A, Bianco AC, Gereben B. Thyroid hormone and the neuroglia: both source and target. J Thyroid Res 2011; 2011:215718. [PMID: 21876836 PMCID: PMC3163027 DOI: 10.4061/2011/215718] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 05/31/2011] [Accepted: 06/17/2011] [Indexed: 12/15/2022] Open
Abstract
Thyroid hormone plays a crucial role in the development and function of the nervous system. In order to bind to its nuclear receptor and regulate gene transcription thyroxine needs to be activated in the brain. This activation occurs via conversion of thyroxine to T3, which is catalyzed by the type 2 iodothyronine deiodinase (D2) in glial cells, in astrocytes, and tanycytes in the mediobasal hypothalamus. We discuss how thyroid hormone affects glial cell function followed by an overview on the fine-tuned regulation of T3 generation by D2 in different glial subtypes. Recent evidence on the direct paracrine impact of glial D2 on neuronal gene expression underlines the importance of glial-neuronal interaction in thyroid hormone regulation as a major regulatory pathway in the brain in health and disease.
Collapse
Affiliation(s)
- Petra Mohácsik
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, H-1083, Hungary
| | | | | | | |
Collapse
|
15
|
Bukur J, Herrmann F, Handke D, Recktenwald C, Seliger B. Identification of E2F1 as an important transcription factor for the regulation of tapasin expression. J Biol Chem 2010; 285:30419-26. [PMID: 20663889 PMCID: PMC2945534 DOI: 10.1074/jbc.m109.094284] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 06/23/2010] [Indexed: 11/06/2022] Open
Abstract
HER-2/neu overexpression in tumor cells caused abnormalities of MHC class I surface expression due to impaired expression of components of the antigen-processing machinery (APM) including the low molecular weight proteins, the transporter associated with antigen processing (TAP), and the chaperone tapasin, whereas the expression of MHC class I heavy chain as well as β(2)-microglobulin was only marginally affected. This oncogene-mediated deficient APM component expression could be reverted by interferon-γ treatment, suggesting a deregulation rather than structural alterations as underlying molecular mechanisms. To determine the level of regulation, the transcriptional activity of APM components was analyzed in HER-2/neu(-) and HER-2/neu(+) cells. All major APM components were transcriptionally down-regulated in HER-2/neu(+) when compared with HER-2/neu(-) cells, which was accompanied by a reduced binding of RNA polymerase II to the APM promoters. Site-directed mutagenesis of the p300- and E2F-binding sites in the APM promoters did not reconstitute the oncogene-mediated decreased transcription rate with the exception of tapasin, which was restored in HER-2/neu(+) cells to levels of wild type tapasin promoter activity in HER-2/neu(-) fibroblasts. The E2F-directed control of tapasin expression was further confirmed by chromatin immunoprecipitation analyses showing that E2F1 and p300 bind to the tapasin and APM promoters in both cell lines. Moreover, siRNA-mediated silencing of E2F1 was associated with an increased tapasin expression, whereas transient overexpression of E2F1 launch a reduced tapasin transcription, suggesting that E2F1 is an essential transcription factor for tapasin.
Collapse
Affiliation(s)
- Juergen Bukur
- From the Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Felix Herrmann
- From the Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Diana Handke
- From the Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Christian Recktenwald
- From the Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Barbara Seliger
- From the Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| |
Collapse
|
16
|
Chong JL, Wenzel PL, Sáenz-Robles MT, Nair V, Ferrey A, Hagan JP, Gomez YM, Sharma N, Chen HZ, Ouseph M, Wang SH, Trikha P, Culp B, Mezache L, Winton DJ, Sansom OJ, Chen D, Bremner R, Cantalupo PG, Robinson ML, Pipas JM, Leone G. E2f1-3 switch from activators in progenitor cells to repressors in differentiating cells. Nature 2010; 462:930-4. [PMID: 20016602 PMCID: PMC2806193 DOI: 10.1038/nature08677] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 11/17/2009] [Indexed: 01/20/2023]
Abstract
In the classic paradigm of mammalian cell cycle control, Rb functions to restrict cells from entering S phase by sequestering E2F activators (E2f1, E2f2 and E2f3), which are invariably portrayed as the ultimate effectors of a transcriptional program that commit cells to enter and progress through S phase1, 2. Using a panel of tissue-specific cre-transgenic mice and conditional E2f alleles we examine the effects of E2f1, E2f2 and E2f3 triple deficiency in murine ES cells, embryos and small intestines. We show that in normal dividing progenitor cells E2F1-3 function as transcriptional activators, but contrary to current dogma, are dispensable for cell division and instead are necessary for cell survival. In differentiating cells they function in complex with Rb as repressors to silence E2F targets and facilitate exit from the cell cycle. The inactivation of Rb in differentiating cells resulted in a switch of E2F1-3 from repressors to activators, leading to the superactivation of E2F responsive targets and ectopic cell divisions, and loss of E2f1-3 completely suppressed these phenotypes. This work contextualizes the activator versus repressor functions of E2F1-3 in vivo, revealing distinct roles in dividing versus differentiating cells and in normal versus cancer-like cell cycles in vivo.
Collapse
Affiliation(s)
- Jean-Leon Chong
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Yurkova N, Shaw J, Blackie K, Weidman D, Jayas R, Flynn B, Kirshenbaum LA. The Cell Cycle Factor E2F-1 Activates Bnip3 and the Intrinsic Death Pathway in Ventricular Myocytes. Circ Res 2008; 102:472-9. [DOI: 10.1161/circresaha.107.164731] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The cell cycle factor E2F-1 is known to regulate a variety of cellular processes including apoptosis. Previously we showed that disruption of Rb–E2F-1 complexes provoked apoptosis of postmitotic adult and neonatal ventricular myocytes; however, the underlying mechanism was undetermined. In this report, we show that E2F-1 provokes cell death of ventricular myocytes through a mechanism that directly impinges on the intrinsic death pathway. Furthermore, we show mechanistically that the hypoxia-inducible death factor Bnip3 is a direct transcriptional target of E2F-1 that is necessary and sufficient for E2F-1–induced cell death. Expression of E2F-1 resulted in a 4.9-fold increase (
P
<0.001) in nucleosomal DNA fragmentation and cell death by Hoechst 33258 dye and vital staining. E2F-1 provoked mitochondrial perturbations that were consistent with permeability transition pore opening. As determined by quantitative real-time PCR analysis, a 6.2-fold increase (
P
<0.001) in endogenous Bnip3 gene transcription was observed in cells expressing wild-type E2F-1 but not in cells expressing a mutation of E2F-1 defective for DNA binding. Rb, the principle regulator of cellular E2F-1 activity, was proteolytically cleaved and inactivated in ventricular myocytes during hypoxia. Consistent with the proteolytic cleavage of Rb, chromatin immunoprecipitation analysis revealed increased binding of E2F-1 to the Bnip3 promoter during hypoxia, a finding concordant with the induction of Bnip3 gene transcription. The Bnip3 homolog Nix/Bnip3L was unaffected in ventricular myocytes by either E2F-1 or hypoxia. Genetic knockdown of E2F-1 or expression of a caspase-resistant form of Rb suppressed basal and hypoxia-inducible Bnip3 gene transcription. Loss-of-function mutations of Bnip3 defective for mitochondrial membrane insertion or small interference RNA directed against Bnip3 suppressed cell death signals elicited by E2F-1. To our knowledge, the data provide the first direct evidence that activation of the intrinsic mitochondrial death pathway by E2F-1 is mutually dependent on and obligatorily linked to the transcriptional activation of Bnip3.
