151
|
Abstract
ABSTRACTThe main objective of this paper is to offer a detailed analysis of mortality change in the United Kingdom at the beginning the 21st century. Starting from an exploration of 20th century mortality trends, focusing in particular on the 1990s, underlying forces driving trends in longevity are discussed. These include the ‘cohort effect’ and the ‘ageing of mortality improvement’. International mortality statistics and trends are also analysed. The pace of medical advances is discussed, with specific focus on research into the ageing process and a potential treatment for cardiovascular disease. The paper also discusses the potential threat from infectious diseases.The analysis of underlying trends suggests that life expectancy in retirement in the U.K. is likely to increase rapidly in the early part of the 21st century. Some scientists are also claiming that we will be seeing the fruits of anti-ageing research within just a few decades.A core theme of the paper is that future projections should be grounded in as good an understanding of the past as possible. Different methods for projecting future rates of mortality are discussed, and it is noted that emphasis should be placed on the uncertainty surrounding projections.The financial impact of using different assumptions for future mortality is explored. Significant differences in the cost of an annuity or pension arise from the use of the various projection bases.Life assurance companies have already declared significant losses as a result of strengthening reserves on annuity portfolios. Taken together, future increases in life expectancy, increasing awareness of the risk of providing longevity insurance, changes in legislation and shortages in market capacity and capital, may well lead to worsening annuity rates.It is difficult to assess the precise impact of future changes in life expectancy on final salary pension schemes. There is a lack of readily available information on the mortality assumptions being used in practice. It is therefore suggested that more disclosure in this area would be helpful. Employers sponsoring final salary schemes are making promises to their employees that extend up to 70 or 80 years into the future. Actuaries should be clear in spelling out to employers and trustees the nature of the risks behind the promises they are making. Future scheme design should reflect the possibility of substantial increases in life expectancy.An over-riding implication of the anticipated increases in life expectancy is that people will remain in work for longer in the future. The age at which people retire will inevitably have to increase, and this trend will necessarily drive changes in all aspects of our society. As actuaries we have a vital role in helping to inform the wider debate.
Collapse
|
152
|
Rao F, Wang T, Li M, Li Z, Hong N, Zhao H, Yan Y, Lu W, Chen T, Wang W, Lim M, Yuan Y, Liu L, Zeng L, Wei Q, Guan G, Li C, Hong Y. Medaka tert produces multiple variants with differential expression during differentiation in vitro and in vivo. Int J Biol Sci 2011; 7:426-39. [PMID: 21547060 PMCID: PMC3088285 DOI: 10.7150/ijbs.7.426] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2011] [Accepted: 04/01/2011] [Indexed: 12/31/2022] Open
Abstract
Embryonic stem (ES) cells have immortality for self-renewal and pluripotency. Differentiated human cells undergo replicative senescence. In human, the telomerase reverse transcriptase (Tert), namely the catalytic subunit of telomerase, exhibits differential expression to regulate telomerase activity governing cellular immortality or senescence, and telomerase activity or tert expression is a routine marker of pluripotent ES cells. Here we have identified the medaka tert gene and determined its expression and telomerase activity in vivo and in vitro. We found that the medaka tert locus produces five variants called terta to terte encoding isoforms TertA to TertE. The longest TertA consists of 1090 amino acid residues and displays a maximum of 34% identity to the human TERT and all the signature motifs of the Tert family. TertB to TertE are novel isoforms and have considerable truncation due to alternative splicing. The terta RNA is ubiquitous in embryos, adult tissues and cell lines, and accompanies ubiquitous telomerase activity in vivo and in vitro as revealed by TRAP assays. The tertb RNA was restricted to the testis, absent in embryos before gastrulation and barely detectable in various cell lines The tertc transcript was absent in undifferentiated ES cells but became evident upon ES cell differentiation, in vivo it was barely detectable in early embryos and became evident when embryogenesis proceeds. Therefore, ubiquitous terta expression correlates with ubiquitous telomerase activity in medaka, and expression of other tert variants appears to delineate cell differentiation in vitro and in vivo.
Collapse
Affiliation(s)
- Feng Rao
- Department of Biological Sciences, National University of Singapore, 10 Kent Ridge Crescent, Singapore 119260
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Majerská J, Sýkorová E, Fajkus J. Non-telomeric activities of telomerase. MOLECULAR BIOSYSTEMS 2011; 7:1013-1023. [PMID: 21283914 DOI: 10.1039/c0mb00268b] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Recent results suggest that telomerase is involved in many more cellular processes than merely telomere elongation. These include telomere-independent anti-apoptotic, cytoprotective and pro-proliferative effects of telomerase or protection of mitochondrial DNA against oxidative stress. Telomerase also participates in DNA repair and its essential subunits, hTR and hTERT, are able to modulate independently the cell's response to DNA damage. Recent high throughput analyses of gene expression showed that hTERT expression modulates expression of about 300 genes, including genes involved in the regulation of cell cycle progression, proliferation and differentiation. Besides the well-known telomerase catalytic activity of RNA-dependent DNA polymerase, its RNA-dependent RNA polymerase activity was recently described in association with the RNA subunit of mitochondrial RNA processing endoribonuclease, thus suggesting involvement of telomerase in RNA interference processes. These recent discoveries open novel possibilities and entirely unexpected research perspectives, branching off from the mainstream telomere and telomerase research.
Collapse
Affiliation(s)
- Jana Majerská
- Department of Functional Genomics and Proteomics, Faculty of Science, Masaryk University and Central European Institute of Technology, Masaryk University, Kamenice 5, CZ-62500 Brno, Czech Republic.
| | | | | |
Collapse
|
154
|
Abstract
The PinX1 protein inhibits telomerase, an enzyme that lengthens telomeres - the structures that protect the ends of chromosomes. Loss of PinX1 leads to increased telomere length along with defects in chromosome dynamics. In this issue of the JCI, Zhou et al. present novel evidence from human tumors and mouse models indicating that PinX1 is a clinically significant tumor suppressor. Importantly, the genome-destabilizing effects of PinX1 loss appear to depend on telomerase activity, raising new models and questions for how telomeres and telomerase contribute to the development of cancer.
Collapse
Affiliation(s)
- F Brad Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
155
|
Stadler G, Chen JC, Wagner K, Robin JD, Shay JW, Emerson CP, Wright WE. Establishment of clonal myogenic cell lines from severely affected dystrophic muscles - CDK4 maintains the myogenic population. Skelet Muscle 2011; 1:12. [PMID: 21798090 PMCID: PMC3156635 DOI: 10.1186/2044-5040-1-12] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 03/08/2011] [Indexed: 12/30/2022] Open
Abstract
Background A hallmark of muscular dystrophies is the replacement of muscle by connective tissue. Muscle biopsies from patients severely affected with facioscapulohumeral muscular dystrophy (FSHD) may contain few myogenic cells. Because the chromosomal contraction at 4q35 linked to FSHD is thought to cause a defect within myogenic cells, it is important to study this particular cell type, rather than the fibroblasts and adipocytes of the endomysial fibrosis, to understand the mechanism leading to myopathy. Results We present a protocol to establish clonal myogenic cell lines from even severely dystrophic muscle that has been replaced mostly by fat, using overexpression of CDK4 and the catalytic component of telomerase (human telomerase reverse transcriptase; hTERT), and a subsequent cloning step. hTERT is necessary to compensate for telomere loss during in vitro cultivation, while CDK4 prevents a telomere-independent growth arrest affecting CD56+ myogenic cells, but not their CD56- counterpart, in vitro. Conclusions These immortal cell lines are valuable tools to reproducibly study the effect of the FSHD mutation within myoblasts isolated from muscles that have been severely affected by the disease, without the confounding influence of variable amounts of contaminating connective-tissue cells.
Collapse
Affiliation(s)
- Guido Stadler
- Department of Cell Biology, UT Southwestern Medical Center at Dallas, Dallas, TX, USA
| | | | | | | | | | | | | |
Collapse
|
156
|
Tabach Y, Kogan-Sakin I, Buganim Y, Solomon H, Goldfinger N, Hovland R, Ke XS, Oyan AM, Kalland KH, Rotter V, Domany E. Amplification of the 20q chromosomal arm occurs early in tumorigenic transformation and may initiate cancer. PLoS One 2011; 6:e14632. [PMID: 21297939 PMCID: PMC3031497 DOI: 10.1371/journal.pone.0014632] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 12/03/2010] [Indexed: 11/18/2022] Open
Abstract
Duplication of chromosomal arm 20q occurs in prostate, cervical, colon, gastric, bladder, melanoma, pancreas and breast cancer, suggesting that 20q amplification may play a causal role in tumorigenesis. According to an alternative view, chromosomal imbalance is mainly a common side effect of cancer progression. To test whether a specific genomic aberration might serve as a cancer initiating event, we established an in vitro system that models the evolutionary process of early stages of prostate tumor formation; normal prostate cells were immortalized by the over-expression of human telomerase catalytic subunit hTERT, and cultured for 650 days till several transformation hallmarks were observed. Gene expression patterns were measured and chromosomal aberrations were monitored by spectral karyotype analysis at different times. Several chromosomal aberrations, in particular duplication of chromosomal arm 20q, occurred early in the process and were fixed in the cell populations, while other aberrations became extinct shortly after their appearance. A wide range of bioinformatic tools, applied to our data and to data from several cancer databases, revealed that spontaneous 20q amplification can promote cancer initiation. Our computational model suggests that 20q amplification induced deregulation of several specific cancer-related pathways including the MAPK pathway, the p53 pathway and Polycomb group factors. In addition, activation of Myc, AML, B-Catenin and the ETS family transcription factors was identified as an important step in cancer development driven by 20q amplification. Finally we identified 13 "cancer initiating genes", located on 20q13, which were significantly over-expressed in many tumors, with expression levels correlated with tumor grade and outcome suggesting that these genes induce the malignant process upon 20q amplification.
