We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Spontaneous reversal of the developmental aging of normal human cells following transcriptional reprogramming

    H Vaziri

    Homayoun Vaziri Ontario Cancer Institute/PMH, University of Toronto, Canada

    ,
    KB Chapman

    Karen B Chapman BioTime, Inc., Alameda, CA 94502, USA

    ,
    A Guigova

    Andriana Guigova Ontario Cancer Institute/PMH, University of Toronto, Canada

    ,
    J Teichroeb

    Jonathan Teichroeb Ontario Cancer Institute/PMH, University of Toronto, Canada

    ,
    MD Lacher

    Markus D Lacher BioTime, Inc., Alameda, CA 94502, USA

    ,
    H Sternberg

    Hal Sternberg BioTime, Inc., Alameda, CA 94502, USA

    ,
    I Singec

    Ilyas Singec Burnham Institute for Medical Research, La Jolla, CA, USA

    ,
    L Briggs

    Laura Briggs Sierra Sciences, Reno NV, USA

    ,
    J Wheeler

    Jessica Wheeler Sierra Sciences, Reno NV, USA

    ,
    J Sampathkumar

    Janani Sampathkumar BioTime, Inc., Alameda, CA 94502, USA

    ,
    R Gonzalez

    Rodolfo Gonzalez The Scripps Research Institute, La Jolla, CA 92037, USA

    ,
    D Larocca

    David Larocca Mandala Biosciences, LLC, La Jolla, CA, USA

    ,
    J Murai

    James Murai BioTime, Inc., Alameda, CA 94502, USA

    ,
    E Snyder

    Evan Snyder Burnham Institute for Medical Research, La Jolla, CA, USA

    ,
    WH Andrews

    William H Andrews Sierra Sciences, Reno NV, USA

    ,
    WD Funk

    Walter D Funk BioTime, Inc., Alameda, CA 94502, USA

    &
    MD West

    † Author for correspondence

    Michael D West BioTime, Inc., Alameda, CA 94502, USA.

    Published Online:https://doi.org/10.2217/rme.10.21

    Aim: To determine whether transcriptional reprogramming is capable of reversing the developmental aging of normal human somatic cells to an embryonic state. Materials & methods: An isogenic system was utilized to facilitate an accurate assessment of the reprogramming of telomere restriction fragment (TRF) length of aged differentiated cells to that of the human embryonic stem (hES) cell line from which they were originally derived. An hES-derived mortal clonal cell strain EN13 was reprogrammed by SOX2, OCT4 and KLF4. The six resulting induced pluripotent stem (iPS) cell lines were surveyed for telomere length, telomerase activity and telomere-related gene expression. In addition, we measured all these parameters in widely-used hES and iPS cell lines and compared the results to those obtained in the six new isogenic iPS cell lines. Results: We observed variable but relatively long TRF lengths in three widely studied hES cell lines (16.09–21.1 kb) but markedly shorter TRF lengths (6.4–12.6 kb) in five similarly widely studied iPS cell lines. Transcriptome analysis comparing these hES and iPS cell lines showed modest variation in a small subset of genes implicated in telomere length regulation. However, iPS cell lines consistently showed reduced levels of telomerase activity compared with hES cell lines. In order to verify these results in an isogenic background, we generated six iPS cell clones from the hES-derived cell line EN13. These iPS cell clones showed initial telomere lengths comparable to the parental EN13 cells, had telomerase activity, expressed embryonic stem cell markers and had a telomere-related transcriptome similar to hES cells. Subsequent culture of five out of six lines generally showed telomere shortening to lengths similar to that observed in the widely distributed iPS lines. However, the clone EH3, with relatively high levels of telomerase activity, progressively increased TRF length over 60 days of serial culture back to that of the parental hES cell line. Conclusion: Prematurely aged (shortened) telomeres appears to be a common feature of iPS cells created by current pluripotency protocols. However, the spontaneous appearance of lines that express sufficient telomerase activity to extend telomere length may allow the reversal of developmental aging in human cells for use in regenerative medicine.

