501
|
Rossi A, Russo G, Puca A, La Montagna R, Caputo M, Mattioli E, Lopez M, Giordano A, Pentimalli F. The antiretroviral nucleoside analogue Abacavir reduces cell growth and promotes differentiation of human medulloblastoma cells. Int J Cancer 2009; 125:235-43. [PMID: 19358275 DOI: 10.1002/ijc.24331] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Abacavir is one of the most efficacious nucleoside analogues, with a well-characterized inhibitory activity on reverse transcriptase enzymes of retroviral origin, and has been clinically approved for the treatment of AIDS. Recently, Abacavir has been shown to inhibit also the human telomerase activity. Telomerase activity seems to be required in essentially all tumours for the immortalization of a subset of cells, including cancer stem cells. In fact, many cancer cells are dependent on telomerase for their continued replication and therefore telomerase is an attractive target for cancer therapy. Telomerase expression is upregulated in primary primitive neuroectodermal tumours and in the majority of medulloblastomas suggesting that its activation is associated with the development of these diseases. Therefore, we decided to test Abacavir activity on human medulloblastoma cell lines with high telomerase activity. We report that exposure to Abacavir induces a dose-dependent decrease in the proliferation rate of medulloblastoma cells. This is associated with a cell accumulation in the G(2)/M phase of the cell cycle in the Daoy cell line, and with increased cell death in the D283-MED cell line, and is likely to be dependent on the inhibition of telomerase activity. Interestingly, both cell lines showed features of senescence after Abacavir treatment. Moreover, after Abacavir exposure we detected, by immunofluorescence staining, increased protein expression of the glial marker glial fibrillary acidic protein and the neuronal marker synaptophysin in both medulloblastoma cell lines. In conclusion, our results suggest that Abacavir reduces proliferation and induces differentiation of human medulloblastoma cells through the downregulation of telomerase activity. Thus, using Abacavir, alone or in combination with current therapies, might be an effective therapeutic strategy for the treatment of medulloblastoma.
Collapse
Affiliation(s)
- Alessandra Rossi
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia PA
| | | | | | | | | | | | | | | | | |
Collapse
|
502
|
Gallardo F, Chartrand P. [Telomerase biogenesis: a journey to the end of chromosomes]. Med Sci (Paris) 2009; 25:232-3. [PMID: 19361383 DOI: 10.1051/medsci/2009253232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
503
|
Marian CO, Shay JW. Prostate tumor-initiating cells: A new target for telomerase inhibition therapy? Biochim Biophys Acta Mol Basis Dis 2009; 1792:289-96. [DOI: 10.1016/j.bbadis.2009.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 02/20/2009] [Accepted: 02/23/2009] [Indexed: 10/21/2022]
|
504
|
Lin T, Jones RJ, Matsui W. Cancer stem cells: relevance to SCT. Bone Marrow Transplant 2009; 43:517-23. [PMID: 19234507 PMCID: PMC2950943 DOI: 10.1038/bmt.2009.19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2008] [Accepted: 12/31/2008] [Indexed: 12/26/2022]
Abstract
The cancer stem cell (CSC) hypothesis suggests that clonogenic growth potential within an individual tumor is restricted to a specific and phenotypically defined cell population. Evidence for CSC in human tumors initially arose from studies of AML, but functionally similar cell populations have been identified in an increasing number of malignancies. Despite these findings, controversy surrounds the CSC hypothesis, especially the generalization that clonogenic tumor cells are rare. Nevertheless, efforts to define the cellular processes regulating self-renewal and resistance to anticancer therapeutics, two of the major properties ascribed to CSC, are likely to provide useful insights into tumor biology as a whole. BMT has been at the forefront of clinically translating basic stem cell concepts starting with the original hypothesis that normal hematopoietic precursors could rescue patients from myeloablative doses of radiation or chemotherapy. Even today, a better understanding of CSC may enhance ongoing efforts to induce specific and effective anti-tumor immune responses in both the allogeneic and autologous setting. It is also likely that new clinical research approaches will be required to accurately evaluate novel CSC-targeting strategies. Owing to the capacity to produce remissions in most diseases, SCT may provide the ideal clinical platform to carry out these investigations by studying the ability of anti-CSC agents to prolong relapse free and overall survival.
