201
|
Mahale AM, Khan ZA, Igarashi M, Nanjangud GJ, Qiao RF, Yao S, Lee SW, Aaronson SA. Clonal Selection in Malignant Transformation of Human Fibroblasts Transduced with Defined Cellular Oncogenes. Cancer Res 2008; 68:1417-26. [DOI: 10.1158/0008-5472.can-07-3021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
202
|
Roy NS, Chandler-Militello D, Lu G, Wang S, Goldman SA. Retrovirally mediated telomerase immortalization of neural progenitor cells. Nat Protoc 2008; 2:2815-25. [PMID: 18007617 DOI: 10.1038/nprot.2007.402] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Traditional methods of generating immortalized lines of both somatic cells and their progenitors have relied on the use of oncogenes. However, the resulting lines are typically anaplastic in vitro and tumorigenic in vivo, and hence of limited utility. The overexpression of telomerase, as mediated by the induced overexpression of human telomerase reverse transcriptase (hTERT), has permitted the generation of stable, non-oncogenic lines of a variety of cell types. This strategy for immortalization has found special utility in the central nervous system, as few stable lines are available for the study of either human neural progenitor cells, or of neurons or glia of restricted phenotype. We describe the use of retroviral hTERT overexpression for generating lines of immortalized human neural progenitor cells, whose neuronal progeny are phenotypically restricted, post-mitotic and functionally competent. Although we focus here on telomerase immortalization of spinal neural progenitors, this is a broadly applicable protocol for using hTERT to immortalize human fetal neural progenitors of any pre-selected phenotype and for characterizing the cell lines thereby generated.
Collapse
Affiliation(s)
- Neeta S Roy
- Department of Neurology, Weill Medical College of Cornell University, New York, New York 10021, USA.
| | | | | | | | | |
Collapse
|
203
|
Forsyth NR, McWhir J. Human Embryonic Stem Cell Telomere Length Impacts Directly on Clonal Progenitor Isolation Frequency. Rejuvenation Res 2008; 11:5-17. [DOI: 10.1089/rej.2007.0567] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Nicholas R. Forsyth
- Department of Gene Function and Development, Roslin Institute, Roslin, United Kingdom
| | - Jim McWhir
- Department of Gene Function and Development, Roslin Institute, Roslin, United Kingdom
| |
Collapse
|
204
|
Abstract
The expression level of the telomerase catalytic subunit (telomerase reverse transcriptase, TERT) positively correlates with cell survival after exposure to several lethal stresses. However, whether the protective role of TERT is independent of telomerase activity has not yet been clearly explored. Here, we genetically evaluated the protective roles of both TERT and telomerase activity against cell death induced by staurosporine (STS) and N-methyl-D-aspartic acid (NMDA). First generation (G1) TERT-deficient mouse embryonic fibroblasts (MEFs) displayed an increased sensitivity to STS, while TERT transgenic MEFs were more resistant to STS-induced apoptosis than wild-type. Deletion of the telomerase RNA component (TERC) failed to alter the sensitivity of TERT transgenic MEFs to STS treatment. Similarly, NMDA-induced excitotoxic cell death of primary neurons was suppressed by TERT, but not by TERC both in vitro and in vivo. Specifically, NMDA accelerated death of TERT-deficient mice, while TERT transgenic mice showed enhanced survival when compared with wild-type littermates after administration of NMDA. In addition, the transgenic expression of TERT protected motor neurons from apoptosis induced by sciatic nerve axotomy. These results indicate that telomerase activity is not essential for the protective function of TERT. This telomerase activity-independent TERT function may contribute to cancer development and aging independently of telomere lengthening.
Collapse
|
205
|
Nakamura H. hTERT-immortalized cells useful for analyzing effects of low-dose-rate radiation on human cells. JOURNAL OF RADIATION RESEARCH 2008; 49:9-15. [PMID: 18160772 DOI: 10.1269/jrr.07088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
To establish immortal human cells, we introduced the cDNA of the human telomere reverse transcriptase (hTERT) gene into skin fibroblast cells obtained from normal and ataxia telangiectasia (AT) individuals of Japanese origin. hTERT-immortalized cells retained their original characteristics and radiosensitivity except for immortalization, suggesting that these cells might be useful for analyzing the effects of radiation on human cells.hTERT-immortalized cells from a normal individual showed a greater resistance after low-dose-rate irradiation than after high-dose-rate irradiation. In contrast, cells from AT patients irradiated at a low-dose rate showed virtually the same survival as those irradiated at a high-dose rate. In hTERT-immortalized normal cells, the genetic effects of low-dose-rate radiation were quantitatively and qualitatively less severe than those of high-dose-rate radiation. In hTERT-immortalized AT cells, some fraction of DNA damage such as DNA double-strand breaks might not be repaired, and AT cells were severely affected by low-dose-rate radiation. The activation of ataxia telangiectasia mutated (ATM) protein was confirmed during low-dose-rate radiation, and may play an important role in repair of DNA damage induced by low-dose-rate radiation. This paper reviews briefly the current research at our laboratory. The hTERT-immortalized cells may be useful in determining the effects of low-dose and low-dose-rate radiation on human cells.
Collapse
Affiliation(s)
- Hideaki Nakamura
- Central Laboratory and Radiation Biology, Aichi Cancer Center Research Institute, Nagoya, Japan.
| |
Collapse
|
206
|
Cairney CJ, Keith WN. Telomerase redefined: Integrated regulation of hTR and hTERT for telomere maintenance and telomerase activity. Biochimie 2008; 90:13-23. [PMID: 17854971 DOI: 10.1016/j.biochi.2007.07.025] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Accepted: 07/23/2007] [Indexed: 10/23/2022]
Abstract
Telomerase activity is dependent on the expression of 2 main core component genes, hTERT, which encodes the catalytic component and hTR (also called TERC), which encodes the RNA component. The correlation between telomerase activity and carcinogenesis has made this molecule of great interest in cancer research, however in order to fully understand the regulation of telomerase the mechanisms controlling both telomerase genes need to be studied. Some of these mechanisms of regulation have begun to emerge, however many more remain to be deciphered. For many years hTERT has been regarded as the limiting component of telomerase and much of the research in this field has focussed on its regulation, however it was clear from an early stage that hTR expression was also tightly regulated in normal cells and disease. More recently evidence from biochemistry, promoter studies and mouse models has been steadily increasing for a role for hTR as a limiting and essential component for telomerase activity and telomere maintenance. Perhaps the time has come to redefine our view of telomerase regulation. Knowledge of the mechanisms controlling both telomerase genes in normal systems and cancer may aid our understanding of the role of telomerase in carcinogenesis or highlight potential areas for therapeutic intervention. Here we review the essential requirement of hTR for telomere maintenance and telomerase activity in normal tissues and disease and focus on recent advances in our understanding of hTR regulation in relation to hTERT.
Collapse
Affiliation(s)
- C J Cairney
- Centre for Oncology and Applied Pharmacology, University of Glasgow, Cancer Research UK Beatson Laboratories, Bearsden, Glasgow G61 1BD, UK
| | | |
Collapse
|
207
|
Shawi M, Autexier C. Telomerase, senescence and ageing. Mech Ageing Dev 2007; 129:3-10. [PMID: 18215413 DOI: 10.1016/j.mad.2007.11.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 11/23/2007] [Accepted: 11/30/2007] [Indexed: 10/22/2022]
Abstract
Telomeres serve to camouflage chromosome ends from the DNA damage response machinery. Telomerase activity is required to maintain telomeres. One consequence of telomere dysfunction is cellular senescence, a permanent growth arrest state. We review the key regulators of cellular senescence and recent in vivo evidence which supports p53-dependent senescence induced by short telomeres as a potent tumor suppressor pathway. The in vivo link between cellular senescence and tumor regression is also discussed. The relationship between short telomere length and ageing or disease states in various cells of the body is increasingly reported. Paradoxically, the introduction of telomerase is proposed as a method to combat ageing via cell therapy and a possible method to regenerate tissue, while telomerase inhibition and telomere shortening is suggested as a possible therapy to defeat cancers with intact p53. Researchers thus face the challenge of understanding the complex processes which regulate the potential benefits of both telomerase inhibition and activation.
Collapse
Affiliation(s)
- May Shawi
- Department of Medicine, Division of Experimental Medicine, McGill University, Canada
| | | |
Collapse
|
208
|
Moldaver MV, Dashinimaev EB, Vishnyakova KS, Chumakov PM, Yegorov YE. Influence of oxygen on three different types of telomerized cells derived from a single donor. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2007. [DOI: 10.1134/s1990747807040034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
209
|
Iancu EM, Speiser DE, Rufer N. Assessing ageing of individual T lymphocytes: mission impossible? Mech Ageing Dev 2007; 129:67-78. [PMID: 18048082 DOI: 10.1016/j.mad.2007.10.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 10/15/2007] [Accepted: 10/23/2007] [Indexed: 12/22/2022]
Abstract
Effector T lymphocytes are the progeny of a limited number of antigen-specific precursor cells and it has been estimated that clonotypic human T cells may expand million fold on their way reaching high cell numbers that are sufficient for immune protection. Moreover, memory T cell responses are characterized by repetitive expansion of antigen-specific T cell clonotypes, and limitations in the proliferative capacity could lead to immune senescence. Because telomeres progressively shorten as a function of cell division, telomere length is a powerful indicator of the replicative in vivo history of human T lymphocytes. In this review, we summarize observations made over the last decade on telomere length dynamics of well-defined T cell populations derived from healthy donors and patients with infectious disease or cancer. We focus on T cell differentiation, T cell ageing, and natural and vaccine induced immune responses. We also discuss the scientific evidence for in vivo replicative senescence of antigen-specific T cells, and evaluate the available methods for measuring telomere lengths and telomerase activity, and their potential and limitations to increase our understanding of T cell physiology.