Collapse
Affiliation(s)
- Yuval Tabach
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ira Kogan-Sakin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Buganim
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hilla Solomon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Naomi Goldfinger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Randi Hovland
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Xi-Song Ke
- The Gade Institute, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Anne M. Oyan
- The Gade Institute, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Karl-H. Kalland
- The Gade Institute, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eytan Domany
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
157
|
Senescence as a modulator of oral squamous cell carcinoma development. Oral Oncol 2010; 46:840-53. [DOI: 10.1016/j.oraloncology.2009.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 09/24/2009] [Accepted: 09/24/2009] [Indexed: 12/25/2022]
|
158
|
New culture technique of human eliminable feeder-assisted target cell sheet production. Biochem Biophys Res Commun 2010; 399:373-8. [PMID: 20659424 DOI: 10.1016/j.bbrc.2010.07.079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 07/22/2010] [Indexed: 11/21/2022]
Abstract
Cultured cell sheets for transplantation generally have been co-cultured with animal feeder cells, which carry risks because of different species and results in non-contact culture between the feeder and target cells. We developed a new technique to produce human eliminable feeder-assisted target cell sheets by novel human-derived genetically modified feeder cells. Three genes (human-derived telomerase reverse transcriptase gene, enhanced green fluorescent protein gene, and herpes simplex virus thymidine kinase gene) were transducted into human stromal cells, which enabled genetically modified feeder cells to be immortalized, labeled, and eliminated as needed. A target cell sheet was produced as one sheet by assisting the genetically modified feeder cells and successfully transplanted in vivo without their contamination. Genetically modified human eliminable feeder cells could be a promising tool for cultivated cell sheet transplantation.
Collapse
|
159
|
Telomerase protects adult rodent olfactory ensheathing glia from early senescence. Exp Neurol 2010; 229:54-64. [PMID: 20736004 DOI: 10.1016/j.expneurol.2010.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 08/07/2010] [Indexed: 11/21/2022]
Abstract
Adult olfactory bulb ensheathing glia (OB-OEG) promote the repair of acute, subacute, and chronic spinal cord injuries and autologous transplantation is a feasible approach. There are interspecies differences between adult rodent and primate OB-OEG related to their longevity in culture. Whereas primate OB-OEG exhibit a relatively long life span, under the same culture conditions rodent OB-OEG divide just three to four times, are sensitive to oxidative stress and become senescent after the third week in vitro. Telomerase is a "physiological key regulator" of the life span of normal somatic cells and also has extratelomeric functions such as increased resistance to oxidative stress. To elucidate whether telomerase has a role in the senescence of rodent OB-OEG, we have introduced the catalytic subunit of telomerase mTERT into cultures of these cells by retroviral infection. Native and modified adult rat OB-OEG behaved as telomerase-competent cells as they divided while expressing mTERT but entered senescence once the gene switched off. After ectopic expression of mTERT, OB-OEG resumed division at a nonsenescent rate, expressed p75 and other OEG markers, and exhibited the morphology of nonsenescent OB-OEG. The nonsenescent period of mTERT-OEG lasted 9weeks and then ectopic mTERT switched off and cells entered senescence again. Our results suggest a role of telomerase in early senescence of adult rodent OB-OEG cultures and a protection from oxidative damage. This article is part of a Special Issue entitled: Understanding olfactory ensheathing glia and their prospect for nervous system repair.
Collapse
|
160
|
Daniels DJ, Clothier C, Swan DC, Saretzki G. Immediate and gradual gene expression changes in telomerase over-expressing fibroblasts. Biochem Biophys Res Commun 2010; 399:7-13. [DOI: 10.1016/j.bbrc.2010.07.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 07/07/2010] [Indexed: 10/19/2022]
|
161
|
Baumer Y, Funk D, Schlosshauer B. Does telomerase reverse transcriptase induce functional de-differentiation of human endothelial cells? Cell Mol Life Sci 2010; 67:2451-65. [PMID: 20352467 PMCID: PMC11115536 DOI: 10.1007/s00018-010-0349-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Revised: 02/26/2010] [Accepted: 03/09/2010] [Indexed: 01/05/2023]
Abstract
By counteracting the shortening of chromosome telomeres, telomerase reverse transcriptase (hTERT) prevents senescence and age-related cell death. Embryonic cells display a high telomerase activity that declines rapidly with cell differentiation. Conversely, de-differentiated tumor cells tend to re-express telomerase. In view of the controversial data on the reciprocal correlation between cell proliferation and differentiation, we questioned whether telomerase overexpression and the resulting immortalization would affect the functional phenotype of human endothelial cells. Our comparative analysis addressed (1) distinct cell adhesion to different ECM-proteins analyzed on miniaturized multisubstrate arrays (MSA), (2) protein expression of diverse markers, (3) the uptake of DiI-Ac-LDL, (4) the inflammatory response based on upregulation of ICAM-1, (5) tube formation, and (6) the barrier properties of cell monolayers in transfilter cultures. Our results, based on some 40 data sets, demonstrate that immortalization of primary endothelial cells by hTERT maintains the typical endothelial characteristics without any sign of functional de-differentiation.
Collapse
Affiliation(s)
- Yvonne Baumer
- NMI, Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstraße 55, 72770 Reutlingen, Germany
| | - Dorothee Funk
- NMI, Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstraße 55, 72770 Reutlingen, Germany
| | - Burkhard Schlosshauer
- NMI, Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstraße 55, 72770 Reutlingen, Germany
| |
Collapse
|
162
|
Hung CJ, Yao CL, Cheng FC, Wu ML, Wang TH, Hwang SM. Establishment of immortalized mesenchymal stromal cells with red fluorescence protein expression for in vivo transplantation and tracing in the rat model with traumatic brain injury. Cytotherapy 2010; 12:455-65. [PMID: 20230225 DOI: 10.3109/14653240903555827] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND AIMS Human mesenchymal stromal cells (hMSC) play a crucial role in tissue engineering and regenerative medicine, and have important clinical potential for cell therapy. However, many hMSC studies have been restricted by limited cell numbers and difficult detection in vivo. To expand the lifespan, hMSC are usually immortalized by virus-mediated gene transfer. However, these genetically modified cells easily lose critical phenotypes and stable genotypes because of insertional mutagenesis. METHODS We used a non-viral transfection method to establish human telomerase reverse transcriptase-immortalized cord blood hMSC (hTERT-cbMSC). We also established red fluorescent protein (RFP)-expressing hTERT-cbMSC (hTERT/RFP-cbMSC) by the same non-viral transfection method, and these cells were injected into a rat model with traumatic brain injury for in vivo detection analysis. RESULTS The hTERT-cbMSC could grow more than 200 population doublings with a stable doubling time and maintained differentiation capacities. hTERT/RFP-cbMSC could proliferate efficiently within 2 weeks at the injury location and could be detected easily under a fluorescent microscope. Importantly, both hTERT-cbMSC and hTERT/RFP-cbMSC showed no chromosomal abnormalities by karyotype analysis and no tumor formation in severe combined immunodeficient (SCID) mice by transplantation assay. CONCLUSIONS We have developed immortalized cbMSC with hTERT expression and RFP expression, which will be useful tools for stem cell research and translational study.
Collapse
Affiliation(s)
- Chi-Jen Hung
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | | | | | | | | | | |
Collapse
|
163
|
Yamaguchi H, Kojima T, Ito T, Kimura Y, Imamura M, Son S, Koizumi JI, Murata M, Nagayama M, Nobuoka T, Tanaka S, Hirata K, Sawada N. Transcriptional control of tight junction proteins via a protein kinase C signal pathway in human telomerase reverse transcriptase-transfected human pancreatic duct epithelial cells. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:698-712. [PMID: 20566751 DOI: 10.2353/ajpath.2010.091226] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In human pancreatic cancer, integral membrane proteins of tight junction claudins are abnormally regulated, making these proteins promising molecular diagnostic and therapeutic targets. However, the regulation of claudin-based tight junctions remains unknown not only in the pancreatic cancer cells but also in normal human pancreatic duct epithelial (HPDE) cells. To investigate the regulation of tight junction molecules including claudins in normal HPDE cells, we introduced the human telomerase reverse transcriptase (hTERT) gene into HPDE cells in primary culture. The hTERT-transfected HPDE (hTERT-HPDE) cells were positive for the pancreatic duct epithelial markers such as CK7, CK19, and carbonic anhydrase isozyme 2 and expressed epithelial tight junction molecules claudin-1, -4, -7 and, -18, occludin, JAM-A, ZO-1, ZO-2, and tricellulin. By treatment with fetal bovine serum or 12-O-tetradecanoylphorbol 13-acetate (TPA), the tight junction molecules were up-regulated at the transcriptional level via a protein kinase C (PKC) signal pathway. A PKC-alpha inhibitor, Gö6976, prevented up-regulation of claudin-4 by TPA. Furthermore, a PKC-delta inhibitor, rottlerin, prevented up-regulation of claudin-7, occludin, ZO-1, and ZO-2 by TPA. By GeneChip analysis, up-regulation of the transcription factor ELF3 was observed in both fetal bovine serum- and TPA-treated cells. Treatment with small interfering RNAs of ELF3 prevented up-regulation of claudin-7 by TPA. These data suggest that tight junctions of normal HPDE cells were at least in part regulated via a PKC signal pathway by transcriptional control.
Collapse
Affiliation(s)
- Hiroshi Yamaguchi
- Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Momin EN, Vela G, Zaidi HA, Quiñones-Hinojosa A. The Oncogenic Potential of Mesenchymal Stem Cells in the Treatment of Cancer: Directions for Future Research. CURRENT IMMUNOLOGY REVIEWS 2010; 6:137-148. [PMID: 20490366 PMCID: PMC2873198 DOI: 10.2174/157339510791111718] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) represent a promising new approach to the treatment of several diseases that are associated with dismal outcomes. These include myocardial damage, graft versus host disease, and possibly cancer. Although the potential therapeutic aspects of MSCs continue to be well-researched, the possible hazards of MSCs, and in particular their oncogenic capacity are poorly understood. This review addresses the oncogenic and tumor-supporting potential of MSCs within the context of cancer treatment. The risk for malignant transformation is discussed for each stage of the clinical lifecycle of MSCs. This includes malignant transformation in vitro during production phases, during insertion of potentially therapeutic transgenes, and finally in vivo via interactions with tumor stroma. The immunosuppressive qualities of MSCs, which may facilitate evasion of the immune system by a tumor, are also addressed. Limitations of the methods employed in clinical trials to date are reviewed, including the absence of long term follow-up and lack of adequate screening methods to detect formation of new tumors. Through discussions of the possible oncogenic and tumor-supporting mechanisms of MSCs, directions for future research are identified which may eventually facilitate the future clinical translation of MSCs for the treatment of cancer and other diseases.