Collapse
Affiliation(s)
- T Lin
- Section of Hematology and Oncology, Department of Internal Medicine, LSU School of Medicine, New Orleans, LA, USA
| | - RJ Jones
- Division of Hematologic Malignancies, Department of Oncology, Johns Hopkins University School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - W Matsui
- Division of Hematologic Malignancies, Department of Oncology, Johns Hopkins University School of Medicine and The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| |
Collapse
|
505
|
Recent insights into the molecular mechanisms involved in aging and the malignant transformation of adult stem/progenitor cells and their therapeutic implications. Ageing Res Rev 2009; 8:94-112. [PMID: 19114129 DOI: 10.1016/j.arr.2008.12.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 12/04/2008] [Indexed: 02/07/2023]
Abstract
Recent advancements in tissue-resident adult stem/progenitor cell research have revealed that enhanced telomere attrition, oxidative stress, ultraviolet radiation exposure and oncogenic events leading to severe DNA damages and genomic instability may occur in these immature and regenerative cells during chronological aging. Particularly, the alterations in key signaling components controlling their self-renewal capacity and an up-regulation of tumor suppressor gene products such as p16(INK4A), p19(ARF), ataxia-telangiectasia mutated (ATM) kinase, p53 and/or the forkhead box O (FOXOs) family of transcription factors may result in their dysfunctions, growth arrest and senescence or apoptotic death during the aging process. These molecular events may culminate in a progressive decline in the regenerative functions and the number of tissue-resident adult stem/progenitor cells, and age-related disease development. Conversely, the telomerase re-activation and accumulation of numerous genetic and/or epigenetic alterations in adult stem/progenitor cells with advancing age may result in their immortalization and malignant transformation into highly leukemic or tumorigenic cancer-initiating cells and cancer initiation. Therefore, the cell-replacement and gene therapies and molecular targeting of aged and dysfunctional adult stem/progenitor cells including their malignant counterpart, cancer-initiating cells, hold great promise for treating and even curing diverse devastating human diseases. These diseases include premature aging diseases, hematopoietic, cardiovascular, musculoskeletal, pulmonary, ocular, urogenital, neurodegenerative and skin disorders and aggressive and recurrent cancers.
Collapse
|
506
|
Abstract
Cancer is a complex collection of distinct genetic diseases united by common hallmarks. Here, we expand upon the classic hallmarks to include the stress phenotypes of tumorigenesis. We describe a conceptual framework of how oncogene and non-oncogene addictions contribute to these hallmarks and how they can be exploited through stress sensitization and stress overload to selectively kill cancer cells. In particular, we present evidence for a large class of non-oncogenes that are essential for cancer cell survival and present attractive drug targets. Finally, we discuss the path ahead to therapeutic discovery and provide theoretical considerations for combining orthogonal cancer therapies.
Collapse
Affiliation(s)
- Ji Luo
- Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
507
|
Cinatl J, Nevels M, Paulus C, Michaelis M. Activation of telomerase in glioma cells by human cytomegalovirus: another piece of the puzzle. J Natl Cancer Inst 2009; 101:441-3. [PMID: 19318630 DOI: 10.1093/jnci/djp047] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
508
|
Proctor A, Brownhill SC, Burchill SA. The promise of telomere length, telomerase activity and its regulation in the translocation-dependent cancer ESFT; clinical challenges and utility. Biochim Biophys Acta Mol Basis Dis 2009; 1792:260-74. [PMID: 19264125 DOI: 10.1016/j.bbadis.2009.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 02/19/2009] [Accepted: 02/20/2009] [Indexed: 01/12/2023]
Abstract
The Ewing's sarcoma family of tumours (ESFT) are diagnosed by EWS-ETS gene translocations. The resulting fusion proteins play a role in both the initiation and maintenance of these solid aggressive malignant tumours, suppressing cellular senescence and increasing cell proliferation and survival. EWS-ETS fusion proteins have altered transcriptional activity, inducing expression of a number of different target genes including telomerase. Up-regulation of hTERT is most likely responsible for the high levels of telomerase activity in primary ESFT, although telomerase activity and expression of hTERT are not predictive of outcome. However levels of telomerase activity in peripheral blood may be useful to monitor response to some therapeutics. Despite high levels of telomerase activity, telomeres in ESFT are frequently shorter than those of matched normal cells. Uncertainty about the role that telomerase and regulators of its activity play in the maintenance of telomere length in normal and cancer cells, and lack of studies examining the relationship between telomerase activity, regulators of its activity and their clinical significance in patient samples have limited their introduction into clinical practice. Studies in clinical samples using standardised assays are critical to establish how telomerase and regulators of its activity might best be exploited for patient benefit.