    Bibliography

    • Weismann A: Essays Upon Heredity and Kindred Biological Problems (Vol. I). Clarendon Press, London, UK (1891).
    • McLaren A: Embryology. The quest for immortality. Nature359,482–483 (1992).
    • McLaren A: Mammalian germ cells: birth, sex, and immortality. Cell Struct. Funct.26,119–122 (2001).
    • Hayflick L, Moorhead PS: The serial cultivation of human diploid cell strains. Exp. Cell Res.25,585–621 (1961).
    • Hayflick L: The limited in vitro lifetime of human diploid cell strains. Exp. Cell Res.37,614–636 (1965).
    • Hayflick L: Aging, longevity, and immortality in vitro. Exp. Gerontol.27,363–368 (1992).
    • Olovnikov AM: Principles of marginotomy in template synthesis of polynucleotides. Dokl. Akad. Nauk SSSR201,1496–1499 (1971).
    • Cooke HJ, Smith BA: Variability at the telomeres of the human X/Y pseudoautosomal region. Cold Spring Harb. Symp. Quant. Biol.51(Pt 1),213–219 (1986).
    • Harley CB, Futcher AB, Greider CW: Telomeres shorten during ageing of human fibroblasts. Nature345,458–460 (1990).
    • 10  Feng J, Funk WD, Wang S-S et al.: The RNA component of human telomerase. Science269,1236–1241 (1995).
    • 11  Nakamura TM, Morin GB, Chapman KB et al.: Telomerase catalytic subunit homologs from fission yeast and human. Science277,955–959 (1997).
    • 12  Bodnar AG, Ouellette M, Frolkis M et al.: Extension of cell life-span by introduction of telomerase into normal human cells. Science279,349–352 (1998).
    • 13  Vaziri H, Benchimol S: Reconstitution of telomerase activity in normal human cells leads to elongation of telomeres and extended replicative life span. Curr. Biol.8,279–282 (1998).
    • 14  West MD: Embryonic stem cells: Prospects of regenerative medicine for the treatment of human aging. In: The Future of Aging. Fahy GM, West MD, Coles LS, Harris SB (Eds). Springer, NY, USA (2010) (In Press).
    • 15  Shawi M, Autexier C: Telomerase, senescence, and ageing. Mech. Ageing Dev.129,3–10 (2008).
    • 16  Shay JW, Wright WE: Use of telomerase to create bioengineered tissues. Ann. NY Acad. Sci.1057,479–491 (2005).
    • 17  Huang Q, Chen M: Improving cell therapy – experiments using transplanted telomerase-immortalized cells in immunodeficient mice. Mech. Ageing Dev.128(1),25–30 (2007).
    • 18  Kim NW, Piatyszek MA, Prowse KR et al.: Specific association of human telomerase activity with immortal cells and cancer. Science266,2011–2014 (1994).
    • 19  Thomson JA, Itskovitx-Eldor J, Shapiro SS et al.: Embryonic stem cell lines derived from human blastocysts. Science282,1145–1147 (1998).
    • 20  Shamblott MJ, Axelman J, Wang S et al.: Derivation of pluripotent stem cells from cultured human primordial germ cells. Proc. Natl Acad. Sci. USA95,13726–13731 (1998).
    • 21  Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH: Viable offspring derived from fetal and adult mammalian cells. Nature385(6619),810–813 (1997).
    • 22  Lanza RP, Cibelli JB, West MD: Prospects for the use of nuclear transfer in human transplantation. Nat. Biotechnol.17(12),1171–1174 (1999).
    • 23  Lanza RP, Cibelli JB, West MD: Human therapeutic cloning. Nat. Med.5(9),975–977 (1999).
    • 24  Shiels PG, Kind AJ, Campbell KH et al.: Analysis of telomere lengths in cloned sheep. Nature399(6734),316–317 (1999).
    • 25  Lanza RP, Cibelli JB, Blackwell C et al.: Extension of cell life-span and telomere length in animals cloned from senescent somatic cells. Science288,665–669 (2000).
    • 26  Wakayama T, Shinkai Y, Tamashiro KL et al.: Cloning of mice to six generations. Nature407(6802),318–319 (2000).