Collapse
Affiliation(s)
- Natalia Yurkova
- From the Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Departments of Physiology (N.Y., K.B., D.W., R.J., B.F., L.A.K.), Pharmacology & Therapeutics (J.S., L.A.K.), Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | - James Shaw
- From the Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Departments of Physiology (N.Y., K.B., D.W., R.J., B.F., L.A.K.), Pharmacology & Therapeutics (J.S., L.A.K.), Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | - Karen Blackie
- From the Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Departments of Physiology (N.Y., K.B., D.W., R.J., B.F., L.A.K.), Pharmacology & Therapeutics (J.S., L.A.K.), Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | - Danielle Weidman
- From the Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Departments of Physiology (N.Y., K.B., D.W., R.J., B.F., L.A.K.), Pharmacology & Therapeutics (J.S., L.A.K.), Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | - Ravi Jayas
- From the Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Departments of Physiology (N.Y., K.B., D.W., R.J., B.F., L.A.K.), Pharmacology & Therapeutics (J.S., L.A.K.), Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | - Bryan Flynn
- From the Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Departments of Physiology (N.Y., K.B., D.W., R.J., B.F., L.A.K.), Pharmacology & Therapeutics (J.S., L.A.K.), Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | - Lorrie A. Kirshenbaum
- From the Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Departments of Physiology (N.Y., K.B., D.W., R.J., B.F., L.A.K.), Pharmacology & Therapeutics (J.S., L.A.K.), Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
18
|
Gao A, Liu B, Shi X, Jia X, Ye M, Jiao S, You B, Huang C. Phosphatidylinositol-3 kinase/Akt/p70S6K/AP-1 signaling pathway mediated benzo(a)pyrene-induced cell cycle alternation via cell cycle regulatory proteins in human embryo lung fibroblasts. Toxicol Lett 2007; 170:30-41. [PMID: 17383120 DOI: 10.1016/j.toxlet.2007.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 02/09/2007] [Accepted: 02/09/2007] [Indexed: 11/17/2022]
Abstract
Benzo(a)pyrene (B(a)P), a potent environmental procarcinogen, has been shown to cause cell cycle alternation. However, the mechanisms involved in this effect are not well understood yet. Our current results demonstrated that B(a)P exposure led to cell proliferation and a 33.5% increase in S phase cells as well as a 26.8% decrease in G1 phase cells in human embryo lung fibroblasts (HELFs). Those cell cycle alternations were accompanied with transactivation of activator protein-1 (AP-1) and phosphorylation of Akt and p70(S6K). These changes were blocked by overexpression of dominant negative mutants of phosphatidylinositol-3 kinase (Deltap85) or Akt (DN-Akt), respectively. Moreover, pretreatment of cells with rapamycin, a specific p70(S6K) inhibitor, inhibited B(a)P-induced AP-1 activation, cell cycle alteration and phosphorylation of p70(S6K), but had no effect on Akt phosphorylation. Our results, therefore, suggest that phosphatidylinositol-3 kinase (PI-3K)/Akt/p70(S6K)/AP-1 pathway participates in B(a)P-induced cell cycle alternations. Furthermore, we explored the effect of this pathway on cell cycle regulatory proteins. B(a)P markedly increases in the expression of cyclin D1 and E2F1 and phosphorylation of retinoblastoma protein (Rb). In addition, we found that inactivation of PI-3K, Akt or p70(S6K) could eliminate those effects on cell cycle regulatory proteins. Collectively, PI-3K/Akt/p70(S6K)/AP-1 pathway mediated B(a)P-induced alternation of cell cycle through regulation of cell cycle regulatory proteins such as cyclin D1, E2F1, and Rb in HELFs.
Collapse
Affiliation(s)
- Ai Gao
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, 29 Nan Wei Road, Beijing 100050, PR China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
de las Cuevas N, Muñoz U, Hermida OG, Martín-Requero A. Altered transcriptional regulators in response to serum in immortalized lymphocytes from Alzheimer's disease patients. Neurobiol Aging 2005; 26:615-24. [PMID: 15708436 DOI: 10.1016/j.neurobiolaging.2004.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2003] [Revised: 05/05/2004] [Accepted: 06/16/2004] [Indexed: 11/30/2022]
Abstract
Cell cycle disturbances may precede neuronal death in Alzheimer's disease (AD). We described alterations, in lymphocytes from AD patients, on the activity of two transcription factors, E2F and NF-kappaB, involved in cell proliferation and survival regulation, demonstrating that cell cycle dysfunction also occurs in peripheral cells. The analysis of E2F-DNA binding activity revealed lower signal intensity of protein-DNA complexes in AD cells, which correlated with increased phosphorylation of retinoblastoma (pRb) related proteins and enhanced proliferation. The calmodulin (CaM) antagonist calmidazolium (CMZ) abrogated the increased activity of AD cells by partially dephosphorylating pRb and p130. The NF-kappaB-DNA binding activity increased as cell progress through the cell cycle. The reduced NF-kappaB activation observed in AD cells appears not to be related to the increased phosphorylation of the pRb family proteins nor with the enhanced proliferative activity of AD cells, but seems to protect them from death induced by the loss of trophic support. Ca2+/CaM antagonists rescue NF-kappaB-DNA binding activity and sensitize AD cells to serum withdrawal. These observations suggest that disruption of Ca2+/CaM signaling pathway could be linked mechanistically to its pro cell survival actions, promoting enhanced proliferation or decreased cell death depending on the presence of growth-stimulatory signals.
Collapse
Affiliation(s)
- Natividad de las Cuevas
- Department of Pathophysiology and Human Molecular Genetics, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040-Madrid, Spain
| | | | | | | |
Collapse
|
20
|
Wharton W. Repression of G0/G1 traverse in human fibroblasts exposed to low levels of ionizing radiation. J Biol Chem 2004; 279:43667-74. [PMID: 15304475 DOI: 10.1074/jbc.m407959200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Quiescent cultures of human fibroblasts were exposed to levels of ionizing radiation sufficient to induce a transient growth delay, while causing only small decreases in long term clonogenicity. Following the mitogenic stimulation of damaged cells, cyclin D-associated kinase activity was induced to levels equivalent to those seen in control cultures. In addition, late G0/G1 E2F-dependent transcriptional and translational activity was observed in restimulated irradiated cells. However, cells became arrested prior to entry into S phase in a manner that paralleled the repression of cdk2-associated kinase activity. Cyclin A/cdk2-associated kinase activity was repressed in a biphasic manner following the irradiation of logarithmically growing cells. The initial rapid decline in activity to levels approximately 50% of those observed in control cultures occurred prior to increases in cellular levels of p21Cip1 protein, was not blocked by the addition of cycloheximide, and was not accompanied by alterations in cdk2 phosphotyrosine content. The subsequent repression to undetectable levels was coincident with the induction of p21Cip1 and was dependent on de novo protein synthesis. Only a subpopulation of cyclin A complexes were associated with p21Cip1 regardless of the magnitude of the repression of catalytic activity, although all cyclin A-cdk2-p21Cip1 complexes were inactive. These data suggest that temporally and functionally distinct mechanisms mediate the repression of cyclin-cdk activity in damaged cells. In addition, we present evidence that irradiated cells are competent to traverse S phase and arrest in G2 in the complete absence of cdk2-associated kinase activity.
