401
|
Abstract
Complex living organisms possess qualities that cannot be reduced to the simple addition of quantities. Among such qualities are a specific form and a specific organization. Thinking about morphological aspects is a prime example of the qualitative approach to biological matters. Such a morphogenetic perspective has been continuously developed, both theoretically and experimentally, along the past century, even though it is now rather marginal within a mainstream dominated by molecular biology. However, the morphogenetic outlook can be applied to the understanding of complex biological phenomena, such as cancer. This phenomenon is currently explained as a cellular problem caused by specific gene mutations and/or specific loss of gene regulation. Nevertheless, cancer is a problem that affects the whole organism. Contemporary research based on the genetic paradigm of cancer causation has led to paradoxes and anomalies that cannot be explained within such a reductionist paradigm. Here it is proposed that real, non-experimental, sporadic cancer may be understood as a conflict between an organized morphology (the organism) and a part of such a morphology that drifts towards an amorphous state (the tumour). Thus, rare, sporadic cancer in children can be the result of early disruption of the developmental constraints before the organism has achieved its morphological maturity. While common sporadic cancer in aged individuals may ensue as a result of the weakening or exhaustion of the developmental constraints that determine the morphological stability of the organism, once the organism is past its reproductive prime.
Collapse
Affiliation(s)
- A Aranda-Anzaldo
- Laboratorio de Biología Molecular, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico.
| |
Collapse
|
402
|
Brookes S, Rowe J, Ruas M, Llanos S, Clark PA, Lomax M, James MC, Vatcheva R, Bates S, Vousden KH, Parry D, Gruis N, Smit N, Bergman W, Peters G. INK4a-deficient human diploid fibroblasts are resistant to RAS-induced senescence. EMBO J 2002; 21:2936-45. [PMID: 12065407 PMCID: PMC126048 DOI: 10.1093/emboj/cdf289] [Citation(s) in RCA: 153] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The CDKN2A tumour suppressor locus encodes two distinct proteins, p16(INK4a) and p14(ARF), both of which have been implicated in replicative senescence, the state of permanent growth arrest provoked in somatic cells by aberrant proliferative signals or by cumulative population doublings in culture. Here we describe primary fibroblasts from a member of a melanoma-prone family who is homozygous for an intragenic deletion in CDKN2A. Analyses of the resultant gene products imply that the cells are p16(INK4a) deficient but express physiologically relevant levels of a frameshift protein that retains the known functions of p14(ARF). Although they have a finite lifespan, the cells are resistant to arrest by oncogenic RAS. Indeed, ectopic expression of RAS and telomerase (hTERT) results in outgrowth of anchorage-independent colonies that have essentially diploid karyotypes and functional p53. We find that in human fibroblasts, ARF is not induced demonstrably by RAS, pointing to significant differences between the proliferative barriers implemented by the CDKN2A locus in different cell types or species.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Radost Vatcheva
- Molecular Oncology and
Human Cytogenetics Laboratories, Cancer Research UK London Research Institute, Lincolns Inn Fields, London WC2A 3PX, UK, NCI-FCRDC, Frederick, MD 21702-1201, DNAX Research Institute, Palo Alto, CA 94304-1104, USA and Department of Dermatology, Leiden University Medical Centre, 2333 AL Leiden, The Netherlands Corresponding author e-mail:
| | - Stewart Bates
- Molecular Oncology and
Human Cytogenetics Laboratories, Cancer Research UK London Research Institute, Lincolns Inn Fields, London WC2A 3PX, UK, NCI-FCRDC, Frederick, MD 21702-1201, DNAX Research Institute, Palo Alto, CA 94304-1104, USA and Department of Dermatology, Leiden University Medical Centre, 2333 AL Leiden, The Netherlands Corresponding author e-mail:
| | - Karen H. Vousden
- Molecular Oncology and
Human Cytogenetics Laboratories, Cancer Research UK London Research Institute, Lincolns Inn Fields, London WC2A 3PX, UK, NCI-FCRDC, Frederick, MD 21702-1201, DNAX Research Institute, Palo Alto, CA 94304-1104, USA and Department of Dermatology, Leiden University Medical Centre, 2333 AL Leiden, The Netherlands Corresponding author e-mail:
| | - David Parry
- Molecular Oncology and
Human Cytogenetics Laboratories, Cancer Research UK London Research Institute, Lincolns Inn Fields, London WC2A 3PX, UK, NCI-FCRDC, Frederick, MD 21702-1201, DNAX Research Institute, Palo Alto, CA 94304-1104, USA and Department of Dermatology, Leiden University Medical Centre, 2333 AL Leiden, The Netherlands Corresponding author e-mail:
| | - Nelleke Gruis
- Molecular Oncology and
Human Cytogenetics Laboratories, Cancer Research UK London Research Institute, Lincolns Inn Fields, London WC2A 3PX, UK, NCI-FCRDC, Frederick, MD 21702-1201, DNAX Research Institute, Palo Alto, CA 94304-1104, USA and Department of Dermatology, Leiden University Medical Centre, 2333 AL Leiden, The Netherlands Corresponding author e-mail:
| | - Nico Smit
- Molecular Oncology and
Human Cytogenetics Laboratories, Cancer Research UK London Research Institute, Lincolns Inn Fields, London WC2A 3PX, UK, NCI-FCRDC, Frederick, MD 21702-1201, DNAX Research Institute, Palo Alto, CA 94304-1104, USA and Department of Dermatology, Leiden University Medical Centre, 2333 AL Leiden, The Netherlands Corresponding author e-mail:
| | - Wilma Bergman
- Molecular Oncology and
Human Cytogenetics Laboratories, Cancer Research UK London Research Institute, Lincolns Inn Fields, London WC2A 3PX, UK, NCI-FCRDC, Frederick, MD 21702-1201, DNAX Research Institute, Palo Alto, CA 94304-1104, USA and Department of Dermatology, Leiden University Medical Centre, 2333 AL Leiden, The Netherlands Corresponding author e-mail:
| | - Gordon Peters
- Molecular Oncology and
Human Cytogenetics Laboratories, Cancer Research UK London Research Institute, Lincolns Inn Fields, London WC2A 3PX, UK, NCI-FCRDC, Frederick, MD 21702-1201, DNAX Research Institute, Palo Alto, CA 94304-1104, USA and Department of Dermatology, Leiden University Medical Centre, 2333 AL Leiden, The Netherlands Corresponding author e-mail:
| |
Collapse
|
403
|
Artandi SE, Alson S, Tietze MK, Sharpless NE, Ye S, Greenberg RA, Castrillon DH, Horner JW, Weiler SR, Carrasco RD, DePinho RA. Constitutive telomerase expression promotes mammary carcinomas in aging mice. Proc Natl Acad Sci U S A 2002; 99:8191-6. [PMID: 12034875 PMCID: PMC123043 DOI: 10.1073/pnas.112515399] [Citation(s) in RCA: 219] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Telomerase is up-regulated in the vast majority of human cancers and serves to halt the progressive telomere shortening that ultimately blocks would-be cancer cells from achieving a full malignant phenotype. In contrast to humans, the laboratory mouse possesses long telomeres and, even in early generation telomerase-deficient mice, the level of telomere reserve is sufficient to avert telomere-based checkpoint responses and to permit full malignant progression. These features in the mouse provide an opportunity to determine whether enforced high-level telomerase activity can serve functions that extend beyond its ability to sustain telomere length and function. Here, we report the generation and characterization of transgenic mice that express the catalytic subunit of telomerase (mTERT) at high levels in a broad variety of tissues. Expression of mTERT conferred increased telomerase enzymatic activity in several tissues, including mammary gland, splenocytes, and cultured mouse embryonic fibroblasts. In mouse embryonic fibroblasts, mTERT overexpression extended telomere lengths but did not prevent culture-induced replicative arrest, thus reinforcing the view that this phenomenon is not related to occult telomere shortening. Robust telomerase activity, however, was associated with the spontaneous development of mammary intraepithelial neoplasia and invasive mammary carcinomas in a significant proportion of aged females. These data indicate that enforced mTERT expression can promote the development of spontaneous cancers even in the setting of ample telomere reserve.
Collapse
Affiliation(s)
- Steven E Artandi
- Department of Adult Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street (M413), Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
404
|
Simonsen JL, Rosada C, Serakinci N, Justesen J, Stenderup K, Rattan SIS, Jensen TG, Kassem M. Telomerase expression extends the proliferative life-span and maintains the osteogenic potential of human bone marrow stromal cells. Nat Biotechnol 2002; 20:592-6. [PMID: 12042863 DOI: 10.1038/nbt0602-592] [Citation(s) in RCA: 624] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human bone marrow stromal cells (hMSCs) were stably transduced by a retroviral vector containing the gene for the catalytic subunit of human telomerase (hTERT). Transduced cells (hMSC-TERTs) had telomerase activity, and the mean telomere length was increased as compared with that of control cells. The transduced cells have now undergone more than 260 population doublings (PD) and continue to proliferate, whereas control cells underwent senescence-associated proliferation arrest after 26 PD. The cells maintained production of osteoblastic markers and differentiation potential during continuous subculturing, did not form tumors, and had a normal karyotype. When implanted subcutaneously in immunodeficient mice, the transduced cells formed more bone than did normal cells. These results suggest that ectopic expression of telomerase in hMSCs prevents senescence-associated impairment of osteoblast functions.
