251
|
Ungrin MD, Harrington L. Strict control of telomerase activation using Cre-mediated inversion. BMC Biotechnol 2006; 6:10. [PMID: 16504006 PMCID: PMC1403769 DOI: 10.1186/1472-6750-6-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Accepted: 02/20/2006] [Indexed: 01/25/2023] Open
Abstract
Background Human cells appear exquisitely sensitive to the levels of hTERT expression, the telomerase reverse transcriptase. In primary cells that do not express hTERT, telomeres erode with each successive cell division, leading to the eventual loss of telomere DNA, an induction of a telomere DNA damage response, and the onset of cellular senescence or crisis. In some instances, an average of less than one appropriately spliced hTERT transcript per cell appears sufficient to restore telomerase activity and telomere maintenance, and overcome finite replicative capacity. Results To underscore this sensitivity, we showed that a widely used system of transcriptional induction involving ecdysone (muristerone) led to sufficient expression of hTERT to immortalize human fibroblasts, even in the absence of induction. To permit tightly regulated expression of hTERT, or any other gene of interest, we developed a method of transcriptional control using an invertible expression cassette flanked by antiparallel loxP recombination sites. When introduced into human fibroblasts with the hTERT cDNA positioned in the opposite orientation relative to a constitutively active promoter, no telomerase activity was detected, and the cell population retained a mortal phenotype. Upon inversion of the hTERT cDNA to a transcriptionally competent orientation via the action of Cre recombinase, cells acquired telomerase activity, telomere DNA was replenished, and the population was immortalized. Further, using expression of a fluorescent protein marker, we demonstrated the ability to repeatedly invert specific transcripts between an active and inactive state in an otherwise isogenic cell background. Conclusion This binary expression system thus provides a useful genetic means to strictly regulate the expression of a given gene, or to control the expression of at least two different genes in a mutually exclusive manner.
Collapse
Affiliation(s)
- Mark D Ungrin
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Terrence Donnelly Centre for Cellular and Biomolecular Research, 160 College Street, Toronto, Ontario, M5S 3E1, USA
| | - Lea Harrington
- Department of Medical Biophysics, University of Toronto, Ontario Cancer Institute, and Campbell Family Institute for Breast Cancer Research, 620 University Avenue, Toronto, ON M5G 2C1, USA
| |
Collapse
|
252
|
Klinger RY, Blum JL, Hearn B, Lebow B, Niklason LE. Relevance and safety of telomerase for human tissue engineering. Proc Natl Acad Sci U S A 2006; 103:2500-5. [PMID: 16477025 PMCID: PMC1413782 DOI: 10.1073/pnas.0508184103] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tissue engineering holds the promise of replacing damaged or diseased tissues and organs. The use of autologous donor cells is often not feasible because of the limited replicative lifespan of cells, particularly those derived from elderly patients. Proliferative arrest can be overcome by the ectopic expression of telomerase via human telomerase reverse transcriptase (hTERT) gene transfection. To study the efficacy and safety of this potentially valuable technology, we used differentiated vascular smooth muscle cells (SMC) and vascular tissue engineering as a model system. Although we previously demonstrated that vessels engineered with telomerase-expressing SMC had improved mechanics over those grown with control cells, it is critical to assess the phenotypic impact of telomerase expression in donor cells, because telomerase up-regulation is observed in >95% of human malignancies. To study the impact of telomerase in tissue engineering, expression of hTERT was retrovirally induced in SMC from eight elderly patients and one young donor. In hTERT SMC, significant lifespan extension beyond that of control was achieved without population doubling time acceleration. Karyotype changes were seen in both control and hTERT SMC but were not clonal nor representative of cancerous change. hTERT cells also failed to show evidence of neoplastic transformation in functional assays of tumorigenicity. In addition, the impact of donor age on cellular behavior, particularly the synthetic capability of SMC, was not affected by hTERT expression. Hence, this tissue engineering model system highlights the impact of donor age on cellular synthetic function that appears to be independent of lifespan extension by hTERT.
Collapse
MESH Headings
- Blood Vessels/cytology
- Blood Vessels/enzymology
- Blood Vessels/physiology
- Cell Culture Techniques/methods
- Cell Transformation, Neoplastic
- Chromosome Aberrations
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Humans
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/physiology
- Retroviridae/genetics
- Telomerase/genetics
- Telomerase/metabolism
- Telomere/enzymology
- Telomere/genetics
- Tissue Engineering/methods
- Transfection
Collapse
Affiliation(s)
- Rebecca Y. Klinger
- *Department of Biomedical Engineering, Duke University, Durham, NC 27708; and
| | - Juliana L. Blum
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
| | - Bevin Hearn
- *Department of Biomedical Engineering, Duke University, Durham, NC 27708; and
| | - Benjamin Lebow
- *Department of Biomedical Engineering, Duke University, Durham, NC 27708; and
| | - Laura E. Niklason
- *Department of Biomedical Engineering, Duke University, Durham, NC 27708; and
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
253
|
Glaviano A, Nayak V, Cabuy E, Baird DM, Yin Z, Newson R, Ladon D, Rubio MA, Slijepcevic P, Lyng F, Mothersill C, Case CP. Effects of hTERT on metal ion-induced genomic instability. Oncogene 2006; 25:3424-35. [PMID: 16449970 DOI: 10.1038/sj.onc.1209399] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is currently a great interest in delayed chromosomal and other damaging effects of low-dose exposure to a variety of pollutants which appear collectively to act through induction of stress-response pathways related to oxidative stress and ageing. These have been studied mostly in the radiation field but evidence is accumulating that the mechanisms can also be triggered by chemicals, especially heavy metals. Humans are exposed to metals, including chromium (Cr) (VI) and vanadium (V) (V), from the environment, industry and surgical implants. Thus, the impact of low-dose stress responses may be larger than expected from individual toxicity projections. In this study, a short (24 h) exposure of human fibroblasts to low doses of Cr (VI) and V (V) caused both acute chromosome damage and genomic instability in the progeny of exposed cells for at least 30 days after exposure. Acutely, Cr (VI) caused chromatid breaks without aneuploidy while V (V) caused aneuploidy without chromatid breaks. The longer-term genomic instability was similar but depended on hTERT positivity. In telomerase-negative hTERT- cells, Cr (VI) and V (V) caused a long lasting and transmissible induction of dicentric chromosomes, nucleoplasmic bridges, micronuclei and aneuploidy. There was also a long term and transmissible reduction of clonogenic survival, with an increased beta-galactosidase staining and apoptosis. This instability was not present in telomerase-positive hTERT+ cells. In contrast, in hTERT+ cells the metals caused a persistent induction of tetraploidy, which was not noted in hTERT- cells. The growth and survival of both metal-exposed hTERT+ and hTERT- cells differed if they were cultured at subconfluent levels or plated out as colonies. Genomic instability is considered to be a driving force towards cancer. This study suggests that the type of genomic instability in human cells may depend critically on whether they are telomerase-positive or -negative and that their sensitivities to metals could depend on whether they are clustered or diffuse.
Collapse
Affiliation(s)
- A Glaviano
- Bristol Implant Research Centre, University of Bristol, Bristol, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
254
|
Gire V. [Senescence: a telomeric limit to immortality or a cellular response to physiologic stresses?]. Med Sci (Paris) 2006; 21:491-7. [PMID: 15885198 DOI: 10.1051/medsci/2005215491] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cells entering a state of senescence undergo a irreversible cell cycle arrest, associated by a set of functional and morphological changes. Senescence occurs following telomeres shortening (replicative senescence) or exposure to other acute or chronic physiologic stress signals (a phenomenon termed stasis: stress or aberrant signaling-induced senescence). In this review, I discuss the pathways of cellular senescence, the mechanisms involved and the role that these pathways have in regulating the initiation and progression of cancer. Telomere-initiated senescence or loss of telomere function trigger focal recruitement of protein sensors of the DNA double-strand breaks leading to the activation of the DNA damage checkpoint responses and the tumour suppressor gene product, p53, which in turn induces the cell-cycle inhibitor, p21(WAF1). Loss of p53 and pRb function allows continued cell division despite increasing telomere dysfunction and eventually entry into telomere crisis. Immortalisation is an essential prerequisite for the formation of a tumour cell. Therefore, a developing tumour cell must circumvent at least two proliferative barriers--cellular senescence and crisis--to achieve neoplastic transformation. These barriers are regulated by telomere shortening and by the p16(INK4a)/Rb and p53 tumour suppressor pathways. Elucidation of the genes and emerging knowledge about the regulatory mechanisms that lead to senescence and determine the pattern of gene expression in senescent cells may lead to more effective treatments for cancer.
Collapse
Affiliation(s)
- Véronique Gire
- Centre de recherche en biochimie macromoléculaire, CNRS FRE 2593, IFR 24, 1919, route de Mende, 34293 Montpellier Cedex 5, France.
| |
Collapse
|
255
|
Rosa J, Canovas P, Islam A, Altieri DC, Doxsey SJ. Survivin modulates microtubule dynamics and nucleation throughout the cell cycle. Mol Biol Cell 2006; 17:1483-93. [PMID: 16407408 PMCID: PMC1382334 DOI: 10.1091/mbc.e05-08-0723] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Survivin is a member of the chromosomal passenger complex implicated in kinetochore attachment, bipolar spindle formation, and cytokinesis. However, the mechanism by which survivin modulates these processes is unknown. Here, we show by time-lapse imaging of cells expressing either green fluorescent protein (GFP)-alpha-tubulin or the microtubule plus-end binding protein GFP-EB1 that depletion of survivin by small interfering RNAs (siRNAs) increased both the number of microtubules nucleated by centrosomes and the incidence of microtubule catastrophe, the transition from microtubule growth to shrinking. In contrast, survivin overexpression reduced centrosomal microtubule nucleation and suppressed both microtubule dynamics in mitotic spindles and bidirectional growth of microtubules in midbodies during cytokinesis. siRNA depletion or pharmacologic inhibition of another chromosomal passenger protein Aurora B, had no effect on microtubule dynamics or nucleation in interphase or mitotic cells even though mitosis was impaired. We propose a model in which survivin modulates several mitotic events, including spindle and interphase microtubule organization, the spindle assembly checkpoint and cytokinesis through its ability to modulate microtubule nucleation and dynamics. This pathway may affect the microtubule-dependent generation of aneuploidy and defects in cell polarity in cancer cells, where survivin is commonly up-regulated.