Collapse
Affiliation(s)
- Andrew Proctor
- Cancer Research UK Clinical Centre, Leeds Institute of Molecular Medicine, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | | | | |
Collapse
|
509
|
Cookson J, Laughton C. The levels of telomere-binding proteins in human tumours and therapeutic implications. Eur J Cancer 2009; 45:536-50. [DOI: 10.1016/j.ejca.2008.11.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 11/10/2008] [Indexed: 12/16/2022]
|
510
|
Blagoev KB. Cell proliferation in the presence of telomerase. PLoS One 2009; 4:e4622. [PMID: 19247450 PMCID: PMC2644786 DOI: 10.1371/journal.pone.0004622] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 01/07/2009] [Indexed: 12/01/2022] Open
Abstract
Background Telomerase, which is active early in development and later in stem and germline cells, is also active in the majority of human cancers. One of the known functions of telomerase is to extend the ends of linear chromosomes, countering their gradual shortening at each cell division due to the end replication problem and postreplication processing. Telomerase concentration levels vary between different cell types as well as between different tumors. In addition variable telomerase concentrations will exist in different cells in the same tumor when telomerase inhibitors are used, because of limitations of drug delivery in tissue. Telomerase extends short telomeres more frequently than long telomeres and the relation between the extension frequency and the telomere length is nonlinear. Methodolgy/Principal Findings Here, the biological data of the nonlinear telomerase-telomere dynamics is incorporated in a mathematical theory to relate the proliferative potential of a cell to the telomerase concentration in that cell. The main result of the paper is that the proliferative capacity of a cell grows exponentially with the telomerase concentration. Conclusions/Significance The theory presented here suggests that long term telomerase inhibition in every cancer progenitor or cancer stem cell is needed for successful telomere targeted cancer treatment. This theory also can be used to plan and asses the results of clinical trials targeting telomerase.
Collapse
Affiliation(s)
- Krastan B Blagoev
- National Science Foundation, Arlington, Virginia, United States of America.
| |
Collapse
|
511
|
Dikmen ZG, Ozgurtas T, Gryaznov SM, Herbert BS. Targeting critical steps of cancer metastasis and recurrence using telomerase template antagonists. Biochim Biophys Acta Mol Basis Dis 2009; 1792:240-7. [PMID: 19419695 DOI: 10.1016/j.bbadis.2009.01.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 01/29/2009] [Accepted: 01/30/2009] [Indexed: 01/11/2023]
Abstract
Metastasis, tumor relapse, and drug resistance remain major obstacles in the treatment of cancer. Therefore, more research on the mechanisms of these processes in disease is warranted for improved treatment options. Recent evidence suggests that the capability to sustain tumor growth and metastasis resides in a subpopulation of cells, termed cancer stem cells or tumor-initiating cells. Continuous proliferation and self-renewal are characteristics of stem/progenitor cells. Telomerase and the maintenance of telomeres are key players in the ability of stem and cancer cells to bypass senescence and be immortal. Therefore, telomerase inhibitors have the therapeutic potential for reducing tumor relapse by targeting cancer stem cells and other processes involved in metastasis. Herein we review the role of telomerase in the immortal phenotype of cancer and cancer stem cells, targeting telomerase in cancer, and discuss other opportunities for telomerase inhibitors to target critical steps in cancer metastasis and recurrence.