Collapse
Affiliation(s)
- Janne L Simonsen
- Department of Endocrinology and Metabolism, University Hospital of Aarhus, DK-8000 Aarhus C, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
405
|
Cordeiro-Stone M, Frank A, Bryant M, Oguejiofor I, Hatch SB, McDaniel LD, Kaufmann WK. DNA damage responses protect xeroderma pigmentosum variant from UVC-induced clastogenesis. Carcinogenesis 2002; 23:959-65. [PMID: 12082017 DOI: 10.1093/carcin/23.6.959] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Lack of DNA polymerase eta and the attendant defect in bypass replication of pyrimidine dimers induced in DNA by ultraviolet light (UV) underlie the enhanced mutagenesis and carcinogenesis observed in xeroderma pigmentosum variant (XP-V). We investigated whether diploid XP-V fibroblasts growing in culture are also more susceptible to UV-induced clastogenesis than normal human fibroblasts (NHF). This study utilized diploid fibroblasts immortalized by the ectopic expression of human telomerase. The cell lines displayed checkpoint responses to DNA damage comparable with those measured in the parental strains. Shortly after exposure to low doses of UVC (< or =4 J/m2), XP-V cells accumulated daughter strand gaps in excess of normal controls (>25-fold). Daughter strand gaps generated in UV-irradiated S phase cells are potential precursors of chromatid-type chromosomal aberrations. Nonetheless, chromatid-type chromosomal aberrations were only 1.5 to 2 times more abundant in XP-V than in NHF exposed to the same UVC dose. XP-V cells, however, displayed S phase delays at lower doses of UVC and for longer periods of time than NHF. These results support the hypothesis that aberrant DNA structures activate S phase checkpoint responses that increase the time available for postreplication repair. Alternatively, cells that cannot be properly repaired remain permanently arrested and never reach mitosis. These responses protect human cells from chromosomal aberrations, especially when other pathways, such as accurate lesion bypass, are lost.
Collapse
Affiliation(s)
- Marila Cordeiro-Stone
- Department of Pathology and Laboratory Medicine, University of NC at Chapel Hill, Chapel Hill, NC 27599-7525, USA.
| | | | | | | | | | | | | |
Collapse
|
406
|
Nakamura H, Fukami H, Hayashi Y, Kiyono T, Nakatsugawa S, Hamaguchi M, Ishizaki K. Establishment of immortal normal and ataxia telangiectasia fibroblast cell lines by introduction of the hTERT gene. JOURNAL OF RADIATION RESEARCH 2002; 43:167-174. [PMID: 12238331 DOI: 10.1269/jrr.43.167] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
To establish immortal human cells, we introduced the human catalytic subunit of telomerase (hTERT) gene into skin fibroblast cells obtained from normal and ataxia telangiectasia (AT) individuals of Japanese origin. After hTERT introduction, these cells continue to grow beyond a population doubling number of 200 while maintaining their original radiosensitivity. Inductions of p53, phosphorylation of Ser15 in p53, and induction of p21 by X-ray irradiation in immortal cells derived from normal individual were not affected by the hTERT introduction. Both normal and AT immortal cells exhibited an apparent inhibition of growth as original primary cells when they reached confluence. Karyotype analysis has revealed that they are in a diploid range. These results suggest that cells immortalized by hTERT introduction retain their original characteristics except for immortalization, and that they may be useful for analyzing various effects of radiation on human cells.
Collapse
Affiliation(s)
- Hideaki Nakamura
- Central Laboratory and Radiation Biology, Aichi Cancer Center Research Institute, Department of Molecular Pathogenesis, Nagoya University School of Medicine, Nagoya, Japan.
| | | | | | | | | | | | | |
Collapse
|
407
|
Carney SA, Tahara H, Swartz CD, Risinger JI, He H, Moore AB, Haseman JK, Barrett JC, Dixon D. Immortalization of human uterine leiomyoma and myometrial cell lines after induction of telomerase activity: molecular and phenotypic characteristics. J Transl Med 2002; 82:719-28. [PMID: 12065682 DOI: 10.1097/01.lab.0000017499.51216.3e] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
In vitro model systems for studying uterine leiomyomas are limited in that human-derived leiomyoma cells grow poorly in culture compared with normal myometrial cells and begin to senesce early, at approximately passage 10 in our studies. To our knowledge, a good in vitro human-derived cell culturing system for leiomyomas does not exist. In an attempt to fill this void, we have immortalized a uterine leiomyoma cell line by inducing telomerase activity, which allows cells to bypass their normal programmed senescence. Telomerase activity was induced by infecting the target (uterine leiomyoma and normal myometrial) cells with a retroviral vector containing hTERT, the gene for the catalytic subunit of telomerase. Subsequent analysis by RT-PCR and the telomeric repeat amplification protocol assay confirmed expression of the inserted gene and induction of telomerase activity in leiomyoma and myometrial cells. Analysis of cells for estrogen receptor-alpha and progesterone receptor proteins by Western blotting showed no change in expression of these proteins between the immortalized and parental leiomyoma and myometrial cells. Both immortalized and parental myometrial and leiomyoma cells expressed the smooth muscle-specific cytoskeletal protein alpha-actin and were negative for mutant p53 protein as evidenced by immunocytochemical staining. The immortalized leiomyoma and myometrial cells showed no anchorage-independent growth, with the exception of a small subpopulation of immortalized leiomyoma cells at a higher passage that did form two to three small colonies (per 50,000 cells) in soft agar. None of the immortalized cells were tumorigenic in nude mice. In conclusion, our data show the successful insertion of the hTERT gene into leiomyoma and myometrial cells and the immortalization of these cell lines without phenotypic alteration from the parental cell types (up to 200 population doublings). These cells should help to advance research in understanding the molecular pathways involved in the conversion of a normal myometrial cell to a leiomyoma cell and the mechanisms responsible for the growth of uterine leiomyomas. Answers to these questions will undoubtedly lead to the development of more effective treatment and intervention regimens for clinical cases of uterine leiomyoma.
Collapse
Affiliation(s)
- Sara A Carney
- Laboratory of Experimental Pathology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
408
|
Shi S, Gronthos S, Chen S, Reddi A, Counter CM, Robey PG, Wang CY. Bone formation by human postnatal bone marrow stromal stem cells is enhanced by telomerase expression. Nat Biotechnol 2002; 20:587-91. [PMID: 12042862 DOI: 10.1038/nbt0602-587] [Citation(s) in RCA: 289] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human postnatal bone marrow stromal stem cells (BMSSCs) have a limited life-span and progressively lose their stem cell properties during ex vivo expansion. Here we report that ectopic expression of human telomerase reverse transcriptase (hTERT) in BMSSCs extended their life-span and maintained their osteogenic potential. In xenogenic transplants, hTERT-expressing BMSSCs (BMSSC-Ts) generated more bone tissue, with a mineralized lamellar bone structure and associated marrow, than did control BMSSCs. The enhanced bone-forming ability of BMSSC-Ts was correlated with a higher and sustained expression of the early pre-osteogenic stem cell marker STRO-1, indicating that telomerase expression helped to maintain the osteogenic stem cell pool during ex vivo expansion. These results show that telomerase expression can overcome critical technical barriers to the ex vivo expansion of BMSSCs, and suggest that telomerase therapy may be a useful strategy for bone regeneration and repair.
Collapse
Affiliation(s)
- Songtao Shi
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
409
|
Roninson IB. Oncogenic functions of tumour suppressor p21(Waf1/Cip1/Sdi1): association with cell senescence and tumour-promoting activities of stromal fibroblasts. Cancer Lett 2002; 179:1-14. [PMID: 11880176 DOI: 10.1016/s0304-3835(01)00847-3] [Citation(s) in RCA: 323] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
p21(Waf1/Cip1/Sdi1) is best known as a broad-specificity inhibitor of cyclin/cyclin-dependent kinase complexes, but p21 also interacts with many other regulators of transcription or signal transduction. p21 induction, which is mediated by p53 and by p53-independent mechanisms, is essential for the onset of cell cycle arrest in damage response and cell senescence. The effects of p21 knockout in mice and its expression patterns in human cancer are consistent with a role for p21 as both a tumour suppressor and an oncogene. Several functions of p21 are likely to promote carcinogenesis and tumour progression. These include endoreduplication and abnormal mitosis that develop in tumour cells after release from p21-induced growth arrest, the ability of p21 to inhibit apoptosis through several different mechanisms, and its ability to stimulate transcription of secreted factors with mitogenic and anti-apoptotic activities. The latter effects of p21 show close resemblance to paracrine activities of senescent cells and to tumour-promoting functions of stromal fibroblasts. Therapeutic strategies targeting the oncogenic consequences of p21 expression may provide a new approach to chemoprevention and treatment of cancer.
Collapse
Affiliation(s)
- Igor B Roninson
- Department of Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607-7170, USA.
| |
Collapse
|
410
|
Biroccio A, Amodei S, Benassi B, Scarsella M, Cianciulli A, Mottolese M, Del Bufalo D, Leonetti C, Zupi G. Reconstitution of hTERT restores tumorigenicity in melanoma-derived c-Myc low-expressing clones. Oncogene 2002; 21:3011-9. [PMID: 12082531 DOI: 10.1038/sj.onc.1205415] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2001] [Revised: 02/01/2002] [Accepted: 02/20/2002] [Indexed: 12/12/2022]
Abstract
c-Myc is involved in the control of telomerase activity through its ability to induce the expression of the catalytic subunit of the enzyme, the human telomerase reverse transcriptase (hTERT). Our aim was to study whether telomerase plays a critical role in c-Myc-dependent tumorigenicity of melanoma cells. By using M14-derived clones, expressing low levels of c-Myc, we demonstrated that the down-regulation of c-Myc reduced cell proliferation rate, cloning efficiency and tumorigenicity and increased the apoptotic rate. Decreased tumorigenic potential correlated with reduced hTERT gene expression, telomerase activity and telomere shortening. Introduction of wild-type hTERT into these cells increased their proliferation rate and partially re-established their tumorigenic potential, at early passages, even though the apoptotic rate of the population remained unaltered. After several in vitro passages, hTERT-mediated cell proliferation made the tumorigenic potential of the c-Myc low-expressing clones comparable to that of the M14 parental line. Over-expression of the mutant biologically inactive hTERT did not drive cells to proliferate. In conclusion, our results demonstrate that the reconstitution of high levels of telomerase activity reverses the low tumorigenicity due to low c-Myc expression.