Collapse
Affiliation(s)
| | - Guillermo Vela
- Department of Neurosurgery and Oncology, The Johns Hopkins School of Medicine, Baltimore, MD
| | - Hasan A. Zaidi
- Department of Neurosurgery and Oncology, The Johns Hopkins School of Medicine, Baltimore, MD
| | | |
Collapse
|
165
|
Molecular modeling study of binding site selectivity of TQMP to G-quadruplex DNA. Eur J Med Chem 2010; 45:983-91. [DOI: 10.1016/j.ejmech.2009.11.040] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 09/06/2009] [Accepted: 11/20/2009] [Indexed: 11/23/2022]
|
166
|
Roig AI, Eskiocak U, Hight SK, Kim SB, Delgado O, Souza RF, Spechler SJ, Wright WE, Shay JW. Immortalized epithelial cells derived from human colon biopsies express stem cell markers and differentiate in vitro. Gastroenterology 2010; 138:1012-21.e1-5. [PMID: 19962984 DOI: 10.1053/j.gastro.2009.11.052] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Revised: 11/16/2009] [Accepted: 11/20/2009] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Long-term propagation of human colonic epithelial cells (HCEC) of adult origin has been a challenge; currently used HCEC lines are of malignant origin and/or contain multiple cytogenetic changes. We sought to immortalize human colon biopsy-derived cells expressing stem cell markers and retaining multilineage epithelial differentiation capability. METHODS We isolated and cultured cells from biopsy samples of 2 patients undergoing routine screening colonoscopy. Cells were immortalized by expression of the nononcogenic proteins cyclin-dependent kinase 4 (Cdk4) and the catalytic component of human telomerase (hTERT) and maintained for more than 1 year in culture. RESULTS The actively proliferating HCECs expressed the mesenchymal markers vimentin and alpha-smooth muscle actin. Upon growth arrest, cells assumed a cuboidal shape, decreased their mesenchymal features, and expressed markers of colonic epithelial cells such as cytokeratin 18, zonula occludens-1, mucins-1 and -2, antigen A33, and dipeptidyl peptidase 4. Immortalized cells expressed stem cell markers that included LGR5, BMI1, CD29, and CD44. When placed in Matrigel in the absence of a mesenchymal feeder layer, individual cells divided and formed self-organizing, cyst-like structures; a subset of cells exhibited mucin-2 or polarized villin staining. CONCLUSIONS We established immortalized HCECs that are capable of self-renewal and multilineage differentiation. These cells should serve as valuable reagents for studying colon stem cell biology, differentiation, and pathogenesis.
Collapse
Affiliation(s)
- Andres I Roig
- Department of Cell Biology, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Hashim M, Sayed M, Samy N, Elshazly S. Prognostic significance of telomerase activity and some tumor markers in non-small cell lung cancer. Med Oncol 2010; 28:322-30. [PMID: 20146105 DOI: 10.1007/s12032-010-9444-0] [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: 01/27/2010] [Accepted: 01/29/2010] [Indexed: 11/30/2022]
Abstract
The prognosis of non small cell lung cancer (NSCLC) has remained disappointing over the last decades even in localized stages. Numerous prognostic factors have been investigated which might select patients for additional treatment. The objective of the current study was to assess the prognostic significance of telomerase activity, serum anti-p53 antibodies (anti-p53a), c-erbB-2 and CEA in patients with NSCLC. The study included 60 patients with histological proven NSCLC besides 60 controls (30 smokers and 30 nonsmokers). Patients were divided into four stages according to their histopathology. All patients were subjected to; determination of telomerase activity by telomeric repeat amplification protocol (TRAP) assay in tumor tissue specimens and adjacent normal lung tissues, also, determination of preoperative serum anti-p53a, c-erbB-2 and CEA. Telomerase activity was detected in 40 of 60 (66.6%) of NSCLC tissue specimens using the TRAP assay. As regard the stages, telomerase activity was positive in 5 of 15 patients (33.3%) with stage I NSCLC, in 11 of 20 patients (55%) with stage II NSCLC, in 9 of 10 patients (90%) with stage III NSCLC and in all patients (100%) with stage IV NSCLC. More cases of positive telomerase activity were observed in the group with advanced disease and in the group with poorly differentiated tumors. Telomerase activity was not detected in any normal lung tissue. The concentrations of serum anti-p53a, c-erbB-2, CEA were significantly higher in patients with NSCLC in comparison to the smoker and nonsmoker controls and their levels increased according to the stage of disease. Logistic regression test showed a relation between telomerase positivity and anti- p53a but no relation with c-erbB2, CEA. Telomerase activity was detected in most of NSCLC tissues; it was detected more frequently in advanced disease than early-stage disease. Anti-p53, c-erbB-2 and CEA were significantly higher in patients with NSCLC than controls and this increment was more evident in late stages of the disease. So, these biological markers might be useful predictors of prognosis. They may be helpful in defining groups of patients with NSCLC who could benefit from adjuvant treatments, also these markers can be used as therapeutic targets.
Collapse
Affiliation(s)
- Maha Hashim
- Biochemistry Department, National Research Centre, Cairo, Egypt
| | | | | | | |
Collapse
|
168
|
Telomeres: protecting chromosomes against genome instability. Nat Rev Mol Cell Biol 2010; 11:171-81. [PMID: 20125188 DOI: 10.1038/nrm2848] [Citation(s) in RCA: 718] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The natural ends of linear chromosomes require unique genetic and structural adaptations to facilitate the protection of genetic material. This is achieved by the sequestration of the telomeric sequence into a protective nucleoprotein cap that masks the ends from constitutive exposure to the DNA damage response machinery. When telomeres are unmasked, genome instability arises. Balancing capping requirements with telomere replication and the enzymatic processing steps that are obligatory for telomere function is a complex problem. Telomeric proteins and their interacting factors create an environment at chromosome ends that inhibits DNA repair; however, the repair machinery is essential for proper telomere function.
Collapse
|
169
|
Fossel M, Flanary B. Telomerase and human disease: the beginnings of the ends? Rejuvenation Res 2010; 12:333-40. [PMID: 19725774 DOI: 10.1089/rej.2009.0873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Michael Fossel
- Department of Medicine, Michigan State University, Ada, Michigan, USA
| | | |
Collapse
|
170
|
Debacq-Chainiaux F, Boilan E, Dedessus Le Moutier J, Weemaels G, Toussaint O. p38(MAPK) in the senescence of human and murine fibroblasts. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 694:126-37. [PMID: 20886761 DOI: 10.1007/978-1-4419-7002-2_10] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oncogenic and environmental stresses, such as reactive oxygen species, UV radiation etc, can induce premature cellular senescence without critical telomere shortening. The role of the Ras/Raf/ERK signal transduction cascade in this process has been previously established, but recent evidence also indicates a critical role of the p38 MAP kinases pathway. Oncogenic and environmental stresses impinge upon the p38(MAPK) pathway, suggesting a major role of this pathway in senescence induced by stresses. Prematurely senescent cells are most likely to appear in several age-relatedpathologies associated with a stressful environment and/or the release of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Florence Debacq-Chainiaux
- University of Namur, Research Unit on Cellular Biology, Rue de Bruxelles, 61, Namur B-5000, Belgium.
| | | | | | | | | |
Collapse
|
171
|
Granic A, Padmanabhan J, Norden M, Potter H. Alzheimer Abeta peptide induces chromosome mis-segregation and aneuploidy, including trisomy 21: requirement for tau and APP. Mol Biol Cell 2009; 21:511-20. [PMID: 20032300 PMCID: PMC2820417 DOI: 10.1091/mbc.e09-10-0850] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Chromosome aneuploidy, especially trisomy 21, arises in both familial and sporadic Alzheimer's disease. Expression of FAD genes or exposure to Aβ peptide induces aneuploidy in tg-mice and cultured cells. The requirement for GSK-3β, calpain, and Tau in Aβ-induced chromosome mis-segregation points to MT dysfunction as contributing to AD pathogenesis. Both sporadic and familial Alzheimer's disease (AD) patients exhibit increased chromosome aneuploidy, particularly trisomy 21, in neurons and other cells. Significantly, trisomy 21/Down syndrome patients develop early onset AD pathology. We investigated the mechanism underlying mosaic chromosome aneuploidy in AD and report that FAD mutations in the Alzheimer Amyloid Precursor Protein gene, APP, induce chromosome mis-segregation and aneuploidy in transgenic mice and in transfected cells. Furthermore, adding synthetic Aβ peptide, the pathogenic product of APP, to cultured cells causes rapid and robust chromosome mis-segregation leading to aneuploid, including trisomy 21, daughters, which is prevented by LiCl addition or Ca2+ chelation and is replicated in tau KO cells, implicating GSK-3β, calpain, and Tau-dependent microtubule transport in the aneugenic activity of Aβ. Furthermore, APP KO cells are resistant to the aneugenic activity of Aβ, as they have been shown previously to be resistant to Aβ-induced tau phosphorylation and cell toxicity. These results indicate that Aβ-induced microtubule dysfunction leads to aneuploid neurons and may thereby contribute to the pathogenesis of AD.