    • 27  Clark AJ, Ferrier P, Aslam S et al.: Proliferative lifespan is conserved after nuclear transfer. Nat. Cell. Biol.5(6),535–538 (2003).
    • 28  Cibelli JB, Kiessling AA, Cunniff K, Richards C, Lanza RP, West MD: Somatic cell nuclear transfer in humans: Pronuclear and early embryonic development. J. Regen. Med.2,25–31 (2001).
    • 29  Takahashi K, Tanabe K, Ohnuki M et al.: Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell131,861–872 (2007).
    • 30  Yu J, Vodyanik MA, Smuga-Otto K et al.: Induced pluripotent stem cell lines derived from human somatic cells. Science318,1917–1920 (2007).
    • 31  Marion RM, Strati K, Li H et al.: Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells. Cell Stem Cell4(2),141–154 (2009).
    • 32  Suhr ST, Chang EA, Rodriguez RM et al.: Telomere dynamics in human cells reprogrammed to pluripotency. PLoS One4(12),e8124 (2009).
    • 33  Feng Q, Lu S-J, Klimanskaya I et al.: Hemangioblastic derivatives from human induced pluripotent stem cells exhibit limited expansion an early senescence. Stem Cells (2010) (Epub ahead of print).
    • 34  Agarwal S, Loh YH, McLoughlin EM et al.: Telomere elongation in induced pluripotent stem cells from dyskeratosis congenital patients. Nature (2010) (Epub ahead of print).
    • 35  Levy MZ, Allsopp RC, Futcher AB, Greider CW, Harley CB: Telomere end-replication problem and cell aging. J. Mol. Biol.225,951–960 (1992).
    • 36  Riethman H, Ambrosini A, Paul S: Human subtelomere structure and variation. Chrom. Res.13(5),505–515 (2005).
    • 37  Rosler ES, Fisk GJ, Ares X et al.: Long-term culture of human embryonic stem cells in feeder-free conditions. Dev. Dyn.229,259–274 (2004).
    • 38  Lund RD, Wang S, Klimanskaya I et al.: Human embryonic stem cell-derived cells rescue visual function in dystrophic RCS rats. Cloning Stem Cells8(3),189–199 (2006).
    • 39  Park IH, Zhao R, West JA et al.: Reprogramming of human somatic cells to pluripotency with defined factors. Nature451(7175),141–146 (2007).
    • 40  Chiu CP: Differential expression of telomerase activity in hematopoietic progenitors from adult human bone marrow. Stem Cells14,239–248 (1996).
    • 41  Mantell LL, Greider CW: Telomerase activity in germline and embryonic cells of Xenopus. EMBO J.13,3211–3217 (1994).
    • 42  Schaetzlein S: Telomere length is reset during early mammalian embryogenesis. Proc. Natl Acad. Sci. USA101,8034–8038 (2004).
    • 43  Wright WE, Piatyszek MA, Rainey WE, Byrd W, Shay JW: Telomerase activity in human germline and embryonic tissues and cells. Dev. Genet.18,173–179 (1996).
    • 44  Shore D, Bianchi A: Telomere length regulation: coupling DNA end processing to feedback regulation of telomerase. EMBO J.28(16),2309–2322 (2009).
    • 45  Gonzalo S, Jaco I, Fraga MF et al.: DNA methyltransferases control telomere length and telomere recombination in mammalian cells. Nat. Cell. Biol.8(4),416–424 (2006).
    • 46  West MD, Sargent RG, Long J et al.: The ACTCellerate Initiative: large-scale combinatorial cloning of novel human embryonic stem cell derivatives. Regen. Med.3(3),287–308 (2008).
    • 47  Vaziri H: Loss of telomeric DNA during aging of normal and trisomy 21 human lymphocytes. Am. J. Hum. Genet.52,661–667 (1993).
    • 48  Allsopp RC: Telomere length predicts replicative capacity of human fibroblasts. Proc. Natl Acad. Sci. USA89,10114–10118 (1992).
    • 49  Shaikh TH, Gai X, Perin JC et al.: High-resolution mapping and analysis of copy number variations in the human genome: a data resource for clinical and research applications. Genome Res.19(9),1682–1690 (2009).