Collapse
Affiliation(s)
- Annamaria Biroccio
- Experimental Chemotherapy Laboratory, Regina Elena Cancer Institute, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
411
|
Abstract
Cancer arises from a stepwise accumulation of genetic changes that liberates neoplastic cells from the homeostatic mechanisms that govern normal cell proliferation. In humans, at least four to six mutations are required to reach this state, but fewer seem to be required in mice. By rationalizing the shared and unique elements of human and mouse models of cancer, we should be able to identify the molecular circuits that function differently in humans and mice, and use this knowledge to improve existing models of cancer.
Collapse
Affiliation(s)
- William C Hahn
- Whitehead Institute for Biomedical Research, Massachusetts 02142, USA.
| | | |
Collapse
|
412
|
Nielsen HL, Rønnov-Jessen L, Villadsen R, Petersen OW. Identification of EPSTI1, a novel gene induced by epithelial-stromal interaction in human breast cancer. Genomics 2002; 79:703-10. [PMID: 11991720 DOI: 10.1006/geno.2002.6755] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
During growth, invasion, and metastasis, tumor cells interact extensively with the surrounding stroma. To identify genes that are upregulated during this process, we compared mRNA pooled from tumor cells and fibroblasts cultured separately to mRNA from cells in coculture. Using differential display (DD), a transcript representing a novel gene, designated epithelial-stromal interaction 1 (breast) (EPSTI1), was identified. EPSTI1 showed no homology to any known gene, but matched a cluster of expressed-sequence tags (ESTs). The full-length cDNA of 1508 bp was generated by 5'-RACE, included an open reading frame (ORF) encoding a putative 307-amino-acid protein, and mapped to chromosome 13q13.3. EPSTI1 was highly upregulated in invasive breast carcinomas compared with normal breast. In a tissue mRNA panel the most prominent expression of EPSTI1 was found in placenta. Thus, EPSTI1 is a novel human gene expressed in tissues characterized by extensive epithelial-stromal interaction, and expression of this gene may be a crucial event in invasion and metastasis of cancer.
Collapse
Affiliation(s)
- Helga Lind Nielsen
- Structural Cell Biology Unit, Department of Medical Anatomy A, the Panum Institute, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | | | | | | |
Collapse
|
413
|
McCaul JA, Gordon KE, Clark LJ, Parkinson EK. Telomerase inhibition and the future management of head-and-neck cancer. Lancet Oncol 2002; 3:280-8. [PMID: 12067805 DOI: 10.1016/s1470-2045(02)00729-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Telomeres are tandem repeats of DNA associated with specific proteins. These structures cap eukaryotic chromosomes and maintain the integrity of the chromosome ends. In the germline, telomeres are maintained by the enzyme telomerase, but in normal somatic cells the enzyme's activity is low or undetectable. Human tumours, including squamous-cell carcinoma of the head and neck (SCCHN), need telomerase to maintain telomere function; inhibition of the enzyme can lead to apoptosis. Furthermore, because most tumour cells have very short telomeres, they are more likely to succumb to telomerase inhibition than normal cells. Telomerase is therefore a potential selective anticancer target. The telomere is also involved in the repair of DNA double strand breaks, and telomere dysfunction provokes radiosensitivity. In this review we consider whether manipulation of telomere function may selectively sensitise SCCHN to radiotherapy and discuss the possible pitfalls. We also assess how some conventional treatments may affect the subsequent use of telomerase inhibitors.
Collapse
Affiliation(s)
- James A McCaul
- Maxillofacial surgery, Beatson Institute for Cancer Research, Glasgow, UK.
| | | | | | | |
Collapse
|
414
|
Abstract
The growth and turnover of blood vessels in the skin is fundamental in normal development, wound repair, hair follicle cycling, tumor cell metastasis, and in many different states of cutaneous pathology. Whereas many investigations are focused on mechanisms of angiogenesis in the skin, the influence of cellular aging and replicative senescence (i.e., the inability, after a critical number of population doublings, to replicate) on microvascular remodeling events has received relatively less attention. In this article, we review the clinical and pathologic relationships associated with cutaneous vascular aging and update current knowledge of endothelial cell survival characteristics. A hypothesis is presented in which endothelial cell aging and survival are linked to molecular mechanisms controlling cell proliferation, quiescence, apoptosis, and cellular senescence. We review recent results demonstrating how activation of telomerase in human dermal microvascular endothelial cells affects their durability both in vitro and in vivo and conclude by linking these studies with current concepts involving endothelial cell precursors, control of postnatal somatic cell telomerase activity, and murine model systems.
Collapse
Affiliation(s)
- Edwin Chang
- Department of Dermatology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | |
Collapse
|
415
|
Kudo Y, Hiraoka M, Kitagawa S, Miyauchi M, Kakuo S, Zhao M, Ide T, Takata T. Establishment of human cementifying fibroma cell lines by transfection with temperature-sensitive simian virus-40 T-antigen gene and hTERT gene. Bone 2002; 30:712-7. [PMID: 11996909 DOI: 10.1016/s8756-3282(02)00689-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Human cementifying fibroma (HCF) is a benign fibro-osseous neoplasm of periodontal ligament (PDL) origin containing varying amounts of mineralized material resembling cementum. In the present study, we established cell lines from HCF, which were detected in the mandible of a 54-year-old Japanese man. To obtain immortalized cell clones, we undertook transfection with temperature-sensitive simian virus-40 (SV40) T-antigen and hTERT into HCF cells. Cells transfected with SV40 T-antigen entered "crisis" state between passages 22 and 35, but activation of telomerase by transfection with hTERT in the SV40-transformed HCF cells resulted in bypass of the crisis and maintenance over passage 200. HCF cell lines decreased the expression of SV40 T-antigen and the activity of cell proliferation at a nonpermissive temperature (39 degrees C) in comparison with that at a permissive temperature (33 degrees C). High activities of alkaline phosphatase and mineralization and the expression of type I collagen, osteocalcin, osteopontin, and bone sialoprotein by reverse transcription-polymerase chain reaction (RT-PCR) were observed in HCF cells at 39 degrees C. Overall, these findings suggest that: (i) HCF cell lines may represent a novel in vitro human cell model for the study of the regulatory mechanism of differentiation and proliferation of the human PDL; and (ii) transfection of plasmids encoding the temperature-sensitive SV40 T-antigen gene and hTERT gene may be useful for obtaining immortalized cell lines from benign human tumor and, probably, nonneoplastic human tissues.
Collapse
Affiliation(s)
- Y Kudo
- Department of Oral Pathology, Faculty of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | | | | | | | | | | | | | | |
Collapse
|
416
|
Fajkus J, Simícková M, Maláska J. Tiptoeing to chromosome tips: facts, promises and perils of today's human telomere biology. Philos Trans R Soc Lond B Biol Sci 2002; 357:545-62. [PMID: 12028791 PMCID: PMC1692969 DOI: 10.1098/rstb.2001.1053] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The past decade has witnessed an explosion of knowledge concerning the structure and function of chromosome terminal structures-telomeres. Today's telomere research has advanced from a pure descriptive approach of DNA and protein components to an elementary understanding of telomere metabolism, and now to promising applications in medicine. These applications include 'passive' ones, among which the use of analysis of telomeres and telomerase (a cellular reverse transcriptase that synthesizes telomeres) for cancer diagnostics is the best known. The 'active' applications involve targeted downregulation or upregulation of telomere synthesis, either to mortalize immortal cancer cells, or to rejuvenate mortal somatic cells and tissues for cellular transplantations, respectively. This article reviews the basic data on structure and function of human telomeres and telomerase, as well as both passive and active applications of human telomere biology.
Collapse
Affiliation(s)
- J Fajkus
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Královopolská 135, CZ-612 65 Brno, Czech Republic.
| | | | | |
Collapse
|
417
|
Akiyama M, Yamada O, Kanda N, Akita S, Kawano T, Ohno T, Mizoguchi H, Eto Y, Anderson KC, Yamada H. Telomerase overexpression in K562 leukemia cells protects against apoptosis by serum deprivation and double-stranded DNA break inducing agents, but not against DNA synthesis inhibitors. Cancer Lett 2002; 178:187-97. [PMID: 11867204 DOI: 10.1016/s0304-3835(01)00838-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Telomeres are specialized DNA/protein structures that act as protective caps to prevent end fusions. The maintenance of telomeres is essential for chromosomal stability. Telomerase is regulated by human telomerase reverse transcriptase (hTERT). c-Myc oncoprotein is also implicated in the positive regulation of hTERT expression. We show here that two clones of hTERT-transfected K562 erythroleukemia cells have elongated telomeres (22.5 and 24.0 kb), whereas telomere length of both c-Myc-transfected K562 cells and parental K562 cells is 6.5 kb. Telomerase activity and hTERT mRNA expression increased in hTERT-transfected K562 cells, while the expression levels of telomerase activity and hTERT in c-Myc-transfected K562 cells were similar to that in parental K562 cells, despite an overexpression of c-Myc. Importantly, we found that hTERT-transfected K562 cells are protected against apoptosis induced by serum deprivation and double-stranded DNA break inducing agents (ionizing irradiation, and etoposide (VP-16)), but not against DNA synthesis inhibitors (1-beta-D-arabinofuranosylcytosine and hydroxyurea). These findings suggest that overexpression of telomerase by transfecting hTERT confers telomere-elongation and resistance to double-stranded DNA break inducing agents.