Collapse
Affiliation(s)
- Emanuela M Iancu
- Division of Experimental Oncology, Multidisciplinary Oncology Center CePO, Avenue Pierre-Decker 4, CH-1005 Lausanne, Switzerland
| | | | | |
Collapse
|
210
|
|
211
|
Campisi J, d'Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol 2007; 8:729-40. [PMID: 17667954 DOI: 10.1038/nrm2233] [Citation(s) in RCA: 3192] [Impact Index Per Article: 177.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cells continually experience stress and damage from exogenous and endogenous sources, and their responses range from complete recovery to cell death. Proliferating cells can initiate an additional response by adopting a state of permanent cell-cycle arrest that is termed cellular senescence. Understanding the causes and consequences of cellular senescence has provided novel insights into how cells react to stress, especially genotoxic stress, and how this cellular response can affect complex organismal processes such as the development of cancer and ageing.
Collapse
Affiliation(s)
- Judith Campisi
- Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, California 94720, USA.
| | | |
Collapse
|
212
|
Abstract
Telomeres are repetitive DNA sequences at the ends of linear chromosomes. Telomerase, a cellular reverse transcriptase, helps maintain telomere length in human stem cells, reproductive cells and cancer cells by adding TTAGGG repeats onto the telomeres. However, most normal human cells do not express telomerase and thus each time a cell divides some telomeric sequences are lost. When telomeres in a subset of cells become short (unprotected), cells enter an irreversible growth arrest state called replicative senescence. Cells in senescence produce a different constellation of proteins compared to normal quiescent cells. This may lead to a change in the homeostatic environment in a tissue-specific manner. In most instances cells become senescent before they can become cancerous; thus, the initial growth arrest induced by short telomeres may be thought of as a potent anti-cancer protection mechanism. When cells can be adequately cultured until they reach telomere-based replicative senescence, introduction of the telomerase catalytic protein component (hTERT) into telomerase-silent cells is sufficient to restore telomerase activity and extend cellular lifespan. Cells with introduced telomerase are not cancer cells, since they have not accumulated the other changes needed to become cancerous. This indicates that telomerase-induced telomere length manipulations may have utility for tissue engineering and for dissecting the molecular mechanisms underlying genetic diseases, including cancer.
Collapse
Affiliation(s)
- J W Shay
- University of Texas Southwestern Medical Center, Department of Cell Biology, Dallas, TX 75390-9039, USA.
| | | |
Collapse
|
213
|
Haker B, Fuchs S, Dierlamm J, Brümmendorf TH, Wege H. Absence of oncogenic transformation despite acquisition of cytogenetic aberrations in long-term cultured telomerase-immortalized human fetal hepatocytes. Cancer Lett 2007; 256:120-7. [PMID: 17630152 DOI: 10.1016/j.canlet.2007.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Revised: 05/10/2007] [Accepted: 06/05/2007] [Indexed: 10/23/2022]
Abstract
As a culture model to study hepatocarcinogenesis, telomerase-immortalized human fetal hepatocytes were monitored for karyotype changes evolving in long-term culture and development of functional defects in DNA damage response. G-banding revealed acquisition of characteristic karyotype abnormalities, e.g., trisomy 7 and monosomy X, in two independently immortalized and cultured populations after 80-100 population doublings. Interestingly, the detected aneuploidies resemble some of the genetic events observed in hepatocellular cancer. However, these genetic changes were not sufficient to induce oncogenic transformation reflected by absence of anchorage-independent growth. Furthermore, long-term cultured telomerase-immortalized cells preserved p53 expression levels and effective p53-mediated damage response.
Collapse
Affiliation(s)
- Björn Haker
- Department of Gastroenterology and Hepatology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | | | | | | | | |
Collapse
|
214
|
Li NF, Broad S, Lu YJ, Yang JS, Watson R, Hagemann T, Wilbanks G, Jacobs I, Balkwill F, Dafou D, Gayther SA. Human ovarian surface epithelial cells immortalized with hTERT maintain functional pRb and p53 expression. Cell Prolif 2007; 40:780-94. [PMID: 17877616 PMCID: PMC6495942 DOI: 10.1111/j.1365-2184.2007.00462.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Accepted: 04/16/2007] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE Cell immortalization is considered to be a prerequisite status for carcinogenesis. Normal human ovarian surface epithelial (OSE) cells, which are thought to be the origin of most of human ovarian carcinomas, have a very limited lifespan in culture. Establishment of immortalized OSE cell lines has, in the past, required inactivation of pRb and p53 functions. However, this often leads to increased chromosome instability during prolonged culture. MATERIALS AND METHODS In this study, we have used a retroviral infection method to overexpress human telomerase reverse transcriptase (hTERT) gene, in primary normal OSE cells, under optimized culture conditions. RESULTS In vitro and in vivo analysis of hTERT-immortalized cell lines confirmed their normal epithelial characteristics. Gene expression profiles and functional analysis of p16(INK4A), p15(INK4B), pRb and p53 confirmed the presence of their intact functions. Our study suggests that inactivation of pRb and p53 is not necessary for OSE immortalization. Furthermore, down-regulation of p15(INK4B) in the immortalized cells may indicate a functional role for this protein in them. CONCLUSION These immortal OSE cell lines are likely to be an important tool for studying human OSE biology and carcinogenesis.
Collapse
Affiliation(s)
- N F Li
- Centre for Translational Oncology, Barts and the London, Queen Mary's School of Medicine and Dentistry, Charterhouse Square, London, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
215
|
Kondoh K, Tsuji N, Asanuma K, Kobayashi D, Watanabe N. Inhibition of estrogen receptor β-mediated human telomerase reverse transcriptase gene transcription via the suppression of mitogen-activated protein kinase signaling plays an important role in 15-deoxy-Δ12,14-prostaglandin J2-induced apoptosis in cancer cells. Exp Cell Res 2007; 313:3486-96. [PMID: 17706193 DOI: 10.1016/j.yexcr.2007.06.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Revised: 06/23/2007] [Accepted: 06/25/2007] [Indexed: 01/05/2023]
Abstract
The nuclear hormone receptor peroxisome proliferator-activated receptor (PPAR)-gamma plays a role in cancer development in addition to its role in glucose metabolism. The natural ligand of PPAR-gamma, namely, 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)), has been shown to possess antineoplastic activity in cancer cells. However, the mechanism underlying its antineoplastic activity remains to be elucidated. Inhibition of the expression of human telomerase reverse transcriptase (hTERT), a major determinant of telomerase activity, reportedly induces rapid apoptosis in cancer cells. In this study, we investigated the effect of 15d-PGJ(2) on hTERT expression. We found that 15d-PGJ(2) induced apoptosis in the MIAPaCa-2 pancreatic cancer cells and dose-dependently decreased hTERT mRNA and protein expression. Down-regulation of hTERT expression by hTERT-specific small inhibitory RNA also induced apoptosis. Furthermore, 15d-PGJ(2) attenuated the DNA binding of estrogen receptor (ER). MIAPaCa-2 expressed only ERbeta, and although its expression did not decrease due to 15d-PGJ(2), its phosphorylation was suppressed. Additionally, a mitogen-activated protein kinase (MAPK) kinase inhibitor decreased ERbeta phosphorylation, and 15d-PGJ(2) attenuated MAPK activity. We conclude that hTERT down-regulation by 15d-PGJ(2) plays an important role in the proapoptotic property of the latter. Furthermore, 15d-PGJ(2) inhibits ERbeta-mediated hTERT gene transcription by suppressing ERbeta phosphorylation via the inhibition of MAP kinase signaling.
Collapse
Affiliation(s)
- Kei Kondoh
- Department of Clinical Laboratory Medicine, Sapporo Medical University School of Medicine, Japan
| | | | | | | | | |
Collapse
|
216
|
Satpathy M, Cao L, Pincheira R, Emerson R, Bigsby R, Nakshatri H, Matei D. Enhanced Peritoneal Ovarian Tumor Dissemination by Tissue Transglutaminase. Cancer Res 2007; 67:7194-202. [PMID: 17671187 DOI: 10.1158/0008-5472.can-07-0307] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tissue transglutaminase (TG2) is involved in Ca(2+)-dependent aggregation and polymerization of proteins. We previously reported that TG2 mRNA is up-regulated in epithelial ovarian cancer (EOC) cells compared with normal ovarian epithelium. Here, we show overexpression of the TG2 protein in ovarian cancer cells and tumors and its secretion in ascites fluid and define its role in EOC. By stable knockdown and overexpression, we show that TG2 enhances EOC cell adhesion to fibronectin and directional cell migration. This phenotype is preserved in vivo, where the pattern of tumor dissemination in the peritoneal space is dependent on TG2 expression levels. TG2 knockdown diminishes dissemination of tumors on the peritoneal surface and mesentery in an i.p. ovarian xenograft model. This phenotype is associated with deficient beta(1) integrin-fibronectin interaction, leading to weaker anchorage of cancer cells to the peritoneal matrix. Highly expressed in ovarian tumors, TG2 facilitates i.p. tumor dissemination by enhancing cell adhesion to the extracellular matrix and modulating beta(1) integrin subunit expression.