Collapse
Affiliation(s)
- Jack Rosa
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | |
Collapse
|
256
|
van Waarde-Verhagen MAWH, Kampinga HH, Linskens MHK. Continuous growth of telomerase-immortalised fibroblasts: How long do cells remain normal? Mech Ageing Dev 2006; 127:85-7. [PMID: 16213574 DOI: 10.1016/j.mad.2005.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2005] [Accepted: 08/24/2005] [Indexed: 11/28/2022]
Abstract
Previously, we reported successful immortalisation following hTERT introduction in primary human fibroblasts, strain VH25. Since one subclone in that study developed some abnormalities, we decided to study eight additional independent immortalised clones to get an indication of the frequency and type of abnormalities that develop after hTERT-mediated immortalisation. We show that although some cell lines can maintain a normal phenotype for 500 population doublings (PDs), in four clones after 150-300PDs changes developed in basal and radiation-induced p53 and p21(WAF-1,CIP-1) levels. Our experiments demonstrate that, after prolonged culture, cells with abnormalities in cell cycle control parameters can take over the population. This calls for caution when working with hTERT-immortalised cells in vitro as well as in vivo.
Collapse
Affiliation(s)
- M A W H van Waarde-Verhagen
- Department of Cell Biology, Section for Radiation & Stress Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|
257
|
Porter PC, Clark DR, McDaniel LD, McGregor WG, States JC. Telomerase-immortalized human fibroblasts retain UV-induced mutagenesis and p53-mediated DNA damage responses. DNA Repair (Amst) 2006; 5:61-70. [PMID: 16140041 DOI: 10.1016/j.dnarep.2005.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Revised: 07/20/2005] [Accepted: 07/21/2005] [Indexed: 12/23/2022]
Abstract
Immortalized cells frequently have disruptions of p53 activity and lack p53-dependent nucleotide excision repair (NER). We hypothesized that telomerase immortalization would not alter p53-mediated ultraviolet light (UV)-induced DNA damage responses. DNA repair proficient primary diploid human fibroblasts (GM00024) were immortalized by transduction with a telomerase expressing retrovirus. Empty retrovirus transduced cells senesced after a few doublings. Telomerase transduced GM00024 cells (tGM24) were cultured continuously for 6 months (>60 doublings). Colony forming ability after UV irradiation was dose-dependent between 0 and 20J/m2 UVC (LD50=5.6J/m2). p53 accumulation was UV dose- and time-dependent as was induction of p48(XPE/DDB2), p21(CIP1/WAF1), and phosphorylation on p53-S15. UV dose-dependent apoptosis was measured by nuclear condensation. UV exposure induced UV-damaged DNA binding as monitored by electrophoretic mobility shift assays using UV irradiated radiolabeled DNA probe was inhibited by p53-specific siRNA transfection. p53-Specific siRNA transfection also prevented UV induction of p48 and improved UV survival measured by colony forming ability. Strand-specific NER of cyclobutane pyrimidine dimers (CPD) within DHFR was identical in tGM24 and GM00024 cells. CPD removal from the transcribed strand was nearly complete in 6h and from the non-transcribed strand was 73% complete in 24h. UV-induced HPRT mutagenesis in tGM24 was indistinguishable from primary human fibroblasts. These wide-ranging findings indicate that the UV-induced DNA damage response remains intact in telomerase-immortalized cells. Furthermore, telomerase immortalization provides permanent cell lines for testing the immediate impact on NER and mutagenesis of selective genetic manipulation without propagation to establish mutant lines.
Collapse
Affiliation(s)
- Paul C Porter
- Department of Pharmacology & Toxicology, University of Louisville, 570 South Preston Street, Rm221, Louisville, KY 40202, USA
| | | | | | | | | |
Collapse
|
258
|
Ouaissi M, Ouaissi A. Histone deacetylase enzymes as potential drug targets in cancer and parasitic diseases. J Biomed Biotechnol 2006; 2006:13474. [PMID: 16883049 PMCID: PMC1510935 DOI: 10.1155/jbb/2006/13474] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2005] [Revised: 03/19/2006] [Accepted: 03/22/2006] [Indexed: 11/18/2022] Open
Abstract
The elucidation of the mechanisms of transcriptional activation and repression in eukaryotic cells has shed light on the important role of acetylation-deacetylation of histones mediated by histone acetyltransferases (HATs) and histone deacetylases (HDACs), respectively. Another group belonging to the large family of sirtuins (silent information regulators (SIRs)) has an (nicotinamide adenine dinucleotide) NAD(+)-dependent HDAC activity. Several inhibitors of HDACs (HDIs) have been shown to exert antitumor effects. Interestingly, some of the HDIs exerted a broad spectrum of antiprotozoal activity. The purpose of this review is to analyze some of the current data related to the deacetylase enzymes as a possible target for drug development in cancer and parasitic diseases with special reference to protozoan infections. Given the structural differences among members of this family of enzymes, development of specific inhibitors will not only allow selective therapeutic intervention, but may also provide a powerful tool for functional study of these enzymes.
Collapse
Affiliation(s)
- Mehdi Ouaissi
- Service de Chirurgie Digestive et Générale, Hôpital Sainte Marguerite,
270 Boulevard de Sainte Marguerite, 13009 Marseille, France
| | - Ali Ouaissi
- IRD UR008 “Pathogénie des Trypanosomatidés,” Centre IRD de Montpellier, Institut de la Recherche pour le Développement, 911
Avenue Agropolis, BP 64501, 34394 Montpellier Cedex 5, France
| |
Collapse
|
259
|
Abstract
Telomeres are specialized DNA-protein structures at the ends of the linear chromosomes. In mammalian cells, they are composed of multifold hexameric TTAGGG repeats and a number of associated proteins. The double-stranded telomeric DNA ends in a 3' single stranded overhang of 150 to 300 base pair (bp) which is believed to be required for a higher order structure (reviewed in (Blackburn, 2001)). One important model is that the telomeres form loop structures, the T-loops, and by invasion of the 3' overhang into the duplex region of the double stranded part protect the DNA against degradation and hinder the cellular machinery to recognize the ends as broken DNA, thus providing chromosomal integrity (Griffith et al, 1999). If telomeres become critically short they loose their capping function, become sticky, and are prone to illegitimate chromosome end-to-end fusions. The resulting dicentric chromosomes are highly unusable and because of bridge-fusion-breakage cycles they give rise to chromosomal translocations, deletions, and amplifications. Thus, critically short telomeres are thought to be responsible for the onset of genomic instability. In addition, we provide evidence that in a length-independent manner telomeres can confer to genomic instability by forming telomericaggregates which through chromosomal dys-locations contribute to chromosomal aberrations.
Collapse
Affiliation(s)
- Petra Boukamp
- German Cancer Research Center, Division of Genetics of Skin Carcinogensis, Im Neuenheimer Feld, Heidelberg, Germany.
| | | | | |
Collapse
|
260
|
Gromley A, Yeaman C, Rosa J, Redick S, Chen CT, Mirabelle S, Guha M, Sillibourne J, Doxsey SJ. Centriolin anchoring of exocyst and SNARE complexes at the midbody is required for secretory-vesicle-mediated abscission. Cell 2005; 123:75-87. [PMID: 16213214 DOI: 10.1016/j.cell.2005.07.027] [Citation(s) in RCA: 349] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2004] [Revised: 06/03/2005] [Accepted: 07/27/2005] [Indexed: 12/28/2022]
Abstract
The terminal step in cytokinesis, called abscission, requires resolution of the membrane connection between two prospective daughter cells. Our previous studies demonstrated that the coiled-coil protein centriolin localized to the midbody during cytokinesis and was required for abscission. Here we show that centriolin interacts with proteins of vesicle-targeting exocyst complexes and vesicle-fusion SNARE complexes. These complexes require centriolin for localization to a unique midbody-ring structure, and disruption of either complex inhibits abscission. Exocyst disruption induces accumulation of v-SNARE-containing vesicles at the midbody ring. In control cells, these v-SNARE vesicles colocalize with a GFP-tagged secreted polypeptide. The vesicles move to the midbody ring asymmetrically from one prospective daughter cell; the GFP signal is rapidly lost, suggesting membrane fusion; and subsequently the cell cleaves at the site of vesicle delivery/fusion. We propose that centriolin anchors protein complexes required for vesicle targeting and fusion and integrates membrane-vesicle fusion with abscission.
Collapse
Affiliation(s)
- Adam Gromley
- Program in Molecular Medicine, University of Massachusetts Medical Center, Worcester, Massachusetts 01605, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
261
|
Sutherland BM, Cuomo NC, Bennett PV. Induction of anchorage-independent growth in primary human cells exposed to protons or HZE ions separately or in dual exposures. Radiat Res 2005; 164:493-6. [PMID: 16187755 DOI: 10.1667/rr3357.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Travelers on space missions will be exposed to a complex radiation environment that includes protons and heavy charged particles. Since protons are present at much higher levels than are heavy ions, the most likely scenario for cellular radiation exposure will be proton exposure followed by a hit by a heavy ion. Although the effects of individual ion species on human cells are being investigated extensively, little is known about the effects of exposure to both radiation types. One useful measure of mammalian cell damage is induction of the ability to grow in a semi-solid agar medium highly inhibitory to the growth of normal human cells, termed neoplastic transformation. Using primary human cells, we evaluated induction of soft-agar growth and survival of cells exposed to protons only or to heavy charged particles (600 MeV/nucleon silicon) only as well as of cells exposed to protons followed after a 4-day interval by silicon ions. Both ions alone efficiently transformed the human cells to anchorage-independent growth. Initial experiments indicate that the dose responses for neoplastic transformation of cells exposed to protons and then after 4 days to silicon ions appear similar to that of cells exposed to silicon ions alone.
Collapse
Affiliation(s)
- B M Sutherland
- Biology Department, Brookhaven National Laboratory, Upton, New York 11973-5000, USA
| | | | | |
Collapse
|
262
|
Abstract
It is thought that a limited investment by the body in many types of maintenance and repair causes aging. Cell turnover is one mechanism of replacing damaged cells, and cell division thus contributes to good repair, but the number of times cells can divide is limited to form a barrier against cancer. Precancerous cells must divide many times to accumulate all of the mutations needed to become malignant. Limiting the number of times they can divide helps prevent cancer. The mechanism for counting cell divisions lies in structures at the ends of the chromosomes called telomeres that shorten with every division, eventually causing cell aging. This shortening can be prevented and cells immortalized using the enzyme telomerase, which can elongate telomeres. Immortalizing all of the cells in the body might increase repair but would remove the barrier to malignancy and would probably cause premature death from cancer in many cases, although the ability to immortalize cells opens up enormous opportunities for using normal cells for therapeutic purposes in localized areas. Eventually, once better controls and treatments for cancer are available, cellular rejuvenation by manipulating telomeres may reduce some of the physiological declines that accompany aging. Such treatments should increase health span, but because replicative aging represents only one of many processes that may contribute to overall human aging, modest increases in life span are expected at best.
