Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer

Bruno Bernardes de Jesus, Elsa Vera, Kerstin Schneeberger, Agueda M Tejera, Eduard Ayuso, Fatima Bosch, Maria A Blasco, Bruno Bernardes de Jesus, Elsa Vera, Kerstin Schneeberger, Agueda M Tejera, Eduard Ayuso, Fatima Bosch, Maria A Blasco

Abstract

A major goal in aging research is to improve health during aging. In the case of mice, genetic manipulations that shorten or lengthen telomeres result, respectively, in decreased or increased longevity. Based on this, we have tested the effects of a telomerase gene therapy in adult (1 year of age) and old (2 years of age) mice. Treatment of 1- and 2-year old mice with an adeno associated virus (AAV) of wide tropism expressing mouse TERT had remarkable beneficial effects on health and fitness, including insulin sensitivity, osteoporosis, neuromuscular coordination and several molecular biomarkers of aging. Importantly, telomerase-treated mice did not develop more cancer than their control littermates, suggesting that the known tumorigenic activity of telomerase is severely decreased when expressed in adult or old organisms using AAV vectors. Finally, telomerase-treated mice, both at 1-year and at 2-year of age, had an increase in median lifespan of 24 and 13%, respectively. These beneficial effects were not observed with a catalytically inactive TERT, demonstrating that they require telomerase activity. Together, these results constitute a proof-of-principle of a role of TERT in delaying physiological aging and extending longevity in normal mice through a telomerase-based treatment, and demonstrate the feasibility of anti-aging gene therapy.

Copyright © 2012 EMBO Molecular Medicine.

Figures

Figure 1. AAV9-mediated mTERT expression
Figure 1. AAV9-mediated mTERT expression
  1. Direct GFP fluorescence in shaved back skin of mice treated with the indicated vectors.

  2. Dark grey bars: fold change in mean ± SEM mTERT mRNA levels in AAV9-mTERT treated mice compared to AAV9-eGFP controls; light grey bars represent mean ± SEM after setting to 1 the control mice. mTERT mRNA values are normalized to actin. At least 5 male mice per group were used. Student's t-test was used for statistical analysis.

  3. TERT Western blots of tissue whole extracts from the 1 year-old group. Top, representative Western blot. Bottom, quantification of mTERT protein from 3 to 6 male mice. Values are normalized to actin. Student's t-test was used for statistical analysis. Data are given as mean ± SD. Molecular weight markers are shown. Controls for TERT antibody are shown in Supporting Information Fig S2.

  4. Telomerase activity (measured through TRAP assay) in several tissues from either AAV9-eGFP or AAV9-mTERT injected mice.

Figure 2. Delayed aging in AAV9-mTERT treated…
Figure 2. Delayed aging in AAV9-mTERT treated mice
  1. Femur bone mineral density (BMD femur) at the indicated times post-treatment with the indicated vectors. Two-way ANOVA was used for statistical analysis demonstrating that both mTERT (p = 0.0002) and age (p = 0.03) contribute significantly to the differences observed. Data are given as mean ± SEM (*p < 0.05; **p < 0.01).

  2. Thickness of the subcutaneous fat layer at the indicated time or at the time of death (in tissues fixed immediately post-mortem – see Materials and Methods Section) in AAV9-mTERT and AAV9-eGFP treated mice. Two-way ANOVA was used for statistical analysis demonstrating a significant effect of mTERT treatment (p = 0.04) and age (p < 0.0001). Data are given as mean ± SEM (*p < 0.05).

  3. Insulin levels at the indicated times post-treatment with the vectors after mice were fasted for at least 8 h. Age-matched female mice were used. Data are given as mean ± SEM. Student's t-test was used for statistical analysis.

  4. IGF-1 levels at the indicated times post-treatment with the vectors. Two-way ANOVA was used for statistical analysis demonstrating a significant interaction between mTERT and age (p < 0.05). Data are given as mean ± SEM (**p < 0.01).