Collapse
Affiliation(s)
- Minati Satpathy
- Department of Medicine, Indiana University School of Medicine, 535 Barnhill Drive, RT 473, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | |
Collapse
|
217
|
Zhdanova NS, Rubtsov NB, Minina YM. Terminal regions of mammal chromosomes: Plasticity and role in evolution. RUSS J GENET+ 2007. [DOI: 10.1134/s1022795407070022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
218
|
Coates SSA, Lehnert BE, Sharma S, Kindell SM, Gary RK. Beryllium induces premature senescence in human fibroblasts. J Pharmacol Exp Ther 2007; 322:70-9. [PMID: 17395767 DOI: 10.1124/jpet.106.118018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
After cells have completed a sufficient number of cell divisions, they exit the cell cycle and enter replicative senescence. Here, we report that beryllium causes proliferation arrest with premature expression of the principal markers of senescence. After young presenescent human fibroblasts were treated with 3 microM BeSO(4) for 24 h, p21 cyclin-dependent kinase inhibitor mRNA increased by >200%. Longer periods of exposure caused mRNA and protein levels to increase for both p21 and p16(Ink4a), a senescence regulator that prevents pRb-mediated cell cycle progression. BeSO(4) also caused dose-dependent induction of senescence-associated beta-galactosidase activity (SA-beta-gal). Untreated cells had 48 relative fluorescence units (RFU)/microg/h of SA-beta-gal, whereas 3 microM BeSO(4) caused activity to increase to 84 RFU/microg/h. In chromatin immunoprecipitation experiments, BeSO(4) caused p53 protein to associate with its DNA binding site in the promoter region of the p21 gene, indicating that p53 transcriptional activity is responsible for the large increase in p21 mRNA elicited by beryllium. Forced expression of human telomerase reverse transcriptase (hTERT) rendered HFL-1 cells incapable of normal replicative senescence. However, there was no difference in the responsiveness of normal HFL-1 fibroblasts (IC(50) = 1.9 microM) and hTERT-immortalized cells (IC(50) = 1.7 microM) to BeSO(4) in a 9-day proliferation assay. The effects of beryllium resemble those of histone deacetylase-inhibiting drugs, which also cause large increases in p21. However, beryllium produced no changes in histone acetylation, suggesting that Be(2+) acts as a novel and potent pharmacological inducer of premature senescence.
Collapse
Affiliation(s)
- Shannon S A Coates
- Department of Chemistry, University of Nevada, Las Vegas, 4505 Maryland Parkway, Las Vegas, NV 89154, USA
| | | | | | | | | |
Collapse
|
219
|
Richardson RM, Nguyen B, Holt SE, Broaddus WC, Fillmore HL. Ectopic telomerase expression inhibits neuronal differentiation of NT2 neural progenitor cells. Neurosci Lett 2007; 421:168-72. [PMID: 17566647 DOI: 10.1016/j.neulet.2007.03.079] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Revised: 02/19/2007] [Accepted: 03/30/2007] [Indexed: 11/21/2022]
Abstract
There is significant interest in the potential use of telomerase-immortalized cells in transplantation to replace neurons lost to neurodegenerative diseases and other central nervous system injuries. Neural progenitor cells (NPCs) transduced with human telomerase reverse transcriptase (hTERT), the catalytic component of telomerase, have the potential both to proliferate indefinitely in vitro and to respond to differentiation signals necessary for generating appropriate cells for transplantation. The purpose of this study was to evaluate the differentiation of neurons from NT2 cells, a model NPC cell line, following hTERT transduction. RT-PCR and telomerase activity data demonstrated that persistent exogenous hTERT expression significantly inhibited the differentiation of neurons from NT2 cells. Following retinoic acid induced differentiation, hTERT-NT2 cells produced only one fourth of the neurons generated by parental and vector-control cells. A differentiation-inhibiting effect of constitutive telomerase activity has not been reported previously in other hTERT-transduced progenitor cell lines, implying a unique role for telomerase in the proliferation and differentiation of NPCs that have tumorigenic potential. Elucidating the mechanism responsible for this effect may aid in understanding the potential role of telomerase activity in the tumorigenicity of NPCs, as well as in optimizing the production of safe, telomerase-engineered, transplantable neurons.
Collapse
Affiliation(s)
- R Mark Richardson
- Department of Neurosurgery, Medical College of Virginia, Hospitals, Virginia Commonwealth University, VA, USA.
| | | | | | | | | |
Collapse
|
220
|
Abstract
During the evolution of linear genomes, it became essential to protect the natural chromosome ends to prevent triggering of the DNA-damage repair machinery and enzymatic attack. Telomeres - tightly regulated complexes consisting of repetitive G-rich DNA and specialized proteins - accomplish this task. Telomeres not only conceal linear chromosome ends from detection and inappropriate repair but also provide a buffer to counteract replication-associated shortening. Lessons from many model organisms have taught us about the complications of maintaining these specialized structures. Here, we discuss how telomeres interact and cooperate with the DNA replication and DNA-damage repair machineries.
Collapse
Affiliation(s)
- Ramiro E Verdun
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037-1099, USA
| | | |
Collapse
|
221
|
Oostingh GJ, Schlickum S, Friedl P, Schön MP. Impaired induction of adhesion molecule expression in immortalized endothelial cells leads to functional defects in dynamic interactions with lymphocytes. J Invest Dermatol 2007; 127:2253-8. [PMID: 17460735 DOI: 10.1038/sj.jid.5700828] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Immortalization should overcome the problem of short lifespan and difficult culture of endothelial cells that limited their use in functional studies. We used four different immortalized endothelial cell lines to study dynamic interactions with lymphocytes. Surprisingly, tumor necrosis factor (TNF)alpha-stimulated human umbilical vein endothelial cells (HUVECs) or human dermal microvascular endothelial cells (HDMECs) readily supported rolling and binding of lymphocytes, whereas none of the immortalized cell lines did. As rolling interactions are primarily mediated by selectins and vascular cell adhesion molecule (VCAM)-1, the endothelial cells were analyzed regarding expression of selectins and other adhesion molecules. Interestingly, cell surface expression of E-selectin could only be detected on HUVEC and HDMEC. Immunocytochemistry showed that some immortalized endothelial cells expressed E-selectin intracellularly following TNFalpha stimulation, suggesting translation but defective post-translational processing or transport of the molecule. In contrast, other immortalized cell lines did not have detectable levels of E-selectin mRNA, suggesting impaired transcription. VCAM-1 could only be induced on normal and human placental microvascular endothelial cell-A2 endothelial cells, whereas all cell lines expressed intercellular adhesion molecule-1 following TNF stimulation. The immortalized endothelial cells tested here have lost functions that are required for dynamic interactions with immune cells and that are common to primary endothelial cells.
Collapse
Affiliation(s)
- Gertie J Oostingh
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine and Department of Dermatology, University of Würzburg, Würzburg, Germany
| | | | | | | |
Collapse
|
222
|
Mikule K, Delaval B, Kaldis P, Jurcyzk A, Hergert P, Doxsey S. Loss of centrosome integrity induces p38-p53-p21-dependent G1-S arrest. Nat Cell Biol 2007; 9:160-70. [PMID: 17330329 DOI: 10.1038/ncb1529] [Citation(s) in RCA: 238] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Centrosomes organize the microtubule cytoskeleton for both interphase and mitotic functions. They are implicated in cell-cycle progression but the mechanism is unknown. Here, we show that depletion of 14 out of 15 centrosome proteins arrests human diploid cells in G1 with reduced Cdk2-cyclin A activity and that expression of a centrosome-disrupting dominant-negative construct gives similar results. Cell-cycle arrest is always accompanied by defects in centrosome structure and function (for example, duplication and primary cilia assembly). The arrest occurs from within G1, excluding contributions from mitosis and cytokinesis. The arrest requires p38, p53 and p21, and is preceded by p38-dependent activation and centrosomal recruitment of p53. p53-deficient cells fail to arrest, leading to centrosome and spindle dysfunction and aneuploidy. We propose that loss of centrosome integrity activates a checkpoint that inhibits G1-S progression. This model satisfies the definition of a checkpoint in having three elements: a perturbation that is sensed, a transducer (p53) and a receiver (p21).
Collapse
Affiliation(s)
- Keith Mikule
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | | | | | |
Collapse
|
223
|
Beliveau A, Bassett E, Lo AT, Garbe J, Rubio MA, Bissell MJ, Campisi J, Yaswen P. p53-dependent integration of telomere and growth factor deprivation signals. Proc Natl Acad Sci U S A 2007; 104:4431-6. [PMID: 17360541 PMCID: PMC1838618 DOI: 10.1073/pnas.0700260104] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Ectopically expressed hTERT enables p16(INK4A)(-) human mammary epithelial cells to proliferate in the absence of growth factors, a finding that has led to the hypothesis that hTERT has growth regulatory properties independent of its role in telomere maintenance. We now show that telomerase can alter the growth properties of cells indirectly through its role in telomere maintenance, without altering growth stimulatory pathways. We find that telomere dysfunction, indicated by 53BP1/phosphorylated histone H2AX foci at chromosome ends, is present in robustly proliferating human mammary epithelial cells long before senescence. These foci correlate with increased levels of active p53. Ectopic expression of hTERT reduces the number of foci and the level of active p53, thereby decreasing sensitivity to growth factor depletion, which independently activates p53. The continuous presence of hTERT is not necessary for this effect, indicating that telomere maintenance, rather than the presence of the enzyme itself, is responsible for the increased ability to proliferate in the absence of growth factors. Our findings provide a previously unrecognized mechanistic explanation for the observation that ectopically expressed hTERT conveys growth advantages to cells, without having to postulate nontelomeric functions for the enzyme.