Collapse
Affiliation(s)
- Z Gunnur Dikmen
- Department of Biochemistry, University of Hacettepe, 06100 Sihhiye, Ankara, Turkey.
| | | | | | | |
Collapse
|
512
|
Folini M, Gandellini P, Zaffaroni N. Targeting the telosome: therapeutic implications. Biochim Biophys Acta Mol Basis Dis 2009; 1792:309-16. [PMID: 19419699 DOI: 10.1016/j.bbadis.2009.01.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 01/28/2009] [Accepted: 01/30/2009] [Indexed: 12/12/2022]
Abstract
Since telomere integrity is required to guarantee the unlimited replicative potential of cancer cells, telomerase, the enzyme responsible for telomere length maintenance in most human tumors, and lately also telomeres themselves have become extremely attractive targets for new anticancer interventions. At the current status of knowledge, it is still not possible to define the best therapeutic target between telomerase and telomeres. It is noteworthy that interfering with telomeres, through direct targeting of telomeric DNA or proteins involved in the telosome complex, could negatively affect the proliferative potential not only of tumors expressing telomerase activity but also of those that maintain their telomeres through alternative lengthening or still unknown mechanisms. This review presents the different therapeutic approaches proposed thus far and developed in preclinical tumor models and discusses the perspectives for their use in the clinical setting.
Collapse
Affiliation(s)
- Marco Folini
- Department of Experimental Oncology and Laboratories, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | | | | |
Collapse
|
513
|
Telomere-dependent and telomere-independent origins of endogenous DNA damage in tumor cells. Aging (Albany NY) 2009; 1:212-8. [PMID: 20157510 PMCID: PMC2806003 DOI: 10.18632/aging.100019] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 02/02/2009] [Indexed: 11/25/2022]
Abstract
Human tumors and
cultured cells contain elevated levels of endogenous DNA damage resulting
from telomere dysfunction, replication and transcription errors, reactive
oxygen species, and genome instability. However, the contribution of
telomere-associated versus telomere-independent endogenous DNA lesions to
this damage has never been examined. In this study, we characterized the
relative amounts of these two types of DNA damage in five tumor cell lines
by noting whether γ-H2AX
foci, generally considered to mark DNA double-strand breaks (DSBs), were on
chromosome arms or at chromosome ends. We found that while the numbers of
non-telomeric DSBs were remarkably similar in these cultures, considerable
variation was detected in the level of telomeric damage. The distinct
heterogeneity in the numbers of γ-H2AX foci in these tumor cell lines
was found to be due to foci associated with uncapped telomeres, and the
amount of total telomeric damage also appeared to inversely correlate with
the telomerase activity present in these cells. These results indicate that
damaged telomeres are the major factor accounting for the variability in
the amount of DNA DSB damage in tumor cells. This characterization of DNA
damage in tumor cells helps clarify the contribution of non-telomeric DSBs
and damaged telomeres to major genomic alterations.
Collapse
|
514
|
Abstract
Protected telomeres ensure normal chromosomal segregation during mitosis but at the same time can endow genetically abnormal cancer cells with immortality. Telomerase has a pivotal role in telomere protection, both in normal and cancer cells. Understanding the functional interplay between telomere shortening and telomerase expression in cancer cells is of critical importance to elucidating the precise mechanisms by which these cells are able to bypass telomere crisis and become immortal.
Collapse
|
515
|
Drewe WC, Nanjunda R, Gunaratnam M, Beltran M, Parkinson GN, Reszka AP, Wilson WD, Neidle S. Rational design of substituted diarylureas: a scaffold for binding to G-quadruplex motifs. J Med Chem 2009; 51:7751-67. [PMID: 19053833 DOI: 10.1021/jm801245v] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The design and synthesis of a series of urea-based nonpolycyclic aromatic ligands with alkylaminoanilino side chains as telomeric and genomic G-quadruplex DNA interacting agents are described. Their interactions with quadruplexes have been examined by means of fluorescent resonance energy transfer melting, circular dichroism, and surface plasmon resonance-based assays. These validate the design concept for such urea-based ligands and also show that they have significant selectivity over duplex DNA, as well as for particular G-quadruplexes. The ligand-quadruplex complexes were investigated by computational molecular modeling, providing further information on structure-activity relationships. Preliminary biological studies using short-term cell growth inhibition assays show that some of the ligands have cancer cell selectivity, although they appear to have low potency for intracellular telomeric G-quadruplex structures, suggesting that their cellular targets may be other, possibly oncogene-related quadruplexes.