Collapse
Affiliation(s)
- Walker Wharton
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque 87131, USA.
| |
Collapse
|
21
|
Cole KA, Harmon AW, Harp JB, Patel YM. Rb regulates C/EBPβ-DNA-binding activity during 3T3-L1 adipogenesis. Am J Physiol Cell Physiol 2004; 286:C349-54. [PMID: 14576085 DOI: 10.1152/ajpcell.00255.2003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Two pathways are initiated upon 3T3-L1 preadipocyte differentiation: the reentry of cells into the cell cycle and the initiation of a cascade of transcriptional events that “prime” the cell for differentiation. The “priming” event involves the synthesis of members of the CCAAT/enhancer binding protein (C/EBP) family of transcription factors. However, the relationship between these two pathways is unknown. Here we report that in the 3T3-L1 preadipocytes induced to differentiate, cell cycle progression and the initiation of differentiation are linked by a cell cycle-dependent Rb-C/EBPβ interaction. Cell cycle arrest in G1 by l-mimosine inhibited differentiation-induced C/EBPβ-DNA-binding activity and Rb phosphorylation. However, cell cycle arrest after the G1/S transition by aphidicolin or nocodazole did not prevent C/EBPβ-DNA-binding activity or Rb phosphorylation. Furthermore, hypophosphorylated Rb and C/EBPβ coimmunoprecipitated, whereas phosphorylated Rb and C/EBPβ did not. Electrophoretic mobility shift assays demonstrated that recombinant hypophosphorylated Rb decreased C/EBPβ-DNA-binding activity and that Rb overexpression inhibited C/EBPβ-induced transcriptional activation of a C/EBPα-promoter-luciferase reporter gene. We conclude that C/EBPβ-DNA-binding activity is regulated by its interaction with hypophosphorylated Rb, thereby linking the progression of the cell cycle to the initiation of differentiation during 3T3-L1 adipogenesis.
Collapse
Affiliation(s)
- Kathryn A Cole
- Department of Nutrition, University of North Carolina, School of Public Health, 2216A McGavran-Greenberg, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
22
|
Abstract
After balloon angioplasty, locally expressed tumor necrosis factor (TNF)-α disrupts endothelial cell (EC) proliferation and reendothelialization of the injured vessel. We have previously reported that TNF inhibits the EC cycle and downregulates the transcription factor E2F1. Ectopic expression of E2F1 at the site of injury improves reendothelialization of the injured vessel. In this study, we report that c-Jun N-terminal kinase (JNK) 1 and p38 mitogen-activated protein kinases (MAPKs) are differentially required for E2F1 expression and activity in ECs. Overexpression of constitutively active JNK1 mimicked TNF-mediated inhibitory events, whereas dominant-negative JNK1 prevented these effects. E2F
cis
elements in the promoter of E2F1 gene mediate suppressive actions of TNF, because removal of these sites rendered E2F1 promoter activity insensitive to TNF. JNK1 physically interacted with E2F1 and inactivated it via direct phosphorylation. Additionally, TNF inhibited Rb phosphorylation and dissociation from E2F1. Overexpression of constitutively active p38 MAPK facilitated Rb-E2F1 dissociation, whereas that of dominant-negative p38 MAPK did not. Taken together, these data suggest a differential requirement of JNK1 and p38 MAPK in TNF regulation of E2F1. Targeted inactivation of JNK1 at arterial injury sites may represent a potential therapeutic intervention for ameliorating TNF-mediated EC dysfunction.
Collapse
Affiliation(s)
- Raj Kishore
- Division of Cardiovascular Research, St Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Mass 02135, USA.
| | | | | | | | | |
Collapse
|
23
|
Ashe M, Pabon-Peña L, Dees E, Price KL, Bader D. LEK1 is a potential inhibitor of pocket protein-mediated cellular processes. J Biol Chem 2003; 279:664-76. [PMID: 14555653 DOI: 10.1074/jbc.m308810200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
LEK1, a member of the LEK family of proteins, is ubiquitously expressed in developing murine tissues. Our current studies are aimed at identifying the role of LEK1 during cell growth and differentiation. Little is known about the function of LEK proteins. Recent studies in our laboratory have focused on the characterization of the LEK1 atypical Rb-binding domain that is conserved among all LEK proteins. Our findings suggest that LEK1 potentially functions as a universal regulator of pocket protein activity. Pocket proteins exhibit distinct expression patterns during development and function to regulate cell cycle, apoptosis, and tissue-specific gene expression. We show that LEK1 interacts with all three pocket proteins, p107, p130, and pRb. Additionally, this interaction occurs specifically between the LEK1 Rb-binding motif and the "pocket domain" of Rb proteins responsible for Rb association with other targets. Analyses of the effects of disruption of LEK1 protein expression by morpholino oligomers demonstrate that LEK1 depletion decreases cell proliferation, disrupts cell cycle progression, and induces apoptosis. Given its expression in developing cells, its association with pocket proteins, and its effects on proliferation, cell cycle, and viability of cells, we suggest that LEK1 functions in a similar manner to phosphorylation to disrupt association of Rb proteins with appropriate binding targets. Thus, the LEK1/Rb interaction serves to retain cells in a pre-differentiative, actively proliferative state despite the presence of Rb proteins during development. Our data suggest that LEK1 is unique among LEK family members in that it specifically functions during murine development to regulate the activity of Rb proteins during cell division and proliferation. Furthermore, we discuss the distinct possibility that a yet unidentified splice variant of the closely related human CENP-F, serves a similar function to LEK1 in humans.
Collapse
Affiliation(s)
- Mabelle Ashe
- Stahlman Cardiovascular Research Laboratories, Program for Developmental Biology and the Division of Cardiovascular Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | | | | | |
Collapse
|
24
|
Gagnon A, Abaiian KJ, Crapper T, Layne MD, Sorisky A. Down-regulation of aortic carboxypeptidase-like protein during the early phase of 3T3-L1 adipogenesis. Endocrinology 2002; 143:2478-85. [PMID: 12072377 DOI: 10.1210/endo.143.7.8875] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aortic carboxypeptidase-like protein (ACLP) is a 175-kDa protein that is expressed in vascular smooth muscle cells and contains a signal peptide sequence, a lysine- and proline-rich repeating motif, a discoidin-like domain with 35% identity to discoidin I, and a carboxypeptidase-like domain that is 39% identical with carboxypeptidase E. It is secreted into the extracellular matrix and may play a role in abdominal wall development and dermal wound healing. ACLP is also expressed in adipose tissue, but at lower levels. In this study we demonstrate that ACLP protein and mRNA are severely down-regulated in the early phase of 3T3-L1 preadipocyte differentiation induced by insulin, dexamethasone, and isobutylmethylxanthine. Neither dexamethasone, isobutylmethylxanthine, nor insulin treatment alone reduced the level of ACLP protein, suggesting that ACLP down-regulation is a differentiation-associated event. ACLP down-regulation coincided with the onset of the postconfluent mitotic clonal expansion phase of adipogenesis. In contrast, subconfluent 3T3-L1 cell proliferation did not alter ACLP expression, suggesting a specific linkage between ACLP and differentiation-induced clonal expansion. Stable overexpression of ACLP had no effect on preadipocyte differentiation assessed by triacylglycerol accumulation and peroxisome proliferator-activated receptor-gamma levels. The role of ACLP and its marked reduction during adipogenesis merit further study.