Collapse
Affiliation(s)
- Alain Beliveau
- *Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
| | - Ekaterina Bassett
- *Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
| | - Alvin T. Lo
- *Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
| | - James Garbe
- *Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
| | - Miguel A. Rubio
- *Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
| | - Mina J. Bissell
- *Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
| | - Judith Campisi
- *Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945
| | - Paul Yaswen
- *Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and
- To whom correspondence should be addressed at:
Lawrence Berkeley National Laboratory, Mailstop 977R225A, 1 Cyclotron Road, Berkeley, CA 94720. E-mail:
| |
Collapse
|
224
|
Cortez-Gonzalez X, Zanetti M. Telomerase immunity from bench to bedside: round one. J Transl Med 2007; 5:12. [PMID: 17324292 PMCID: PMC1839079 DOI: 10.1186/1479-5876-5-12] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2006] [Accepted: 02/26/2007] [Indexed: 11/10/2022] Open
Abstract
Telomerase, a reverse transcriptase primarily devoted to the elongation of telomeres in mammalian cells, is also the first bona fide common tumor antigen. In fact, telomerase is over-expressed in > 85% of tumor cells irrespective of origin and histological type. In the past seven years, there has been considerable interest in assessing telomerase as substrate for vaccination in cancer patients to induce CD8 T cell responses. Because the activation of T cells is restricted by the MHC molecules on antigen presenting cells or tumor cells, the identification of telomerase peptides immunogenic for humans is tightly linked with HLA types. To date, a handful of peptides have been identified through a variety of screening procedures, including bioinformatics prediction, in vivo immunization of HLA transgenic mice, in vitro immunization of PBMC from normal donors and cancer patients, and processing in human tumor cells. Currently, there exist putative peptides for five major HLA types (A2, A1, A3, A24 and B7). Due to the complexity of the HLA system, trials have been performed focusing on the most prevalent HLA type, HLA-A2. Here, we summarize this collective effort and highlight results obtained in Phase 1 trials including a Phase 1 trial performed at the UCSD Cancer Center.
Collapse
Affiliation(s)
- Xochtil Cortez-Gonzalez
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0837, USA.
| | | |
Collapse
|
225
|
Mallette FA, Gaumont-Leclerc MF, Ferbeyre G. The DNA damage signaling pathway is a critical mediator of oncogene-induced senescence. Genes Dev 2007; 21:43-8. [PMID: 17210786 PMCID: PMC1759898 DOI: 10.1101/gad.1487307] [Citation(s) in RCA: 319] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Here we report that RNA interference against ATM inhibited p53 accumulation in cells expressing oncogenic STAT5 and cooperated with Rb inactivation to suppress STAT5A-induced senescence. Knocking down ATM was also effective to bypass E2F1-induced senescence and in combination with Rb inactivation, inhibited RasV12-induced senescence. Cells that senesced in response to ca-STAT5A or RasV12 accumulated DNA damage foci and activated ATM, ATR, Chk1, and Chk2, indicating that aberrant oncogene activation induces a DNA damage signaling response. Intriguingly, bypassing oncogene-induced senescence by inactivation of p53 and Rb did not eliminate the accumulation of oncogene-induced DNA damage foci (ODDI), suggesting a mechanism that may limit transformation in immortalized cells.
Collapse
Affiliation(s)
| | | | - Gerardo Ferbeyre
- Département de Biochimie, Université de Montréal, Montréal, Québec H3C 3J7, Canada
- Corresponding author.E-MAIL ; FAX (514) 343-2210
| |
Collapse
|
226
|
Huang Q, Chen M, Liang S, Acha V, Liu D, Yuan F, Hawks CL, Hornsby PJ. Improving cell therapy--experiments using transplanted telomerase-immortalized cells in immunodeficient mice. Mech Ageing Dev 2007; 128:25-30. [PMID: 17123586 PMCID: PMC1797893 DOI: 10.1016/j.mad.2006.11.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cell therapy is the use of stem cells and other types of cells in various therapies for age-related diseases. Two issues that must be addressed before cell therapy could be used routinely in medicine are improved efficacy of the transplanted cells and demonstrated long-term safety. Desirable genetic modifications that could be made to cells to be used for cell therapy include immortalization with human telomerase reverse transcriptase (hTERT). We have used a model for cell therapy in which transplantation of adrenocortical cells restores glucocorticoid and mineralocorticoid hormone levels in adrenalectomized immunodeficient mice. In this model, clones of cells that had been immortalized with hTERT were shown to be able to replace the function of the animals' adrenal glands by forming vascularized tissue structures when cells were transplanted beneath the capsule of the kidney. hTERT-modified cells showed no tendency for neoplastic changes. Moreover, a series of experiments showed that hTERT does not cooperate with known oncoproteins in tumorigenesis either in adrenocortical cells or in human fibroblasts. Nevertheless, hTERT was required for tumorigenesis when cells were implanted subcutaneously rather than in the subrenal capsule space. Changes in gene expression make hTERT-modified cells more robust. Understanding these changes is important so as to be able to separately control immortalization and other desirable properties of cells that could be used in cell therapy. Alternatively, desirable properties of transplants might be provided by co-transplanted mesenchymal cells: mesenchymal cell-assisted cell therapy. For both hTERT modification and mesenchymal cell-assisted cell therapy, genomics approaches will be needed to define what genetic modifications are desirable and safe in cells used in cell therapy.
Collapse
Affiliation(s)
- Qin Huang
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78245, USA
| | | | | | | | | | | | | | | |
Collapse
|
227
|
Malkas LH, Herbert BS, Abdel-Aziz W, Dobrolecki LE, Liu Y, Agarwal B, Hoelz D, Badve S, Schnaper L, Arnold RJ, Mechref Y, Novotny MV, Loehrer P, Goulet RJ, Hickey RJ. A cancer-associated PCNA expressed in breast cancer has implications as a potential biomarker. Proc Natl Acad Sci U S A 2006; 103:19472-7. [PMID: 17159154 PMCID: PMC1697829 DOI: 10.1073/pnas.0604614103] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Two isoforms of proliferating cell nuclear antigen (PCNA) have been observed in breast cancer cells. Commercially available antibodies to PCNA recognize both isoforms and, therefore, cannot differentiate between the PCNA isoforms in malignant and nonmalignant breast epithelial cells and tissues. We have developed a unique antibody that specifically detects a PCNA isoform (caPCNA) associated with breast cancer epithelial cells grown in culture and breast-tumor tissues. Immunostaining studies using this antibody suggest that the caPCNA isoform may be useful as a marker of breast cancer and that the caPCNA-specific antibody could potentially serve as a highly effective detector of malignancy. We also report here that the caPCNA isoform functions in breast cancer-cell DNA replication and interacts with DNA polymerase delta. Our studies indicate that the caPCNA isoform may be a previously uncharacterized detector of breast cancer.
Collapse
Affiliation(s)
- Linda H. Malkas
- *Division of Hematology and Oncology, Department of Medicine, and
- Indiana Cancer Center, Indianapolis, IN 46202
- To whom correspondence may be addressed at:
Division of Hematology and Oncology, Department of Medicine, Indiana University School of Medicine, Indiana University Cancer Center, 1044 West Walnut Street, Room R4-171, Indianapolis, IN 46202. E-mail:
or
| | | | - Waleed Abdel-Aziz
- *Division of Hematology and Oncology, Department of Medicine, and
- Indiana Cancer Center, Indianapolis, IN 46202
| | | | - Yang Liu
- *Division of Hematology and Oncology, Department of Medicine, and
| | - Beamon Agarwal
- Department of Pathology, Howard University College of Medicine, Washington, DC 20059
| | - Derek Hoelz
- *Division of Hematology and Oncology, Department of Medicine, and
- Indiana Cancer Center, Indianapolis, IN 46202
| | - Sunil Badve
- Departments of Pathology
- Molecular Genetics, and
| | - Lauren Schnaper
- **Comprehensive Breast Care Center, Greater Baltimore Medical Center, Baltimore, MD 21204; and
| | - Randy J. Arnold
- Department of Chemistry, Indiana University, Bloomington, IN 47405
| | - Yehia Mechref
- Department of Chemistry, Indiana University, Bloomington, IN 47405
| | - Milos V. Novotny
- Indiana Cancer Center, Indianapolis, IN 46202
- Department of Chemistry, Indiana University, Bloomington, IN 47405
| | - Patrick Loehrer
- *Division of Hematology and Oncology, Department of Medicine, and
- Indiana Cancer Center, Indianapolis, IN 46202
| | - Robert J. Goulet
- Surgery, Indiana University School of Medicine, and
- Indiana Cancer Center, Indianapolis, IN 46202
| | - Robert J. Hickey
- *Division of Hematology and Oncology, Department of Medicine, and
- Indiana Cancer Center, Indianapolis, IN 46202
- To whom correspondence may be addressed at:
Division of Hematology and Oncology, Department of Medicine, Indiana University School of Medicine, Indiana University Cancer Center, 1044 West Walnut Street, Room R4-171, Indianapolis, IN 46202. E-mail:
or
| |
Collapse
|
228
|
Abstract
The cell phenotypes of senescence and crisis operate to circumscribe the proliferative potential of mammalian cells, suggesting that both are capable of operating in vivo to suppress the formation of tumors. The key regulators of these phenotypes are the telomeres, which are located at the ends of chromosomes and operate to protect the chromosomes from end-to-end fusions. Telomere erosion below a certain length can trigger crisis. The relationship between senescence and telomere function is more complex, however: Cell-physiological stresses as well as dysfunction of the complex molecular structures at the ends of telomeric DNA can trigger senescence. Cells can escape senescence by inactivating the Rb and p53 tumor suppressor proteins and can surmount crisis by activating a telomere maintenance mechanism. The resulting cell immortalization is an essential component of the tumorigenic phenotype of human cancer cells. Here we discuss how telomeres are monitored and maintained and how loss of a functional telomere influences biological functions as diverse as aging and carcinogenesis.