Collapse
Affiliation(s)
- William C Drewe
- The Cancer Research UK Biomolecular Structure Group, The School of Pharmacy, University of London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | | | | | | | | | | | | | | |
Collapse
|
516
|
Mennuni C, Ugel S, Mori F, Cipriani B, Iezzi M, Pannellini T, Lazzaro D, Ciliberto G, La Monica N, Zanovello P, Bronte V, Scarselli E. Preventive vaccination with telomerase controls tumor growth in genetically engineered and carcinogen-induced mouse models of cancer. Cancer Res 2009; 68:9865-74. [PMID: 19047167 DOI: 10.1158/0008-5472.can-08-1603] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The telomerase reverse transcriptase, TERT, is an attractive target for human cancer vaccination because its expression is reactivated in a conspicuous fraction of human tumors. Genetic vaccination with murine telomerase (mTERT) could break immune tolerance in different mouse strains and resulted in the induction of both CD4+ and CD8+ telomerase-specific T cells. The mTERT-derived immunodominant epitopes recognized by CD8+ T cells were further defined in these mouse strains and used to track immune responses. Antitumor efficacy of telomerase-based vaccination was investigated in two cancer models closely resembling human diseases: the TRAMP transgenic mice for prostate cancer and a carcinogen-induced model for colon cancer. TERT overexpression in tumor lesions was shown in both models by immunohistochemistry, thus reinforcing the similarity of these tumors to their human counterparts. Repeated immunizations with mTERT-encoding DNA resulted in a significant delay of tumor formation and progression in both the prostate cancer and the colon cancer models. Moreover, evaluation of the intratumoral infiltrate revealed the presence of telomerase-specific T cells in vaccinated mice. The safety of vaccination was confirmed by the absence of histomorphologic changes on postnecropsy analysis of several organs and lack of adverse effects on blood cell counts. These results indicate that TERT vaccination can elicit antigen-specific immunosurveillance and imply this antigen as a potential candidate for preventive cancer vaccines.
Collapse
|
517
|
Lança V, Zee RY, Rivera A, Romero JR. Quantitative telomerase activity in circulating human leukocytes: utility of real-time telomeric repeats amplification protocol (RQ-TRAP) in a clinical/epidemiological setting. Clin Chem Lab Med 2009; 47:870-3. [DOI: 10.1515/cclm.2009.188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
518
|
Marie-Egyptienne DT, Brault ME, Nimmo GAM, Londoño-Vallejo JA, Autexier C. Growth defects in mouse telomerase RNA-deficient cells expressing a template-mutated mouse telomerase RNA. Cancer Lett 2008; 275:266-76. [PMID: 19056167 DOI: 10.1016/j.canlet.2008.10.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 10/15/2008] [Accepted: 10/17/2008] [Indexed: 01/03/2023]
Abstract
Cellular viability requires telomere maintenance, which, in mammals, is mainly mediated by the reverse transcriptase telomerase. Telomerase core components are a catalytic subunit TERT and an RNA subunit TR (hTR in humans, mTR in mouse) that carries the template to generate telomeres de novo. Telomere dysfunction can lead to senescence or apoptosis and impairs the continued growth of immortal cancerous cell lines. The introduction of a template-mutated hTR in telomerase-positive and telomerase-negative human cell lines results in dramatic growth defects. No study has addressed the consequences of expressing a template-mutated mTR in mouse immortal cell lines. Therefore, we analyzed the effects of long-term expression of a template-mutated mTR in the telomerase-positive and telomerase-negative murine cell lines CB17 and DKO301, respectively. Whereas the CB17 clones expressing the template-mutated mTR did not demonstrate any growth impairment, many of the DKO301 clones expressing the template-mutated mTR underwent growth and cell cycle defects and eventual cell death. These results suggest that in the absence of wild-type telomerase, the expression of the template-mutated mTR likely perturbs telomere function, leading to decreased cellular viability. Furthermore, whereas the expression of template-mutated hTR in telomerase-negative human cell lines leads to immediate cellular toxicity, the expression of the template-mutated mTR in the telomerase-negative mouse cell line did not.