Collapse
Affiliation(s)
- Antoneta Granic
- Eric Pfeiffer Suncoast Alzheimer's Center, University of South Florida, Tampa FL, 33613, USA
| | | | | | | |
Collapse
|
172
|
Crameri G, Todd S, Grimley S, McEachern JA, Marsh GA, Smith C, Tachedjian M, De Jong C, Virtue ER, Yu M, Bulach D, Liu JP, Michalski WP, Middleton D, Field HE, Wang LF. Establishment, immortalisation and characterisation of pteropid bat cell lines. PLoS One 2009; 4:e8266. [PMID: 20011515 PMCID: PMC2788226 DOI: 10.1371/journal.pone.0008266] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 11/12/2009] [Indexed: 11/19/2022] Open
Abstract
Background Bats are the suspected natural reservoir hosts for a number of new and emerging zoonotic viruses including Nipah virus, Hendra virus, severe acute respiratory syndrome coronavirus and Ebola virus. Since the discovery of SARS-like coronaviruses in Chinese horseshoe bats, attempts to isolate a SL-CoV from bats have failed and attempts to isolate other bat-borne viruses in various mammalian cell lines have been similarly unsuccessful. New stable bat cell lines are needed to help with these investigations and as tools to assist in the study of bat immunology and virus-host interactions. Methodology/Findings Black flying foxes (Pteropus alecto) were captured from the wild and transported live to the laboratory for primary cell culture preparation using a variety of different methods and culture media. Primary cells were successfully cultured from 20 different organs. Cell immortalisation can occur spontaneously, however we used a retroviral system to immortalise cells via the transfer and stable production of the Simian virus 40 Large T antigen and the human telomerase reverse transcriptase protein. Initial infection experiments with both cloned and uncloned cell lines using Hendra and Nipah viruses demonstrated varying degrees of infection efficiency between the different cell lines, although it was possible to infect cells in all tissue types. Conclusions/Significance The approaches developed and optimised in this study should be applicable to bats of other species. We are in the process of generating further cell lines from a number of different bat species using the methodology established in this study.
Collapse
Affiliation(s)
- Gary Crameri
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
- Australian Biosecurity Cooperative Research Centre for Emerging Infectious Diseases, Brisbane, Australia
| | - Shawn Todd
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Samantha Grimley
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
- Monash Immunology and Stem Cell Laboratories, Monash University, Melbourne, Australia
| | - Jennifer A. McEachern
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
- Australian Biosecurity Cooperative Research Centre for Emerging Infectious Diseases, Brisbane, Australia
| | - Glenn A. Marsh
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Craig Smith
- Australian Biosecurity Cooperative Research Centre for Emerging Infectious Diseases, Brisbane, Australia
- Queensland Primary Industries and Fisheries, Biosecurity Queensland, Brisbane, Australia
| | - Mary Tachedjian
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
- Australian Biosecurity Cooperative Research Centre for Emerging Infectious Diseases, Brisbane, Australia
| | - Carol De Jong
- Australian Biosecurity Cooperative Research Centre for Emerging Infectious Diseases, Brisbane, Australia
- Queensland Primary Industries and Fisheries, Biosecurity Queensland, Brisbane, Australia
| | - Elena R. Virtue
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
- Australian Biosecurity Cooperative Research Centre for Emerging Infectious Diseases, Brisbane, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| | - Meng Yu
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
- Australian Biosecurity Cooperative Research Centre for Emerging Infectious Diseases, Brisbane, Australia
| | - Dieter Bulach
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
- Australian Biosecurity Cooperative Research Centre for Emerging Infectious Diseases, Brisbane, Australia
| | - Jun-Ping Liu
- Central Clinical School, Monash University, Melbourne, Australia
| | - Wojtek P. Michalski
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
- Monash Immunology and Stem Cell Laboratories, Monash University, Melbourne, Australia
| | - Deborah Middleton
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
| | - Hume E. Field
- Australian Biosecurity Cooperative Research Centre for Emerging Infectious Diseases, Brisbane, Australia
- Queensland Primary Industries and Fisheries, Biosecurity Queensland, Brisbane, Australia
| | - Lin-Fa Wang
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Australia
- Australian Biosecurity Cooperative Research Centre for Emerging Infectious Diseases, Brisbane, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
- * E-mail:
| |
Collapse
|
173
|
Lund TC, Glass TJ, Tolar J, Blazar BR. Expression of telomerase and telomere length are unaffected by either age or limb regeneration in Danio rerio. PLoS One 2009; 4:e7688. [PMID: 19893630 PMCID: PMC2766636 DOI: 10.1371/journal.pone.0007688] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Accepted: 10/13/2009] [Indexed: 12/26/2022] Open
Abstract
Background The zebrafish is an increasingly popular model for studying many aspects of biology. Recently, ztert, the zebrafish homolog of the mammalian telomerase gene has been cloned and sequenced. In contrast to humans, it has been shown that the zebrafish maintains telomerase activity for much of its adult life and has remarkable regenerative capacity. To date, there has been no longitudinal study to assess whether this retention of telomerase activity equates to the retention of chromosome telomere length through adulthood. Methodology/Principal Findings We have systematically analyzed individual organs of zebrafish with regard to both telomere length and telomerase activity at various time points in its adult life. Heart, gills, kidney, spleen, liver, and intestine were evaluated at 3 months, 6 months, 9 months, and 2 years of age by Southern blot analysis. We found that telomeres do not appreciably shorten throughout the lifespan of the zebrafish in any organ. In addition, there was little difference in telomere lengths between organs. Even when cells were under the highest pressure to divide after fin-clipping experiments, telomere length was unaffected. All aged (2 year old) tissues examined also expressed active amounts of telomerase activity as assessed by TRAP assay. Conclusions/Significance In contrast to several other species including humans, the retention of lifelong telomerase and telomeres, as we have reported here, would be necessary in the zebrafish to maintain its tremendous regenerative capacity. The ongoing study of the zebrafish's ability to maintain telomerase activity may be helpful in unraveling the complexity involved in the maintenance (or lack thereof) of telomeres in other species such the mouse or human.
Collapse
Affiliation(s)
- Troy C Lund
- Division of Pediatric Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, MN, USA.
| | | | | | | |
Collapse
|
174
|
Schmelzer E, Reid LM. Human telomerase activity, telomerase and telomeric template expression in hepatic stem cells and in livers from fetal and postnatal donors. Eur J Gastroenterol Hepatol 2009; 21:1191-8. [PMID: 19240645 PMCID: PMC2743773 DOI: 10.1097/meg.0b013e32832973fc] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Although telomerase activity has been analyzed in various normal and malignant tissues, including liver, it is still unknown to what extent telomerase can be associated with specific maturational lineage stages. METHODS We assessed human telomerase activity, protein and gene expression for the telomerase reverse transcriptase, as well as expression of the telomeric template RNA hTER in hepatic stem cells and in various developmental stages of the liver from fetal to adult. In addition, the effect of growth factors on telomerase activity was analyzed in hepatic stem cells in vitro. RESULTS Telomerase was found to be highly active in fetal liver cells and was significantly higher than in hepatic stem cells, correlating with gene and protein expression levels. Activity in postnatal livers from all donor ages varied considerably and did not correlate with age or gene expression levels. The hter expression could be detected throughout the development. A short stimulation by growth factors of cultured hepatic stem cells did not increase telomerase activity. CONCLUSION Telomerase is considerably active in fetal liver and variably in postnatal livers. Although telomerase protein is present at varying levels in liver cells of all donor ages, gene expression is solely associated with fetal liver cells.
Collapse
Affiliation(s)
- Eva Schmelzer
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Lola M. Reid
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Biomedical Engineering, Program in Molecular Biology and Biotechnology, Lineberger Cancer Center and Center for Gastrointestinal and Biliary Disease Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
175
|
Straszewski-Chavez SL, Abrahams VM, Alvero AB, Aldo PB, Ma Y, Guller S, Romero R, Mor G. The isolation and characterization of a novel telomerase immortalized first trimester trophoblast cell line, Swan 71. Placenta 2009; 30:939-48. [PMID: 19766308 DOI: 10.1016/j.placenta.2009.08.007] [Citation(s) in RCA: 196] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Revised: 08/20/2009] [Accepted: 08/22/2009] [Indexed: 11/17/2022]
Abstract
Studies using first trimester trophoblast cells may be limited by the inability to obtain patient samples and/or adequate cell numbers. First trimester trophoblast cell lines have been generated by SV40 transformation or similar methods, however, this approach is known to induce phenotypic and karyotypic abnormalities. The introduction of telomerase has been proposed to be a viable alternative for the immortalization of primary human cells. To investigate whether telomerase-induced immortalization might be a more feasible approach for the generation of first trimester trophoblast cell lines, we isolated primary trophoblast cells from a 7-week normal placenta and infected the cells with human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase. Although this hTERT-infected first trimester trophoblast cell line, which we have named Swan 71, has been propagated for more than 100 passages, it still has attributes that are characteristic of primary first trimester trophoblast cells. The Swan 71 cells are positive for the expression of cytokeratin 7, vimentin and HLA-G, but do not express CD45, CD68 or the Fibroblast Specific Antigen (FSA), CD90/Thy-1. In addition, we also demonstrated that the Swan 71 cells secrete fetal fibronectin (FFN) as well as low levels of human Chorionic Gonadotrophin (hCG). Moreover, the Swan 71 cells exhibit a cytokine and growth factor profile that is similar to primary trophoblast cells and are resistant to Fas, but not TNF-alpha-induced apoptosis. This suggests that the Swan 71 cells may represent a valuable model for future in vitro trophoblast studies.
Collapse
Affiliation(s)
- S L Straszewski-Chavez
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | | | | | | | | | | | | | | |
Collapse
|
176
|
Nakahara H, Misawa H, Hayashi T, Kondo E, Yuasa T, Kubota Y, Seita M, Kawamoto H, Hassan WARA, Hassan RARA, Javed SM, Tanaka M, Endo H, Noguchi H, Matsumoto S, Takata K, Tashiro Y, Nakaji S, Ozaki T, Kobayashi N. Bone repair by transplantation of hTERT-immortalized human mesenchymal stem cells in mice. Transplantation 2009; 88:346-53. [PMID: 19667936 DOI: 10.1097/tp.0b013e3181ae5ba2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Human mesenchymal stem cells (hMSCs) are multipotent stem cells found in the adult bone marrow that have the capacity to differentiate into various mesenchymal cell types. The hMSCs may provide a potential therapy to restore damaged tissues or organs of mesenchymal origin; however, a drawback is their limited life span in vitro. METHODS We immortalized normal hMSCs with retrovirally transmitted human telomerase reverse transcriptase cDNA. One of the immortalized clones (YKNK-12) was established, and the biological characteristics were investigated in vitro and in vivo. RESULTS YKNK-12 cells were capable of differentiating adipocytes, osetoblasts, and chondrocytes. Osteogenically differentiated YKNK-12 cells produced significant levels of growth factors BMP4, BMP6, FGF6, FGF7, transforming growth factor-beta1, and transforming growth factor-beta3.. Microcomputer tomography T and soft X-ray assays showed an excellent calvarial bone healing in mice after transplantation of osteogenically differentiated YKNK-12 cells. These cells expressed human-specific osteocalcin and increased the gene expression of runt-related transcription factor 2, alkaline phosphatase, osteocalcin, and osterix in the bone regenerating area. YKNK-12 cell transplant corrected the bone defect without inducing any adverse effects. CONCLUSIONS We conclude that hMSCs immortalized by transduction with human telomerase reverse transcriptase may provide an unlimited source of cells for therapeutic use in bone regeneration.