Collapse
Affiliation(s)
- AnneMarie Gagnon
- Department of Medicine, Ottawa Health Research Institute, University of Ottawa, Canada K1Y 4E9
| | | | | | | | | |
Collapse
|
25
|
Abstract
Cell growth is under the control of a variety of positive and negative signals. An imbalance of such signals results in deregulation of cell behavior. Recessive oncogenes or tumor suppressor genes, opposite to dominant oncogenes, encode important cellular proteins which could function as negative regulators of the cell cycle, i.e., cell cycle brakes. Inactivation of recessive oncogenes, by allelic deletion, loss of expression, mutation, or functional inactivation by interacting with oncogene products of DNA tumor viruses or with amplified cellular binding proteins, will lead to uncontrolled cell growth or tumor formation. Besides the classic suppressor genes such as the p53 and RB, a growing number of novel tumor suppressor genes have been identified in recent years. While some tumor suppressor genes have been found to be important for the development of a large number of human malignancies (e.g., the p53 gene), others are more tumor type-specific (e.g., the NF-1 gene). Many human cancer types showed abnormalities of multiple tumor suppressor genes, offering strong support to the concept that tumorigenesis and progression result from an accumulation of multiple genetic alterations. In this review, we will begin with an overview (gene, transcript, protein and mechanisms of action) of the tumor suppressor genes (the RB, p53, DCC, APC, MCC, WT1, VHL, MST1, and BRCA1 genes) identified to date and then discuss the specific involvement of tumor suppressor genes in human malignancies including prostate cancer. Various chromosomal regions which potentially may contain tumor suppressor genes also will be reviewed.
Collapse
Affiliation(s)
- Xiang Gao
- Wayne State University, School of Medicine, Department of Radiation Oncology, Detroit, USA
| | | |
Collapse
|
26
|
Abstract
Sialylation represents one of the most frequently occurring terminations of the oligosaccharide chains of glycoproteins and glycolipids. Sialic acid is commonly found alpha2,3- or alpha2,6-linked to galactose (Gal), alpha2,6-linked to N-acetylgalactosamine (GalNAc) or alpha2,8-linked to another sialic acid. The biosynthesis of the various linkages is mediated by the different members of the sialyltransferase family. The addition of sialic acid in alpha2,6-linkage to the galactose residue of lactosamine (type 2 chains) is catalyzed by beta-galactoside alpha2,6-sialyltransferase (ST6Gal.I). Although expressed by a single gene, this enzyme shows a complex pattern of regulation which allows its tissue- and stage-specific modulation. The cognate oligosaccharide structure, NeuAcalpha2,6Galbeta1,4GlcNAc, is widely distributed among tissues and is involved in biological processes such as the regulation of the immune response and the progression of colon cancer. This review summarizes the current knowledge on the biochemistry of ST6Gal.I and on the functional role of the sialyl-alpha2,6-lactosaminyl structure.
Collapse
Affiliation(s)
- F Dall'Olio
- Dipartimento di Patologia Sperimentale, Università di Bologna, Italy
| |
Collapse
|
27
|
Patel YM, Lane MD. Mitotic clonal expansion during preadipocyte differentiation: calpain-mediated turnover of p27. J Biol Chem 2000; 275:17653-60. [PMID: 10749891 DOI: 10.1074/jbc.m910445199] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Evidence is presented that calpain, a calcium-activated protease, degrades the cyclin-dependent kinase inhibitor, p27, during the mitotic clonal expansion phase of 3T3-L1 preadipocyte differentiation. Calpain activity is required during an early stage of the adipocyte differentiation program. Thus, inhibition of calpain with N-acetyl-Leu-Leu-norleucinal (ALLN) blocks clonal expansion and acquisition of the adipocyte phenotype only when added between 12 and 24 h after the induction of differentiation. Likewise, inhibition of calpain by overexpression of calpastatin, the specific endogenous inhibitor of calpain, prevents 2-day post-confluent preadipocytes from reentering the cell cycle triggered by the differentiation inducers. Inhibition of calpain with ALLN causes preadipocytes to arrest just prior to S phase and prevents phosphorylation of the retinoblastoma gene product, DNA replication, clonal expansion, and subsequent adipocyte differentiation but does not affect the expression of immediate early genes (i.e. fos, jun, C/EBPbeta, and C/EBPdelta). Inhibition of calpain by either ALLN or by overexpression of calpastatin blocks the degradation of p27. p27 is degraded in vitro by cell-free extracts from clonally expanding preadipocytes that contain "active" calpain but not by extracts from pre-mitotic preadipocytes that do not. This action is inhibited by calpastatin or ALLN. Likewise, p27 in preadipocyte extracts is a substrate for purified calpain; this proteolytic action was inhibited by heat inactivation, EGTA, or ALLN. Thus, extracellular signals from the differentiation inducers appear to activate calpain, which degrades p27 allowing density-dependent inhibited preadipocytes to reenter the cell cycle and undergo mitotic clonal expansion.
Collapse
Affiliation(s)
- Y M Patel
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
28
|
Abstract
Human papillomaviruses (HPVs) are common DNA viruses in humans. Recently, epithelial cancers associated with HPV infection have been used as models of virus-induced carcinogenesis. HPVs can be divided into two groups, mucosal and cutaneous. HPV-16 is the most frequent mucosal type associated with cervical cancer. Although the molecular mechanisms of carcinogenesis by HPV-16 have not been completely elucidated, it is apparent that HPV infection is the major risk factor in cervical carcinogenesis. Two viral early genes, E6 and E7, and an upstream regulatory region (URR) are preserved in cervical carcinoma cell lines as well as in clinical samples of cervical cancer, indicating that these regions are important in cancer development. E6 and E7 function as transforming genes. E6 protein binds to and promotes degradation of the tumor suppressor protein, p53, while E7 protein complexes and inactivates the Rb protein; together, they disrupt cell cycle regulation. E6 and E7 are transcribed from a promoter, P97. P97 is regulated by complex interactions between multiple, positive and negative, cellular factors and the viral E2 product. E2 disruption caused by the integration into the cellular genome may induce overexpression of E6 and E7. The E6 and E7 proteins are thought to act as critical factors in cervical carcinogenesis by inactivating the two tumor suppressor proteins, p53 and Rb, which are commonly mutated in other human cancers.