Collapse
Affiliation(s)
- Woodring E Wright
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Building K2.206, MC 9039, Dallas, TX 75235, USA.
| | | |
Collapse
|
263
|
Abstract
This paper reports on the application of quantum mechanical (QM) energy calculations, QM optimisations and MD simulations to explore the stability of a human telomeric guanine quadruplex, containing potassium and sodium cations. G-quadruplexes are of great biological interest as it has been suggested that they offer a novel path to cancer inhibition. By understanding the stability and geometry of these DNA features gives us the ability to design ligands which can bind and stabilise the G-quadruplex. There are significant structural differences between the potassium containing crystal structure of human telomeric G-quadruplex and the sodium containing NMR structure; in this paper, we investigate the energetics and dynamics of the potassium derived crystal structure and a model for the sodium containing structure. QM investigations upon the 12 G-quadruplex core, extracted from the human potassium quadruplex crystal structure, indicate that replacement of the potassium cations with sodium yields an energetically more favourable structure. However, attempts to geometry optimise both structures at the QM level proved unsuccessful, the structure of the partially optimised potassium containing G-quadruplex retains significant structural integrity with respect to the original crystal structure, whilst the sodium containing G-quadruplex shows significant structural distortion. QM investigation of the 12 G-quadruplex core containing no cations unsurprisingly yields a highly unfavourable energetic structure. MD simulations on the complete quadruplex structure, containing potassium cations, yields a remarkably stable structure after 4ns of simulation, the most significant deviation from the original crystal structure being the loss of the capping potassium cation from the structure. MD simulation of the sodium containing quadruplex for 4ns show significant structural reorganisation compared with the original potassium containing crystal structure.
Collapse
Affiliation(s)
- Emma H Clay
- Biological and Biophysical Chemistry, Department of Chemistry, Imperial College, London, UK
| | | |
Collapse
|
264
|
Gagos S, Irminger-Finger I. Chromosome instability in neoplasia: chaotic roots to continuous growth. Int J Biochem Cell Biol 2005; 37:1014-33. [PMID: 15743675 DOI: 10.1016/j.biocel.2005.01.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2004] [Revised: 01/06/2005] [Accepted: 01/07/2005] [Indexed: 12/31/2022]
Abstract
Multiple rearrangements of chromosome number and structure are common manifestations of genomic instability encountered in mammalian tumors. In neoplasia, in continuous immortalized growth in vitro, and in animal models, the accumulation of various defects on DNA repair and telomere maintenance machineries, mitotic spindle abnormalities, and breakage-fusion-bridge cycles, deteriorate the precise mitotic distribution of the genomic content, thus producing various types of chromosomal anomalies. These lesions generate tremendous genomic imbalances, which are evolutionary selected, since they force the function of the whole genome towards continuous growth. For more than a century chromosomal rearrangements and aneuploidy in neoplasia have been discussed and a vast number of genes and pathways, directly or indirectly implicated, have been described. In this review, we focus on the biological mechanisms that generate numerical or structural deviations of the normal diploid chromosomal constitution in epithelial neoplasia. There is growing evidence that chromosomal instability is both an epiphenomenon and a leading cause of cancer. We will discuss the roles of genes, chromosome structure, and telomere dysfunction in the initiation of chromosomal instability. We will explore research strategies that can be applied to identify rates of chromosomal instability in a specimen, and the putative biological consequences of karyotypic heterogeneity. Finally, we will re-examine the longstanding hypothesis of the generation of aneuploidy in the context of telomere dysfunction and restoration.
Collapse
Affiliation(s)
- Sarantis Gagos
- Laboratory of Genetics, Foundation for Biomedical Research of the Academy of Athens Greece, Soranou Efessiou 4, Athens 11527, Greece.
| | | |
Collapse
|
265
|
Crea F, Sarti D, Falciani F, Al-Rubeai M. Over-expression of hTERT in CHO K1 results in decreased apoptosis and reduced serum dependency. J Biotechnol 2005; 121:109-23. [PMID: 16144725 DOI: 10.1016/j.jbiotec.2005.07.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2005] [Revised: 06/14/2005] [Accepted: 07/04/2005] [Indexed: 12/16/2022]
Abstract
The enzyme telomerase plays a crucial role in cellular proliferation. By adding hexameric repeats to the chromosome ends, it prevents telomeric loss and, thus entry into senescence of limited life span cells. It is unclear, however, what would be the effect of over-expressing telomerase in an immortalised cell line, characterised by unlimited life span and high levels of apoptosis under sub-optimal growth conditions. In order to address this question, we have transfected the immortal cell line CHO K1 with the human telomerase reverse transcriptase (hTERT) catalytic subunit. Differences in the growth profile and apoptosis levels between the cells over-expressing hTERT (Telo) and the cells containing mock vector were found under standard growth conditions. Similarly, the Telo cells showed lower levels of apoptosis, greater attachment tendency and higher viable cell density under serum-deprived conditions compared to the control cell line, suggesting a major role for hTERT over-expression in stressed cultures. Using a mouse cDNA microarray, the collagen type III and V genes were shown to have at least a 10-fold higher expression in the Telo cells than the control cells, suggesting a role of hTERT in the cell attachment pathways.
Collapse
Affiliation(s)
- Francesco Crea
- Department of Chemical Engineering, School of Engineering, University of Birmingham, Birmingham B15 2TT, UK
| | | | | | | |
Collapse
|
266
|
Bayne S, Liu JP. Hormones and growth factors regulate telomerase activity in ageing and cancer. Mol Cell Endocrinol 2005; 240:11-22. [PMID: 16005142 DOI: 10.1016/j.mce.2005.05.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Accepted: 05/23/2005] [Indexed: 01/05/2023]
Abstract
Telomerase is a specialised reverse transcriptase that synthesises and preserves telomeres (the ends of chromosomes), thereby playing a key role in regulating the lifespan of cell proliferation. Telomerase activity is critically involved in cell development, ageing and tumourigenesis. Activation of telomerase to maintain telomeres is required for self renewal and proliferative expansion of a number of cell types, including stem cells, activated lymphocytes and cancerous cells. However, recent studies show that the safeguard mechanisms and the modes of regulation of telomerase are more revealing than thought under various physiological and pathological conditions. Considerable evidence suggests that hormones and growth factors are crucially involved in regulating telomerase activity and gene expression of telomerase reverse transcriptase (TERT). This review briefly summarises our current understanding of how hormones and growth factors regulate the telomerase and telomere network and how deregulation can induce ageing and related diseases such as cancer.
Collapse
Affiliation(s)
- Sharyn Bayne
- Molecular Signalling Laboratory, Department of Immunology, Monash University, AMREP, Prahran, Melbourne, Vic., Australia
| | | |
Collapse
|
267
|
Herbert BS, Gellert GC, Hochreiter A, Pongracz K, Wright WE, Zielinska D, Chin AC, Harley CB, Shay JW, Gryaznov SM. Lipid modification of GRN163, an N3'-->P5' thio-phosphoramidate oligonucleotide, enhances the potency of telomerase inhibition. Oncogene 2005; 24:5262-8. [PMID: 15940257 DOI: 10.1038/sj.onc.1208760] [Citation(s) in RCA: 189] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The vast majority of human cancers express telomerase activity, while most human somatic cells do not have detectable telomerase activity. Since telomerase plays a critical role in cell immortality, it is an attractive target for a selective cancer therapy. Oligonucleotides complementary to the RNA template region of human telomerase (hTR) have been shown to be effective inhibitors of telomerase and, subsequently, cancer cell growth in vitro. We show here that a lipid-modified N3'-->P5' thio-phosphoramidate oligonucleotide (GRN163L) inhibits telomerase more potently than its parental nonconjugated thio-phosphoramidate sequence (GRN163). Cells were treated with both the first- (GRN163) and second-generation (GRN163L) oligonucleotides, including a mismatch control, with or without a transfection enhancer reagent. GRN163L inhibited telomerase activity effectively in a dose-dependent manner, even without the use of a transfection reagent. The IC50 values for GRN163 in various cell lines were on average sevenfold higher than for GRN163L. GRN163L inhibition of telomerase activity resulted in a more rapid loss of telomeres and cell growth than GRN163. This report is the first to show that lipid modification enhanced the potency of the novel GRN163 telomerase inhibitor. These results suggest that the lipid-conjugated thio-phosphoramidates could be important for improved pharmacodynamics of telomerase inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Brittney-Shea Herbert
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9039, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
268
|
von Zglinicki T, Martin-Ruiz CM, Saretzki G. Telomeres, cell senescence and human ageing. ACTA ACUST UNITED AC 2005. [DOI: 10.1002/sita.200400049] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
269
|
Lee KM, Yasuda H, Hollingsworth MA, Ouellette MM. Notch 2-positive progenitors with the intrinsic ability to give rise to pancreatic ductal cells. J Transl Med 2005; 85:1003-12. [PMID: 15924149 DOI: 10.1038/labinvest.3700298] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pancreatic adenocarcinomas display foci of duct-like structures that are positive for markers of pancreatic ductal cells. The development of these tumors is promoted by conditions leading to acinar-to-ductal metaplasia, a process by which acinar cells are replaced by ductal cells. Acinar-to-ductal metaplasia has recently been shown to proceed through intermediary cells expressing Nestin. To create an in vitro system to study pancreatic adenocarcinomas, we had used an hTERT cDNA to immortalize primary cells of the human pancreas. In this report, we show that the immortalized cells, termed hTERT-HPNE cells, have the ability to differentiate to pancreatic ductal cells. Exposing hTERT-HPNE cells to sodium butyrate and 5-aza-2'-deoxycytidine lead to the formation of pancreatic ductal cells marked by the expression of MDR-1, carbonic anhydrase II, and the cytokeratins 7, 8, and 19. hTERT-HPNE cells were found to have properties of the intermediary cells formed during acinar-to-ductal metaplasia, which included their undifferentiated phenotype, expression of Nestin, evidence of active Notch signaling, and ability to differentiate to pancreatic ductal cells. These results provide further evidence for the presence in the adult pancreas of a precursor of ductal cells. hTERT-HPNE cells should provide a useful model to study acinar-to-ductal metaplasia and the role played by this process in pancreatic cancer development.