Collapse
Affiliation(s)
- Hiroyuki Nakahara
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Vardar-Sengul S, Arora S, Baylas H, Mercola D. Expression profile of human gingival fibroblasts induced by interleukin-1beta reveals central role of nuclear factor-kappa B in stabilizing human gingival fibroblasts during inflammation. J Periodontol 2009; 80:833-49. [PMID: 19405838 DOI: 10.1902/jop.2009.080483] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Interleukin (IL)-1beta is a key cytokine in the pathogenesis of periodontitis, and it induces inflammatory mediators in periodontal diseases. We developed immortalized human gingival fibroblasts (HGFs), investigated the effects of IL-1beta on the gene expression using expression arrays containing approximately 40,000 genes, and tested the role of nuclear factor-kappa B (NF-kappaB) in maintaining an activated HGF population. METHODS Total RNA was isolated from IL-1beta-induced and mock-induced control cells. Gene expression analyses were performed using expression arrays and confirmed by quantitative real-time polymerase chain reaction. Western blot analysis to show inhibitor of kappa B-alpha (IkappaBalpha) phosphorylation and immunostaining of cells for NF-kappaB nuclear translocation were performed. Apoptosis was confirmed by assay of poly ADP-ribose polymerase (PARP) cleavage. RESULTS A total of 382 probe sets corresponding to 254 genes were differentially expressed in IL-1beta-induced cells (P <0.001). A total of 215 genes were upregulated, and 39 genes were downregulated. Most notable NF-kappaB pathway members (NFkappaB1, NFkappaB2, IkappaBalpha, IkappaBepsilon, IkappaBzeta, REL, RELB, and TA-NFKBH) were upregulated. IkappaBalpha was phosphorylated, and NF-kappaB accumulated in the nucleus. An IL-1beta-induced set of 27 genes was downregulated by an NF-kappaB inhibitor, leading to a decreased number of viable cells and suggesting an antiapoptotic role for NF-kappaB. CONCLUSIONS IL-1beta leads to a large number of significant expression changes consistent with a pathologic role in periodontitis, including enhancement of inflammatory cytokines, chemokines, transcription factors, matrix metalloproteinases, adhesion molecules, and especially NF-kappaB-dependent antiapoptotic genes. NF-kappaB activation blocks apoptosis, thereby stabilizing the HGF population in inflammation.
Collapse
Affiliation(s)
- Saynur Vardar-Sengul
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | | | | | | |
Collapse
|
178
|
Andersson U, Osterman P, Sjöström S, Johansen C, Henriksson R, Brännström T, Broholm H, Christensen HC, Ahlbom A, Auvinen A, Feychting M, Lönn S, Kiuru A, Swerdlow A, Schoemaker M, Roos G, Malmer B. MNS16A minisatellite genotypes in relation to risk of glioma and meningioma and to glioblastoma outcome. Int J Cancer 2009; 125:968-72. [PMID: 19405125 DOI: 10.1002/ijc.24363] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The human telomerase reverse transcriptase (hTERT) gene is upregulated in a majority of malignant tumours. A variable tandem repeat, MNS16A, has been reported to be of functional significance for hTERT expression. Published data on the clinical relevance of MNS16A variants in brain tumours have been contradictory. The present population-based study in the Nordic countries and the United Kingdom evaluated brain-tumour risk and survival in relation to MNS16A minisatellite variants in 648 glioma cases, 473 meningioma cases and 1,359 age, sex and geographically matched controls. By PCR-based genotyping all study subjects with fragments of 240 or 271 bp were judged as having short (S) alleles and subjects with 299 or 331 bp fragments as having long (L) alleles. Relative risk of glioma or meningioma was estimated with logistic regression adjusting for age, sex and country. Overall survival was analysed using Kaplan-Meier estimates and equality of survival distributions using the log-rank test and Cox proportional hazard ratios. The MNS16A genotype was not associated with risk of occurrence of glioma, glioblastoma (GBM) or meningioma. For GBM there were median survivals of 15.3, 11.0 and 10.7 months for the LL, LS and SS genotypes, respectively; the hazard ratio for having the LS genotype compared with the LL was significantly increased HR 2.44 (1.56-3.82) and having the SS genotype versus the LL was nonsignificantly increased HR 1.46 (0.81-2.61). When comparing the LL versus having one of the potentially functional variants LS and SS, the HR was 2.10 (1.41-3.1). However, functionality was not supported as there was no trend towards increasing HR with number of S alleles. Collected data from our and previous studies regarding both risk and survival for the MNS16A genotypes are contradictory and warrant further investigations.
Collapse
Affiliation(s)
- Ulrika Andersson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Abstract
The LDL receptor (LDLR) mediates efficient endocytosis of VLDL, VLDL remnants, and LDL. As part of the uptake process, the LDLR releases lipoproteins in endosomes. Released lipoproteins are subsequently trafficked to lysosomes for degradation, while the LDLR recycles back to the cell surface for further rounds of uptake. Endosomes have at least two features that can promote lipoprotein release: an acidic pH and low concentrations of free calcium. The relative contributions of acidic pH and low free calcium to lipoprotein release are not known. Here, we generated fibroblasts that express either normal LDLR or an LDLR variant that is unable to employ the acid-dependent release mechanism to determine the relative contributions of acidic pH and low free calcium on lipoprotein release. We show that endosomal concentrations of free calcium can drive lipoprotein release at rates that are similar to those of acid-dependent release and that the calcium-dependent and acid-dependent mechanisms can cooperate during lipoprotein release. Assessment of lipoprotein uptake by these two cell lines showed that LDL uptake requires the acid-dependent mechanism, while uptake of the VLDL remnant, beta-VLDL, does not. We propose that endosomes use both the acid-dependent and calcium-dependent release mechanisms to drive lipoprotein release and that the acid-dependent process is only required for LDL release.
Collapse
Affiliation(s)
- Zhenze Zhao
- Department of Cell Biology at the University of Texas Southwestern Medical Center, Dallas, Texas 75390-9039
| | - Peter Michaely
- Department of Cell Biology at the University of Texas Southwestern Medical Center, Dallas, Texas 75390-9039
| |
Collapse
|
180
|
Samalecos A, Reimann K, Wittmann S, Schulte HM, Brosens JJ, Bamberger AM, Gellersen B. Characterization of a novel telomerase-immortalized human endometrial stromal cell line, St-T1b. Reprod Biol Endocrinol 2009; 7:76. [PMID: 19619280 PMCID: PMC2719639 DOI: 10.1186/1477-7827-7-76] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Accepted: 07/20/2009] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Coordinated differentiation of the endometrial compartments in the second half of the menstrual cycle is a prerequisite for the establishment of pregnancy. Endometrial stromal cells (ESC) decidualize under the influence of ovarian progesterone to accommodate implantation of the blastocyst and support establishment of the placenta. Studies into the mechanisms of decidualization are often hampered by the lack of primary ESC. Here we describe a novel immortalized human ESC line. METHODS Primary ESC were immortalized by the transduction of telomerase. The resultant cell line, termed St-T1b, was characterized for its morphological and biochemical properties by immunocytochemistry, RT-PCR and immunoblotting. Its progestational response was tested using progesterone and medroxyprogesterone acetate with and without 8-Br-cAMP, an established inducer of decidualization in vitro. RESULTS St-T1b were positive for the fibroblast markers vimentin and CD90 and negative for the epithelial marker cytokeratin-7. They acquired a decidual phenotype indistinguishable from primary ESC in response to cAMP stimulation. The decidual response was characterized by transcriptional activation of marker genes, such as PRL, IGFBP1, and FOXO1, and enhanced protein levels of the tumor suppressor p53 and the metastasis suppressor KAI1 (CD82). Progestins alone had no effect on St-T1b cells, but medroxyprogesterone acetate greatly enhanced the cAMP-stimulated expression of IGFBP-1 after 3 and 7 days. Progesterone, albeit more weakly, also augmented the cAMP-induced IGFBP-1 production but only after 7 days of treatment. The cell line remained stable in continuous culture for more than 150 passages. CONCLUSION St-T1b express the appropriate phenotypic ESC markers and their decidual response closely mimics that of primary cultures. Decidualization is efficiently induced by cAMP analog and enhanced by medroxyprogesterone acetate, and, to a lesser extent, by natural progesterone. St-T1b cells therefore serve as a useful model for primary ESC.
Collapse
Affiliation(s)
| | | | | | | | - Jan J Brosens
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London W12 ONN, UK
| | - Ana-Maria Bamberger
- Section on Endocrinology and Ageing, University Clinic Hamburg-Eppendorf, 20251 Hamburg, Germany
| | | |
Collapse
|
181
|
Kuruvilla L, T R S, Kartha CC. Immortalization and Characterization of Porcine Ventricular Endocardial Endothelial Cells. ACTA ACUST UNITED AC 2009; 14:35-43. [PMID: 17364895 DOI: 10.1080/10623320601177353] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Endocardial endothelial cells (EECs), which form the inner lining of the cavities of the heart, are a distinct cell population whose dysfunction can be critical in pathological conditions of heart. Insights into the role and organization of these cells in pathological states of the heart are limited mainly due to a dearth of experimental models. To date no endocardial endothelial cell line is available. The authors attempted to immortalize porcine ventricular EECs by transfecting the cells with human telomerase reverse transcriptase (hTERT). EECs immortalized by ectopic expression of hTERT exhibit phenotypic and functional characteristics similar to primary EECs. The EE cell line could be useful for the study of mechanisms involved in the interaction of EECs with the underlying myocardium and cardiac interstitium and as useful tools in understanding their role in diseased states of heart.