Collapse
Affiliation(s)
- T Ishiji
- Department of Dermatology, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
29
|
Kashanchi F, Agbottah ET, Pise-Masison CA, Mahieux R, Duvall J, Kumar A, Brady JN. Cell cycle-regulated transcription by the human immunodeficiency virus type 1 Tat transactivator. J Virol 2000; 74:652-60. [PMID: 10623726 PMCID: PMC111584 DOI: 10.1128/jvi.74.2.652-660.2000] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cyclin-dependent kinases are required for the Tat-dependent transition from abortive to productive elongation. Further, the human immunodeficiency virus type 1 (HIV-1) Vpr protein prevents proliferation of infected cells by arresting them in the G(2) phase of the cell cycle. These findings suggest that the life cycle of the virus may be integrally related to the cell cycle. We now demonstrate by in vitro transcription analysis that Tat-dependent transcription takes place in a cell cycle-dependent manner. Remarkably, Tat activates gene expression in two distinct stages of the cell cycle. Tat-dependent long terminal repeat activation is observed in G(1). This activation is TAR dependent and requires a functional Sp1 binding site. A second phase of transactivation by Tat is observed in G(2) and is TAR independent. This later phase of transcription is enhanced by a natural cell cycle blocker of HIV-1, vpr, which arrests infected cells at the G(2)/M boundary. These studies link the HIV-1 Tat protein to cell cycle-specific biological functions.
Collapse
Affiliation(s)
- F Kashanchi
- Virus Tumor Biology Section, Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Lam EW, Glassford J, van der Sman J, Banerji L, Pizzey AR, Shaun N, Thomas B, Klaus GG. Modulation of E2F activity in primary mouse B cells following stimulation via surface IgM and CD40 receptors. Eur J Immunol 1999; 29:3380-9. [PMID: 10540350 DOI: 10.1002/(sici)1521-4141(199910)29:10<3380::aid-immu3380>3.0.co;2-c] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Since signals via CD40 and the B cell receptor are known to synergize to induce B cell activation, we have analyzed the pocket protein/E2F complexes in mouse B lymphocytes following stimulation by anti-IgM, anti-CD40, alone or together. We find that E2F4 and DP1 form the predominant E2F heterodimers in the G0 and G1 phases of the cell cycle, complexed with hypophosphorylated p130. During late G1 and S phase this complex is replaced by at least three different E2F complexes, one of which is an E2F complex containing p107 or pRB as well as two "free" E2F complexes consisting of E2F4/DP1 and E2F1-3/DP1. These effects were mirrored by the levels and phosphorylation status of the three pocket proteins. We also observed an increase in electrophoretic mobility of DP1 and E2F4 as B cells progressed from G0 into early G1, resulting from their dephosphorylation. This is known to correlate with a decrease in DNA binding capacity of these proteins and could also be important for derepression of genes negatively regulated through E2F sites in their promoters. These results therefore indicate that the pRB/E2F pathway integrates proliferative signals emanating from the sIgM and CD40 receptors.
Collapse
Affiliation(s)
- E W Lam
- Ludwig Institute for Cancer Research and Section of Virology and Cell Biology Imperial College School of Medicine at St Mary's, London, GB.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Tommasi S, Pfeifer GP. In vivo structure of two divergent promoters at the human PCNA locus. Synthesis of antisense RNA and S phase-dependent binding of E2F complexes in intron 1. J Biol Chem 1999; 274:27829-38. [PMID: 10488129 DOI: 10.1074/jbc.274.39.27829] [Citation(s) in RCA: 36] [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
Proliferating cell nuclear antigen (PCNA) synthesis is strictly regulated during the cell cycle. To investigate PCNA transcriptional regulation, we have analyzed protein-DNA interactions at the promoter region and in the first intron in quiescent fibroblasts and following serum stimulation. Twenty putative protein-binding sites, distributed in two divergent promoters at the PCNA locus, were identified in vivo by genomic footprinting. These elements bind transcription factors continuously throughout the cell cycle with the exception of one E2F consensus site, located in the first intron at position +583. This E2F site becomes strongly occupied 18 h after serum stimulation, implying that an E2F activator complex plays a role in activation of the PCNA gene at the onset of S phase. We detected a 500-600-base pair-long antisense transcript by Northern blot analysis. This RNA has no apparent coding capacity and is constitutively transcribed from a promoter located within the first intron. We suggest that silencing of the PCNA gene is accomplished through base pairing between sense pre-mRNA and antisense RNA. The binding of S phase-specific E2F complexes at the +583 element may help to overcome the negative effect of the antisense transcript, which results in up-regulation of PCNA expression in proliferating cells.
Collapse
Affiliation(s)
- S Tommasi
- Department of Biology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California 91010, USA.
| | | |
Collapse
|
32
|
van der Sman J, Thomas NS, Lam EW. Modulation of E2F complexes during G0 to S phase transition in human primary B-lymphocytes. J Biol Chem 1999; 274:12009-16. [PMID: 10207023 DOI: 10.1074/jbc.274.17.12009] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The pocket protein-E2F complexes are convergence points for cell cycle signaling. In the present report, we identified and monitored the pocket protein-E2F complexes in human primary B-lymphocytes after activation by phorbol 12-myristate 13-acetate. Consistent with previous data from human and mouse fibroblasts and T-lymphocytes, E2F4 and DP1 form the predominant E2F heterodimers both in G0 and G1 phases of the human B-lymphocyte cell cycle, whereas E2F1 and -3 are first detected in late G1, and their expression levels increase towards S phase. Intriguingly, the major E2F complex that we detected in quiescent human B-lymphocytes is consisted of pRB, E2F4, and DP1. Though the levels of DP1 and -2 increase when cells progress from G0 to S, the proportion of DP1 to DP2 remains relatively constant during the cell cycle. We also observed an increase in electrophoretic mobility of the predominant E2F components, DP1 and E2F4, as B-lymphocytes progressed from G0 into early G1. This increase in mobility was attributable to dephosphorylation, as lambda phosphatase treatment could convert the slower migrating forms into the corresponding faster mobility forms. We further demonstrated that this change in phosphorylation status correlates with a decrease in DNA binding activity. This modulation of DNA binding activity mediated through the dephosphorylation of DP1 and E2F4 could help to explain the lack of in vivo DNA footprinting in late G1 and S phases of gene promoters negatively regulated through E2F sites and suggests a novel mechanism for controlling E2F transcriptional activity during the transition from quiescence to proliferation.
Collapse
Affiliation(s)
- J van der Sman
- Ludwig Institute for Cancer Research and Section of Virology and Cell Biology, Imperial College School of Medicine at St Mary's, London W2 1PG, United Kingdom
| | | | | |
Collapse
|
33
|
Lai A, Marcellus RC, Corbeil HB, Branton PE. RBP1 induces growth arrest by repression of E2F-dependent transcription. Oncogene 1999; 18:2091-100. [PMID: 10321733 DOI: 10.1038/sj.onc.1202520] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Growth arrest and cell cycle progression are regulated by the retinoblastoma tumour suppressor pRB and related proteins p130 and p107 that bind to and inhibit the E2F family of transcription factors. Although the precise mechanism of this inhibition remains to be established, previous studies indicated the presence of transcriptional repression activity in the 'pocket' of RB family members. We show here that RBP1, a known pRB pocket-binding protein, possesses transcriptional repression activity and associates with p130-E2F and pRB-E2F complexes specifically during growth arrest. Overexpression of RBP1 both inhibited E2F-dependent gene expression and suppressed cell growth. Thus repression of E2F-dependent transcription by RBP1 via RB family members may play a central role in inducing growth arrest.