Collapse
Affiliation(s)
- Sheila A Stewart
- Departments of Cell Biology and Physiology and of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | |
Collapse
|
229
|
Canales BK, Li Y, Thompson MG, Gleason JM, Chen Z, Malaeb B, Corey DR, Herbert BS, Shay JW, Koeneman KS. Small molecule, oligonucleotide-based telomerase template inhibition in combination with cytolytic therapy in an in vitro androgen-independent prostate cancer model. Urol Oncol 2006; 24:141-51. [PMID: 16520278 DOI: 10.1016/j.urolonc.2005.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE Determine the efficacy and timing of small molecule oligonucleotide-based inhibitors to the enzyme telomerase in an in vitro model of androgen-independent, osseous prostate cancer. MATERIALS AND METHODS Telomerase was inhibited in prostate cancer cell lines C4-2/C4-2B and in controls by using small molecule antisense oligonucleotide-based inhibitors alone or in various combinations of small-dose Taxotere (sanofi-aventis, Bridgewater, NJ) and/or conditionally replication competent adenovirus (AD-BSP-E1a). After transfection and proliferation, telomerase telomeric repeat amplification protocol and telomere restriction fragment assays were performed, with specific times for evaluating telomere length. Specimens were stained for analysis with hematoxylin and eosin (H&E), terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL), and prostate-specific antigen (PSA). RESULTS C4-2/C4-2B cell lines had the shortest initial mean telomere length (approximately 2.5 kilobase [kb]) compared to PC-3 (approximately 5.5 kb). Dose-dependent inhibition of telomerase activity was seen using match oligonucleotide-based inhibitors to telomerase (50% inhibitory concentration 3-5 nm), whereas mismatch compound showed no telomerase inhibition. Significant growth delay and apoptosis in cell lines occurred after > 50 days of treatment. Cells treated with combination "triple therapy" (i.e., telomerase inhibitors, adenovirus, and Taxotere) had the highest amount of apoptosis. Compared to controls, all combination treatment groups had statistically significant reductions in prostate-specific antigen in the conditioned media. CONCLUSIONS Combining cytotoxic regimens with small molecule inhibitors to telomerase with oligonucleotide-based agents could be beneficial in controlling osseous hormone refractory prostate cancer, as evidenced by these in vitro, preclinical investigations. Telomerase inhibition needs to move into in vivo models and human studies.
Collapse
Affiliation(s)
- Benjamin K Canales
- Department of Urology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Varella-Garcia M, Chen L, Zheng X, Yu L, Dixon D. Karyotypic characteristics of human uterine leiomyoma and myometrial cell lines following telomerase induction. ACTA ACUST UNITED AC 2006; 170:71-5. [PMID: 16965960 DOI: 10.1016/j.cancergencyto.2006.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Accepted: 05/12/2006] [Indexed: 12/21/2022]
Abstract
Uterine leiomyomas are the most frequently diagnosed tumors of the female genital tract and the primary cause of hysterectomy in premenopausal women in the United States. In vitro model systems for studying these tumors are limited, due to poor culture growth. For the present study, myometrial (UtSMC) and uterine leiomyoma (UtLM) cell lines and their human telomerase reverse transcriptase (hTERT) immortalized counterparts were evaluated by GTL-banding and spectral karyotyping. UtSMC, at passage 9 showed the normal female karyotype, 46,XX. UtSMC-hTERT at passage 13 (population doubling [PD] 22 post immortalization) had two cell clones: 46,XX,der(13)t(10;13)(q11.2;p13)]/46,XX. UtLM, at passage 14, and the immortalized counterpart UtLM-hTERT (passage 13, PD 20 post immortalization), had the reciprocal translocation t(12;14)(q14;q23) in all cells and monosomy X in a fraction of cells. UtLM also displayed genomic instability with 15% of cells showing structural abnormalities involving chromosome arms 1p and 5q. UtLM-hTERT was karyotypically more stable than the parental line, thereby reflecting the inhibition of the accumulation of cytogenetic abnormalities by the maintenance of telomere integrity. This phenomenon was not observed in the UtSMC cells.
Collapse
Affiliation(s)
- Marileila Varella-Garcia
- Department of Medicine, Division of Medical Oncology, University of Colorado Health Sciences Center, Aurora, 80045, USA
| | | | | | | | | |
Collapse
|
231
|
Menzel O, Migliaccio M, Goldstein DR, Dahoun S, Delorenzi M, Rufer N. Mechanisms Regulating the Proliferative Potential of Human CD8+ T Lymphocytes Overexpressing Telomerase. THE JOURNAL OF IMMUNOLOGY 2006; 177:3657-68. [PMID: 16951325 DOI: 10.4049/jimmunol.177.6.3657] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In human somatic cells, including T lymphocytes, telomeres progressively shorten with each cell division, eventually leading to a state of cellular senescence. Ectopic expression of telomerase results in the extension of their replicative life spans without inducing changes associated with transformation. However, it is yet unknown whether somatic cells that overexpress telomerase are physiologically indistinguishable from normal cells. Using CD8+ T lymphocyte clones overexpressing telomerase, we investigated the molecular mechanisms that regulate T cell proliferation. In this study, we show that early passage T cell clones transduced or not with human telomerase reverse transcriptase displayed identical growth rates upon mitogenic stimulation and no marked global changes in gene expression. Surprisingly, reduced proliferative responses were observed in human telomerase reverse transcriptase-transduced cells with extended life spans. These cells, despite maintaining high expression levels of genes involved in the cell cycle progression, also showed increased expression in several genes found in common with normal aging T lymphocytes. Strikingly, late passage T cells overexpressing telomerase accumulated the cyclin-dependent inhibitors p16Ink4a and p21Cip1 that have largely been associated with in vitro growth arrest. We conclude that alternative growth arrest mechanisms such as those mediated by p16Ink4a and p21Cip1 still remained intact and regulated the growth potential of cells independently of their telomere status.
Collapse
Affiliation(s)
- Olivier Menzel
- National Center of Competence in Research Molecular Oncology, Swiss Institute for Experimental Cancer Research, Epalinges, Switzerland
| | | | | | | | | | | |
Collapse
|
232
|
Berger KL, Barriga F, Lace MJ, Turek LP, Zamba GJ, Domann FE, Lee JH, Klingelhutz AJ. Cervical keratinocytes containing stably replicating extrachromosomal HPV-16 are refractory to transformation by oncogenic H-Ras. Virology 2006; 356:68-78. [PMID: 16945398 PMCID: PMC1993897 DOI: 10.1016/j.virol.2006.07.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2006] [Revised: 05/25/2006] [Accepted: 07/27/2006] [Indexed: 10/24/2022]
Abstract
Ras expression in human epithelial cells with integrated HPV genomes has been shown to cause tumorigenic transformation. The effects of Ras in cells representing early stage HPV-associated disease (i.e., when HPV is extrachromosomal and the oncogenes are under control of native promoters) have not been examined. Here, we used human cervical keratinocyte cell lines containing stably replicating extrachromosomal HPV-16 and present the novel finding that these cells resist transformation by oncogenic H-Ras. Ras expression consistently diminished anchorage-independent growth (AI), reduced E6 and E7 expression, and caused p53 induction in these cells. Conversely, AI was enhanced or maintained in Ras-transduced cervical cells that were immortalized with a 16E6/E7 retrovirus, and minimal effects on E6 and E7 expression were observed. Ras expression with either episomal HPV-16 or LXSN-E6/E7 was insufficient for tumorigenic growth suggesting that other events are needed for tumorigenic transformation. In conclusion, our results indicate that Ras-mediated transformation depends on the context of HPV oncogene expression and that this is an important point to address when developing HPV tumor models.
Collapse
Affiliation(s)
- Kristi L. Berger
- Department of Microbiology, Holden Comprehensive Cancer Research Center, University of Iowa, 375 Newton Road, 2202 MERF, Iowa City, IA 52242, USA
| | - Felicia Barriga
- Department of Microbiology, Holden Comprehensive Cancer Research Center, University of Iowa, 375 Newton Road, 2202 MERF, Iowa City, IA 52242, USA
| | | | | | - Gideon J. Zamba
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 52242, USA
| | - Frederick E. Domann
- Department of Radiology, Free Radical and Radiation Biology Program, University of Iowa, Iowa City, IA 52242, USA
| | - John H. Lee
- Department of Otolaryngology, Head and Neck Surgery, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Aloysius J. Klingelhutz
- Department of Microbiology, Holden Comprehensive Cancer Research Center, University of Iowa, 375 Newton Road, 2202 MERF, Iowa City, IA 52242, USA
- Corresponding author. Fax: +1 319 353 4556, E-mail address: (A.J. Klingelhutz)
| |
Collapse
|
233
|
Brümmendorf TH, Balabanov S. Telomere length dynamics in normal hematopoiesis and in disease states characterized by increased stem cell turnover. Leukemia 2006; 20:1706-16. [PMID: 16888616 DOI: 10.1038/sj.leu.2404339] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Telomeres both reflect and limit the replicative lifespan of normal somatic cells. Immature sub-populations of human CD34+38- hematopoietic stem cell (HSC) can be identified in vitro based on their growth kinetics and telomere length. Fluorescence in situ hybridization and flow cytometry (flow-FISH) has been used to characterize telomere length dynamics as a surrogate marker for HSC turnover in vivo. Investigations in normal steady-state hematopoiesis provided the basis for follow-up studies in model scenarios characterized by increased HSC turnover. Disorders with underlying malignant transformation of HSC (e.g., chronic myeloid leukemia (CML)) can be discriminated from disease states with increased HSC turnover rates secondary to depletion of the stem cell compartment, for example, as in defined bone marrow failure syndromes. In some of these model scenarios, the degree of telomere shortening can be correlated with disease duration, disease stage and severity as well as with response to disease-modifying treatment strategies. Whether increased telomere shortening represents a causal link between HSC turnover, replicative senescence and/or the induction of genetic instability in acquired HSC disorders remains to be shown. However, data from congenital disorders, like dyskeratosis congenita (DKC), suggest that disturbed telomere maintenance may play a role for replicative exhaustion of the HSC pool in vivo.