    • 50  Wang J, Xie LY, Allan S, Beach D, Hannon GJ: Myc activates telomerase. Genes Dev.12(12),1769–1774 (1998).
    • 51  van Steensel B, de Lange T: Control of telomere length by the human telomeric protein TRF1. Nature385,740–743 (1997).
    • 52  Sullivan EJ, Kasinathan S, Kasinathan P, Robl JM, Collas P: Cloned calves from chromatin remodeled in vitro. Biol. Reprod.70(1),146–153 (2004).
    • 53  Freberg CT, Dahl JA, Timoskainen S, Collas P: Epigenetic reprogramming of OCT4 and NANOG regulatory regions by embryonal carcinoma cell extract. Mol. Biol. Cell18,1543–1553 (2007).
    • 54  Binet R, Ythier D, Robles AI et al.: WNT16B is a new marker of cellular senescence that regulates p53 activity and the phosphoinositide 3-kinase/AKT pathway. Cancer Res.69(24),9183–9191 (2009).
    • 55  Hu BY, Weick JP, Yu J et al.: Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency. Proc. Natl Acad. Sci. USA107(9),4335–4340 (2010).
    • 56  Lee HW, Blasco MA, Gottlieb GJ, Horner JW 2nd, Greider CW, DePinho RA: Essential role of mouse telomerase in highly proliferative organs. Nature392,569–574 (1998).
    • 57  Halliday TR: Reptiles & Amphibians. Adler K, Halliday TR (Eds). Torstar Books, NY, USA 101 (1986).
    • 58  Jang G, Hong SG, Oh HJ et al.: A cloned toy poodle produced from somatic cells derived from an aged female dog. Theriogenology69(5),556–563 (2008).
    • 59  Maynard Smith J: Review lectures on senescence: I. The causes of ageing. Proc. R. Soc. Lond. B1962157, 115–127 (1962).
    • 60  Drmanac R, Sparks AB, Callow MJ et al.: Human genome sequencing using unchained base reads on self-assembling DNA nanoarrays. Science327(5961),78–81 (2010).
    • 61  Cibelli JB, Campbell KH, Seidel GE, West MD, Lanza RP: The health profile of cloned animals. Nat. Biotech.20,13–14 (2002).
    • 62  Kishigami S, Wakayama S, Hosoi Y, Iritani A, Wakayama T: Somatic cell nuclear transfer: Infinite reproduction of a unique diploid genome. Exp. Cell Res.314,1945–1950 (2008).
    • 63  Friedman DS, O’Colmain BJ, Munoz B et al.: Prevalence of age-related macular degeneration in the United States. Arch. Ophthalmol.122,564–572 (2004).
    • 64  Cawthon RM, Smith KR, O’Brien E, Sivatchenko A, Kerber RA: Association between telomere length in blood and mortality in people aged 60 years or older. Lancet361,393–395 (2003).
    • 65  American Heart Association: Heart disease and stroke statistics – 2006 update. A report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation113,e85–e151 (2008).
    • 66  Butler RN: The Longevity Revolution: The Benefits and Challenges of Living a Long Life. Public Affairs, NY, USA (2008).
    • 67  Klimanskaya I, Hipp J, Rezai KA, West M, Atala A, Lanza R: Derivation and comparative assessment of retinal pigment epithelium from human embryonic stem cells using transcriptomics. Cloning Stem Cells6(3),217–245 (2004).
    • 68  Roy NS, Cleren C, Singh SK, Yang L, Beal MF, Goldman SA: Functional emgraftment of human ES cell-derived dopaminergic neurons enriched by coculture with telomerase-immortalized midbrain astrocytes. Nat. Med.12,1259–1268 (2006).
    • 69  Pipes BL, Tsang T, Peng SX, Fiederlein R, Graham M, Harris DT: Telomere length changes after umbilical cord blood transplant. Transfusion46(6),1038–1043 (2006).
    • 70  Schatteman GC, Ma N: Old bone marrow cells inhibit skin wound vascularization. Stem Cells24(3),717–721 (2006).
    • 71  Le Douarin NM: Ontogeny of the peripheral nervous system from the neural crest and the placodes: a developmental model studied on the basis of the quail–chick chimaera system. Harvey Lect.80,137–186 (1984).