Collapse
Affiliation(s)
- A Lai
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
34
|
Abstract
The cell cycle is driven by the sequential activation of a family of cyclin-dependent kinases (cdk), which phosphorylate and activate proteins that execute events critical to cell cycle progression. In mammalian cells cdk2-cyclin A has a role in S phase. Many replication proteins are potential substrates for this cdk kinase, suggesting that initiation, elongation and checkpoint control of replication could all be regulated by cdk2. The association of PCNA, a replication protein, with cdk-cyclins during G-1 to S phase transition and with cdk-cyclin inhibitors, adds an interesting complexity to regulation of DNA replication.
Collapse
Affiliation(s)
- R Fotedar
- Institut de Biologie Structurale J.-P. Ebel, Grenoble, France
| | | |
Collapse
|
35
|
Nead MA, Baglia LA, Antinore MJ, Ludlow JW, McCance DJ. Rb binds c-Jun and activates transcription. EMBO J 1998; 17:2342-52. [PMID: 9545246 PMCID: PMC1170577 DOI: 10.1093/emboj/17.8.2342] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The retinoblastoma protein (Rb) acts as a critical cell-cycle regulator and loss of Rb function is associated with a variety of human cancer types. Here we report that Rb binds to members of the AP-1 family of transcription factors, including c-Jun, and stimulates c-Jun transcriptional activity from an AP-1 consensus sequence. The interaction involves the leucine zipper region of c-Jun and the B pocket of Rb as well as a C-terminal domain. We also present evidence that the complexes are found in terminally differentiating keratinocytes and cells entering the G1 phase of the cell cycle after release from serum starvation. The human papillomavirus type 16 E7 protein, which binds to both c-Jun and Rb, inhibits the ability of Rb to activate c-Jun. The results provide evidence of a role for Rb as a transcriptional activator in early G1 and as a potential modulator of c-Jun expression during keratinocyte differentiation.
Collapse
Affiliation(s)
- M A Nead
- Departments of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|
36
|
Trimarchi JM, Fairchild B, Verona R, Moberg K, Andon N, Lees JA. E2F-6, a member of the E2F family that can behave as a transcriptional repressor. Proc Natl Acad Sci U S A 1998; 95:2850-5. [PMID: 9501179 PMCID: PMC19658 DOI: 10.1073/pnas.95.6.2850] [Citation(s) in RCA: 172] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The E2F family of proteins is required to establish the correct cell-cycle-dependent transcription of genes that direct the process of cell division. All previously identified E2F proteins can act in a similar manner; depending on whether or not they are associated with the cell cycle inhibitors the retinoblastoma protein (pRB), p107, or p130, they can either repress or activate the transcription of E2F-responsive genes. We now report the cloning and characterization of another E2F family member, E2F-6, whose structure is reminiscent of the dominant inhibitors of other transcription factor families. The dimerization and DNA binding properties of E2F-6 are similar to those of the other E2F family members. However, it is not regulated by pRB, p107, or p130, and it is unable to activate transcription. Instead, it can act to repress the transcription of E2F responsive genes by countering the activity of the other E2F complexes via a pRB-, p107-, or p130-independent mechanism.
Collapse
Affiliation(s)
- J M Trimarchi
- Center for Cancer Research, Massachusetts Institute of Technology, Building E17-517B, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | | | | | | | | | | |
Collapse
|
37
|
Morita T, Mitsialis SA, Koike H, Liu Y, Kourembanas S. Carbon monoxide controls the proliferation of hypoxic vascular smooth muscle cells. J Biol Chem 1997; 272:32804-9. [PMID: 9407056 DOI: 10.1074/jbc.272.52.32804] [Citation(s) in RCA: 267] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Excess vascular smooth muscle cell (VSMC) proliferation and contractility are key events in the pathophysiology of vascular disorders induced by hypoxia. We have recently reported that carbon monoxide (CO), produced by VSMC under conditions of hypoxia, can be a modulator of cGMP levels in both endothelial and smooth muscle cells. In this respect, some of the physiologic effects of CO in the vasculature parallel those of nitric oxide (NO), a well characterized regulator of vascular tone. We report here that under hypoxia, VSMC-derived CO is an important regulator of VSMC proliferation. Inhibiting CO formation or scavenging CO with hemoglobin increased VSMC proliferation in response to serum or to mitogens such as endothelin, whereas increasing CO production or exposing cells to exogenous CO lead to a markedly attenuated growth response. The effects of CO on VSMC proliferation correlated with changes in E2F-1 expression, the prototype member of a family of transcription factors that participate in the control of cell cycle progression. CO significantly suppressed E2F-1 expression, whereas, removal of CO from the cultures with hemoglobin lead to increased E2F-1 gene transcription, mRNA, and protein production as well as mRNA levels of c-myc, a target gene of E2F-1. Moreover, the actions of CO were mediated by the second messenger molecule, cGMP. Limiting VSMC growth by increasing the release of CO may represent a key event in the body's compensatory responses to hypoxia.
Collapse
Affiliation(s)
- T Morita
- Joint Program in Neonatology, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
38
|
Verona R, Moberg K, Estes S, Starz M, Vernon JP, Lees JA. E2F activity is regulated by cell cycle-dependent changes in subcellular localization. Mol Cell Biol 1997; 17:7268-82. [PMID: 9372959 PMCID: PMC232584 DOI: 10.1128/mcb.17.12.7268] [Citation(s) in RCA: 174] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
E2F directs the cell cycle-dependent expression of genes that induce or regulate the cell division process. In mammalian cells, this transcriptional activity arises from the combined properties of multiple E2F-DP heterodimers. In this study, we show that the transcriptional potential of individual E2F species is dependent upon their nuclear localization. This is a constitutive property of E2F-1, -2, and -3, whereas the nuclear localization of E2F-4 is dependent upon its association with other nuclear factors. We previously showed that E2F-4 accounts for the majority of endogenous E2F species. We now show that the subcellular localization of E2F-4 is regulated in a cell cycle-dependent manner that results in the differential compartmentalization of the various E2F complexes. Consequently, in cycling cells, the majority of the p107-E2F, p130-E2F, and free E2F complexes remain in the cytoplasm. In contrast, almost all of the nuclear E2F activity is generated by pRB-E2F. This complex is present at high levels during G1 but disappears once the cells have passed the restriction point. Surprisingly, dissociation of this complex causes little increase in the levels of nuclear free E2F activity. This observation suggests that the repressive properties of the pRB-E2F complex play a critical role in establishing the temporal regulation of E2F-responsive genes. How the differential subcellular localization of pRB, p107, and p130 contributes to their different biological properties is also discussed.