Collapse
Affiliation(s)
- Kwang M Lee
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | |
Collapse
|
270
|
Schreurs MWJ, Hermsen MAJA, Geltink RIK, Scholten KBJ, Brink AATP, Kueter EWM, Tijssen M, Meijer CJLM, Ylstra B, Meijer GA, Hooijberg E. Genomic stability and functional activity may be lost in telomerase-transduced human CD8+ T lymphocytes. Blood 2005; 106:2663-70. [PMID: 16002425 DOI: 10.1182/blood-2004-09-3742] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
To obtain the large amount of T cells required for adoptive immunotherapy in a clinical setting, T-cell lifespan extension by human telomerase reverse transcriptase (hTERT) transduction is of particular interest. However, constitutive expression of hTERT is associated with malignant transformation and thus warrants a detailed evaluation of the safety of hTERT-transduced T cells before clinical application. In view of this, we performed an extensive cytogenetic analysis of hTERT-transduced MART-1 (melanoma antigen recognized by T cell 1)-and human papillomavirus type 16 (HPV16) E7-specific human CD8+ cytotoxic T lymphocytes (CTLs), reactive against melanoma and cervical carcinoma, respectively. Our results, obtained by (spectral) karyotyping and array comparative genomic hybridization, showed the development of minor chromosomal aberrations in an hTERT-transduced MART-1-specific CTL clone, whereas severe clonal aberrations were detected in an hTERT-transduced HPV16 E7-specific CTL clone. Furthermore, hTERT transduction did not protect CTLs from immunosenescence, because the HPV16 E7-specific, hTERT-transduced CTL clone showed a decreased functional activity on prolonged culture. Although the general frequency of major chromosomal aberrations in hTERT-transduced CTLs and the in vivo significance of our observations remain still unclear at this point, the currently available data suggest that clinical application of hTERT-transduced CTLs should proceed with caution.
Collapse
Affiliation(s)
- Marco W J Schreurs
- Department of Pathology, VU University Medical Center, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
271
|
Barbier CS, Becker KA, Troester MA, Kaufman DG. Expression of Exogenous Human Telomerase in Cultures of Endometrial Stromal Cells Does Not Alter Their Hormone Responsiveness1. Biol Reprod 2005; 73:106-14. [PMID: 15772261 DOI: 10.1095/biolreprod.104.035063] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In the human endometrium, stromal cells mediate the proliferative response of epithelial cells to the steroid hormones estrogen and progesterone. These stromal-epithelial interactions are readily studied in vitro by coculture of both cell types. A major impediment to such studies is the rapid senescence of normal stromal cells. To circumvent this problem, we tested whether human endometrial stromal cells immortalized by expressing a transduced human telomerase reverse transcriptase (TERT) subunit retained the ability to mediate hormonal control of epithelial proliferation in the coculture assay. We found that the telomerized stromal cells were very similar to the parental strain from which they were derived according to criteria of proliferation, karyotype, cellular localization of cytoskeletal markers and nuclear staining, and basal gene expression based on microarray analysis. We also showed that expression of estrogen and progesterone receptors, as assessed by immunodetection, was similar in both telomerized and parental stromal cells. Importantly, the telomerized stromal cells were shown in coculture assay to be as effective as normal stromal cells in regulating the proliferation of endometrial epithelial cells in response to estrogen or progesterone. The availability of these long-lived stromal cells may advance studies addressing the mechanistic, regulatory, and cell structural basis of stromal-epithelial interactions and hormonal responses in normal, preneoplastic, and neoplastic human endometrial tissue.
Collapse
Affiliation(s)
- Claire S Barbier
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 27599, USA
| | | | | | | |
Collapse
|
272
|
Endoh T, Tsuji N, Asanuma K, Yagihashi A, Watanabe N. Survivin enhances telomerase activity via up-regulation of specificity protein 1- and c-Myc-mediated human telomerase reverse transcriptase gene transcription. Exp Cell Res 2005; 305:300-11. [PMID: 15817155 DOI: 10.1016/j.yexcr.2004.12.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2004] [Revised: 12/16/2004] [Accepted: 12/20/2004] [Indexed: 01/01/2023]
Abstract
Suppression of apoptosis is thought to contribute to carcinogenesis. Survivin, a member of the inhibitor-of-apoptosis family, blocks apoptotic signaling activated by various cellular stresses. Since elevated expression of survivin observed in human cancers of varied origin was associated with poor patient survival, survivin has attracted growing attention as a potential target for cancer treatment. Immortalization of cells also is required for carcinogenesis; telomere length maintenance by telomerase is required for cancer cells to proliferate indefinitely. Yet how cancer cells activate telomerase remains unclear. We therefore examined possible interrelationships between survivin expression and telomerase activity. Correlation between survivin and human telomerase reverse transcriptase (hTERT) expression was observed in colon cancer tissues, and overexpression of survivin enhanced telomerase activity by up-regulation of hTERT expression in LS180 human colon cancer cells. DNA-binding activities of specificity protein 1 (Sp1) and c-Myc to the hTERT core promoter were increased in survivin gene transfectant cells. Phosphorylation of Sp1 and c-Myc at serine and threonine residues was enhanced by survivin, while total amounts of these proteins were unchanged. Further, "knockdown" of survivin by a small inhibitory RNA decreased Sp1 and c-Myc phosphorylation. Thus survivin participates not only in inhibition of apoptosis, but also in prolonging cellular lifespan.
Collapse
Affiliation(s)
- Teruo Endoh
- Department of Clinical Laboratory Medicine, Sapporo Medical University, School of Medicine, South-1, West-16, Sapporo 060-8543, Japan
| | | | | | | | | |
Collapse
|
273
|
Kang SK, Putnam L, Dufour J, Ylostalo J, Jung JS, Bunnell BA. Expression of telomerase extends the lifespan and enhances osteogenic differentiation of adipose tissue-derived stromal cells. Stem Cells 2005; 22:1356-72. [PMID: 15579653 DOI: 10.1634/stemcells.2004-0023] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Expression of TERT, the catalytic protein subunit of the telomerase complex, can be used to generate cell lines that expand indefinitely and retain multilineage potential. We have created immortal adipose stromal cell lines (ATSCs) by stably transducing nonhuman primate-derived ATSCs with a retroviral vector expressing TERT. Transduced cells (ATSC-TERT) had an increased level of telomerase activity and increased mean telomere length in the absence of malignant cellular transformation. Long-term culture of the ATSC-TERT cells demonstrated that the cells retain the ability to undergo differentiation along multiple lineages such as adipogenic, chondrogenic, and neurogenic. Untransduced cells demonstrated markedly reduced multilineage and self-renewal potentials after 12 passages in vitro. To determine the functional role of telomerase during osteogenesis, we examined osteogenic differentiation potential of ATSC-TERT cells in vitro. Compared with naive ATSCs, which typically begin to accumulate calcium after 3-4 weeks of induction by osteogenic differentiation medium, ATSC-TERT cells were found to accumulate significant amounts of calcium after only 1 week of culture in osteogenic induction medium. The cells have increased production of osteoblastic markers, such as AP2, osteoblast-specific factor 2, chondroitin sulfate proteoglycan 4, and the tumor necrosis factor receptor superfamily, compared with control ATSCs, indicating that telomerase expression may aid in maintaining the osteogenic stem cell pool during in vitro expansion. These results show that ectopic expression of the telomerase gene in nonhuman primate ATSCs prevents senescence-associated impairment of osteoblast functions and that telomerase therapy may be a useful strategy for bone regeneration and repair.
Collapse
Affiliation(s)
- Soo Kyung Kang
- Division of Gene Therapy, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA 70433, USA
| | | | | | | | | | | |
Collapse
|
274
|
Abstract
Telomerase is a ribonucleoprotein that directs the synthesis of telomeric sequence. It is detected in majority of malignant tumors, but not in most normal somatic cells. Because telomerase plays a critical role in cell immortality and tumor formation, it has been one of the targets for anti-cancer and regeneration drug development. In this review, we will discuss therapeutic approaches based mainly on small molecules that have been developed to inhibit telomerase activity, modulate telomerase expression, and telomerase directed gene therapy.
Collapse
Affiliation(s)
- Yi-hsin Hsu
- Institute of Biopharmaceutical Science, National Yang-Ming University, Taipei, Taiwan, China
| | | |
Collapse
|
275
|
Bates SE, Zhou NY, Federico LE, Xia L, O'Connor TR. Repair of cyclobutane pyrimidine dimers or dimethylsulfate damage in DNA is identical in normal or telomerase-immortalized human skin fibroblasts. Nucleic Acids Res 2005; 33:2475-85. [PMID: 15863724 PMCID: PMC1087900 DOI: 10.1093/nar/gki542] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The progression of a normal cell to senescence in vivo and in vitro is accompanied by a reduction in the length of the telomeres, the chromosome capping segments at the end of each linkage group. However, overexpression of the reverse transcriptase subunit (HTERT) of the ribonucleoprotein telomerase restores telomere length and delays cellular senescence. Although some data exist in the literature with respect to survival, no molecular data have shown that DNA repair in telomerase-immortalized cells is normal. Several telomerase-immortalized human skin fibroblast cell lines were constructed from a primary human fibroblast cell line. The primary line and the telomerase-immortalized cell lines were treated with either ultraviolet (UV) radiation or dimethylsulfate (DMS). UV radiation principally produces cyclobutane pyrimidine dimers that are repaired by nucleotide excision repair, whereas DMS introduces mainly N-methylpurines repaired by base excision repair. Here, we show that repair of both types of damage in the telomerase-immortalized human skin fibroblast cell lines is identical to repair observed in normal skin fibroblasts. Thus, telomerase expression and consequent immortalization of skin fibroblasts do not alter nucleotide or base excision repair in human cells.