Collapse
|
519
|
Mimeault M, Batra SK. Recent advances in the development of novel anti-cancer drugs targeting cancer stem/progenitor cells. Drug Dev Res 2008. [DOI: 10.1002/ddr.20273] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
520
|
Wong VCH, Ma J, Hawkins CE. Telomerase inhibition induces acute ATM-dependent growth arrest in human astrocytomas. Cancer Lett 2008; 274:151-9. [PMID: 18945545 DOI: 10.1016/j.canlet.2008.09.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 07/15/2008] [Accepted: 09/09/2008] [Indexed: 11/18/2022]
Abstract
The purpose of the study was to examine the degree of hTERT, the catalytic subunit of telomerase, expression in paediatric high-grade astrocytoma and to explore the potential of telomerase inhibition as a therapy for these tumours. hTERT was expressed at high levels in 36 of 44 paediatric astrocytomas. Telomerase inhibition induced acute DNA damage and ATM-pathway-dependent G2/M cell cycle arrest in astrocytomas in vitro, both occurring prior to telomere shortening itself. Our data suggest that telomerase inhibition could be a useful adjuvant therapy for high-grade astrocytomas, potentially inducing tumour growth arrest following short-term treatment.
Collapse
Affiliation(s)
- Vincent C H Wong
- Division of Pathology, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, 555 University Avenue, Toronto, Ont., Canada M5G 1X8
| | | | | |
Collapse
|
521
|
Niu H, Leung DTM, Ma CH, Law ECY, Tam FCH, Lim PL. Cells that produce deleterious autoreactive antibodies are vulnerable to suicide. THE JOURNAL OF IMMUNOLOGY 2008; 181:2246-57. [PMID: 18641365 DOI: 10.4049/jimmunol.181.3.2246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
It is puzzling how autoreactive B cells that escape self-tolerance mechanisms manage to produce Abs that target vital cellular processes without succumbing themselves to the potentially deleterious effects of these proteins. We report that censorship indeed exists at this level: when the Ab synthesis in the cell is up-regulated in IL-6-enriched environments (e.g., adjuvant-primed mouse peritoneum), the cell dies of the increased intracellular binding between the Ab and the cellular autoantigen. In the case in which telomerase is the autoantigen, mouse hybridoma cells synthesizing such an autoantibody, which appeared to grow well in culture, could not grow in syngeneic BALB/c mice to form ascites, but grew nevertheless in athymic siblings. Culture experiments demonstrated that peritoneal cell-derived IL-6 (and accessory factors) affected the growth and functions of the hybridoma cells, including the induction of mitochondria-based apoptosis. Electron microscopy revealed an abundance of Abs in the nuclear chromatin of IL-6-stimulated cells, presumably piggy-backed there by telomerase from the cytosol. This nuclear presence was confirmed by light microscopy analysis of isolated nuclei. In two other cases, hybridoma cells synthesizing an autoantibody to GTP or osteopontin also showed similar growth inhibition in vivo. In all cases, Ab function was crucial to the demise of the cells. Thus, autoreactive cells, which synthesize autoantibodies to certain intracellular Ags, live delicately between life and death depending on the cytokine microenvironment. Paradoxically, IL-6, which is normally growth-potentiating for B cells, is proapoptotic for these cells. The findings reveal potential strategies and targets for immunotherapy.