Collapse
Affiliation(s)
- Leena Kuruvilla
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| | | | | |
Collapse
|
182
|
Wolbank S, Stadler G, Peterbauer A, Gillich A, Karbiener M, Streubel B, Wieser M, Katinger H, van Griensven M, Redl H, Gabriel C, Grillari J, Grillari-Voglauer R. Telomerase immortalized human amnion- and adipose-derived mesenchymal stem cells: maintenance of differentiation and immunomodulatory characteristics. Tissue Eng Part A 2009; 15:1843-54. [PMID: 19125642 PMCID: PMC3092731 DOI: 10.1089/ten.tea.2008.0205] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cell banking of mesenchymal stem cells (SCs) from various human tissues has significantly increased the feasibility of SC-based therapies. Sources such as adipose tissue and amnion offer outstanding possibilities for allogeneic transplantation due to their high differentiation potential and their ability to modulate immune reaction. Limitations, however, concern the reduced replicative potential as a result of progressive telomere erosion, which hampers scaleable production and long-term analysis of these cells. Here we report the establishment and characterization of two human amnion-derived and two human adipose-derived SC lines immortalized by ectopic expression of the catalytic subunit of human telomerase (hTERT). hTERT overexpression resulted in continuously growing SC lines that were largely unaltered concerning surface marker profile, morphology, karyotype, and immunosuppressive capacity with similar or enhanced differentiation potential for up to 87 population doublings. While all generated lines showed equal immunomodulation compared to the parental cells, one of the amnion-derived immortalized lines resulted in significantly increased immunogenicity. Although telomerase proves as important tool for immortalizing cells, our data emphasize the need for careful and standardized characterization of each individual cell population for cell banks.
Collapse
Affiliation(s)
- Susanne Wolbank
- Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
- Austrian Cluster for Tissue Regeneration, Vienna/Linz, Austria
| | - Guido Stadler
- Austrian Cluster for Tissue Regeneration, Vienna/Linz, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Center, Vienna/Linz, Austria
- Bio-Products & Bio-Engineering AG, Vienna, Austria
| | - Anja Peterbauer
- Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
- Austrian Cluster for Tissue Regeneration, Vienna/Linz, Austria
| | - Astrid Gillich
- Department of Biotechnology, Institute of Applied Microbiology, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | - Michael Karbiener
- Department of Biotechnology, Institute of Applied Microbiology, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | - Berthold Streubel
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Matthias Wieser
- Department of Biotechnology, Institute of Applied Microbiology, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | - Hermann Katinger
- Department of Biotechnology, Institute of Applied Microbiology, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | - Martijn van Griensven
- Austrian Cluster for Tissue Regeneration, Vienna/Linz, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Center, Vienna/Linz, Austria
| | - Heinz Redl
- Austrian Cluster for Tissue Regeneration, Vienna/Linz, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Center, Vienna/Linz, Austria
| | - Christian Gabriel
- Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
- Austrian Cluster for Tissue Regeneration, Vienna/Linz, Austria
| | - Johannes Grillari
- Department of Biotechnology, Institute of Applied Microbiology, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | - Regina Grillari-Voglauer
- Department of Biotechnology, Institute of Applied Microbiology, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| |
Collapse
|
183
|
Dwyer JM, Liu JP. Ets2 transcription factor, telomerase activity and breast cancer. Clin Exp Pharmacol Physiol 2009; 37:83-7. [PMID: 19566835 DOI: 10.1111/j.1440-1681.2009.05236.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1. The enzyme telomerase maintains telomeres (ends of chromosomes) by synthesizing telomeric DNA at each end of the chromosomes. Its association with telomeres has implicated telomerase in cell immortalization. 2. Numerous studies have shown significant levels of telomerase activity in 85% of various types of cancer. Transcriptional control of the catalytic subunit, telomerase reverse transcriptase (TERT), dominates regulation of telomerase. Although several major factors have been identified in regulating TERT, they cannot explain all the transcriptional activity of the hTERT gene. 3. The Ets transcription factor (TF) family is becoming a regular feature in tumourigenesis, particularly in breast cancer. However, the roles and mechanisms of different Ets TFs are largely unknown. 4. The present minireview discusses the research that identified Ets as a regulator of telomerase required for breast cancer cell survival and proliferation, highlighting the discoveries central to understanding the molecular acts used by Ets TFs to mediate TERT gene transcription.
Collapse
Affiliation(s)
- Julie M Dwyer
- Department of Immunology, Molecular Signalling Laboratory, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | | |
Collapse
|
184
|
Moon DO, Kim MO, Lee JD, Choi YH, Kim GY. Butein suppresses c-Myc-dependent transcription and Akt-dependent phosphorylation of hTERT in human leukemia cells. Cancer Lett 2009; 286:172-9. [PMID: 19560862 DOI: 10.1016/j.canlet.2009.05.028] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 05/12/2009] [Accepted: 05/25/2009] [Indexed: 01/21/2023]
Abstract
Telomerase, a ribonucleoprotein that plays an important role in neoplastic immortality, is up-regulated in approximately 85% of cancers, especially in leukemia. The polyphenol, butein, has potent effects against various types of cancer cells, but its effects on telomerase activity have not been well characterized. In this study, we show that butein causes a down-regulation of hTERT gene expression and a concomitant decrease of telomerase activity. Butein also suppresses expression of c-Myc at the transcriptional level and down-regulates DNA-binding activity, regardless of cell type specificity, in leukemia cells. DNA-binding activities of c-Myc to the hTERT core promoter were decreased in butein-treated cells, as seen by chromatin immunoprecipitation assay. Treatment with butein also suppressed the activation of Akt, thereby inhibiting hTERT phosphorylation and translocation into the nucleus. In this process, butein also up-regulated the surface expression of CD11b in leukemia cells. Inhibition of telomerase activity by butein was followed by loss of proliferative capacity, induction of apoptosis, and differentiation. These findings demonstrate the effectiveness of butein at inhibiting telomerase activity by down-regulating hTERT gene expression in human leukemia cells.
Collapse
Affiliation(s)
- Dong-Oh Moon
- Department of Marine Life Science, Jeju National University and Jeju Regional Cancer Center, Jeju 690-756, Republic of Korea
| | | | | | | | | |
Collapse
|
185
|
Simian virus 40 small T antigen activates AMPK and triggers autophagy to protect cancer cells from nutrient deprivation. J Virol 2009; 83:8565-74. [PMID: 19515765 DOI: 10.1128/jvi.00603-09] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
As tumors grow larger, they often experience an insufficient supply of oxygen and nutrients. Hence, cancer cells must develop mechanisms to overcome these stresses. Using an in vitro transformation model where the presence of the simian virus 40 (SV40) small T (ST) antigen has been shown to be critical for tumorigenic transformation, we investigated whether the ST antigen has a role to play in regulating the energy homeostasis of cancer cells. We find that cells expressing the SV40 ST antigen (+ST cells) are more resistant to glucose deprivation-induced cell death than cells lacking the SV40 ST antigen (-ST cells). Mechanistically, we find that the ST antigen mediates this effect by activating a nutrient-sensing kinase, AMP-activated protein kinase (AMPK). The basal level of active, phosphorylated AMPK was higher in +ST cells than in -ST cells, and these levels increased further in response to glucose deprivation. Additionally, inhibition of AMPK in +ST cells increased the rate of cell death, while activation of AMPK in -ST cells decreased the rate of cell death, under conditions of glucose deprivation. We further show that AMPK mediates its effects, at least in part, by inhibiting mTOR (mammalian target of rapamycin), thereby shutting down protein translation. Finally, we show that +ST cells exhibit a higher percentage of autophagy than -ST cells upon glucose deprivation. Thus, we demonstrate a novel role for the SV40 ST antigen in cancers, where it functions to maintain energy homeostasis during glucose deprivation by activating AMPK, inhibiting mTOR, and inducing autophagy as an alternate energy source.
Collapse
|
186
|
He Y, Wu Y, He X, Liu F, He X, Zhang Y. An immortalized goat mammary epithelial cell line induced with human telomerase reverse transcriptase (hTERT) gene transfer. Theriogenology 2009; 71:1417-24. [DOI: 10.1016/j.theriogenology.2009.01.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2008] [Revised: 12/14/2008] [Accepted: 01/13/2009] [Indexed: 11/26/2022]
|
187
|
Kapanadze B, Morris E, Smith E, Trojanowska M. Establishment and characterization of scleroderma fibroblast clonal cell lines by introduction of the hTERT gene. J Cell Mol Med 2009; 14:1156-65. [PMID: 19432820 PMCID: PMC3822752 DOI: 10.1111/j.1582-4934.2009.00773.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Lack of an adequate experimental model has hindered the ability to fully understand scleroderma (SSc) pathogenesis. Current SSc research is based on the study of cultured fibroblasts from skin biopsies. In depth characterization of the SSc fibroblast phenotype is hindered by the limited lifespan and heterogeneity of these cells. The goal of this study was to isolate high collagen-producing fibroblasts from SSc biopsies and extend their lifespan with hTERT immortalization to enable characterization of their phenotype. Fibroblasts from two pairs of closely matched normal and SSc biopsies were infected with an hTERT lentivirus. Infected colonies were isolated, cultured into clonal cell lines and analysed with respect to profibrotic gene expression. The mRNA levels of nine profibrotic genes were measured by quantitative real-time PCR. Protein levels were assessed by Western blot. The hTERT SSc clones were heterogeneous with regards to expression of the profibrotic genes measured. A subset of the SSc clones showed elevated expression levels of collagen I, connective tissue growth factor and thrombospondin 1 mRNA, while expression of other genes was not significantly changed. Elevated expression of collagen I protein and mRNA was correlative with elevated expression of connective tissue growth factor. Several hTERT clones expressed high levels of pSmad1, Smad1 and TGF-βRI indicative of altered TGF-β signalling. A portion of SSc clones expressed several profibrotic genes. This study demonstrates that select characteristics of the SSc phenotype are expressed in a subset of activated fibroblasts in culture. The clonal SSc cell lines may present a new and useful model to investigate the mechanisms involved in SSc fibrosis.