Collapse
Affiliation(s)
- Masaharu Akiyama
- Department of Pediatrics, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo 105-8461, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
418
|
Harvey SA, Jensen KO, Elmore LW, Holt SE. Pharmacological approaches to defining the role of chaperones in aging and prostate cancer progression. Cell Stress Chaperones 2002; 7:230-4. [PMID: 12380692 PMCID: PMC514822 DOI: 10.1379/1466-1268(2002)007<0230:patdtr>2.0.co;2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Sarah A Harvey
- Department of Pharmacology and Toxicology, Medical College of Virginia at Virginia Commonwealth University, Richmond 23298-0662, USA
| | | | | | | |
Collapse
|
419
|
Holyoake TL, Jiang X, Drummond MW, Eaves AC, Eaves CJ. Elucidating critical mechanisms of deregulated stem cell turnover in the chronic phase of chronic myeloid leukemia. Leukemia 2002; 16:549-58. [PMID: 11960331 DOI: 10.1038/sj.leu.2402444] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2001] [Accepted: 10/29/2001] [Indexed: 01/31/2023]
Abstract
Chronic myeloid leukemia (CML) has been studied intensively for many years; yet its treatment remains problematic and its biology remains elusive. In chronic phase, the leukemic clone appears to be maintained by a small number of BCR-ABL-positive hematopoietic stem cells that differentiate normally and amplify slowly. In contrast, as these cells enter the intermediate stages of lineage restriction, their progeny are selectively expanded and generate an enlarged pool of neoplastic progenitors. Recent analyses of purified subsets of primitive CML cells have provided a coherent explanation for this dichotomous behavior of BCR-ABL-positive stem and progenitor cells based on the discovery of an unusual autocrine IL-3/G-CSF mechanism activated in them. This only partially counteracts in vivosignals that maintain normal stem cells in a quiescent state but, when active in CML stem cells, promotes their differentiation in favor of their self-renewal. In more differentiated CML progenitors, the same mechanism has a more potent mitogenic effect which is then extinguished when the cells enter the terminal stages of differentiation. Thus, further expansion of the clone is limited until inevitably additional mutations are acquired that further distort or override the regulatory mechanisms still operative in the chronic phase.
Collapse
Affiliation(s)
- T L Holyoake
- Departments of Medicine and Hematology, Glasgow Royal Infirmary, Glasgow, UK
| | | | | | | | | |
Collapse
|
420
|
Hahn WC, Dessain SK, Brooks MW, King JE, Elenbaas B, Sabatini DM, DeCaprio JA, Weinberg RA. Enumeration of the simian virus 40 early region elements necessary for human cell transformation. Mol Cell Biol 2002; 22:2111-23. [PMID: 11884599 PMCID: PMC133688 DOI: 10.1128/mcb.22.7.2111-2123.2002] [Citation(s) in RCA: 475] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2001] [Revised: 11/29/2001] [Accepted: 01/07/2002] [Indexed: 11/20/2022] Open
Abstract
While it is clear that cancer arises from the accumulation of genetic mutations that endow the malignant cell with the properties of uncontrolled growth and proliferation, the precise combinations of mutations that program human tumor cell growth remain unknown. The study of the transforming proteins derived from DNA tumor viruses in experimental models of transformation has provided fundamental insights into the process of cell transformation. We recently reported that coexpression of the simian virus 40 (SV40) early region (ER), the gene encoding the telomerase catalytic subunit (hTERT), and an oncogenic allele of the H-ras gene in normal human fibroblast, kidney epithelial, and mammary epithelial cells converted these cells to a tumorigenic state. Here we show that the SV40 ER contributes to tumorigenic transformation in the presence of hTERT and oncogenic H-ras by perturbing three intracellular pathways through the actions of the SV40 large T antigen (LT) and the SV40 small t antigen (ST). LT simultaneously disables the retinoblastoma (pRB) and p53 tumor suppressor pathways; however, complete transformation of human cells requires the additional perturbation of protein phosphatase 2A by ST. Expression of ST in this setting stimulates cell proliferation, permits anchorage-independent growth, and confers increased resistance to nutrient deprivation. Taken together, these observations define the elements of the SV40 ER required for the transformation of human cells and begin to delineate a set of intracellular pathways whose disruption, in aggregate, appears to be necessary to generate tumorigenic human cells.
Collapse
Affiliation(s)
- William C Hahn
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
| | | | | | | | | | | | | | | |
Collapse
|
421
|
de Nigris F, Cerutti J, Morelli C, Califano D, Chiariotti L, Viglietto G, Santelli G, Fusco A. Isolation of a SIR-like gene, SIR-T8, that is overexpressed in thyroid carcinoma cell lines and tissues. Br J Cancer 2002; 86:917-23. [PMID: 11953824 PMCID: PMC2364158 DOI: 10.1038/sj.bjc.6600156] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2001] [Accepted: 12/14/2001] [Indexed: 11/19/2022] Open
Abstract
We used subtractive library screening to identify the changes that occur in gene expression during thyroid cell neoplastic transformation. Complementary DNA from normal thyroid cells (HTC 2) was subtracted from a complementary DNA library constructed from a human thyroid papillary carcinoma cell line. The library was screened for genes upregulated in human thyroid papillary carcinoma cell line cells, and several cDNA clones were isolated. One of these clones has a sirtuin core and high homology with the human silent information regulator protein family. This clone, designated "SIR-T8", was overexpressed in human thyroid carcinoma cell lines and tissues, but not in adenomas. The human SIR-T8 protein has a molecular weight of 39 kDa and is primarily located in the cytoplasm under the nuclear membrane. The SIR-T8 gene is located on chromosome 17q25-1.
Collapse
Affiliation(s)
- F de Nigris
- Servizio di Oncologia Sperimentale "E", Istituto per lo Studio e la Cura dei Tumori, Fondazione G. Pascale, 80131 Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
422
|
Helder MN, Wisman GBA, van der Zee GJ. Telomerase and telomeres: from basic biology to cancer treatment. Cancer Invest 2002; 20:82-101. [PMID: 11855380 DOI: 10.1081/cnv-120000370] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The limited capacity to divide is one of the major differences between normal somatic cells and cancerous cells. This 'finite life span' of somatic cells is closely linked to loss of telomeric DNA at telomeres, the 'chromosome caps' consisting of repeated (7TAGGG) sequences., In more than 85% of advanced cancers, this telomeric attrition is compensated by telomerase, 'the immortality enzyme', implying that telomerase inhibition may restore mortality in tumor cells. This review discusses the progress in research on the structure and function of telomeres and the telomerase holoenzyme. In addition, new developments in telomere/telomerase targeting compounds such as antisense oligonucleotides and G-quadruplex stabilizing substances, but also new telomerase expression-related strategies such as telomerase promoter-driven suicide gene therapy and telomerase immunotherapy will be presented. It will be discussed how these data can be implemented in telomerase-directed therapies.
Collapse
Affiliation(s)
- Marco N Helder
- Department of Gynaecological Oncology, University Hospital Groningen, The Netherlands
| | | | | |
Collapse
|
423
|
Piera-Velazquez S, Jimenez SA, Stokes D. Increased life span of human osteoarthritic chondrocytes by exogenous expression of telomerase. ARTHRITIS AND RHEUMATISM 2002; 46:683-93. [PMID: 11920404 DOI: 10.1002/art.10116] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To extend the life span of human osteoarthritic (OA) articular chondrocytes by introduction of the catalytic component of human telomerase while preserving the chondrocyte-specific phenotype. METHODS Human articular chondrocytes were isolated from the femoral head and tibial plateau of patients undergoing knee joint replacement for OA. The chondrocytes were cultured as monolayers and infected with a retroviral telomerase expression construct followed by selection with G418 for 10-14 days. Telomeric-repeat amplification protocol assays and telomere terminal restriction fragment length assays were performed on pools of transduced cells in order to measure telomerase activity and telomere length. Growth kinetics and population doubling capacity were assessed by passaging the cells in monolayer culture. Redifferentiation of the monolayer chondrocyte cultures was induced by transfer to suspension culture on poly-(2-hydroxyethyl-methacrylate) (polyHEMA)-coated dishes. Induction of the chondrocyte-specific phenotype was monitored by analysis of gene expression utilizing reverse transcription-polymerase chain reaction. RESULTS OA chondrocytes isolated from 3 different donors (ages 41, 69, and 75 years) were transduced with a retroviral construct expressing telomerase. After selection, pooled populations of cells from all donors and a clonal cell line from 1 donor expressed telomerase activity and exhibited lengthening of telomeres. Chondrocytes expressing telomerase showed an increase of 5-9 population doublings over 234 days of culture in monolayer. The telomerase-transduced cells recovered a chondrocyte-specific gene expression pattern following culture on polyHEMA-coated dishes. CONCLUSION The exogenous expression of telomerase may represent a way to expand human OA chondrocytes while allowing maintenance of the chondrocyte-specific phenotype. These cells have the potential to be used for restoration of the articular cartilage defects occurring in this disease.
Collapse
|
424
|
Izumi H, Hara T, Oga A, Matsuda K, Sato Y, Naito K, Sasaki K. High telomerase activity correlates with the stabilities of genome and DNA ploidy in renal cell carcinoma. Neoplasia 2002; 4:103-11. [PMID: 11896565 PMCID: PMC1550322 DOI: 10.1038/sj.neo.7900205] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2001] [Accepted: 08/20/2001] [Indexed: 11/09/2022]
Abstract
Malignant tumors have telomerase activity, which is thought to play a critical role in tumor growth. However, the relation between telomerase activity and genomic DNA status in tumor cells is poorly understood. In the present study, we examined telomerase activity in 13 clear cell type renal cell carcinomas (CRCCs) with similar clinicopathologic features by telomeric repeat amplification protocol assay (TRAP). Based on TRAP assay results, we divided the CRCCs into two groups: a high telomerase activity group and a low/no telomerase activity group. We then analyzed genomic aberration, DNA ploidy, and telomere status in these two groups by comparative genomic hybridization (CGH), laser scanning cytometry (LSC), and telomere-specific fluorescence in situ hybridization (T-FISH), respectively. CGH showed the high telomerase activity group to have fewer genomic changes than the low/no telomerase activity group, which had many genomic aberrations. Moreover, with LSC, DNA diploid cells were found more frequently in the high telomerase activity group than in the low/no telomerase activity group. In addition, T-FISH revealed strong telomere signal intensity in the high telomerase activity group compared with that of the low/no telomerase activity group. These results suggest that telomerase activity is linked to genomic DNA status and that high telomerase activity is associated with genomic stability, DNA ploidy, and telomere length in CRCC.