Collapse
Affiliation(s)
| | | | | | | | - Timothy R. O'Connor
- To whom correspondence should be addressed. Tel: +1 626 301 8220; Fax: +1 626 930 5366;
| |
Collapse
|
276
|
Gil J, Kerai P, Lleonart M, Bernard D, Cigudosa JC, Peters G, Carnero A, Beach D. Immortalization of primary human prostate epithelial cells by c-Myc. Cancer Res 2005; 65:2179-85. [PMID: 15781629 DOI: 10.1158/0008-5472.can-03-4030] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A significant percentage of prostate tumors have amplifications of the c-Myc gene, but the precise role of c-Myc in prostate cancer is not fully understood. Immortalization of human epithelial cells involves both inactivation of the Rb/p16INK4a pathway and telomere maintenance, and it has been recapitulated in culture by expression of the catalytic subunit of telomerase, hTERT, in combination with viral oncoproteins. Here, we show the immortalization of human prostate epithelial cells (HPrEC) by a single genetic event, the expression of the c-Myc oncogene. Myc stabilizes telomere length in HPrEC through up-regulation of hTERT expression and overrides the accumulation of cell cycle inhibitory proteins, such as p16INK4a. Overall, HPrECs expressing c-Myc retain many characteristics of normal cells, such as the induction of a senescence-like growth arrest in response to oncogenic Ras, an intact p53 response, and an absence of gross karyotypic abnormalities. However, HPrECs expressing c-Myc lack a Rb/p16INK4a checkpoint and can be transformed without the need for additional genetic lesions in that pathway. These results give a partial explanation for the physiologic role of c-Myc overexpression in prostate cancer.
Collapse
Affiliation(s)
- Jesús Gil
- Molecular Oncology Laboratory, Cancer Research UK, London Research Institute, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
277
|
Wang J, Feng H, Huang XQ, Xiang H, Mao YW, Liu JP, Yan Q, Liu WB, Liu Y, Deng M, Gong L, Sun S, Luo C, Liu SJ, Zhang XJ, Liu Y, Li DWC. Human telomerase reverse transcriptase immortalizes bovine lens epithelial cells and suppresses differentiation through regulation of the ERK signaling pathway. J Biol Chem 2005; 280:22776-87. [PMID: 15849192 DOI: 10.1074/jbc.m500032200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Telomerase is a specialized reverse transcriptase that extends telomeres of eukaryotic chromosomes. The functional telomerase complex contains a telomerase reverse transcriptase catalytic subunit and a telomerase template RNA. We have previously demonstrated that human telomerase reverse transcriptase (hTERT) catalytic subunit is functionally compatible with a telomerase template RNA from rabbit. In this study, we show that hTERT is also functionally compatible with a telomerase template RNA from bovine. Introduction of hTERT into bovine lens epithelial cells (BLECs) provides the transfected cells telomerase activity. The expressed hTERT in BLECs supports normal growth of the transfected cells for 108 population doublings so far, and these cells are still extremely healthy in both morphology and growth. In contrast, the vector-transfected cells display growth crisis after 20 population doublings. These cells run into cellular senescence due to shortening of the telomeres and also commit differentiation as indicated by the accumulation of the differentiation markers, beta-crystallin and filensin. hTERT prevents the occurrence of both events. By synthesizing new telomere, hTERT prevents replicative senescence, and through regulation of MEK/ERK, protein kinase C, and protein kinase A and eventual suppression of the MEK/ERK signaling pathway, hTERT inhibits differentiation of BLECs. Our finding that hTERT can suppress RAS/RAF/MEK/ERK signaling pathway to prevent differentiation provides a novel mechanism to explain how hTERT regulates cell differentiation.
Collapse
Affiliation(s)
- Juan Wang
- College of Life Sciences, Hunan Normal University, Changsha, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
278
|
Burns JS, Abdallah BM, Guldberg P, Rygaard J, Schrøder HD, Kassem M. Tumorigenic Heterogeneity in Cancer Stem Cells Evolved from Long-term Cultures of Telomerase-Immortalized Human Mesenchymal Stem Cells. Cancer Res 2005; 65:3126-35. [PMID: 15833842 DOI: 10.1158/0008-5472.can-04-2218] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Long-term cultures of telomerase-transduced adult human mesenchymal stem cells (hMSC) may evolve spontaneous genetic changes leading to tumorigenicity in immunodeficient mice (e.g., hMSC-TERT20). We wished to clarify whether this unusual phenotype reflected a rare but dominant subpopulation or if the stem cell origin allowed most cells to behave as cancer stem cells. Cultures of the hMSC-TERT20 strain at population doubling 440 were highly clonogenic (94%). From 110 single-cell clones expanded by 20 population doublings, 6 underwent detailed comparison. Like the parental population, each clone had approximately 1.2 days doubling time with loss of contact inhibition. All retained 1,25-(OH)(2) vitamin D(3)-induced expression of osteoblastic markers: collagen type I, alkaline phosphatase, and osteocalcin. All shared INK4a/ARF gene locus deletion and epigenetic silencing of the DBCCR1 tumor suppressor gene. Despite in vitro commonality, only four of six clones shared the growth kinetics and 100% tumorigenicity of the parental population. In contrast, one clone consistently formed latent tumors and the other established tumors with only 30% penetrance. Changing the in vitro microenvironment to mimic in vivo growth aspects revealed concordant clonal heterogeneity. Latent tumor growth correlated with extracellular matrix entrapment of multicellular spheroids and high procollagen type III expression. Poor tumorigenicity correlated with in vitro serum dependence and high p27(Kip1) expression. Aggressive tumorigenicity correlated with good viability plus capillary morphogenesis on serum starvation and high cyclin D1 expression. Thus, hMSC-TERT20 clones represent cancer stem cells with hierarchical tumorigenicity, providing new models to explore the stem cell hypothesis for cancer.
Collapse
Affiliation(s)
- Jorge S Burns
- Department of Endocrinology and Metabolism and Institute of Pathology, Odense University Hospital, Medical Biotechnology Center, Winsløwparken 25, DK-5000 Odense C, Denmark.
| | | | | | | | | | | |
Collapse
|
279
|
Xu C, Jiang J, Sottile V, McWhir J, Lebkowski J, Carpenter MK. Immortalized fibroblast-like cells derived from human embryonic stem cells support undifferentiated cell growth. ACTA ACUST UNITED AC 2005; 22:972-80. [PMID: 15536188 DOI: 10.1634/stemcells.22-6-972] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human embryonic stem cells (hESCs) have the potential to generate multiple cell types and hold promise for future therapeutic applications. Although undifferentiated hESCs can proliferate indefinitely, hESC derivatives significantly downregulate telomerase and have limited replication potential. In this study we examine whether the replicative lifespan of hESC derivatives can be extended by ectopic expression of human telomerase reverse transcriptase (hTERT), the catalytic component of the telomerase complex. To this end, we have derived HEF1 cells, a fibroblast-like cell type, differentiated from hESCs. Infection of HEF1 cells with a retrovirus expressing hTERT extends their replicative capacity, resulting in immortal human HEF1-hTERT cells. HEF1-hTERT cells can be used to produce conditioned medium (CM) capable of supporting hESC growth under feeder-free conditions. Cultures maintained in HEF1-CM show characteristics similar to mouse embryonic fibroblast CM control cultures, including morphology, surface marker and transcription factor expression, telomerase activity, differentiation, and karyotypic stability. In addition, HEF1-hTERT cells have the capacity to differentiate into cells of the osteogenic lineage. These results suggest that immortalized cell lines can be generated from hESCs and that cells derived from hESCs can be used to support their own growth, creating a genotypically homogeneous system for the culture of hESCs.
Collapse
Affiliation(s)
- Chunhui Xu
- Geron Corporation, 230 Constitution Drive, Menlo Park, California 94025, USA.
| | | | | | | | | | | |
Collapse
|
280
|
Abstract
Long-standing difficulties in the in vitro transformation of human cells have been overcome. Using telomerase, several successful oncogene-mediated transformations of human cells have been reported and the following cellular requirements for human cell transformation have been proposed: the maintenance of telomere sequences, the inactivation of Rb and p53 pathways, the perturbation of protein phosphatase 2A (PP2A) and the expression of activated Ras. Even when all of these requirements are fulfilled, however, the transformed phenotypes of human cells seem to be much less malignant than those of rodent cells meeting the same requirements. This suggests the existence of undefined cell-autonomous mechanisms that render human cells resistant to malignant transformation.
Collapse
Affiliation(s)
- Tsuyoshi Akagi
- Laboratory of Molecular Oncology, Osaka Bioscience Institute, 6-2-4 Furuedai, Suita, Osaka 565-0874, Japan.
| |
Collapse
|
281
|
Nakamura H, Fukami H, Hayashi Y, Tachibana A, Nakatsugawa S, Hamaguchi M, Ishizaki K. Cytotoxic and Mutagenic Effects of Chronic Low-Dose-Rate Irradiation onTERT-Immortalized Human Cells. Radiat Res 2005; 163:283-8. [PMID: 15733035 DOI: 10.1667/rr3310] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
To analyze the genetic effects of low-dose-rate radiation on human cells, we used human telomere reverse transcriptase (TERT)-immortalized fibroblast cells obtained from normal individuals. We studied the effect of low-dose-rate (0.3 mGy/ min) and high-dose-rate (2 Gy/min) radiation on cells in a confluent state. Survival and micronucleus induction frequency showed higher resistance after irradiation at low dose rate than at high dose rate. The survival after 5 Gy of high-dose-rate radiation was 0.01 compared to 0.3 after low-dose-rate irradiation at the same dose. In accordance with this, the level of HPRT mutation induction by low-dose-rate radiation decreased to approximately one-eighth that for high-dose-rate radiation. We then characterized the mutants by multiplex PCR analysis, which showed that the fraction of deletion mutations was lower in the mutant cells induced at low dose rate than at high dose rate. Furthermore, the size of the deletions in mutant cells induced by low-dose-rate radiation appeared to be smaller than those in mutant cells irradiated at high dose rate. Only a few exons were deleted in the former mutants while all exons were deleted in most of the latter mutants. The present study indicates that the genetic effects of low-dose-rate radiation on nonproliferating normal human cells are quantitatively and qualitatively less severe than the effect of high-dose-rate radiation.