Collapse
Affiliation(s)
- Bagrat Kapanadze
- Division of Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
188
|
Kogan-Sakin I, Cohen M, Paland N, Madar S, Solomon H, Molchadsky A, Brosh R, Buganim Y, Goldfinger N, Klocker H, Schalken JA, Rotter V. Prostate stromal cells produce CXCL-1, CXCL-2, CXCL-3 and IL-8 in response to epithelia-secreted IL-1. Carcinogenesis 2009; 30:698-705. [DOI: 10.1093/carcin/bgp043] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
189
|
Li NF, Kocher HM, Salako MA, Obermueller E, Sandle J, Balkwill F. A novel function of colony-stimulating factor 1 receptor in hTERT immortalization of human epithelial cells. Oncogene 2009; 28:773-80. [PMID: 18997822 DOI: 10.1038/onc.2008.412] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 10/02/2008] [Accepted: 10/07/2008] [Indexed: 01/27/2023]
Abstract
The receptor for macrophage colony-stimulating factor 1 receptor (CSF1R) is a product of the proto-oncogene c-fms and a member of the class III transmembrane tyrosine kinase receptor family. Earlier, we described increased mRNA expression of CSF1R in human telomerase reverse transcriptase (hTERT) immortalized human ovarian surface epithelial (IOSE) cell lines derived from a single donor. Here, we further describe that CSF1R is upregulated at both the mRNA and protein level in hTERT immortalized human normal OSE cells from two different donors and in hTERT immortalized human pancreatic ductal epithelial cells. CSF1R was not upregulated in hTERT immortalized epithelial clones that subsequently underwent senescence or in immortalized fibroblasts. Upon stimulation by the CSF1R ligand CSF1, the immortalized epithelial cell lines showed rapid internalization of CSF1R with concomitant down-modulation and colocalization of phosphorylated NFkappaBp65 with hTERT protein, hTERT translocation into the nucleus and the binding of c-Myc to the hTERT promoter region. Reducing the expression of CSF1R using short hairpin interfering RNA abolished these effects and also decreased cell survival and the number of population doublings under suboptimal culture conditions. The telomerase inhibitor GRN163L confirmed a role for telomerase in the cleavage of the intracellular domain of CSF1R. On the basis of these findings, we suggest that CSF1R may be a critical factor facilitating hTERT immortalization of epithelial cells.
Collapse
Affiliation(s)
- N F Li
- The Centre for Cancer and Inflammation, Institute of Cancer and the Cancer Research-UK Clinical Centre, Barts and The London School of Medicine and Dentistry, London, UK
| | | | | | | | | | | |
Collapse
|
190
|
Murasawa S, Asahara T. Cardiogenic potential of endothelial progenitor cells. Ther Adv Cardiovasc Dis 2009; 2:341-8. [PMID: 19124432 DOI: 10.1177/1753944708096283] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Transplantation of endothelial progenitor cells (EPCs) are one of the promising strategies to recover the capillary flow in ischaemic diseases such as ischaemic heart disease and peripheral artery disease (PAD) in the leg. However, our previous and another group's works suggested the scarcity of the number of EPCs in peripheral blood might cause insufficient effect for the ischaemic diseases. There are several strategies to overcome this issue, such as (1) in vivo EPC expansion; (2) ex vivo EPC expansion; (3) local (not systemic) EPC injection; and (4) modification of EPC by gene transfer. Recent publications from our own and other groups have reported the possibility of cardiogenic potential of EPCs. We would like to focus on the strategies of EPC transplantation and cardiomyogenesis of EPCs in this review.
Collapse
Affiliation(s)
- Satoshi Murasawa
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, Kobe, Japan.
| | | |
Collapse
|
191
|
Lleonart ME, Artero-Castro A, Kondoh H. Senescence induction; a possible cancer therapy. Mol Cancer 2009; 8:3. [PMID: 19133111 PMCID: PMC2631463 DOI: 10.1186/1476-4598-8-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Accepted: 01/08/2009] [Indexed: 12/13/2022] Open
Abstract
Cellular immortalization is a crucial step during the development of human cancer. Primary mammalian cells reach replicative exhaustion after several passages in vitro, a process called replicative senescence. During such a state of permanent growth arrest, senescent cells are refractory to physiological proliferation stimuli: they have altered cell morphology and gene expression patterns, although they remain viable with preserved metabolic activity. Interestingly, senescent cells have also been detected in vivo in human tumors, particularly in benign lesions. Senescence is a mechanism that limits cellular lifespan and constitutes a barrier against cellular immortalization. During immortalization, cells acquire genetic alterations that override senescence. Tumor suppressor genes and oncogenes are closely involved in senescence, as their knockdown and ectopic expression confer immortality and senescence induction, respectively. By using high throughput genetic screening to search for genes involved in senescence, several candidate oncogenes and putative tumor suppressor genes have been recently isolated, including subtypes of micro-RNAs. These findings offer new perspectives in the modulation of senescence and open new approaches for cancer therapy.
Collapse
Affiliation(s)
- Matilde E Lleonart
- Pathology Department, Fundació Institut de Recerca Hospital Vall d'Hebron, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain.
| | | | | |
Collapse
|
192
|
The LIM-only protein FHL2 mediates ras-induced transformation through cyclin D1 and p53 pathways. PLoS One 2008; 3:e3761. [PMID: 19018287 PMCID: PMC2583050 DOI: 10.1371/journal.pone.0003761] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Accepted: 11/03/2008] [Indexed: 01/26/2023] Open
Abstract
Background Four and a half LIM-only protein 2 (FHL2) has been implicated in multiple signaling pathways that regulate cell growth and tissue homeostasis. We reported previously that FHL2 regulates cyclin D1 expression and that immortalized FHL2-null mouse embryo fibroblasts (MEFs) display reduced levels of cyclin D1 and low proliferative activity. Methodology/Principal Findings Here we address the contribution of FHL2 in cell transformation by investigating the effects of oncogenic Ras in FHL2-null context. We show that H-RasV12 provokes cell cycle arrest accompanied by accumulation of p53 and p16INK4a in immortalized FHL2−/− MEFs. These features contrast sharply with Ras transforming activity in wild type cell lines. We further show that establishment of FHL2-null cell lines differs from conventional immortalization scheme by retaining functional p19ARF/p53 checkpoint that is required for cell cycle arrest imposed by Ras. However, after serial passages of Ras-expressing FHL2−/− cells, dramatic increase in the levels of D-type cyclins and Rb phosphorylation correlates with the onset of cell proliferation and transformation without disrupting the p19ARF/p53 pathway. Interestingly, primary FHL2-null cells overexpressing cyclin D1 undergo a classical immortalization process leading to loss of the p19ARF/p53 checkpoint and susceptibility to Ras transformation. Conclusions/Significance Our findings uncover a novel aspect of cellular responses to mitogenic stimulation and illustrate a critical role of FHL2 in the signalling network that implicates Ras, cyclin D1 and p53.
Collapse
|
193
|
Mao Z, Bozzella M, Seluanov A, Gorbunova V. Comparison of nonhomologous end joining and homologous recombination in human cells. DNA Repair (Amst) 2008; 7:1765-71. [PMID: 18675941 PMCID: PMC2695993 DOI: 10.1016/j.dnarep.2008.06.018] [Citation(s) in RCA: 489] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Revised: 06/25/2008] [Accepted: 06/25/2008] [Indexed: 11/17/2022]
Abstract
The two major pathways for repair of DNA double-strand breaks (DSBs) are homologous recombination (HR) and nonhomologous end joining (NHEJ). HR leads to accurate repair, while NHEJ is intrinsically mutagenic. To understand human somatic mutation it is essential to know the relationship between these pathways in human cells. Here we provide a comparison of the kinetics and relative contributions of HR and NHEJ in normal human cells. We used chromosomally integrated fluorescent reporter substrates for real-time in vivo monitoring of the NHEJ and HR. By examining multiple integrated clones we show that the efficiency of NHEJ and HR is strongly influenced by chromosomal location. Furthermore, we show that NHEJ of compatible ends (NHEJ-C) and NHEJ of incompatible ends (NHEJ-I) are fast processes, which can be completed in approximately 30 min, while HR is much slower and takes 7h or longer to complete. In actively cycling cells NHEJ-C is twice as efficient as NHEJ-I, and NHEJ-I is three times more efficient than HR. Our results suggest that NHEJ is a faster and more efficient DSB repair pathway than HR.
Collapse
Affiliation(s)
- Zhiyong Mao
- Department of Biology, University of Rochester, Rochester, NY 14627-0211, USA
| | - Michael Bozzella
- Department of Biology, University of Rochester, Rochester, NY 14627-0211, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY 14627-0211, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY 14627-0211, USA
| |
Collapse
|
194
|
Shao G, Balajee AS, Hei TK, Zhao Y. p16INK4a downregulation is involved in immortalization of primary human prostate epithelial cells induced by telomerase. Mol Carcinog 2008; 47:775-83. [PMID: 18383581 DOI: 10.1002/mc.20434] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Prostate cancer is a major cause of cancer death among male population. Therefore, development of appropriate model systems is critical for understanding the molecular basis of prostate cancer progression. In this study, introduction of human telomerase (hTERT) into normal human prostate epithelial cells (PrECs) renders them higher telomerase activity, elongated telomere length and an extended proliferative lifespan. The immortal mass culture of PrEC-hTERT cell line with stabilized telomere length has been established using hTERT transfection. However, activation of hTERT alone appears to be insufficient for immortalization of PrEC cells because methylation of p16(INK4a) promoter has been found to be involved in the immortalization process. p53 was functionally intact and no mutations of p53 gene were identified in the immortalized PrECs. In addition, the immortal PrECs show a near diploid complement of chromosomes albeit a few reciprocal and non-reciprocal translocations are identified. They are anchorage dependent and do not form tumors in immunosuppressed host animals. Therefore, premalignantly transformed human PrECs provide a valuable model for prostate cancer research.