Collapse
Affiliation(s)
- Hideki Izumi
- of Pathology, Yamaguchi University School of Medicine, 1-1-1, Minami Kogushi, Ube-city, Yamaguchi, 755-8505, Japan
| | | | | | | | | | | | | |
Collapse
|
425
|
Schnabl B, Choi YH, Olsen JC, Hagedorn CH, Brenner DA. Immortal activated human hepatic stellate cells generated by ectopic telomerase expression. J Transl Med 2002; 82:323-33. [PMID: 11896211 DOI: 10.1038/labinvest.3780426] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Telomere shortening controls the entry of cells into senescence. Functional expression of the telomerase catalytic subunit (human telomerase reverse transcriptase or hTERT) stabilizes telomere length and extends the life span of various normal human cells. Our aim was to assess the role of telomerase activity and telomere maintenance in regulating the proliferation of activated human hepatic stellate cells (HSCs), to establish an immortal human HSC cell line. Human HSCs were isolated from surgical specimens of normal liver and infected with a retrovirus expressing hTERT. Ectopic expression of hTERT reconstituted telomerase activity and maintained telomere length in human HSCs. Control human HSCs, which were either not infected or infected with a retroviral vector containing only the neomycin resistance gene, showed no detectable telomerase activity and had slightly shortened telomeres. These telomerase-negative HSCs entered a nondividing state after about 9 to 15 passages and senesced. In contrast, telomerase-positive HSCs to date have undergone 69 passages. Telomerase-positive HSCs did not undergo oncogenic transformation and exhibit morphologic and functional characteristics of activated HSCs. Microarray and RT-PCR analysis showed that mRNA expression patterns in telomerase-positive HSCs are very similar to those in activated human HSCs. Plating telomerase-positive HSCs on a basement membrane-like matrix reverts them toward a more quiescent phenotype. In conclusion, introduction of hTERT into activated human HSCs immortalizes them and maintains their activated phenotype. This newly developed cell line will be a useful tool to study the cell biology of human HSCs in culture.
Collapse
Affiliation(s)
- Bernd Schnabl
- Departments of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | |
Collapse
|
426
|
Abstract
Shortening of the telomeric DNA at the chromosome ends is presumed to limit the lifespan of human cells and elicit a signal for the onset of cellular senescence. To continually proliferate across the senescent checkpoint, cells must restore and preserve telomere length. This can be achieved by telomerase, which has the reverse transcriptase activity. Telomerase activity is negative in human normal somatic cells but can be detected in most tumor cells. The enzyme is proposed to be an essential factor in cell immortalization and cancer progression. In this review we discuss the structure and function of telomere and telomerase and their roles in cell immortalization and oncogenesis. Simultaneously the experimental studies of telomerase assays for cancer detection and diagnosis are reviewed. Finally, we discuss the potential use of inhibitors of telomerase in anti-cancer therapy.
Collapse
Affiliation(s)
- Jiao Mu
- International Joint Cancer Institute, Second Military Medical University, Shanghai, China.
| | | |
Collapse
|
427
|
Barradas M, Gonos ES, Zebedee Z, Kolettas E, Petropoulou C, Delgado MD, León J, Hara E, Serrano M. Identification of a candidate tumor-suppressor gene specifically activated during Ras-induced senescence. Exp Cell Res 2002; 273:127-37. [PMID: 11822868 DOI: 10.1006/excr.2001.5434] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Normal cells display protective responses against oncogenes. Notably, oncogenic Ras triggers an irreversible proliferation arrest that is reminiscent of replicative senescence and that is considered a relevant tumor-suppressor mechanism. Here, we have used microarrayed filters to identify genes specifically upregulated in Ras-senescent human fibroblasts. Among the initial set of genes selected from the microarrays, we found the cell-cycle inhibitor p21(Cip1/Waf1), thus validating the potency of the screening to identify markers and mediators of Ras-senescence. A group of six genes, formed by those more highly upregulated during Ras-senescence, was analyzed in further detail to evaluate their specificity. In particular, we examined their expression in cells overexpressing Ras but rendered resistant to Ras-senescence by the viral oncoprotein E1a; also, we have studied their expression during replicative senescence, organismal aging, H(2)O(2)-induced senescence, and DNA damage. In this manner, we have identified a novel gene, RIS1 (for Ras-induced senescence 1), which is not upregulated in association to any of the above-mentioned processes, but exclusively during Ras-senescence. Furthermore, RIS1 is also upregulated by the transcriptional factor Ets2, which is a known mediator of Ras-induced senescence. Interestingly, RIS1 is located at chromosomal position 3p21.3 and, more specifically, it is included in a short segment of just 1 Mb previously defined by other investigators for its tumor-suppressor activity. In summary, we report the identification of a novel gene, RIS1, as a highly specific marker of Ras-induced senescence and a candidate tumor-suppressor gene.
Collapse
Affiliation(s)
- Marta Barradas
- Department of Immunology and Oncology, Spanish National Center of Biotechnology (CSIC), Campus de Cantoblanco, Madrid E-28049, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
428
|
Mergny JL, Riou JF, Mailliet P, Teulade-Fichou MP, Gilson E. Natural and pharmacological regulation of telomerase. Nucleic Acids Res 2002; 30:839-65. [PMID: 11842096 PMCID: PMC100331 DOI: 10.1093/nar/30.4.839] [Citation(s) in RCA: 276] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2001] [Revised: 11/29/2001] [Accepted: 11/29/2001] [Indexed: 01/14/2023] Open
Abstract
The extremities of eukaryotic chromosomes are called telomeres. They have a structure unlike the bulk of the chromosome, which allows the cell DNA repair machinery to distinguish them from 'broken' DNA ends. But these specialised structures present a problem when it comes to replicating the DNA. Indeed, telomeric DNA progressively erodes with each round of cell division in cells that do not express telomerase, a specialised reverse transcriptase necessary to fully duplicate the telomeric DNA. Telomerase is expressed in tumour cells but not in most somatic cells and thus telomeres and telomerase may be proposed as attractive targets for the discovery of new anticancer agents.
Collapse
Affiliation(s)
- Jean-Louis Mergny
- Laboratoire de Biophysique, Muséum National d'Histoire Naturelle, INSERM U 201, CNRS UMR 8646, 43 rue Cuvier, F-75005 Paris, France.
| | | | | | | | | |
Collapse
|
429
|
Venetsanakos E, Mirza A, Fanton C, Romanov SR, Tlsty T, McMahon M. Induction of tubulogenesis in telomerase-immortalized human microvascular endothelial cells by glioblastoma cells. Exp Cell Res 2002; 273:21-33. [PMID: 11795943 DOI: 10.1006/excr.2001.5424] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To facilitate the study of human endothelial cells we have used a replication defective retrovirus encoding the catalytic subunit of telomerase (hTERT) to derive populations of telomerase-immortalized human microvascular endothelial (TIME) cells. Whereas parental HMVECs became senescent on average within 35-45 population doublings (PDs), TIME cells have continued to proliferate for at least 200 PDs. TIME cells express readily detectable telomerase activity but display only a modest increase in telomere length. Karyotypic analysis reveals the cells to have a normal complement of human chromosomes with no evidence of gross genetic abnormalities. Furthermore, TIME cells retain many of the characteristics of the primary endothelial cells from which they were derived. For example, they express a panel of characteristic endothelial cell surface marker proteins such as CD31/PECAM-1 and alpha(v)beta3-integrin. In addition, TIME cells express receptors for low-density lipoprotein (LDL) receptor as they are competent for receptor-mediated endocytosis of fluorescent acetylated LDL. Importantly, when plated on matrigel, TIME cells undergo tubule formation. Moreover, when cocultured in the presence of human glioma cells, but not primary human astrocytes, TIME cells are induced to form stable tubules. Detachment of TIME cells from extracellular matrix leads to a form of programmed cell death known as anoikis. Conditional activation of the protein kinase Akt (Akt:ER*) significantly inhibited the onset of TIME cell anoikis under these conditions. We believe that the ability of hTERT to immortalize primary human endothelial cells, and the fact that such cells retain the endothelial characteristics of the cells from which they were derived, will greatly facilitate the analysis of human endothelial cell biology in vitro.