Collapse
Affiliation(s)
- T H Brümmendorf
- Department of Oncology and Hematology with Sections Bone Marrow Transplantation and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | | |
Collapse
|
234
|
Buser R, Montesano R, Garcia I, Dupraz P, Pepper MS. Bovine microvascular endothelial cells immortalized with human telomerase. J Cell Biochem 2006; 98:267-86. [PMID: 16408275 DOI: 10.1002/jcb.20715] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Primary cultures of bovine microvascular endothelial cells (BME) isolated from the adrenal cortex, are commonly used to study vascular endothelium, but have a limited life span. To circumvent these limitations, we have immortalized BME cells with either simian virus 40 (SV40) or with a retrovirus containing the coding region of human telomerase reverse transcriptase (hTERT), and have investigated whether the clonal populations obtained, maintain differentiated properties characteristic of microvascular endothelium. Immortalized cells were characterized for maintenance of typical endothelial morphology, marker expression, and functional characteristics including uptake of Acetylated low-density lipoprotein (Ac-LDL), capillary-like tube formation in three-dimensional collagen gels, as well as metalloproteinase (MMP) and plasminogen activator (PA)-mediated extracellular proteolysis. Whilst immortalization of BME cells with SV40 was associated with loss of endothelial-specific properties, hTERT-BME exhibited an endothelial phenotype similar to that of wild-type endothelial cells. Specifically, they showed a typical cobblestone morphology, were contact-inhibited, expressed endothelial cell-specific markers (e.g., CD31, vWF) and both fibroblast growth factor receptor 1 (FGFR-1) and vascular endothelial growth factor receptor-2 (VEGFR-2). In addition, they expressed receptors for LDL. Importantly, when grown on collagen gels, hTERT-BME cells underwent MMP-dependent tube-like structure formation in response to VEGFR-2 activation. In a collagen gel sandwich assay, hTERT-BME formed tubular structures in the absence of exogenously added angiogenic cytokines. Sustained tube formation was induced by VEGF-A alone or in combination with FGF-2. From 17 sub-clones that displayed a non-transformed phenotype, a high proliferative capacity and tubulogenic properties in three-dimensional collagen gels, we isolated two distinct subpopulations that display a highly specific response to VEGF-A or to FGF-2. We have generated hTERT-BME cells that maintain endothelial-specific properties and function and have isolated clones that respond differentially to VEGF-A or FGF-2. These immortalized cell lines will facilitate the study of endothelial cell biology.
Collapse
Affiliation(s)
- Raphaële Buser
- Department of Cell Physiology and Metabolism, University Medical Center, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
235
|
Tatsumi Y, Sugimoto N, Yugawa T, Narisawa-Saito M, Kiyono T, Fujita M. Deregulation of Cdt1 induces chromosomal damage without rereplication and leads to chromosomal instability. J Cell Sci 2006; 119:3128-40. [PMID: 16835273 DOI: 10.1242/jcs.03031] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The activity of human Cdt1 is negatively regulated by multiple mechanisms. This suggests that Cdt1 deregulation may have a deleterious effect. Indeed, it has been suggested that overexpression of Cdt1 can induce rereplication in cancer cells and that rereplication activates Ataxia-telangiectasia-mutated (ATM) kinase and/or ATM- and Rad3-related (ATR) kinase-dependent checkpoint pathways. In this report, we highlight a new and interesting aspect of Cdt1 deregulation: data from several different systems all strongly indicate that unregulated Cdt1 overexpression at pathophysiological levels can induce chromosomal damage other than rereplication in non-transformed cells. The most important finding in these studies is that deregulated Cdt1 induces chromosomal damage and activation of the ATM-Chk2 DNA damage checkpoint pathway even in quiescent cells. These Cdt1 activities are negatively regulated by cyclin A/Cdks, probably through modification by phosphorylation. Furthermore, we found that deregulated Cdt1 induces chromosomal instability in normal human cells. Since Cdt1 is overexpressed in cancer cells, this would be a new molecular mechanism leading to carcinogenesis.
Collapse
Affiliation(s)
- Yasutoshi Tatsumi
- Virology Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuohku, Tokyo 104-0045, Japan
| | | | | | | | | | | |
Collapse
|
236
|
Jefford CE, Irminger-Finger I. Mechanisms of chromosome instability in cancers. Crit Rev Oncol Hematol 2006; 59:1-14. [PMID: 16600619 DOI: 10.1016/j.critrevonc.2006.02.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2005] [Revised: 02/22/2006] [Accepted: 02/22/2006] [Indexed: 12/31/2022] Open
Abstract
Most tumours arise through clonal selection and waves of expansion of a somatic cell that has acquired genetic alterations in essential genes either controlling cell death or cell proliferation. Furthermore, stability of the genome in cancer cells becomes precarious and compromised because several cancer-predisposing mutations affect genes that are responsible for maintaining the integrity and number of chromosomes during cell division. Consequently, the archetypical transformation in tumour cells results in aneuploidy. Indeed, almost all tumour cells display a host of karyotype alterations, showing translocations, gains or losses of entire or large parts of chromosomes. Cancers do not necessarily have a higher mutation rate than normal tissue at the nucleotide level, unless they have gained a mutator phenotype through exposure to environmental stress, but rather exhibit gross chromosomal changes. Therefore, it appears that the main mechanism of tumour progression stems from chromosome instability. Chromosomal instability prevailing in tumour cells arises through several different pathways and is probably controlled by hundreds of genes. Therefore, this review describes the main factors that control chromosome stability through telomere maintenance, mechanisms of cell division, and the mitotic checkpoints that govern centrosome duplication and correct chromosome segregation.
Collapse
Affiliation(s)
- Charles Edward Jefford
- Biology of Aging Laboratory, Department of Geriatrics, University Hospitals Geneva HUG, Switzerland.
| | | |
Collapse
|
237
|
Rea MA, Zhou L, Qin Q, Barrandon Y, Easley KW, Gungner SF, Phillips MA, Holland WS, Gumerlock PH, Rocke DM, Rice RH. Spontaneous immortalization of human epidermal cells with naturally elevated telomerase. J Invest Dermatol 2006; 126:2507-15. [PMID: 16778798 DOI: 10.1038/sj.jid.5700424] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This work explores spontaneous immortalization in keratinocytes, derived from two skin samples, that display naturally elevated telomerase activity. Serially passaged with 3T3 feeder layer support, the keratinocytes were examined for colony-forming ability, telomerase activity, telomere length, and finally gene expression using Affymetrix DNA microarrays. The cells initially exhibited normal karyotypes and low colony-forming efficiencies typical of normal epidermal cells, but after 40 passages (approximately 400 generations) colony-forming ability increased markedly, yielding immortalized lines exhibiting a small number of chromosomal aberrations and functionally normal p53. An improved protocol for analysis of microarray data permitted detection of 707 transcriptional changes accompanying immortalization including reduced p16(INK4A) mRNA. Telomerase activity was clearly elevated in cells even at low passage from both samples, and telomerase catalytic subunit mRNA was greatly elevated in those with elevated colony-forming ability. The data raise the possibility of an unusual natural phenotype in which aberrant telomerase regulation extends keratinocyte lifespan until rare variants evade senescence. In addition to revealing a potential tumor-prone syndrome, the findings emphasize the desirability of carefully minimizing the degree or timing of elevated expression of telomerase used to immortalize cells for therapeutic purposes.
Collapse
Affiliation(s)
- Miguel A Rea
- Department of Environmental Toxicology, University of California, Davis, California 95616-8588, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
An FQ, Folarin HM, Compitello N, Roth J, Gerson SL, McCrae KR, Fakhari FD, Dittmer DP, Renne R. Long-term-infected telomerase-immortalized endothelial cells: a model for Kaposi's sarcoma-associated herpesvirus latency in vitro and in vivo. J Virol 2006; 80:4833-46. [PMID: 16641275 PMCID: PMC1472065 DOI: 10.1128/jvi.80.10.4833-4846.2006] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman's disease. Most KS tumor cells are latently infected with KSHV and are of endothelial origin. While PEL-derived cell lines maintain KSHV indefinitely, all KS tumor-derived cells to date have lost viral genomes upon ex vivo cultivation. To study KSHV latency and tumorigenesis in endothelial cells, we generated telomerase-immortalized human umbilical vein endothelial (TIVE) cells. TIVE cells express all KSHV latent genes 48 h postinfection, and productive lytic replication could be induced by RTA/Orf50. Similar to prior models, infected cultures gradually lost viral episomes. However, we also obtained, for the first time, two endothelial cell lines in which KSHV episomes were maintained indefinitely in the absence of selection. Long-term KSHV maintenance correlated with loss of reactivation in response to RTA/Orf50 and complete oncogenic transformation. Long-term-infected TIVE cells (LTC) grew in soft agar and proliferated under reduced-serum conditions. LTC, but not parental TIVE cells, formed tumors in nude mice. These tumors expressed high levels of the latency-associated nuclear antigen (LANA) and expressed lymphatic endothelial specific antigens as found in KS (LYVE-1). Furthermore, host genes, like those encoding interleukin 6, vascular endothelial growth factor, and basic fibroblast growth factor, known to be highly expressed in KS lesions were also induced in LTC-derived tumors. KSHV-infected LTCs represent the first xenograft model for KS and should be of use to study KS pathogenesis and for the validation of anti-KS drug candidates.