  5. Coordination and balance using a Rota-Rod wheel. Three trials were measured per mice. Two-way ANOVA was used for statistical analysis demonstrating that both mTERT (p = 0.01) and age (p = 0.01) contribute significantly to the differences observed. Data are given as mean ± SEM (*p < 0.05).

  6. Neuromuscular coordination using the tightrope test at the indicated times post-treatment with the indicated vectors. Fischer's exact test was used for statistical analysis.

Figure 3. Increased median and maximum longevity…
Figure 3. Increased median and maximum longevity of AAV9-mTERT treated mice
  1. Kaplan–Meier survival curves of the indicated mouse cohorts (50% males and 50% females). Alive mice are plotted as a vertical line. The Log rank test was used for statistical analysis.

  2. Average and 90th percentile lifespan of 2 year-old mice treated with AAV9-mTERT or AAV9-eGFP vectors. One-way ANOVA was used for statistical analysis (*p < 0.05; **p < 0.01). The floating bars represent the minimum-to-maximum age of death and the middle bar corresponds to the mean.

  3. Percentage of mice with the indicated tumours at their time of death.

Figure 4. AAV9-mTERT treatment decreases the abundance…
Figure 4. AAV9-mTERT treatment decreases the abundance of short telomeres and increases active β-catenin and cyclinD1-positive cells in AAV9-mTERT treated mice
  1. Percentage of short telomeres (percentage of telomeres presenting an intensity below 50% of the mean intensity of the corresponding control [AAV9-eGFP 1 year old]) 1 month post-treatment with the indicated vectors. Data are given as mean ± SEM. Student's t-test was used for statistical analysis. At least three independent mice were used per condition.

  2. Representative Western blots of tissue whole extracts of 2 year-old male mice treated either vector, 1 month post-treatment.

  3. Quantification of active β-catenin levels after correcting for β-catenin total levels and actin. Data are mean intensity ± SD. Student's t-test was used for statistical analysis.

  4. Quantification of cyclinD1-positive cells with respect to total cells scored in tissues from mice treated with the indicated vectors, 1 month post-treatment. (brain – cerebral cortex [7 × 105 cells scored per mouse]; heart – myocytes from myocardium [5 × 105 cells scored per mouse]; kidney – medulla [2 × 106 cells scored per mouse]; liver – [2 × 106 cells scored per mouse]; muscle – muscle fibers [4 × 105 cells scored per mouse]; lung – [8 × 105 cells scored per mouse]). Student's t-test was used for statistical assessments.

Figure 5. Catalytic activity of mTERT is…
Figure 5. Catalytic activity of mTERT is needed for increased healthspan and life-span extension in AAV9-mTERT treated mice
  1. Scheme of mTERT-wt and mTERT-DN, a catalytically inactive form of telomerase previously generated and described by us (see Materials and Methods Section).

  2. Fold change in mTERT mRNA levels in AAV9-mTERT or AAV9-mTERT-DN treated mice compared to age-matched AAV9-eGFP controls. Values are normalized for actin and are represented as the change in mean ± SEM relative to samples of age-matched AAV9-eGFP mice (set to 1). Student's t-test was used for statistical analysis.

  3. mRNA levels of Wnt target gene CyclinD1 in the liver of mice treated with AAV9-eGFP, AAV9-mTERT or AAV9-mTERT-DN vectors. Values are normalized for actin and are represented as the change in mean ± SEM relative to samples of age-matched AAV9-eGFP mice (set to 1). Student's t-test was used for statistical analysis (*p < 0.05; **p < 0.01).

  4. Expression of p16 mRNA in the liver of mice treated with AAV9-eGFP, AAV9-mTERT or AAV9-mTERT-DN vectors. Data was normalized for actin and are represented as the change in mean ± SEM relative to samples of age-matched AAV9-eGFP mice (set to 1). Student's t-test was used for statistical analysis (*p < 0.05).