Collapse
MESH Headings
- Apoptosis
- Blotting, Western
- Brain Neoplasms/blood supply
- Brain Neoplasms/pathology
- Cells, Cultured
- Coculture Techniques
- DNA-Binding Proteins
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/pathology
- Glioblastoma/blood supply
- Glioblastoma/pathology
- Humans
- In Situ Hybridization, Fluorescence
- Karyotyping
- Lipoproteins, LDL
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- Protein Serine-Threonine Kinases
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt
- RNA, Messenger/metabolism
- Receptors, LDL/metabolism
- Receptors, Vitronectin/metabolism
- Retroviridae/genetics
- Telomerase/genetics
- Telomerase/metabolism
- Telomere/metabolism
Collapse
Affiliation(s)
- Eleni Venetsanakos
- Department of Neurological Surgery, Cancer Research Institute, San Francisco, California 94143, USA
| | | | | | | | | | | |
Collapse
|
430
|
Ducrest AL, Szutorisz H, Lingner J, Nabholz M. Regulation of the human telomerase reverse transcriptase gene. Oncogene 2002; 21:541-52. [PMID: 11850779 DOI: 10.1038/sj.onc.1205081] [Citation(s) in RCA: 155] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Most somatic human cells lack telomerase activity because they do not express the telomerase reverse transcriptase (hTERT) gene. Conversely, most cancer cells express hTERT and are telomerase positive. For most tumors it is not clear whether hTERT expression is due to their origin from telomerase positive stem cells or to reactivation of the gene during tumorigenesis. Telomerase negative cells lack detectable cytoplasmic and nuclear hTERT transcripts; in telomerase positive cells 0.2 to 6 mRNA molecules/cell can be detected. This suggests that expression is regulated by changes in the rate of hTERT gene transcription. In tumor cell lines hTERT expression behaves like a recessive trait, indicating that lack of expression in normal cells is due to one or several repressors. Studies with monochromosomal hybrids indicate that several chromosomes may code for such repressors. A number of transcription factors, tumor suppressors, cell cycle inhibitors, cell fate determining molecules, hormone receptors and viral proteins have been implicated in the control of hTERT expression; but these studies have not yet provided a clear explanation for the tumor specific expression of the hTERT gene, and the cis-acting elements which are the targets of repression in normal cells still have to be identified.
Collapse
Affiliation(s)
- Anne-Lyse Ducrest
- Swiss Institute for Experimental Cancer Research (ISREC), CH-1066 Epalinges, Switzerland
| | | | | | | |
Collapse
|
431
|
Abstract
Telomeres are distinctive structures, composed of a repetitive DNA sequence and associated proteins, that cap the ends of linear chromosomes. Telomeres are essential for maintaining the integrity and stability of eukaryotic genomes. In addition, under some circumstances, telomeres can influence cellular gene expression. In mammals, the length, structure, and function of telomeres have been proposed to contribute to cellular and organismal phenotypes associated with cancer and aging. Here, we discuss what is known about the basis for the links between telomeres, aging and cancer, and some of the known and proposed consequences of telomere dysfunction and maintenance for mammalian cells and organisms.
Collapse
Affiliation(s)
- Sahn-ho Kim Sh
- Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, California, CA 94720, USA
| | | | | |
Collapse
|
432
|
Abstract
The molecular cloning of telomerase and telomere components has enabled the analysis and precise manipulation of processes that regulate telomere length maintenance. In mammalian cells and in other organisms, we now recognize that disruption of telomere integrity via any one of a number of perturbations induces chromosome instability and the activation of DNA damage responses. Thus, telomere dysfunction may represent a physiological trigger of the DNA damage or apoptotic response in an analogous fashion to other genotoxic insults that introduce chromosome breaks. Initial studies in mice lacking the murine telomerase RNA and in cells expressing a dominant negative version of the telomere binding protein TRF2 revealed a strong p53-dependent response to telomere dysfunction. Yet, telomere dysfunction exhibits p53-independent effects as well, an observation supported by p53-independent responses to telomere dysfunction in p53 mutant human tumor cell lines and mouse cells. As most tumors are compromised for p53 function, examination of this p53-independent response warrants closer attention. A better understanding of this p53-independent response may prove critical for determining the ultimate utility of telomerase inhibitors in the clinic. This review will summarize our current understanding of the molecular responses to telomere dysfunction in mammalian cells.
Collapse
Affiliation(s)
- Lea Harrington
- Ontario Cancer Institute/Amgen Research Institute, Department of Medical Biophysics/University of Toronto, 620 University Avenue, Toronto, ON, M5G 2C1, Canada
| | | |
Collapse
|
433
|
Toouli CD, Huschtscha LI, Neumann AA, Noble JR, Colgin LM, Hukku B, Reddel RR. Comparison of human mammary epithelial cells immortalized by simian virus 40 T-Antigen or by the telomerase catalytic subunit. Oncogene 2002; 21:128-39. [PMID: 11791183 DOI: 10.1038/sj.onc.1205014] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2001] [Revised: 09/29/2001] [Accepted: 10/01/2001] [Indexed: 12/17/2022]
Abstract
We directly compared two methods of immortalizing human mammary epithelial cells (HMECs). Cells were transfected with an expression plasmid either for hTERT, the catalytic subunit of telomerase, or for the simian virus 40 (SV40) early region genes. Under standard culture conditions, HMECs were not immortalized by hTERT unless they had spontaneously ceased expression of the p16(INK4a) tumor suppressor gene. Untransfected HMECs had low levels of telomerase expression, and immortalization by both methods was associated with an increase in telomerase activity and prevention of telomere shortening. SV40-induced immortalization was accompanied by aberrant differentiation, loss of DNA damage response, karyotypic instability and, in some cases, tumorigenicity. hTERT-immortalized cells had fewer karyotypic changes, but had intact DNA damage responses, and features of normal differentiation. Although SV40-immortalized cells are useful for studies of carcinogenesis, hTERT-immortalized cells retain more properties of normal cells.
Collapse
Affiliation(s)
- Christian D Toouli
- Children's Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, Australia
| | | | | | | | | | | | | |
Collapse
|
434
|
González-Quevedo R, Iniesta P, Morán A, de Juan C, Sánchez-Pernaute A, Fernández C, Torres A, Díaz-Rubio E, Balibrea JL, Benito M. Cooperative role of telomerase activity and p16 expression in the prognosis of non-small-cell lung cancer. J Clin Oncol 2002; 20:254-62. [PMID: 11773177 DOI: 10.1200/jco.2002.20.1.254] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Telomerase activity and p16 expression can be considered two of the most important molecular markers implicated in tumorigenesis. Our main aim was to study the cooperative role of both molecular alterations in the prognosis of patients surgically resected for non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS We have determined telomerase activity and p16 expression in a series of 98 prospectively collected NSCLC specimens obtained from patients who had undergone surgery without other treatment. Telomerase activity was investigated by a telomeric repeat amplification protocol enzyme-linked immunosorbent assay-based procedure, and p16 expression was examined by Western blot. Associations with survival were evaluated. RESULTS Positive results for telomerase activity were found in 82% of the cases, and this variable correlated with poor differentiation and recurrence of tumors. Lack of p16 expression was observed in 61% of tumors, and a significant association with tumor recurrence was also observed. By univariate analysis, both negative telomerase activity and p16-positive expression were significantly correlated with a better prognosis. Moreover, statistics for equality of survival distributions for telomerase, adjusted for p16, indicated a positive interaction between both parameters. For telomerase-positive tumors, p16 expression emerged as a significant independent protective variable, as indicated by Cox multivariate analysis (relative risk [RR], 0.214; P =.014). This protective effect was maintained only for stage I and II tumors (RR, 0.108; P =.046). CONCLUSION These results suggest that the combined telomerase activity and p16 expression analyses may be of prognostic importance in NSCLC, especially for patients affected by stage I and II tumors.
Collapse
Affiliation(s)
- Rosa González-Quevedo
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense
| | | | | | | | | | | | | | | | | | | |
Collapse
|
435
|
Abstract
The telomere-telomerase hypothesis is the science of cellular aging (senescence) and cancer. The ends of chromosomes, telomeres, count the number of divisions a cell can undergo before entering permanent growth arrest. As divisions are being counted, events occur on the cellular and molecular level, which may either delay or hasten this arrest. As humans age, a particular concern is the accumulation of events that lead to the progression of cancer. Telomerase is a mechanism that most normal cells do not possess, but almost all cancer cells acquire, to overcome their mortality and extend their lifespan. This review aims to provide a comprehensive understanding of the role of telomerase in cancer development, progression, diagnosis, and in the future, treatment. The ultimate goal of telomerase research is to use our understanding to develop anti-telomerase therapies, an almost universal tumor target.
Collapse
Affiliation(s)
- Meaghan P Granger
- Department of Cell Biology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9039, USA.
| | | | | |
Collapse
|
436
|
Duan J, Zhang Z, Tong T. Senescence delay of human diploid fibroblast induced by anti-sense p16INK4a expression. J Biol Chem 2001; 276:48325-31. [PMID: 11606567 DOI: 10.1074/jbc.m104814200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
p16(INK4a), a tumor suppressor gene that inhibits cyclin-dependent kinase 4 and cyclin-dependent kinase 6, is also implicated in the mechanisms underlying replicative senescence, because its RNA and protein accumulate as cells approach their finite number of population doublings in tissue culture. To further explore the involvement of p16(INK4a) in replicative senescence, we constructed a retroviral vector containing antisense p16(INK4a), pDOR-ASp16, and introduced it into early passages of human diploid fibroblasts. The introduction of this construct significantly suppressed the expression of wild-type p16(INK4a). It also imposed a finite increase in proliferative life span and significant delay of several other cell senescent features, such as cell flattening, cell cycle arrest, and senescence-associated beta-galactosidase positivity. Moreover, telomere shortening and decline in DNA repair capacity, which normally accompany cell senescence, are also postponed by the ASp16 transfection. The life span of fibroblasts was significantly extended, but the onset of replicative senescence could not be totally prevented. Telomerase could not be activated even though telomere shortening was slowed. These observations suggest that the telomere pathway of senescence cannot be bypassed by ASp16 expression. These data not only strongly support a role for p16(INK4a) in replicative senescence but also raise the possibility of using the antisense p16(INK4a) therapeutically.