Collapse
Affiliation(s)
- Haitao Niu
- Clinical Immunology Unit, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong
| | | | | | | | | | | |
Collapse
|
522
|
Controversial issue: is it safe to employ mesenchymal stem cells in cell-based therapies? Exp Gerontol 2008; 43:1018-23. [PMID: 18694815 DOI: 10.1016/j.exger.2008.07.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 07/11/2008] [Accepted: 07/15/2008] [Indexed: 12/18/2022]
Abstract
The prospective clinical use of multipotent mesenchymal stromal stem cells (MSC) holds enormous promise for the treatment of a large number of degenerative and age-related diseases. However, the challenges and risks for cell-based therapies are multifaceted. The risks for patients receiving stem cells, which have been expanded in vitro in the presence of xenogenic compounds, can hardly be anticipated and methods for the culture and manipulation of "safe" MSC ex vivo are being investigated. During in vitro expansion, stem cells experience a long replicative history and are thus subject to damage from intracellular and extracellular influences. While murine MSC are prone to cellular transformation in culture, human MSC do not transform. One reason for this striking difference is that during long-term culture, human MSC finally become replicatively senescent. In consequence, this greatly restricts their proliferation and differentiation efficiency. It however also limits the yield of sufficient numbers of cells needed for therapy. Another issue is to eliminate contamination of expanding cells with serum-bound pathogenic agents in order to reduce the risks for infection. A recent technical advancement, which applies human serum platelet lysates as an alternative source for growth factors and essential supplements, allows the unimpaired proliferation of MSC in the absence of animal sera. Here, we present an update regarding cellular senescence of MSC and recent insights concerning potential risks associated with their clinical use.
Collapse
|
523
|
Abstract
Long-lived organisms such as humans have evolved several intrinsic tumour suppressor mechanisms to combat the slew of oncogenic somatic mutations that constantly arise in proliferating stem-cell compartments. One of these anticancer barriers is the telomere, a specialized nucleoprotein complex that caps the ends of eukaryotic chromosome. Impaired telomere function activates the canonical DNA damage response pathway that engages p53 to initiate apoptosis or replicative senescence. Here, we discuss how p53-dependent senescence induced by dysfunctional telomeres may be as potent as apoptosis in suppressing tumorigenesis in vivo.
Collapse
Affiliation(s)
- Yibin Deng
- Department of Cancer Genetics, Box 1010, The UT M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Suzanne Chan
- Department of Cancer Genetics, Box 1010, The UT M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
| | - Sandy Chang
- Department of Cancer Genetics, Box 1010, The UT M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
- Department of Hematopathology, The UT M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030 USA
- Correspondence:
| |
Collapse
|
524
|
Hombach-Klonisch S, Paranjothy T, Wiechec E, Pocar P, Mustafa T, Seifert A, Zahl C, Gerlach KL, Biermann K, Steger K, Hoang-Vu C, Schulze-Osthoff K, Los M. Cancer stem cells as targets for cancer therapy: selected cancers as examples. Arch Immunol Ther Exp (Warsz) 2008; 56:165-80. [PMID: 18512024 DOI: 10.1007/s00005-008-0023-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 05/17/2008] [Indexed: 12/16/2022]
Abstract
It is becoming increasingly evident that cancer constitutes a group of diseases involving altered stem-cell maturation/differentiation and the disturbance of regenerative processes. The observed malignant transformation is merely a symptom of normal differentiation processes gone astray rather than the primary event. This review focuses on the role of cancer stem cells (CSCs) in three common but also relatively under-investigated cancers: head and neck, ovarian, and testicular cancer. For didactic purpose, the physiology of stem cells is first introduced using hematopoietic and mesenchymal stem cells as examples. This is followed by a discussion of the (possible) role of CSCs in head and neck, ovarian, and testicular cancer. Aside from basic information about the pathophysiology of these cancers, current research results focused on the discovery of molecular markers specific to these cancers are also discussed. The last part of the review is largely dedicated to signaling pathways active within various normal and CSC types (e.g. Nanog, Nestin, Notch1, Notch2, Oct3 and 4, Wnt). Different elements of these pathways are also discussed in the context of therapeutic opportunities for the development of targeted therapies aimed at CSCs. Finally, alternative targeted anticancer therapies arising from recently identified molecules with cancer-(semi-)selective capabilities (e.g. apoptin, Brevinin-2R) are considered.
Collapse
|