Collapse
Affiliation(s)
- Hideaki Nakamura
- Central Laboratory and Radiation Biology, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan.
| | | | | | | | | | | | | |
Collapse
|
282
|
Chai W, Shay JW, Wright WE. Human telomeres maintain their overhang length at senescence. Mol Cell Biol 2005; 25:2158-68. [PMID: 15743814 PMCID: PMC1061618 DOI: 10.1128/mcb.25.6.2158-2168.2005] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2004] [Revised: 10/18/2004] [Accepted: 12/08/2004] [Indexed: 11/20/2022] Open
Abstract
Normal human cells in culture enter replicative senescence after a finite number of population doublings. The exact molecular mechanisms triggering the growth arrest are poorly understood. A recent report on the disappearance of the G-rich 3' telomeric overhang in senescent cells led to the hypothesis that loss of the 3' G-rich overhang is the molecular signal that triggers senescence. Here, we describe a quantitative assay to measure the length of the G-rich 3' telomeric overhangs from cultured cells. Using both this assay and the conventional nondenaturing hybridization assay for measuring G-rich overhangs, we show that normal human fibroblasts can maintain their overhangs at senescence. Furthermore, cells do not lose their overhangs when they bypass senescence after the inactivation of p53 and Rb. We thus conclude that a global reduction in overhang length is not the molecular signal that triggers replicative senescence.
Collapse
Affiliation(s)
- Weihang Chai
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9039, USA
| | | | | |
Collapse
|
283
|
Röth A, Baerlocher GM, Schertzer M, Chavez E, Dührsen U, Lansdorp PM. Telomere loss, senescence, and genetic instability in CD4+ T lymphocytes overexpressing hTERT. Blood 2005; 106:43-50. [PMID: 15741219 PMCID: PMC1895130 DOI: 10.1182/blood-2004-10-4144] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Little is known about the long-term consequences of overexpression of the human telomerase reverse transcriptase (hTERT) gene in T lymphocytes. To address this issue, we transduced polyclonal as well as clonally derived populations of naive and memory CD44 T cells from 2 healthy donors (aged 24 and 34 years) with retroviral vectors encoding green fluorescence protein (GFP) and hTERT (GFP-hTERT) or GFP alone. After transduction, cells were sorted on the basis of GFP expression and cultured in vitro until senescence. T cells transduced with hTERT exhibited high stable telomerase activity throughout the culture period. Relative to GFP controls, minor changes in overall gene expression were observed yet the proliferative lifespan of the hTERT-transduced populations was significantly increased and the rate of telomere loss was lower. Nevertheless, hTERT-transduced cells showed progressive telomere loss and had shorter telomeres at senescence than controls (2.3 +/- 0.3 kilobase [kb] versus 3.4 +/- 0.1 kb). Furthermore, a population of cells with 4N DNA consisting of binucleated cells with connected nuclei emerged in the hTERT-transduced cells prior to senescence. We conclude that overexpression of hTERT in CD4+ T cells provides a proliferative advantage independent of the average telomere length but does not prevent eventual genetic instability and replicative senescence.
Collapse
Affiliation(s)
- Alexander Röth
- Terry Fox Laboratory, British Columbia Cancer Agency, 12th floor, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | | | | | | | | | | |
Collapse
|
284
|
Zanetti M, Hernandez X, Langlade-Demoyen P. Telomerase reverse transcriptase as target for anti-tumor T cell responses in humans. ACTA ACUST UNITED AC 2005; 27:87-104. [PMID: 15711953 DOI: 10.1007/s00281-004-0197-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2004] [Accepted: 12/15/2004] [Indexed: 10/25/2022]
Abstract
Human telomerase reverse transcriptase, a ribonucleoprotein intimately connected with the process of cell immortalization, is overexpressed in the vast majority of cancer cells, irrespective of their histological origin. Telomerase is currently viewed as the first antigen with the characteristics of common tumor antigen in humans, and it constitutes a potentially valuable target for attempts to control tumors through CD8 T cell immunity. Telomerase is a self antigen, making it possible that self tolerance imposes severe restrictions on our ability to generate effective anti-tumor immune responses in humans. In this article we review current studies on the antigenic and immunogenic properties of the human telomerase reverse transcriptase, placing them in the context of self tolerance and the size of the available CD8 T cell repertoire restricted by the HLA A2 molecule.
Collapse
Affiliation(s)
- Maurizio Zanetti
- The Laboratory of Immunology, Department of Medicine and Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0837, USA.
| | | | | |
Collapse
|
285
|
Fu XH, Zhang JS, Zhang N, Zhang YD. Combination of telomerase antisense oligonucleotides simultaneously targeting hTR and hTERT produces synergism of inhibition of telomerase activity and growth in human colon cancer cell line. World J Gastroenterol 2005; 11:785-90. [PMID: 15682468 PMCID: PMC4250584 DOI: 10.3748/wjg.v11.i6.785] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate synergism of inhibition of telomerase activity and proliferation of human colon cancer cells by combination of telomerase antisense oligonucleotides (ASODNs) simultaneously targeting human telomerase RNA (hTR) and human telomerase reverse transcriptase (hTERT) in vitro.
METHODS: ASODN of hTR and ASODN of hTERT were transfected into human colon cancer SW480 cells by liposomal transfection reagents. Telomerase activity of SW480 cells was examined using telomeric repeat amplification protocol (TRAP)-enzyme-linked immunosorbent assay (PCR-ELISA). Proliferation activity of SW480 cells was tested by methyl thiazolyl tetrazolium assay. Apoptosis and cell cycle were analyzed by flow cytometry.
RESULTS: The telomerase activity and cell survival rate in SW480 cells transfected with 0.2 µmol/L of ASODN of hTR or ASODN of hTERT for 24-72 h were significantly decreased in a time-dependent manner compared with those after treatment with sense oligonucleotides and untreated (telomerase activity: 24 h, 73%, 74% vs 99%, 98%; 48 h, 61%, 55% vs 98%, 99%; 72 h, 41%, 37% vs 99%, 97%; P<0.01; cell survival rate: 24 h, 88%, 86% vs 94%, 98%; 48 h, 49%, 47% vs 94%, 97%; 72 h, 44%, 42% vs 92%, 96%; P<0.01). Moreover, the telomerase activity and the cell survival rate in SW480 cells treated by the combination of telomerase anti-hTR and anti-hTERT were more significantly suppressed than single anti-hTR or anti-hTERT (telomerase activity: 24 h, 59% vs 73%, 74%; 48 h, 43% vs 61%, 55%; 72 h, 18% vs 41%, 37%; P<0.01; cell survival rate: 24 h, 64% vs 88%, 86%; 48 h, 37% vs 49%, 47%; 72 h, 25% vs 44%, 42%; P<0.01). Meanwhile, the apoptosis rates in the combination group were markedly increased compared with those in the single group (24 h, 18.0% vs 7.2%, 7.4%; 48 h, 23.0% vs 13.0%, 14.0%; 72 h, 28.6% vs 13.2%, 13.75; P<0.01). Cells in combination group were arrested at G0/G1 phase.
CONCLUSION: Telomerase anti-hRT and anti-hTERT suppress telomerase activity, and inhibit growth of human colon cancer cells probably via induction of apoptosis and retardation of cell cycle. Additionally, combined use of telomerase ASODNs targeting both hTR and hTERT yields synergistic action selective for human colon cancer.
Collapse
Affiliation(s)
- Xiao-Hua Fu
- Medical College, Hunan Normal University, Changsha 410006, Hunan Province, China
| | | | | | | |
Collapse
|
286
|
del Arroyo AG, Peters G. The Ink4a/Arf network--cell cycle checkpoint or emergency brake? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2005; 570:227-47. [PMID: 18727503 DOI: 10.1007/1-4020-3764-3_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
287
|
Smelick C, Ahmed S. Achieving immortality in the C. elegans germline. Ageing Res Rev 2005; 4:67-82. [PMID: 15619471 DOI: 10.1016/j.arr.2004.09.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2004] [Accepted: 09/21/2004] [Indexed: 02/04/2023]
Abstract
Germline immortality is a topic that has intrigued theoretical biologists interested in aging for over a century. The germ cell lineage can be passed from one generation to the next, indefinitely. In contrast, somatic cells are typically only needed for a single generation and are then discarded. Germ cells may, therefore, harbor rejuvenation mechanisms that enable them to proliferate for eons. Such processes are thought to be either absent from or down-regulated in somatic cells, although cell non-autonomous forms of rejuvenation are formally possible. A thorough description of mechanisms that foster eternal youth in germ cells is lacking. The mysteries of germline immortality are being addressed in the nematode Caenorhabditis elegans by studying mutants that reproduce normally for several generations but eventually become sterile. The mortal germline mutants probably become sterile as a consequence of accumulating various forms of heritable cellular damage. Such mutants are abundant, indicating that several different biochemical pathways are required to rejuvenate the germline. Thus, forward genetics should help to define mechanisms that enable the germline to achieve immortality.
Collapse
Affiliation(s)
- Chris Smelick
- Department of Biology, Coker Hall, University of North Carolina, Chapel Hill, NC 27599-3280, USA
| | | |
Collapse
|
288
|
Forsyth NR, Morales CP, Damle S, Boman B, Wright WE, Kopelovich L, Shay JW. Spontaneous immortalization of clinically normal colon-derived fibroblasts from a familial adenomatous polyposis patient. Neoplasia 2004; 6:258-65. [PMID: 15153338 PMCID: PMC1502101 DOI: 10.1593/neo.4103] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Normal human diploid cells do not spontaneously immortalize in culture, but instead enter replicative senescence after a finite number of population doublings. Ablation of key checkpoint arrest or cancer-suppressor genes, through dominantly inherited germline mutation (p53+/-, Li-Fraumeni) or viral oncogene expression (SV40 large T, HPV16/18, and E6/E7) can lead to escape from senescence, additional doublings, and entrance into crisis phase, where immortal clones emerge at low frequency. In the vast majority of cases, telomerase is reactivated and telomeres are stabilized. Here we describe the spontaneous immortalization of clinically normal fibroblasts derived from colonic stroma of a familial adenomatous polyposis (FAP) patient. The preimmortal (C26C) and the spontaneously immortalized derivative (C26Ci) cells are heterozygous for a characterized germline mutation in exon 15 of the adenomatous polyposis coli gene. Immortalization was accompanied by spontaneous reactivation of endogenous telomerase and establishment of telomeres at presenescent lengths. Normal checkpoint behavior is retained and a diploid karyotype is maintained. These cells provide a valuable new addition to the limited number of spontaneously immortalized human cell types, particularly fibroblast cells, and will be useful in experimentally determining the functional pathways in neoplastic development and in the identification of potential molecular targets for cancer chemoprevention.