Collapse
Affiliation(s)
- J Duan
- Department of Biochemistry and Molecular Biology, Health Science Center, Peking University, Beijing 100083, People's Republic of China
| | | | | |
Collapse
|
437
|
Gerland LM, Ffrench M, Magaud JP. [Cyclin dependent kinase inhibitors and replicative senescence]. PATHOLOGIE-BIOLOGIE 2001; 49:830-9. [PMID: 11776695 DOI: 10.1016/s0369-8114(01)00249-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Replicative senescence is defined for human diploid fibroblasts in culture as a cell growth arrest appearing beyond 50 +/- 10 population doublings and associated with telomeres' shortening. This phenomenon shows an increased expression of growth cell inhibitors: p21Waf1 described as an universal CDK inhibitor and p16INK4a as a specific inhibitor for both G1 phase kinases CDK4 and CDK6. The cell proliferation inhibitor p14ARF, product of INK4a/ARF locus is involved in replicative senescence too. Overexpression or homozygotic deletion of these inhibitors demonstrated their role in senescence induction. These proteins are involved in two different metabolic pathways, the first including p53, represented by E2F, ARF, MDM2, p53, p21Waf1, and the second concerning pRb and p16INK4a. These two pathways present numerous interactions and the polymerase (PARP) in relation with p53 and activated by telomere shortening might represent via p21Waf1 a link between this shortening and cell cycle control. An another metabolic pathway involving PTEN and p27KIP1 is discussed in senescent-like phenotype induction, but its activity in replicative senescent is uncertain.
Collapse
Affiliation(s)
- L M Gerland
- Laboratoire de cytologie analytique (Inserm U 453), faculté de médecine, 69373 Lyon, France
| | | | | |
Collapse
|
438
|
Ouellette MM, Lee K. Telomerase: diagnostics, cancer therapeutics and tissue engineering. Drug Discov Today 2001; 6:1231-1237. [PMID: 11722875 DOI: 10.1016/s1359-6446(01)02052-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The enzyme telomerase has a key role in controlling the lifespan of human cells. It is absent from most somatic tissues but is reactivated in more than 85% of cancers, making the enzyme ideal as a marker of cancer cells and as a therapeutic target. In the context of normal human cells, the enzyme can extend cellular lifespan without causing cancer-associated changes or altering phenotypic properties. This capability could solve a major obstacle in the use of normal human cells for tissue engineering, that is, the induction of cellular senescence.
Collapse
Affiliation(s)
- Michel M. Ouellette
- The Eppley Institute for Research in Cancer and Allied Diseases, The University of Nebraska Medical Center, 986805 Nebraska Medical Center, 68198, tel: +1 402 559 5556 fax: +1 402 559 4651, Omaha, NE, USA
| | | |
Collapse
|
439
|
Aviv H, Khan MY, Skurnick J, Okuda K, Kimura M, Gardner J, Priolo L, Aviv A. Age dependent aneuploidy and telomere length of the human vascular endothelium. Atherosclerosis 2001; 159:281-7. [PMID: 11730807 DOI: 10.1016/s0021-9150(01)00506-8] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
RATIONALE Aneuploidy and telomere length are two major parameters that have been associated with cellular senescence in vitro. In order to explore the role of aneuploidy and telomere length in aging of the human vasculature, we studied these two parameters in direct preparations of endothelial cells of the human abdominal aorta. METHODS Using fluorescent in situ hybridization on 'touch prep' slides, we evaluated aneuploidy of two autosomes (chromosomes 6 and 16) and sex chromosomes in non cultured endothelial cells of the abdominal aorta as a function of the donor's age. RESULTS We found that the frequency of aneuploidy of vascular endothelial cells significantly increased with age. This was expressed by age-dependent tetrasomy (r(s)=0.56, P=0.006 for chromosome 6; and r(s)=0.54, P=0.008 for chromosome 16), and age dependent loss of the Y chromosome (r(s)=0.85, P=0.0003). In addition, we found that telomere length was inversely correlated with age (r=-0.38, P=0.008). DATA INTERPRETATION: These findings suggest that indicators of cellular senescence, earlier observed in vitro, are also expressed in the human vascular endothelium in vivo. Aneuploidy and telomere attrition might thus play a role in the aging of the human vasculature.
Collapse
Affiliation(s)
- H Aviv
- The Center of Human and Molecular Genetics, UMDNJ-New Jersey Medical School, 185 South Orange Avenue, Newark, NJ 07103, USA
| | | | | | | | | | | | | | | |
Collapse
|
440
|
Abstract
Cultivation of primary cells over many generations eventually results in a reproducible loss of proliferative potential that has been termed 'replicative senescence'. Recent work has revealed the heterogeneity of senescence. Importantly, the analysis of the various aspects and types of senescence has turned out to be very informative about numerous in vivo processes, and particularly about carcinogenesis.
Collapse
Affiliation(s)
- M Serrano
- Department of Immunology and Oncology, National Center of Biotechnology (CSIC), Campus de Cantoblanco, Madrid E-28049, Spain.
| | | |
Collapse
|
441
|
Yasunaga Y, Nakamura K, Ko D, Srivastava S, Moul JW, Sesterhenn IA, McLeod DG, Rhim JS. A novel human cancer culture model for the study of prostate cancer. Oncogene 2001; 20:8036-41. [PMID: 11753687 DOI: 10.1038/sj.onc.1205002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2001] [Revised: 08/22/2001] [Accepted: 09/17/2001] [Indexed: 11/08/2022]
Abstract
Research into molecular and genetic mechanisms underlying prostate carcinogenesis would be greatly advanced by in vitro models of prostate tumors representing primary tumors. We have successfully established an immortalized human prostate epithelial (HPE) cell culture derived from a primary tumor with telomerase. The actively proliferating early passaged RC-58T cells were transduced through infection with a retrovirus vector expressing the human telomerase catalytic subunit (hTERT). A high level of telomerase was detected in RC-58T/hTERT cells but not RC-58T cells. RC-58T/hTERT cells are currently growing well at passage 50, whereas RC-58T cells senesced at passage 7. RC-58T/hTERT cells exhibit transformed morphology. More importantly, these immortalized cells showed anchorage-independent growth as they formed colonies in soft agar and grew above the agar layer. Expression of androgen-regulated prostate specific gene NKX3.1 and epithelial specific cytokeratin 8 (CK8) but not prostate specific antigen (PSA) and androgen receptor was detected in RC-58T/hTERT cells. Prostate stem cell antigen (PSCA) and p16 were also expressed in this cell line. RC-58T/hTERT cells showed growth inhibition when exposed to retinoic acid and transforming growth factor (TGF)-beta1 known potent inhibitors of prostate epithelial cell growth. A number of chromosome alterations were observed including the loss of chromosomes Y, 3p, 10p, 17p, 18q and the gain of chromosomes 16 and 20. These results demonstrate that this primary tumor-derived HPE cell line retained its transformed phenotypes and should allow studies to elucidate molecular and genetic alterations involved in prostate cancer. This is the first documented case of an established human prostate cancer cell line from a primary tumor of a prostate cancer patient with telomerase.
Collapse
Affiliation(s)
- Y Yasunaga
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, MD 20814, USA
| | | | | | | | | | | | | | | |
Collapse
|
442
|
Abstract
Defining the ability of simian virus 40 (SV40) to transform human cells has become of even greater importance with the increased understanding that this virus may play a role in some human malignancies. This report documents the requirement for viral small-t (ST) antigen in large-T (LT)-driven transformation of primary fibroblasts, a requirement that cannot be met by a well-known oncogene, c-Ha-ras (EJ-ras), which can cooperate with LT in rodent systems. The cellular gene telomerase is not essential for transformation, although transformed clones are not immortal without it. Similarly, an immortal mesothelial cell line has been developed using LT and telomerase. Immortalized mesothelial cells are morphologically normal, but can be transformed by introduction of ST, or ST + ras, but not by ras alone. It is likely that ST will be required along with LT for transformation of most human cell types.
Collapse
Affiliation(s)
- J Yu
- Department of Microbiology-Immunology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 303 East Chicago Avenue, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
443
|
Keith WN, Jeffry Evans TR, Glasspool RM. Telomerase and cancer: time to move from a promising target to a clinical reality. J Pathol 2001; 195:404-14. [PMID: 11745671 DOI: 10.1002/path.1001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The past 25 years have seen unparalleled advances in our understanding of the molecular basis of cancer. As a result, novel molecular targets have been identified that provide great potential for the development of new cancer diagnostics and therapies. Four key features of cancer cells distinguish them from their normal counterparts: loss of cell-cycle regulation, loss of control over invasion and metastasis, failure of apoptotic mechanisms, and bypass of senescence. This review examines our understanding of the bypass of senescence and the process of immortalization during carcinogenesis. In addition, the realistic opportunities for telomerase in cancer diagnostics and the challenges faced in clinical trial design for telomerase therapeutics are discussed.
Collapse
Affiliation(s)
- W N Keith
- CRC Department of Medical Oncology, University of Glasgow, CRC Beatson Laboratories, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| | | | | |
Collapse
|
444
|
Abstract
Telomeres are the repetitive DNA sequences and specialized proteins that form the distinctive structure that caps the ends of linear chromosomes. Telomeres allow cells to distinguish the chromosome ends from double strand DNA breaks. The telomeric structure prevents the degradation or fusion of chromosome ends, and thus is essential for maintaining the integrity and stability of eukaryotic genomes. In addition, and perhaps less widely appreciated, telomeres may also indirectly influence gene expression. The length, structure and organization of telomeres are regulated by a host of telomere-associated proteins, and can be influenced by basic cellular processes such as cell proliferation, differentiation, and DNA damage. In mammalian cells, telomere length and/or telomere structure have been linked to both cancer and aging. Here, we briefly review what is known about mammalian telomeres and the proteins that associate with them, and discuss the cellular and organismal consequences of telomere dysfunction and the evidence that cells with dysfunctional telomeres can contribute to cancer and aging phenotypes.