Collapse
Affiliation(s)
- R Verona
- Center for Cancer Research, Massachusetts Institute of Technology, Cambridge 02139, USA
| | | | | | | | | | | |
Collapse
|
39
|
Tommasi S, Pfeifer GP. Constitutive protection of E2F recognition sequences in the human thymidine kinase promoter during cell cycle progression. J Biol Chem 1997; 272:30483-90. [PMID: 9374541 DOI: 10.1074/jbc.272.48.30483] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The sequences responsible for S phase-specific induction of the human thymidine kinase (TK) gene have been mapped to a small region that contains putative E2F binding sites. We have analyzed protein-DNA interactions at the TK promoter during cell cycle progression in human fibroblasts using an in vivo footprinting approach. We found 14 protein binding sites that were occupied in vivo. All of the sites (among them two inverted CCAAT boxes and several Sp1 sites) bound transcription factors constitutively throughout the cell cycle, i.e. none of the factor binding was cell cycle-dependent. An E2F-like site located between nucleotides -97 and -89 relative to the major transcription start site was protected in G0, G1, S, and G2 phases. This cell cycle-independent protection of E2F sequences in the TK promoter differs from the G0/G1-restricted binding of E2F complexes observed for genes in which the E2F sites function as repressor elements (Tommasi, S., and Pfeifer, G. P. (1995) Mol. Cell. Biol. 15, 6901-6913; Zwicker, J., Liu, N., Engeland, K., Lucibello, F. C., and Müller, R. (1996) Science 271, 1595-1597). A comparison of several genes containing E2F motifs indicates that E2F sites located in proximity to the transcription initiation site (-50 to +20) in TATA-less promoters predominantly function as repressor elements, while in other genes constitutively bound E2F complexes located further upstream mediate activation presumably in conjunction with a functional TATA box.
Collapse
Affiliation(s)
- S Tommasi
- Department of Biology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California 91010, USA.
| | | |
Collapse
|
40
|
Jensen DE, Black AR, Swick AG, Azizkhan JC. Distinct roles for Sp1 and E2F sites in the growth/cell cycle regulation of the DHFR promoter. J Cell Biochem 1997; 67:24-31. [PMID: 9328836 DOI: 10.1002/(sici)1097-4644(19971001)67:1<24::aid-jcb3>3.0.co;2-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Dihydrofolate reductase activity is required for many biosynthetic pathways including nucleotide synthesis. Its expression is therefore central to cellular growth, and it has become a key target for cancer chemotherapy. Transcription of the dihydrofolate reductase gene is regulated with growth, being expressed maximally in late G1/early S phase following serum stimulation of quiescent cells. This regulation is directed by a promoter which contains binding sites for only the transcription factors Sp1 and E2F. In this study, the role of these promoter elements in growth/cell cycle regulation of dihydrofolate transcription was addressed directly by transient transfection of Balb/c 3T3 cells with mutant promoter-reporter gene constructs. The E2F sites were found to repress transcription in G0 and early G1 but did not contribute to the level of transcription in late G1/S phase. In contrast, Sp1 sites were able to mediate induction of transcription from the dihydrofolate reductase promoter, as well as a heterologous promoter, following serum stimulation of quiescent cells. These findings add dihydrofolate reductase to a growing list of genes at which E2F sites are primarily repressive elements and delineate a role for Sp1 sites in the growth/cell cycle regulation of transcription.
Collapse
Affiliation(s)
- D E Jensen
- Department of Experimental Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | | | |
Collapse
|
41
|
Svendsen PC, Yee HA, Winkfein RJ, van de Sande JH. The mouse uracil-DNA glycosylase gene: isolation of cDNA and genomic clones and mapping ung to mouse chromosome 5. Gene X 1997; 189:175-81. [PMID: 9168124 DOI: 10.1016/s0378-1119(96)00797-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Uracil-DNA glycosylase (UDG) is the enzyme responsible for the first step in the base-excision repair pathway that specifically removes uracil from DNA. Here we report the isolation of the cDNA and genomic clones for the mouse uracil-DNA glycosylase gene (ung) homologous to the major placental uracil-DNA glycosylase gene (UNG) of humans. The complete characterization of the genomic organization of the mouse uracil-DNA glycosylase gene shows that the entire mRNA coding region for the 1.83-kb cDNA of the mouse ung gene is contained in an 8.2-kb SstI genomic fragment which includes six exons and five introns. The cDNA encodes a predicted uracil-DNA glycosylase (UDG) protein of 295 amino acids (33 kDa) that is highly similar to a group of UDGs that have been isolated from a wide variety of organisms. The mouse ung gene has been mapped to mouse chromosome 5 using fluorescence in situ hybridization (FISH).
Collapse
Affiliation(s)
- P C Svendsen
- Department of Medical Biochemistry, University of Calgary, Health Sciences Centre, Alberta, Canada
| | | | | | | |
Collapse
|
42
|
|
43
|
Affiliation(s)
- J Zwicker
- Institut für Molekularbiologie und Tumorforschung (IMT), Philipps-Universität Marburg, Germany.
| | | |
Collapse
|
44
|
Turek LP, Smith EM. The genetic program of genital human papillomaviruses in infection and cancer. Obstet Gynecol Clin North Am 1996; 23:735-58. [PMID: 8989774 DOI: 10.1016/s0889-8545(05)70275-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Human papillomavirus (HPV) infection has been recognized as the major cause of cervical cancer. This article summarizes the functions of HPV gene products that cause abnormal cell growth--E6 and E7--and reviews how cellular and viral factors influence their synthesis. E6 and E7 inactivate two cellular tumor-suppressor gene products, p53 and RB. In cervical cancer, E6-E7 gene control is deranged by mutations in viral control sequences and in integrated HPV fragments by the disruption of the viral repressor E2. Elimination of this sequence makes E6-E7 mRNAs unstable, and deranges cellular regulation at the integration site. It is apparent that an intricate interplay of cellular and viral factors determines whether the outcome is active papillomavirus infection, viral latency, or ultimately, genital cancer.
Collapse
Affiliation(s)
- L P Turek
- Department of Pathology, Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | | |
Collapse
|
45
|
Moberg K, Starz MA, Lees JA. E2F-4 switches from p130 to p107 and pRB in response to cell cycle reentry. Mol Cell Biol 1996; 16:1436-49. [PMID: 8657117 PMCID: PMC231128 DOI: 10.1128/mcb.16.4.1436] [Citation(s) in RCA: 274] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The E2F transcription factor couples the coordinate expression of cell cycle proteins to their appropriate transition points. Its activity is controlled by the cell cycle regulators pRB, p107, and p130. These bind to E2F at defined but distinct stages of the cell cycle. Using specific antisera, we have identified the DP and E2F components of each of these species. Although present at very different levels, DP-1 and DP-2 are evenly distributed among each of these complexes. In contrast, the individual E2Fs have distinctly different binding profiles. Consistent with previous studies, E2F-1, E2F-2, and E2F-3 bind specifically to the retinoblastoma protein. In each case, their expression and DNA binding activity are restricted to post-G1/S fractions. Surprisingly, E2F-1 and E2F-3 make unequal contributions to the pRB-associated and free E2F activity, suggesting that these proteins perform different cell cycle functions. Most significantly, this study showed E2F-4 accounts for the vast majority of the endogenous E2F activity. In arrested cells, E2F-4 is sequestered by the p130 protein. However, as the cells pass the G1-to-S transition, the levels of pRB and p107 increase and E2F-4 now associates with both of these regulators. Despite this, a considerable amount of E2F-4 exists as free E2F. In G1 cells, this accounts for almost all of the free activity. Once the cells enter S phase, free E2F is composed of an equal mixture of E2F-4 and E2F-1.