Collapse
Affiliation(s)
- Feng-Qi An
- Department of Molecular Genetics and Microbiology, University of Florida, Shands Cancer Center, Gainesville, FL 32610-0232, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Affiliation(s)
- Peter J Hornsby
- Department of Physiology, Sam and Ann Barshop Center for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, Texas, USA
| |
Collapse
|
240
|
Kogan I, Goldfinger N, Milyavsky M, Cohen M, Shats I, Dobler G, Klocker H, Wasylyk B, Voller M, Aalders T, Schalken JA, Oren M, Rotter V. hTERT-immortalized prostate epithelial and stromal-derived cells: an authentic in vitro model for differentiation and carcinogenesis. Cancer Res 2006; 66:3531-40. [PMID: 16585177 DOI: 10.1158/0008-5472.can-05-2183] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostate cancer is the most commonly diagnosed type of cancer in men, and there is no available cure for patients with advanced disease. In vitro model systems are urgently required to permit the study of human prostate cell differentiation and malignant transformation. Unfortunately, human prostate cells are particularly difficult to convert into continuously growing cultures. We report here the successful immortalization without viral oncogenes of prostate epithelial cells and, for the first time, prostate stromal cells. These cells exhibit a significant pattern of authentic prostate-specific features. In particular, the epithelial cell culture is able to differentiate into glandular buds that closely resemble the structures formed by primary prostate epithelial cells. The stromal cells have typical characteristics of prostate smooth muscle cells. These immortalized cultures may serve as a unique experimental platform to permit several research directions, including the study of cell-cell interactions in an authentic prostate microenvironment, prostate cell differentiation, and most significantly, the complex multistep process leading to prostate cell transformation.
Collapse
Affiliation(s)
- Ira Kogan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Wright LS, Prowse KR, Wallace K, Linskens MHK, Svendsen CN. Human progenitor cells isolated from the developing cortex undergo decreased neurogenesis and eventual senescence following expansion in vitro. Exp Cell Res 2006; 312:2107-20. [PMID: 16631163 DOI: 10.1016/j.yexcr.2006.03.012] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2005] [Revised: 03/08/2006] [Accepted: 03/10/2006] [Indexed: 12/11/2022]
Abstract
Isolation of a true self-renewing stem cell from the human brain would be of great interest as a reliable source of neural tissue. Here, we report that human fetal cortical cells grown in epidermal growth factor expressed low levels of telomerase and telomeres in these cultures shortened over time leading to growth arrest after 30 weeks. Following leukemia inhibitory factor (LIF) supplementation, growth rates and telomerase expression increased. This was best demonstrated following cell cycle synchronization and staining for telomerase using immunocytochemistry. This increase in activity resulted in the maintenance of telomeres at approximately 7 kb for more than 60 weeks in vitro. However, all cultures displayed a lack of oligodendrotye production, decreases in neurogenesis over time and underwent replicative senescence associated with increased expression of p21 before 70 weeks in vitro. Thus, under our culture conditions, these cells are not stable, multipotent, telomerase expressing self-renewing stem cells. They may be more accurately described as human neural progenitor cells (hNPC) with limited lifespan and bi-potent potential (neurons/astrocytes). Interestingly, hNPC follow a course of proliferation, neuronal production and growth arrest similar to that seen during expansion and development of the human cortex, thus providing a possible model neural system. Furthermore, due to their high expansion potential and lack of tumorogenicity, these cells remain a unique and safe source of tissue for clinical transplantation.
Collapse
Affiliation(s)
- Lynda S Wright
- Waisman Center and Departments of Neurology and Anatomy University of Wisconsin-Madison, WI 53705-2280, USA
| | | | | | | | | |
Collapse
|
242
|
Carvalho L, Lipay M, Belfort F, Santos I, Andrade J, Haddad A, Brunstein F, Ferreira L. Telomerase activity in prognostic histopathologic features of melanoma. J Plast Reconstr Aesthet Surg 2006; 59:961-8. [PMID: 16920589 DOI: 10.1016/j.bjps.2006.01.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2005] [Revised: 12/21/2005] [Accepted: 01/01/2006] [Indexed: 11/23/2022]
Abstract
BACKGROUND Telomerase activity (TA) is believed to play a role in the regulation of senescence and to limit the number of cell divisions. The deregulation of telomerase appears to contribute to oncogenesis and the formation of immortal cell lines. As a result, it is believed that it could be used as a prognostic marker in melanoma. METHODS TA was assayed by the polymerase chain reaction PCR-ELISA-based telomeric repeat amplification protocol (TRAP assay). One hundred and eight samples were distributed in four histological groups: 30 samples from primary cutaneous melanomas, 24 from peritumoural skin sites, 28 from benign melanocytic lesions, and 26 from normal skin sites as a control. RESULTS TA was different among the four tested groups (Kruskall-Wallis test p<0.001), and increasing values of TA were observed progressing from normal skin to benign and then to malignant lesions. Among melanoma samples, there was a significant association between TA and ulceration (p=0.025), TA and vascular invasion (p=0.018) and TA and mitotic rate (p=0.029) (Mann-Whitney test). A linear regression analysis showed significant associations between the increase of TA with Breslow thickness (p=0.004) and the presence of satellites (p=0.002). CONCLUSIONS We observed that TA had increased from control skin to peritumoural skin, and then to benign melanocytic lesions and finally to melanoma, suggesting tumour progression. TA showed higher values in the presence of some important histopathologic parameters related to poor prognosis in cutaneous melanoma such as ulceration, vascular invasion, satellites, high rates of mitosis, and in thicker tumours.
Collapse
Affiliation(s)
- Lucia Carvalho
- Surgery Department, Plastic Surgery Division, UNIFESP, Federal University of São Paulo, São Paulo, SP, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
243
|
Brachner A, Sasgary S, Pirker C, Rodgarkia C, Mikula M, Mikulits W, Bergmeister H, Setinek U, Wieser M, Chin SF, Caldas C, Micksche M, Cerni C, Berger W. Telomerase- and Alternative Telomere Lengthening–Independent Telomere Stabilization in a Metastasis-Derived Human Non–Small Cell Lung Cancer Cell Line: Effect of Ectopic hTERT. Cancer Res 2006; 66:3584-92. [PMID: 16585183 DOI: 10.1158/0008-5472.can-05-2839] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the majority of human malignancies, maintenance of telomeres is achieved by reactivation of telomerase, whereas a smaller fraction uses an alternative telomere lengthening (ALT) mechanism. Here, we used 16 non-small cell lung cancer (NSCLC) cell lines to investigate telomere stabilization mechanisms and their effect on tumor aggressiveness. Three of 16 NSCLC cell lines (VL-9, SK-LU-1, and VL-7) lacked telomerase activity, correlating with significantly reduced tumorigenicity in vitro and in vivo. Of the three telomerase-negative cell lines, only SK-LU-1 displayed characteristics of an ALT mechanism (i.e., highly heterogeneous telomeres and ALT-associated promyelocytic leukemia bodies). VL-9 cells gained telomerase during in vitro propagation, indicating incomplete immortalization in vivo. In contrast, NSCLC metastasis-derived VL-7 cells remained telomerase and ALT negative up to high passage numbers and following transplantation in severe combined immunodeficient mice. Telomeres of VL-7 cells were homogeneously short, and chromosomal instability (CIN) was comparable with most telomerase-positive cell lines. This indicates the presence of an efficient telomere stabilization mechanism different from telomerase and ALT in VL-7 cells. To test the effect of ectopic telomerase reverse transcriptase (hTERT) in these unique ALT- and telomerase-negative tumor backgrounds, hTERT was transfected into VL-7 cells. The activation of telomerase led to an excessively rapid gain of telomeric sequences resulting in very long ( approximately 14 kb), uniform telomeres. Additionally, hTERT expression induced a more aggressive growth behavior in vitro and in vivo without altering the level of CIN. These data provide further evidence for a direct oncogenic activity of hTERT not based on the inhibition of CIN.
Collapse
Affiliation(s)
- Andreas Brachner
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Gu Y, Li H, Miki J, Kim KH, Furusato B, Sesterhenn IA, Chu WS, McLeod DG, Srivastava S, Ewing CM, Isaacs WB, Rhim JS. Phenotypic characterization of telomerase-immortalized primary non-malignant and malignant tumor-derived human prostate epithelial cell lines. Exp Cell Res 2006; 312:831-43. [PMID: 16413016 DOI: 10.1016/j.yexcr.2005.11.029] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2005] [Revised: 10/25/2005] [Accepted: 11/30/2005] [Indexed: 10/25/2022]
Abstract
In vitro human prostate cell culture models are critical for clarifying the mechanism of prostate cancer progression and for testing preventive and therapeutic agents. Cell lines ideal for the study of human primary prostate tumors would be those derived from spontaneously immortalized tumor cells; unfortunately, explanted primary prostate cells survive only short-term in culture, and rarely immortalize spontaneously. Therefore, we recently have generated five immortal human prostate epithelial cell cultures derived from both the benign and malignant tissues of prostate cancer patients with telomerase, a gene that prevents cellular senescence. Examination of these cell lines for their morphologies and proliferative capacities, their abilities to grow in low serum, to respond to androgen stimulation, to grow above the agar layer, to form tumors in SCID mice, suggests that they may serve as valid, useful tools for the elucidation of early events in prostate tumorigenesis. Furthermore, the chromosome alterations observed in these immortalized cell lines expressing aspects of the malignant phenotypes imply that these cell lines accurately recapitulate the genetic composition of primary tumors. These novel in vitro models may offer unique models for the study of prostate carcinogenesis and also provide the means for testing both chemopreventive and chemotherapeutic agents.