Collapse
Affiliation(s)
- J Campisi
- Life Sciences Division, Lawrence Berkeley National Laboratory, Mailstop 84-171, 1 Cyclotron Road, Berkeley, CA 94720, USA.
| | | | | | | |
Collapse
|
445
|
Roninson IB, Broude EV, Chang BD. If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells. Drug Resist Updat 2001; 4:303-13. [PMID: 11991684 DOI: 10.1054/drup.2001.0213] [Citation(s) in RCA: 541] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Inhibition of the program of apoptosis has been reported to have little or no effect on clonogenic survival after treatment with drugs or radiation in several tumor cell lines. A decrease in apoptosis is compensated in such cell lines by an increase in the fractions of cells that undergo permanent growth arrest with phenotypic features of cell senescence, or die through the process of mitotic catastrophe. Most of the tested tumor cell lines have retained the capacity of normal cells to undergo accelerated senescence after treatment with DNA-interactive drugs, ionizing radiation, or cytostatic agents. p53 and p21(Waf1/Cip1/Sdi1) act as positive regulators of treatment-induced senescence, but they are not required for this response in tumor cells. The senescent phenotype distinguishes tumor cells that survived drug exposure but lost the ability to form colonies from those that recover and proliferate after treatment. Although senescent cells do not proliferate, they are metabolically active and may produce secreted proteins with potential tumor-promoting activities. The expression of such proteins is mediated at least in part by the induction of p21(Waf1/Cip1/Sdi1). The other anti-proliferative response of tumor cells is mitotic catastrophe, a form of cell death that results from abnormal mitosis and leads to the formation of interphase cells with multiple micronuclei. Mitotic catastrophe is induced by different classes of cytotoxic agents, but the pathways of abnormal mitosis differ depending on the nature of the inducer and the status of cell-cycle checkpoints. Mitotic catastrophe can also develop as a consequence of aberrant reentry of tumor cells into cell cycle after prolonged growth arrest. Elucidation of the factors that regulate different aspects of treatment-induced senescence and mitotic catastrophe should assist in improving the efficacy and decreasing side effects of cancer therapy.
Collapse
Affiliation(s)
- I B Roninson
- Department of Molecular Genetics, University of Illinois at Chicago, Chicago 60607-7170, USA.
| | | | | |
Collapse
|
446
|
Prasad KN, Hovland AR, Nahreini P, Cole WC, Hovland P, Kumar B, Prasad KC. Differentiation genes: are they primary targets for human carcinogenesis? Exp Biol Med (Maywood) 2001; 226:805-13. [PMID: 11568302 DOI: 10.1177/153537020122600901] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In spite of extensive research in molecular carcinogenesis, genes that can be considered primary targets in human carcinogenesis remain to be identified. Mutated oncogenes or cellular growth regulatory genes, when incorporated into normal human epithelial cells, failed to immortalize or transform these cells. Therefore, they may be secondary events in human carcinogenesis. Based on some experimental studies we have proposed that downregulation of a differentiation gene may be the primary event in human carcinogenesis. Such a gene could be referred to as a tumor-initiating gene. Downregulation of a differentiation gene can be accomplished by a mutation in the differentiation gene, by activation of differentiation suppressor genes, and by inactivation of tumor suppressor genes. Downregulation of a differentiation gene can lead to immortalization of normal cells. Mutations in cellular proto-oncogenes, growth regulatory genes, and tumor suppressor genes in immortalized cells can lead to transformation. Such genes could be called tumor-promoting genes. This hypothesis can be documented by experiments published on differentiation of neuroblastoma (NB) cells in culture. The fact that terminal differentiation can be induced in NB cells by adenosine 3',5'-cyclic monophosphate (cAMP) suggests that the differentiation gene in these cells is not mutated, and thus can be activated by an appropriate agent. The fact that cAMP-resistant cells exist in NB cell populations suggests that a differentiation gene is mutated in these cancer cells, or that differentiation regulatory genes have become unresponsive to cAMP. In addition to cAMP, several other differentiating agents have been identified. Our proposed hypothesis of carcinogenesis can also be applied to other human tumors such as melanoma, pheochromocytoma, medulloblastoma, glioma, sarcoma, and colon cancer.
Collapse
Affiliation(s)
- K N Prasad
- Center for Vitamins and Cancer Research, Department of Radiology, School of Medicine, University of Colorado Health Sciences Center, Denver, CO 80262, USA.
| | | | | | | | | | | | | |
Collapse
|
447
|
Wei W, Hemmer RM, Sedivy JM. Role of p14(ARF) in replicative and induced senescence of human fibroblasts. Mol Cell Biol 2001; 21:6748-57. [PMID: 11564860 PMCID: PMC99853 DOI: 10.1128/mcb.21.20.6748-6757.2001] [Citation(s) in RCA: 175] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Following a proliferative phase of variable duration, most normal somatic cells enter a growth arrest state known as replicative senescence. In addition to telomere shortening, a variety of environmental insults and signaling imbalances can elicit phenotypes closely resembling senescence. We used p53(-/-) and p21(-/-) human fibroblast cell strains constructed by gene targeting to investigate the involvement of the Arf-Mdm2-p53-p21 pathway in natural as well as premature senescence states. We propose that in cell types that upregulate p21 during replicative exhaustion, such as normal human fibroblasts, p53, p21, and Rb act sequentially and constitute the major pathway for establishing growth arrest and that the telomere-initiated signal enters this pathway at the level of p53. Our results also revealed a number of significant differences between human and rodent fibroblasts in the regulation of senescence pathways.
Collapse
Affiliation(s)
- W Wei
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912, USA
| | | | | |
Collapse
|
448
|
Samper E, Flores JM, Blasco MA. Restoration of telomerase activity rescues chromosomal instability and premature aging in Terc-/- mice with short telomeres. EMBO Rep 2001; 2:800-7. [PMID: 11520856 PMCID: PMC1084029 DOI: 10.1093/embo-reports/kve174] [Citation(s) in RCA: 205] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Reconstitution of telomerase activity is proposed as a potential gene therapy to prevent, or rescue, age-related diseases produced by critical telomere shortening. However, it is not known whether or not short telomeres are irreversibly damaged. We addressed this by re-introducing telomerase in late generation telomerase-deficient mice, Terc-/-, which have short telomeres and show severe proliferative defects. For this, we have crossed these mice with Terc+/- mice and analyzed telomere length, chromosomal instability and premature aging of the progeny. The Terc-/- progeny had one set of chromosomes with normal telomeres, whereas the other set remained with critically short telomeres; these mice presented chromosomal instability and premature aging. In contrast, Terc+/- progeny showed all chromosomes with detectable telomeres, and did not show chromosomal instability or premature aging. These results prove that critically short telomeres can be rescued by telomerase, and become fully functional, thus rescuing premature aging. This has important implications for the future design of telomerase-based gene therapy of age-related diseases.
Collapse
Affiliation(s)
- E Samper
- Department of Immunology and Oncology, National Centre of Biotechnology, E-28049 Madrid, Spain
| | | | | |
Collapse
|
449
|
Lorenz M, Saretzki G, Sitte N, Metzkow S, von Zglinicki T. BJ fibroblasts display high antioxidant capacity and slow telomere shortening independent of hTERT transfection. Free Radic Biol Med 2001; 31:824-31. [PMID: 11557321 DOI: 10.1016/s0891-5849(01)00664-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Human foreskin BJ fibroblasts are well protected against oxidative stress as shown by their low intracellular peroxide content, low levels of protein carbonyls, and low steady-state lipofuscin content as compared to other primary human fibroblasts. This correlates with a long replicative life span of the parental cells of about 90 population doublings and a telomere-shortening rate of only 15-20 bp/PD. This value might define the upper limit of a telomere-shortening rate that can still be explained by the end replication problem alone. In BJ clones immortalized by transfection with hTERT, the catalytic subunit of telomerase, the same telomere-shortening rate as in parental cells is observed over a long time despite strong telomerase activity. Hyperoxia, which induces oxidative stress and accelerates telomere shortening in a variety of human fibroblast strains, does not do so in BJ cells. It is possible that the high antioxidative capacity of BJ cells, by minimizing the accumulation of genomic damage, is instrumental in the successful immortalization of these cells by telomerase.
Collapse
Affiliation(s)
- M Lorenz
- Institute of Pathology, Charite, Humboldt University, Berlin, Germany
| | | | | | | | | |
Collapse
|
450
|
Anisimov VN. Mutant and genetically modified mice as models for studying the relationship between aging and carcinogenesis. Mech Ageing Dev 2001; 122:1221-55. [PMID: 11438116 DOI: 10.1016/s0047-6374(01)00262-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Increased interest is emerging in using mouse models to assess the genetics of aging and age-related diseases, including cancer. However, only limited information is available regarding the relationship between aging and spontaneous tumor development in genetically modified mice. Analysis of various transgenic and knockout rodent models with either a shortened or an extended life span, provides a unique opportunity to evaluate interactions of genes involved in the aging process and carcinogenesis. There are only a few models which show life span extension. Ames dwarf mutant mice, p66(-/-) knockout mice, alpha MUPA and MGMT transgenic mice live longer than wild-type strains. The incidence of spontaneous tumors in these mutant mice was usually similar to those in controls, whereas the latent period of tumor development was increased. Practically all models of accelerated aging showed increased incidence and shorter latency of tumors. This phenomenon has been observed in animals which display a phenotype that more closely resembles natural aging, and in animals which manifest only some features of the normal aging process. These observations are in agreement with an earlier established positive correlation between tumor incidence and the rate of tumor incidence increase associated with aging and the aging rate in a population. Thus, genetically modified animals are a valuable tool in unravelling mechanisms underlying aging and cancer. Systemic evaluation of newly generated models should include onco-gerontological studies.
Collapse
Affiliation(s)
- V N Anisimov
- Department of Carcinogenesis and Oncogerontology, N.N. Petrov Research Institute of Oncology, Pesochny-2, 197758, St Petersburg, Russia.
| |
Collapse
|