Collapse
Affiliation(s)
- Genze Shao
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania, Pennsylvania, USA
| | | | | | | |
Collapse
|
195
|
Mao Z, Bozzella M, Seluanov A, Gorbunova V. DNA repair by nonhomologous end joining and homologous recombination during cell cycle in human cells. Cell Cycle 2008; 7:2902-6. [PMID: 18769152 PMCID: PMC2754209 DOI: 10.4161/cc.7.18.6679] [Citation(s) in RCA: 480] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
DNA double-strand breaks (DSBs) are dangerous lesions that can lead to potentially oncogenic genomic rearrangements or cell death. The two major pathways for repair of DSBs are nonhomologous end joining (NHEJ) and homologous recombination (HR). NHEJ is an intrinsically error-prone pathway while HR results in accurate repair. To understand the origin of genomic instability in human cells it is important to know the contribution of each DSB repair pathway. Studies of rodent cells and human cancer cell lines have shown that the choice between NHEJ or HR pathways depends on cell cycle stage. Surprisingly, cell cycle regulation of DSB repair has not been examined in normal human cells with intact cell cycle checkpoints. Here we measured the efficiency of NHEJ and HR at different cell cycle stages in hTERT-immortalized diploid human fibroblasts. We utilized cells with chromosomally-integrated fluorescent reporter cassettes, in which a unique DSB is introduced by a rare-cutting endonuclease. We show that NHEJ is active throughout the cell cycle, and its activity increases as cells progress from G1 to G2/M (G1 < S < G2/M). HR is nearly absent in G1, most active in the S phase, and declines in G2/M. Thus, in G2/M NHEJ is elevated, while HR is on decline. This is in contrast to a general belief that NHEJ is most active in G1, while HR is active in S, G2 and M. The overall efficiency of NHEJ was higher than HR at all cell cycle stages. We conclude that human somatic cells utilize error-prone NHEJ as the major DSB repair pathway at all cell cycle stages, while HR is used, primarily, in the S phase.
Collapse
Affiliation(s)
| | | | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, New York 14627, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, New York 14627, USA
| |
Collapse
|
196
|
Bizzarri M, Cucina A, Conti F, D’Anselmi F. Beyond the oncogene paradigm: understanding complexity in cancerogenesis. Acta Biotheor 2008; 56:173-96. [PMID: 18288572 DOI: 10.1007/s10441-008-9047-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Accepted: 02/06/2008] [Indexed: 12/13/2022]
Abstract
In the past decades, an enormous amount of precious information has been collected about molecular and genetic characteristics of cancer. This knowledge is mainly based on a reductionistic approach, meanwhile cancer is widely recognized to be a 'system biology disease'. The behavior of complex physiological processes cannot be understood simply by knowing how the parts work in isolation. There is not solely a matter how to integrate all available knowledge in such a way that we can still deal with complexity, but we must be aware that a deeply transformation of the currently accepted oncologic paradigm is urgently needed. We have to think in terms of biological networks: understanding of complex functions may in fact be impossible without taking into consideration influences (rules and constraints) outside of the genome. Systems Biology involves connecting experimental unsupervised multivariate data to mathematical and computational approach than can simulate biologic systems for hypothesis testing or that can account for what it is not known from high-throughput data sets. Metabolomics could establish the requested link between genotype and phenotype, providing informations that ensure an integrated understanding of pathogenic mechanisms and metabolic phenotypes and provide a screening tool for new targeted drug.
Collapse
|
197
|
Tauchi T, Ohyashiki JH, Ohyashiki K. Telomerase inhibition combined with other chemotherapeutic reagents to enhance anti-cancer effect. Methods Mol Biol 2008; 405:181-9. [PMID: 18369825 DOI: 10.1007/978-1-60327-070-0_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Genetic experiments using a dominant-negative form of human telomerase (DN-hTERT) demonstrated that telomerase inhibition can result in telomeric shortening followed by proliferation arrest and cell death by apoptosis. Neoplastic cells from telomerase RNA null (mTERC-/-) mice showed enhanced chemosensitivity to doxorubicin or related double-strand DNA break (DSB)-inducing agents. Telomerase dysfunction, rather than telomerase inhibition, is proposed to be the principal determinant governing chemosensitivity specifically to DSB-inducing agents. We observed that imatinib and vincristine (VCR), in addition to DSB-inducing agents, also enhanced chemosensitivity in telomestatin-treated K562 cells. This observation suggests that combined use of telomerase inhibitors and imatinib or other chemotherapeutic agents may be a very useful approach to treatment of BCR-ABL-positive leukemia.
Collapse
Affiliation(s)
- Tetsuzo Tauchi
- First Department of Internal Medicine, Tokyo Medical University, Tokyo, Japan
| | | | | |
Collapse
|
198
|
Unusually stable abnormal karyotype in a highly aggressive melanoma negative for telomerase activity. Mol Cytogenet 2008; 1:20. [PMID: 18718029 PMCID: PMC2533344 DOI: 10.1186/1755-8166-1-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Accepted: 08/22/2008] [Indexed: 12/27/2022] Open
Abstract
Malignant melanomas are characterized by increased karyotypic complexity, extended aneuploidy and heteroploidy. We report a melanoma metastasis to the peritoneal cavity with an exceptionally stable, abnormal pseudodiploid karyotype as verified by G-Banding, subtelomeric, centromeric and quantitative Fluorescence in Situ Hybridization (FISH). Interestingly this tumor had no detectable telomerase activity as indicated by the Telomere Repeat Amplification Protocol. Telomeric Flow-FISH and quantitative telomeric FISH on mitotic preparations showed that malignant cells had relatively short telomeres. Microsatellite instability was ruled out by the allelic pattern of two major mononucleotide repeats. Our data suggest that a combination of melanoma specific genomic imbalances were sufficient and enough for this fatal tumor progression, that was not accompanied by genomic instability, telomerase activity, or the engagement of the alternative recombinatorial telomere lengthening pathway.
Collapse
|
199
|
Zhao Z, Michaely P. The epidermal growth factor homology domain of the LDL receptor drives lipoprotein release through an allosteric mechanism involving H190, H562, and H586. J Biol Chem 2008; 283:26528-37. [PMID: 18677035 DOI: 10.1074/jbc.m804624200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The low density lipoprotein (LDL) receptor (LDLR) mediates efficient endocytosis of VLDL, VLDL remnants, and LDL. As part of the endocytic process, the LDLR releases lipoproteins in endosomes. The release process correlates with an acid-dependent conformational change in the receptor from an extended, "open" state to a compact, "closed" state. The closed state has an intramolecular contact involving H190, H562, and H586. The current model for lipoprotein release holds that protonation of these histidines drives the conformational change that is associated with release. We tested the roles of H190, H562, and H586 on LDLR conformation and on lipoprotein binding, uptake, and release using variants in which the three histidines were replaced with alanine (AAA variant) or in which the histidines were replaced with charged residues that can form ionic contacts at neutral pH (DRK variant). Contrary to expectation, both the AAA and the DRK variants exhibited normal acid-dependent transitions from open to closed conformations. Despite this similarity, both the AAA and DRK mutations modulated lipoprotein release, indicating that H190, H562, and H586 act subsequent to the conformational transition. These observations also suggest that the intramolecular contact does not drive release through a competitive mechanism. In support of this possibility, mutagenesis experiments showed that beta-VLDL binding was inhibited by mutations at D203 and E208, which are exposed in the closed conformation of the LDLR. We propose that H190, H562, and H586 are part of an allosteric mechanism that drives lipoprotein release.
Collapse
Affiliation(s)
- Zhenze Zhao
- Department of Cell Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9039, USA
| | | |
Collapse
|
200
|
Mazzucchelli GD, Gabelica V, Smargiasso N, Fléron M, Ashimwe W, Rosu F, De Pauw-Gillet MC, Riou JF, De Pauw E. Proteome alteration induced by hTERT transfection of human fibroblast cells. Proteome Sci 2008; 6:12. [PMID: 18419814 PMCID: PMC2386453 DOI: 10.1186/1477-5956-6-12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Accepted: 04/17/2008] [Indexed: 01/15/2023] Open
Abstract
Background Telomerase confers cellular immortality by elongating telomeres, thereby circumventing the Hayflick limit. Extended-life-span cells have been generated by transfection with the human telomerase reverse transcriptase (hTERT) gene. hTERT transfected cell lines may be of outstanding interest to monitor the effect of drugs targeting the telomerase activity. The incidence of hTERT gene transfection at the proteome level is a prerequisite to that purpose. The effect of the transfection has been studied on the proteome of human fibroblast (WI38). Cytosolic and nuclear fractions of WI38 cells, empty vector transfected WI38 (WI38-HPV) and hTERT WI38 cells were submitted to a 2D-DIGE (Two-Dimensional Differential In-Gel Electrophoresis) analysis. Only spots that had a similar abundance in WI38 and WI38-HPV, but were differentially expressed in WI38 hTERT were selected for MS identification. This method directly points to the proteins linked with the hTERT expression. Number of false positive differentially expressed proteins has been excluded by using control WI38-HPV cells. The proteome alteration induced by hTERT WI38 transfection should be taken into account in subsequent use of the cell line for anti-telomerase drugs evaluation. Results 2D-DIGE experiment shows that 57 spots out of 2246 are significantly differentially expressed in the cytosolic fraction due to hTERT transfection, and 38 were confidently identified. In the nuclear fraction, 44 spots out of 2172 were selected in the differential proteome analysis, and 14 were identified. The results show that, in addition to elongating telomeres, hTERT gene transfection has other physiological roles, among which an enhanced ER capacity and a potent cell protection against apoptosis. Conclusion We show that the methodology reduces the complexity of the proteome analysis and highlights proteins implicated in other processes than telomere elongation. hTERT induced proteome changes suggest that telomerase expression enhances natural cell repair mechanisms and stress resistance probably required for long term resistance of immortalized cells. Thus, hTERT transfected cells can not be only consider as an immortal equivalent to parental cells but also as cells which are over-resistant to stresses. These findings are the prerequisite for any larger proteomics aiming to evaluate anti-telomerase drugs proteome alteration and thus therapeutics induced cell reactions.
Collapse
|