Collapse
Affiliation(s)
- Nicholas R Forsyth
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | | | | | |
Collapse
|
289
|
Santos JH, Meyer JN, Skorvaga M, Annab LA, Van Houten B. Mitochondrial hTERT exacerbates free-radical-mediated mtDNA damage. Aging Cell 2004; 3:399-411. [PMID: 15569357 DOI: 10.1111/j.1474-9728.2004.00124.x] [Citation(s) in RCA: 187] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Telomerase is often re-activated in human cancers and is widely used to immortalize cells in culture. In addition to the maintenance of telomeres, telomerase has been implicated in cell proliferation, genomic instability and apoptosis. Here we show that human telomerase reverse transcriptase (hTERT) is targeted to the mitochondria by an N-terminal leader sequence, and that mitochondrial extracts contain telomerase activity. In seven different human cell lines, mitochondrial telomerase increases hydrogen-peroxide-mediated mitochondrial DNA damage. hTERT expression did not alter the rate of hydrogen peroxide breakdown or endogenous cellular levels. Because the damaging effects of hydrogen peroxide are mediated by divalent metal ions (Fenton chemistry), we examined the levels of bioavailable metals. In all cases, higher levels of chelatable metals were found in hTERT-expressing cells. These results suggest that mitochondrial telomerase sensitizes cells to oxidative stress, which can lead to apoptotic cell death, and imply a novel function of telomerase in mitochondrial DNA transactions.
Collapse
Affiliation(s)
- Janine Hertzog Santos
- Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences, Natinoal Institutes of Health, 111, Alexander Drive, MD D3-01, Research Triangle Park, NC 27709, USA
| | | | | | | | | |
Collapse
|
290
|
Telomerase-transduced osteoarthritic fibroblast-like synoviocyte cell line. Biochem Biophys Res Commun 2004; 323:1287-92. [PMID: 15451436 DOI: 10.1016/j.bbrc.2004.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2004] [Indexed: 10/26/2022]
Abstract
To examine whether the life span of fibroblast-like synoviocytes (FLSs) can be extended and to establish FLS cell lines that preserve the characteristics of primary FLSs, we introduced human catalytic subunit of telomerase (hTERT) gene into human osteoarthritic (OA) FLSs. Two hTERT-transduced clonal cell lines were established and one line, hTERT-OA FLS 13A, was characterized. The hTERT-OA FLS 13A cells have a morphology similar to that of the parental untransduced cells and a population-doubling time similar to that of the parental cells of early passages. While the parental untransduced OA FLSs reached senescence after 100 days in culture, the hTERT-OA FLS 13A cells continued to grow at a population-doubling rate of once in about every 2-3 days. The hTERT-OA 13A cells have so far grown in culture beyond 450 days and maintained the same growth rate. Furthermore, the hTERT-OA FLS 13A cells preserved their sensitivity and response to the treatment with basic calcium phosphate crystals and interleukin-1beta. In conclusion, exogenous expression of telomerase represents a way to extend the life span of human FLSs and telomerase-transduced FLS cells offer a promising tool for gene regulation, cell-based assay, cell transplantation-based gene therapy, and tissue engineering research and development.
Collapse
|
291
|
Jurczyk A, Gromley A, Redick S, San Agustin J, Witman G, Pazour GJ, Peters DJM, Doxsey S. Pericentrin forms a complex with intraflagellar transport proteins and polycystin-2 and is required for primary cilia assembly. ACTA ACUST UNITED AC 2004; 166:637-43. [PMID: 15337773 PMCID: PMC2172416 DOI: 10.1083/jcb.200405023] [Citation(s) in RCA: 144] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Primary cilia are nonmotile microtubule structures that assemble from basal bodies by a process called intraflagellar transport (IFT) and are associated with several human diseases. Here, we show that the centrosome protein pericentrin (Pcnt) colocalizes with IFT proteins to the base of primary and motile cilia. Immunogold electron microscopy demonstrates that Pcnt is on or near basal bodies at the base of cilia. Pcnt depletion by RNA interference disrupts basal body localization of IFT proteins and the cation channel polycystin-2 (PC2), and inhibits primary cilia assembly in human epithelial cells. Conversely, silencing of IFT20 mislocalizes Pcnt from basal bodies and inhibits primary cilia assembly. Pcnt is found in spermatocyte IFT fractions, and IFT proteins are found in isolated centrosome fractions. Pcnt antibodies coimmunoprecipitate IFT proteins and PC2 from several cell lines and tissues. We conclude that Pcnt, IFTs, and PC2 form a complex in vertebrate cells that is required for assembly of primary cilia and possibly motile cilia and flagella.
Collapse
Affiliation(s)
- Agata Jurczyk
- Department of Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Biotech II, Suite 210, Worcester, MA 01605, USA
| | | | | | | | | | | | | | | |
Collapse
|
292
|
Abstract
Stem cell research is a burgeoning field with an alluring potential for therapeutic intervention, and thus begs a critical understanding of the long-term consequences of stem cell replacement. Operationally, a stem cell may be defined as a rarely dividing cell with the capacity for self-renewal throughout the lifetime of the organism, and an ability to reconstitute its appropriate lineages via proliferation and differentiation. In many differentiated normal and cancer cell types, the maintenance of telomeres plays a pivotal role in their continued division potential. Taken together with the presence of the enzymatic activity responsible for telomere addition, telomerase, in several progenitor cell lineages, it is presumed that telomere maintenance will be critical for the replenishment of stem cells or their successors. The purpose of this review is to discuss the role of telomere length maintenance in self-renewal, and the consequent challenges and potential pitfalls to the manipulation of normal and cancer-derived stem cells.
Collapse
Affiliation(s)
- Lea Harrington
- Department of Medical Biophysics, Ontario Cancer Institute, University of Toronto, 620 University Avenue, Canada M5G 2C1.
| |
Collapse
|
293
|
Abstract
The field of stem cell biology is currently being redefined. Stem cell (hematopoietic and non-hematopoietic) differentiation has been considered hierarchical in nature, but recent data suggest that there is no progenitor/stem cell hierarchy, but rather a reversible continuum. The stem cell (hematopoietic and non-hematopoietic) phenotype, the total differentiation capacity (hematopoietic and non-hematopoietic), gene expression as well as other stem cell functional characteristics (homing, receptor and adhesion molecule expression) vary throughout a cell-cycle transit widely. This seems to be dependent on shifting chromatin and gene expression with cell-cycle transit. The published data on DNA methylation, histone acetylation, and also RNAi, the major regulators of gene expression, conjoins very well and provides an explanation for the major issues of stem cell biology. Those features of stem cells mentioned above can be rather difficult to apprehend when a classical hierarchy biology view is applied, but they become clear and easier to understand once they are correlated with the underlining epigenetic changes. We are entering a new era of stem cell biology the era of "chromatinomics." We are one step closer to the practical use of cellular therapy for degenerative diseases.
Collapse
Affiliation(s)
- Jan Cerny
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
| | | |
Collapse
|
294
|
Abstract
Telomeres are essential for genome stability in all eukaryotes. Changes in telomere functions and the associated chromosomal abnormalities have been implicated in human aging and cancer. Telomeres are composed of repetitive sequences that can be maintained by telomerase, a complex containing a reverse transcriptase (hTERT in humans and Est2 in budding yeast), a template RNA (hTERC in humans and Tlc1 in yeast), and accessory factors (the Est1 proteins and dyskerin in humans and Est1, Est3, and Sm proteins in budding yeast). Telomerase is regulated in cis by proteins that bind to telomeric DNA. This regulation can take place at the telomere terminus, involving single-stranded DNA-binding proteins (POT1 in humans and Cdc13 in budding yeast), which have been proposed to contribute to the recruitment of telomerase and may also regulate the extent or frequency of elongation. In addition, proteins that bind along the length of the telomere (TRF1/TIN2/tankyrase in humans and Rap1/Rif1/Rif2 in budding yeast) are part of a negative feedback loop that regulates telomere length. Here we discuss the details of telomerase and its regulation by the telomere.
Collapse
|
295
|
Shay JW, Wright WE. Senescence and immortalization: role of telomeres and telomerase. Carcinogenesis 2004; 26:867-74. [PMID: 15471900 DOI: 10.1093/carcin/bgh296] [Citation(s) in RCA: 505] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Telomere dynamics are a critical component of both aging and cancer. Telomeres progressively shorten in almost all dividing cells and most human cells do not express or maintain sufficient telomerase activity to fully maintain telomeres. There is accumulating evidence that when only a few telomeres are short, they form end-associations, leading to a DNA damage signal resulting in replicative senescence (a cellular growth arrest, also called the M1 stage). In the absence of cell-cycle checkpoint pathways (e.g. p53 and or p16/Rb), cells bypass M1 senescence and telomeres continue to shorten eventually resulting in crisis (also called the M2 stage). M2 is characterized by many 'uncapped' chromosome ends, end-fusions, chromosome breakage fusion-bridge cycles, mitotic catastrophe and a high fraction of apoptotic cells. In a rare M2 cell, telomerase (a cellular reverse transcriptase) can be reactivated or up-regulated, resulting in indefinite cell proliferation. This cellular immortalization is a potentially rate-limiting step in carcinogenesis that is important for the continuing evolution of most advanced cancers. In this perspective we will present our views on the evidence for telomere dysfunction in aging and in cancer progression. We will argue that telomere shortening in the absence of other alterations may be a potent tumor suppressor mechanism and we will discuss the evidence for and against the major molecular mechanisms proposed to initiate replicative senescence.
Collapse
Affiliation(s)
- Jerry W Shay
- University of Texas Southwestern Medical Center at Dallas, Department of Cell Biology, 5323 Harry Hines Boulevard, Dallas, TX 75390-9039, USA.
| | | |
Collapse
|
296
|
Argilla D, Chin K, Singh M, Hodgson JG, Bosenberg M, de Solórzano CO, Lockett S, DePinho RA, Gray J, Hanahan D. Absence of telomerase and shortened telomeres have minimal effects on skin and pancreatic carcinogenesis elicited by viral oncogenes. Cancer Cell 2004; 6:373-85. [PMID: 15488760 DOI: 10.1016/j.ccr.2004.08.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Revised: 06/16/2004] [Accepted: 08/30/2004] [Indexed: 01/25/2023]
Abstract
The telomere-stabilizing enzyme telomerase is induced in tumors and functionally associated with unlimited replicative potential. To further explore its necessity, transgenic mice expressing SV40 or HPV16 oncogenes, which elicit carcinomas in pancreas and skin, respectively, were rendered telomerase-deficient. Absence of telomerase had minimal impact on tumorigenesis, even in terc(-/-) generations (G5-7) exhibiting shortened telomeres and phenotypic abnormalities in multiple organs. Analyses of chromosomal aberrations were not indicative of telomere dysfunction or increased genomic instability in tumors. Quantitative image analysis of telomere repeat intensities comparing biopsies of skin hyperplasia, dysplasia, and carcinoma revealed that telomere numbers and relative lengths were maintained during progression, implicating a means for preserving telomere repeats and functionality in the absence of telomerase.