Collapse
Affiliation(s)
- Yongpeng Gu
- Center for Prostate Disease Research, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Hertzog RG. Ancestral telomere shortening: a countdown that will increase mean life span? Med Hypotheses 2006; 67:157-60. [PMID: 16530337 DOI: 10.1016/j.mehy.2006.01.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Revised: 12/22/2005] [Accepted: 01/04/2006] [Indexed: 11/25/2022]
Abstract
Like cells, all mammals have a limited life span. Among cells there are a few exceptions (e.g., immortal cells), among mammals not, even if some of them live longer. Many in vitro and in vivo studies support the consensus that telomere length is strongly correlated with life span. At the somatic cellular level, long telomeres have been associated with longer life span. A different situation can be seen in immortal cells, such as cancer, germ and stem cells, where telomeres are maintained by telomerase, a specialized reverse transcriptase that is involved in synthesis of telomeres. Irrespective of telomere length, if telomerase is active, telomeres can be maintained at a sufficient length to ensure cell survival. To the contrary, telomeres shorten progressively with each cell division and when a critical telomere length (Hayflick limit) is reached, the cells undergo senescence and subsequently apoptosis. In mammals, those with the longest telomeres (e.g., mice) have the shortest life span. Furthermore, the shorter the mean telomere length, the longer the mean life span, as observed in humans (10-14 kpb) and bowhead-whales (undetermined telomere length), which have the longest mean life span among mammals. Over the past centuries, human average life span has increased. The hypothesis presented here suggests that this continual increase in the mean life span could be due to a decrease of mean telomere length over the last hundreds years. Actually, the life span is not directly influenced by length of telomeres, but rather by telomere length - dependent gene expression pattern. According to Greider, "rather than average telomere length, it is the shortest telomere length that makes the biggest difference to a cell". In the context of fast-growing global elderly population due to increase in life expectancy, it also seem to be an age related increase in cancer incidence. Nevertheless, extending healthy life span could depend on how good cells achieve, during the prenatal period and few years after birth, the equilibrium between telomere length and telomerase activity, as seen in germ cells. After all, I suggest that decrease in mean telomere length might result in, on the one hand, an increased life span and, on the other, a higher risk of tumorigenesis.
Collapse
Affiliation(s)
- Radu G Hertzog
- Army Center for Medical Research, CA Rosetti 37, 020012 Bucharest, Romania.
| |
Collapse
|
246
|
Xi L, Chen G, Zhou J, Xu G, Wang S, Wu P, Zhu T, Zhang A, Yang W, Xu Q, Lu Y, Ma D. Inhibition of telomerase enhances apoptosis induced by sodium butyrate via mitochondrial pathway. Apoptosis 2006; 11:789-98. [PMID: 16554963 DOI: 10.1007/s10495-006-5701-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Telomerase activation represents an early step in carcinogenesis. Increased telomerase activity in cervical cancer suggests a potential target for the development of novel therapeutic drugs. The aim of this study is to investigate the impact of telomerase activity on the biological features of HeLa cells and the possible mechanisms of enhanced apoptosis rate induced by sodium butyrate after telomerase inhibition. We introduced vectors encoding dominate negative (DN)-hTERT, wild-type (WT)-hTERT, or a control vector expressing only a drug-resistance marker into HeLa cells. Thus we assessed the biological effects of telomerase activity on telomere length, cell proliferation, chemosensitivity and radiosensitivity. In order to understand the mechanisms in which DN-hTERT enhances the apoptosis induced by sodium butyrate, we detected the release status of cytochrome c and apoptosis inducing factor (AIF) from mitochondria. Ectopic expression of DN-hTERT resulted in inhibition of telomerase activity, reduction of telomere length, decreased colony formation ability, and loss of tumorigenicity in nude mice. Moreover, DN-hTERT transfected HeLa cells with shortened telomeres were more susceptible to multiple chemotherapeutic agents and radiation. WT-hTERT transfected HeLa cells with longer telomeres exhibited resistance to radiation and chemotherapeutic agents. Our data demonstrate that elevated release level of cytochrome c and AIF from mitochondria might contribute to the enhanced apoptosis in DN-hTERT transfected HeLa cells after treatment with sodium butyrate. Inhibition of telomerase might serve as a promising adjunctive therapy combined with conventional therapy in cervical cancer.
Collapse
Affiliation(s)
- L Xi
- Cancer Biology Research Center, TongJi Hospital, TongJi Medical School, Huazhong University of Science & Technology, Wuhan, Hubei 430030, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Boiko AD, Porteous S, Razorenova OV, Krivokrysenko VI, Williams BR, Gudkov AV. A systematic search for downstream mediators of tumor suppressor function of p53 reveals a major role of BTG2 in suppression of Ras-induced transformation. Genes Dev 2006; 20:236-52. [PMID: 16418486 PMCID: PMC1356114 DOI: 10.1101/gad.1372606] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Factors that mediate p53 tumor suppressor activity remain largely unknown. In this study we describe a systematic approach to identify downstream mediators of tumor suppressor function of p53, consisting of global gene expression profiling, focused short hairpin RNA (shRNA) library creation, and functional selection of genetic elements cooperating with oncogenic Ras in cell transformation. This approach is based on our finding that repression of gene expression is a major event, occurring in response to p53 inactivation during transformation and immortalization of primary cells. Functional analysis of the subset of genes universally down-regulated in the cells that lacked functional p53 revealed BTG2 as a major downstream effector of p53-dependent proliferation arrest of mouse and human fibroblasts transduced with oncogenic Ras. shRNA-mediated knockdown of BTG2 cooperates with oncogenic Ras to transform primary mouse fibroblasts containing wild-type transcriptionally active p53. Repression of BTG2 results in up-regulation of cyclins D1 and E1 and phosphorylation of Rb and, in cooperation with other oncogenic elements, induces neoplastic transformation of primary human fibroblasts. BTG2 expression was found to be significantly reduced in a large proportion of human kidney and breast carcinomas, suggesting that BTG2 is a tumor suppressor that links p53 and Rb pathways in human tumorigenesis.
Collapse
Affiliation(s)
- Alexander D Boiko
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | | | |
Collapse
|
248
|
Zhang DH, Zhou B, Huang Y, Xu LX, Zhou JQ. The human Pif1 helicase, a potential Escherichia coli RecD homologue, inhibits telomerase activity. Nucleic Acids Res 2006; 34:1393-404. [PMID: 16522649 PMCID: PMC1390689 DOI: 10.1093/nar/gkl029] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Telomeres, the protein-DNA complexes at the ends of eukaryotic chromosomes, are essential for chromosome stability, and their maintenance is achieved by the specialized reverse transcriptase activity of telomerase or the homologous recombination pathway in most eukaryotes. Here, we identified a human helicase, hPif1 that inhibits telomerase activity. The primary sequence and biochemical analysis suggest that hPif1 is a potential homologue of Escherichia coli RecD, an ATP-dependent 5' to 3' DNA helicase. Ectopic expression of wild-type, but not the ATPase/helicase-deficient hPif1, causes telomere shortening in HT1080 cells. hPif1 reduces telomerase processivity and unwinds DNA/RNA duplex in vitro. hPif1 preferentially binds telomeric DNA in vitro and in vivo. We propose that the mechanism of hPif1's inhibition on telomerase involves unwinding of the DNA/RNA duplex formed by telomerase RNA and telomeric DNA, and RecD homologues in eukaryotes may have evolved gaining additional functions.
Collapse
Affiliation(s)
| | | | | | | | - Jin-Qiu Zhou
- To whom correspondence should be addressed. Tel: 86 21 54921078; Fax: 86 21 54921076;
| |
Collapse
|
249
|
|
250
|
Flanary BE, Streit WJ. Effects of axotomy on telomere length, telomerase activity, and protein in activated microglia. J Neurosci Res 2006; 82:160-71. [PMID: 16175576 DOI: 10.1002/jnr.20636] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The adult central nervous system (CNS) is generally thought of as a postmitotic organ. However, DNA labeling studies have shown that one major population of nonneuronal cells, called microglia, retain significant mitotic potential. Microglial cell division is prominent during acute CNS injury involving neuronal damage or death. Prior work from this laboratory has shown that purified microglia maintained in vitro with continual mitogenic stimulation exhibit telomere shortening before entering senescence. In the current study, we sought to investigate whether telomere shortening occurs in dividing microglia in vivo. For this purpose, we used a nerve injury model that is known to trigger localized microglial proliferation in a well-defined CNS region, the facial motor nucleus. Adult Sprague-Dawley rats underwent facial nerve axotomy, and facial motor nuclei were microdissected after 1, 4, 7, and 10 days. Whole tissue samples were subjected to measurements of telomere length, telomerase activity, and telomerase protein. Results revealed a tendency for all of these parameters to be increased in lesioned samples. In addition, microglial cells isolated directly from axotomized facial nuclei with fluorescence-activated cell sorting (FACS) showed increased telomerase activity relative to unoperated controls, suggesting that microglia are the primary cell type responsible for the increases observed in whole tissue samples. Overall, the results show that microglia activated by injury are capable of maintaining telomere length via telomerase during periods of high proliferation in vivo. We conclude that molecular mechanisms pertaining to telomere maintenance are active in the injured CNS.
Collapse
Affiliation(s)
- Barry E Flanary
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | | |
Collapse
|