Collapse
Affiliation(s)
- K Moberg
- Center for Cancer Research, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | | | | |
Collapse
|
46
|
Lemaitre JM, Buckle RS, Méchali M. c-Myc in the control of cell proliferation and embryonic development. Adv Cancer Res 1996; 70:95-144. [PMID: 8902055 DOI: 10.1016/s0065-230x(08)60873-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
47
|
Margolis MJ, Pajovic S, Wong EL, Wade M, Jupp R, Nelson JA, Azizkhan JC. Interaction of the 72-kilodalton human cytomegalovirus IE1 gene product with E2F1 coincides with E2F-dependent activation of dihydrofolate reductase transcription. J Virol 1995; 69:7759-67. [PMID: 7494286 PMCID: PMC189718 DOI: 10.1128/jvi.69.12.7759-7767.1995] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Three polypeptides are produced from the major immediate-early (IE) region of human cytomegalovirus by alternative splicing. The IE gene products regulate subsequent viral and cellular gene expression. We previously reported that cotransfection of a genomic clone of the major IE region stimulated transient expression of chloramphenicol acetyltransferase driven by the dihydrofolate reductase (DHFR) promoter and that an intact E2F site was required for the trans activation (M. Wade, T. F. Kowalik, M. Mudryj, E.-S. Huang, and J. C. Azizkhan, Mol. Cell. Biol. 12:4364-4374, 1992). With the availability of cDNA clones for the individual major IE proteins, we sought to determine which of these proteins exerted this effect and whether the IE protein(s) interacted with E2F. In this study, we use cotransfection to demonstrate that the 55- and 86-kDa major IE proteins from the IE2 region can each moderately trans activate the DHFR promoter and that the 72-kDa IE1 protein stimulates DHFR transcription to a much higher level. Furthermore, trans activation through the 72-kDa IE1 protein is in part E2F dependent, while activation by the 55- and 86-kDa IE proteins is E2F independent. We also demonstrate by in vitro pull-down assays that the 72-kDa IE1 protein can specifically interact with the DNA binding domain of E2F1 (amino acids 88 to 191) in the presence of nuclear extract. Moreover, antibodies to either E2F1 or IE72 will immunoprecipitate both E2F and IE72 from cells that stably express IE72, and antibody to E2F1 will immunoprecipitate IE72 from normal human fibroblast cells infected with human cytomegalovirus.
Collapse
Affiliation(s)
- M J Margolis
- Department of Experimental Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Tommasi S, Pfeifer GP. In vivo structure of the human cdc2 promoter: release of a p130-E2F-4 complex from sequences immediately upstream of the transcription initiation site coincides with induction of cdc2 expression. Mol Cell Biol 1995; 15:6901-13. [PMID: 8524257 PMCID: PMC230945 DOI: 10.1128/mcb.15.12.6901] [Citation(s) in RCA: 145] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In quiescent cells, cdc2 mRNA is almost undetectable. Stimulation of cells to reenter the cell cycle results in induction of cdc2 expression, beginning at the G1-to-S transition and reaching maximum levels during late S and G2 phases. To investigate cdc2 transcriptional regulation throughout cell cycle progression, we monitored protein-DNA interactions by in vivo footprinting along 800 bp of the human cdc2 promoter in quiescent fibroblasts and at different time points following serum stimulation. We found 11 in vivo protein-binding sites, but no protein binding was observed at a high-affinity E2F site that had previously been implicated in cdc2 regulation. Nine of the identified in vivo binding sites (among them were two inverted CCAAT boxes, two Sp1 sites, and one ets-2 site) bind transcription factors constitutively throughout the cell cycle. However, at two elements located at positions -60 and -20 relative to the transcription start site, the binding pattern changes significantly as the cells are entering S phase. A G0- and G1-specific protein complex disappears at the -20 element at the beginning of S phase. This sequence deviates at one base position from known E2F consensus binding sites. We found that the major E2F activity in human fibroblasts contains E2F-4 and p130. The -20 element of the cdc2 gene specifically interacts with a subset of E2F-4-p130 complexes present in G0 cells but does not interact with S-phase-specific E2F complexes. Transient-transfection experiments with wild-type and mutant cdc2 promoter constructs indicate that the -20 element is involved in suppressing cdc2 activity in quiescent cells. We suggest that the presence of the p130-E2F-4 complex in G0/G1 blocks access of components of the basal transcription machinery or prevents transaction by the constitutively bound upstream activator proteins.
Collapse
Affiliation(s)
- S Tommasi
- Department of Biology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California 91010, USA
| | | |
Collapse
|
49
|
Datta PK, Raychaudhuri P, Bagchi S. Association of p107 with Sp1: genetically separable regions of p107 are involved in regulation of E2F- and Sp1-dependent transcription. Mol Cell Biol 1995; 15:5444-5452. [PMID: 7565695 PMCID: PMC230794 DOI: 10.1128/mcb.15.10.5444] [Citation(s) in RCA: 85] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The retinoblastoma-related protein p107 has been shown to be a regulator of the transcription factor E2F. p107 associates with E2F via its pocket region and represses E2F-dependent transcription. In this study, we provide evidence for a novel interaction between p107 and the transcription factor Sp1. We show that p107 can be found endogenously associated with Sp1 in the extracts of several different cell lines. Moreover, in transient transfection assays, expression of p107 represses Sp1-dependent transcription. This repression of Sp1-dependent transcription does not require the DNA-binding domain of Sp1. Transcription driven by a chimeric protein containing the Ga14 DNA-binding domain and the Sp1 activation domains is inhibited by p107. Interestingly, unlike the repression of E2F-dependent transcription, the repression of Sp1-dependent transcription does not depend on an intact pocket region. We show that distinct regions of p107 are involved in the control of Sp1 and E2F.
Collapse
Affiliation(s)
- P K Datta
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago 60612, USA
| | | | | |
Collapse
|
50
|
Chadee DN, Taylor WR, Hurta RA, Allis CD, Wright JA, Davie JR. Increased phosphorylation of histone H1 in mouse fibroblasts transformed with oncogenes or constitutively active mitogen-activated protein kinase kinase. J Biol Chem 1995; 270:20098-105. [PMID: 7650028 DOI: 10.1074/jbc.270.34.20098] [Citation(s) in RCA: 87] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
We compared the nucleosomal organization, histone H1 subtypes, and histone H1 phosphorylated isoforms of ras-transformed and parental 10T1/2 mouse fibroblasts. In agreement with previous studies, we found that ras-transformed mouse fibroblasts have a less condensed chromatin structure than normal fibroblasts. ras-transformed and parental 10T1/2 cells had similar amounts of H1 subtypes, proteins that have a key role in the compaction of chromatin. However, labeling studies with 32P and Western blot experiments with an antiphosphorylated H1 antibody show that interphase ras-transformed cells have higher levels of phosphorylated H1 isoforms than parental cells. G1/S phase-arrested ras-transformed cells had higher amounts of phosphorylated H1 than G1/S phase-arrested parental cells. Mouse fibroblasts transformed with fes, mos, raf, myc, or constitutively active mitogen-activated protein (MAP) kinase kinase had increased levels of phosphorylated H1. These observations suggest that increased phosphorylation of H1 is one of the consequences of the persistent activation of the mitogen-activated protein kinase signal transduction pathway. Indirect immunofluorescent studies show that phosphorylated H1b is localized in centers of RNA splicing in the nucleus, suggesting that this modified H1 subtype is complexed to transcriptionally active chromatin.
Collapse
Affiliation(s)
- D N Chadee
- Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, Canada
| | | | | | | | | | | |
Collapse
|