Collapse
MESH Headings
- Anaphase
- Animals
- Carcinoma, Squamous Cell/enzymology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Cell Division
- Cell Transformation, Neoplastic
- Chromosomal Instability
- Chromosomes, Mammalian/genetics
- Chromosomes, Mammalian/metabolism
- Disease Progression
- Hybridization, Genetic
- In Situ Hybridization, Fluorescence
- Mice
- Mice, Knockout
- Oncogene Proteins, Viral/genetics
- Pancreatic Neoplasms/enzymology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Phenotype
- Skin Neoplasms/enzymology
- Skin Neoplasms/genetics
- Skin Neoplasms/pathology
- Telomerase/deficiency
- Telomerase/genetics
- Telomerase/metabolism
- Telomere/genetics
- Telomere/metabolism
Collapse
Affiliation(s)
- David Argilla
- Department of Biochemistry and Biophysics, Diabetes Center, and Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
297
|
Magnenat L, Blancafort P, Barbas CF. In vivo selection of combinatorial libraries and designed affinity maturation of polydactyl zinc finger transcription factors for ICAM-1 provides new insights into gene regulation. J Mol Biol 2004; 341:635-49. [PMID: 15288776 DOI: 10.1016/j.jmb.2004.06.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2004] [Revised: 06/11/2004] [Accepted: 06/14/2004] [Indexed: 11/20/2022]
Abstract
Zinc finger DNA-binding domains can be combined to create new proteins of desired DNA-binding specificity. By shuffling our repertoire of modified zinc finger domains to create randomly generated polydactyl zinc finger proteins with transcriptional regulatory domains, we developed large combinatorial libraries of zinc finger transcription factors (TFZFs). Millions of TFZFs can then be simultaneously screened in mammalian cells. Here, we successfully isolated specific TFZFs that significantly positively and negatively modulate the transcription of the ICAM-1 gene in primary and cancer cells, which are relevant to ICAM-1 biology and tumor development. We show that TFZFs can work in a general and in a cell-type specific manner depending on the regulatory domain and the zinc finger protein. We show that a TFZF that interacts directly with the ICAM-1 promoter at an overlapping NF-kappaB binding enhancer can overcome or synergistically cooperate with NF-kappaB induction of ICAM-1. For this TFZF, rational design was used to optimize the binding of the zinc finger protein to its DNA element and the resulting TFZF demonstrated a direct correlation between increased affinity and efficiency of target gene regulation. Thus, combining library and affinity maturation approaches generated superior TFZFs that may find further applications in therapeutic research and in ICAM-1 biology, and also provided novel mechanistic insights into the biology of transcription factors. Transcription factor libraries provide genome-wide approaches that can be applied towards the development of TFZFs specific for virtually any gene or desired phenotype and may lead to the discovery of new genetic functions and pathways.
Collapse
Affiliation(s)
- Laurent Magnenat
- The Skaggs Institute for Chemical Biology and the Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
298
|
Wise SS, Elmore LW, Holt SE, Little JE, Antonucci PG, Bryant BH, Wise JP. Telomerase-mediated lifespan extension of human bronchial cells does not affect hexavalent chromium-induced cytotoxicity or genotoxicity. Mol Cell Biochem 2004; 255:103-11. [PMID: 14971651 DOI: 10.1023/b:mcbi.0000007266.82705.d9] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Hexavalent chromium (Cr(VI)) is a metal of increasing public health concern, as exposure to it is widespread and it is a well-established cause of human bronchial carcinomas and fibrosarcomas. The water-insoluble Cr(VI) salts are potent carcinogens compared to the water soluble salts; yet the genotoxic mechanisms of both may be mediated by soluble Cr(VI) ions. Currently, these mechanisms are poorly understood. Emerging evidence suggests that initial cell culture models used to study the general toxicity of Cr(VI) may be suboptimal for investigating mechanisms specific to human bronchial cells. Accordingly, we have developed a new model system of human bronchial cells by introducing hTERT, the catalytic subunit of human telomerase, into primary human bronchial fibroblasts (PHBF). We have isolated a stable, clonally derived cell line, WHTBF-6, that demonstrate reconstitution of telomerase activity and maintenance of telomere lengths with increasing culture age. WHTBF-6 has been characterized as having an extended in vitro lifespan, a normal growth rate, a normal diploid karyotype that is maintained over time, and exhibits serum-dependent contact-inhibited anchorage-dependent growth. Moreover, we find that both particulate and soluble hexavalent chromium induce a pattern and degree of cytotoxicity and clastogenicity in WHTBF-6 that is similar to the parental PHBF cells. Because telomerase does not compromise growth or the response to Cr(VI), our results indicate that this is an excellent system for studying the mechanisms of Cr(VI) and potentially other carcinogens implicated in the development of lung cancer.
Collapse
Affiliation(s)
- Sandra S Wise
- Laboratory of Environmental and Genetic Toxicology, Department of Epidemiology and Public Health, Division of Environmental Health Sciences, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | | |
Collapse
|
299
|
Nisato RE, Harrison JA, Buser R, Orci L, Rinsch C, Montesano R, Dupraz P, Pepper MS. Generation and characterization of telomerase-transfected human lymphatic endothelial cells with an extended life span. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:11-24. [PMID: 15215158 PMCID: PMC1618539 DOI: 10.1016/s0002-9440(10)63271-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The study of lymphatic endothelial cells and lymphangiogenesis has, in the past, been hampered by the lack of lymphatic endothelial-specific markers. The recent discovery of several such markers has permitted the isolation of lymphatic endothelial cells (LECs) from human skin. However, cell numbers are limited and purity is variable with the different isolation procedures. To overcome these problems, we have transfected human dermal microvascular endothelial cells (HDMVECs) with a retrovirus containing the coding region of human telomerase reverse transcriptase (hTERT), and have produced a cell line, hTERT-HDLEC, with an extended lifespan. hTERT-HDLEC exhibit a typical cobblestone morphology when grown in culture, are contact-inhibited, and express endothelial cell-specific markers. hTERT-HDLEC also express the recognized lymphatic markers, Prox-1, LYVE-1 and podoplanin, as well as integrin alpha9, but do not express CD34. They also form tube-like structures in three-dimensional collagen gels when stimulated with vascular endothelial growth factors -A and -C. Based on these currently recognized criteria, these cells are LEC. Surprisingly, we also found that the widely studied HMEC-1 cell line expresses recognized lymphatic markers; however, these cells are also CD34-positive. In summary, the ectopic expression of hTERT increases the life span of LECs and does not affect their capacity to form tube-like structures in a collagen matrix. The production and characterization of hTERT-HDLEC will facilitate the study of the properties of lymphatic endothelium in vitro.
Collapse
MESH Headings
- Biomarkers
- Cell Division/drug effects
- Cell Line
- Cellular Senescence
- Coculture Techniques
- Collagen Type I/metabolism
- DNA-Binding Proteins
- Endothelium, Lymphatic/cytology
- Endothelium, Lymphatic/enzymology
- Endothelium, Lymphatic/immunology
- Endothelium, Lymphatic/metabolism
- Endothelium, Lymphatic/ultrastructure
- Endothelium, Vascular/cytology
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/ultrastructure
- Fibroblast Growth Factor 2/pharmacology
- Gels
- Humans
- Immunohistochemistry
- Lymphatic Vessels/cytology
- Matrix Metalloproteinases/analysis
- Matrix Metalloproteinases/metabolism
- Recombinant Proteins/pharmacology
- Retroviridae/genetics
- Skin/cytology
- Telomerase/genetics
- Telomerase/metabolism
- Vascular Endothelial Growth Factor A/pharmacology
Collapse
Affiliation(s)
- Riccardo E Nisato
- Department of Morphology, University Medical Center, 1 rue Michel Servet, 1211 Geneva 4, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
300
|
de Magalhães JP, Chainiaux F, de Longueville F, Mainfroid V, Migeot V, Marcq L, Remacle J, Salmon M, Toussaint O. Gene expression and regulation in H2O2-induced premature senescence of human foreskin fibroblasts expressing or not telomerase. Exp Gerontol 2004; 39:1379-89. [PMID: 15489061 DOI: 10.1016/j.exger.2004.06.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2004] [Revised: 05/06/2004] [Accepted: 06/01/2004] [Indexed: 10/26/2022]
Abstract
We compared the DNA-binding activity of transcription factors and gene expression patterns in BJ human diploid fibroblasts (HDFs) expressing or not telomerase (hTERT) in stress-induced premature senescence (SIPS). Senescent BJ cells were also studied. Hydrogen peroxide (H2O2)-induced SIPS modulated gene expression in both BJ and hTERT-BJ1 cells. Increased p21(WAF-1) mRNA level was amongst the common gene expression changes in BJ and hTERT-BJ1 cells induced by SIPS. Telomerase expression markedly changed gene expression in non-stressful conditions. Expression patterns of senescent BJ cells partially overlapped those of BJ and hTERT-BJ1 cells in SIPS. The basal levels of DNA-binding activity of NF-kappaB and phosphorylated ATF-2 were different in BJ and hTERT-BJ1 cells. Both cell lines displayed a higher DNA-binding activity of p53 and HIF-1 72 h after H2O2 exposure. Our results indicate that similar mechanisms involving p21(WAF-1) and probably p53 are at work in BJ and hTERT-BJ1 HDFs under H2O2-induced SIPS, suggesting that generalized DNA damage rather than telomere length/telomerase plays a crucial role in H2O2induced SIPS. We propose that H2O2-induced SIPS involves a rearrangement of proliferative and apoptotic pathways. The marked changes in gene expression induced by telomerase suggest that apart from immortalization of HDFs, telomerase also alters the normal cellular functions but does not protect against SIPS.
Collapse
Affiliation(s)
- João Pedro de Magalhães
- Research Unit on Cellular Biology (URBC), Department of Biology, University of Namur (FUNDP), Rue de Bruxelles, 61, B-5000 Namur, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|