  5. mRNA levels of Wnt target gene CyclinD1 in the brain of mice treated with AAV9-eGFP, AAV9-mTERT or AAV9-mTERT-DN vectors. Values are normalized for actin and are represented as the change in mean ± SEM relative to samples of age-matched AAV9-eGFP mice (set to 1). Student's t-test was used for statistical analysis (*p < 0.05).

  6. Percentage of short telomeres (percentage of telomeres presenting an intensity below 50% of the mean intensity of the corresponding control [AAV9-eGFP 1 year old]) 1 month post-treatment with the indicated vectors or in age-matched non-treated mice (control). Data are given as mean ± SEM. One-way ANOVA (with Dunnett's post hoc test against AAV9-eGFP) was used for statistical analysis (*p < 0.05; **p < 0.005).

  7. Femur bone mineral density (BMD femur) measured 6 months post injection with the indicated vectors or in age-matched non-treated mice (control). Data are given as mean ± SEM. One-way ANOVA (with Dunnett's post hoc test against AAV9-eGFP) was used for statistical analysis (**p < 0.01).

  8. Insulin levels measured 3 months post injection with the indicated vectors or in age-matched non-treated mice (control). Age-matched female mice were used. Data are given as mean ± SEM. One-way ANOVA (with Dunnett's post hoc test against AAV9-eGFP) was used for statistical analysis (*p < 0.05).

  9. Survival curves of the indicated mouse cohorts. Alive mice are plotted as a vertical line.

References

    1. Armanios MY, Chen JJ, Cogan JD, Alder JK, Ingersoll RG, Markin C, Lawson WE, Xie M, Vulto I, Phillips JA, III, et al. Telomerase mutations in families with idiopathic pulmonary fibrosis. N Engl J Med. 2007;356:1317–1326.
    1. Artandi SE, Alson S, Tietze MK, Sharpless NE, Ye S, Greenberg RA, Castrillon DH, Horner JW, Weiler SR, Carrasco RD, et al. Constitutive telomerase expression promotes mammary carcinomas in aging mice. Proc Natl Acad Sci USA. 2002;99:8191–8196.
    1. Ayuso E, Mingozzi F, Montane J, Leon X, Anguela XM, Haurigot V, Edmonson SA, Africa L, Zhou S, High KA, et al. High AAV vector purity results in serotype- and tissue-independent enhancement of transduction efficiency. Gene Ther. 2010;17:503–510.
    1. Bailey CJ, Flatt PR. Hormonal control of glucose homeostasis during development and ageing in mice. Metabolism. 1982;31:238–246.
    1. Bevins RA, Besheer J. Object recognition in rats and mice: a one-trial non-matching-to-sample learning task to study ‘recognition memory’. Nat Protoc. 2006;1:1306–1311.
    1. Blasco MA, Lee HW, Hande MP, Samper E, Lansdorp PM, DePinho RA, Greider CW. Telomere shortening and tumor formation by mouse cells lacking telomerase RNA. Cell. 1997;91:25–34.
    1. Blasco MA, Rizen M, Greider CW, Hanahan D. Differential regulation of telomerase activity and telomerase RNA during multi-stage tumorigenesis. Nat Genet. 1996;12:200–204.
    1. Bluher M, Kahn BB, Kahn CR. Extended longevity in mice lacking the insulin receptor in adipose tissue. Science. 2003;299:572–574.
    1. Bodnar AG, Ouellette M, Frolkis M, Holt SE, Chiu CP, Morin GB, Harley CB, Shay JW, Lichtsteiner S, Wright WE. Extension of life-span by introduction of telomerase into normal human cells. Science. 1998;279:349–352.
    1. Buning H, Perabo L, Coutelle O, Quadt-Humme S, Hallek M. Recent developments in adeno-associated virus vector technology. J Gene Med. 2008;10:717–733.
    1. Canela A, Martin-Caballero J, Flores JM, Blasco MA. Constitutive expression of tert in thymocytes leads to increased incidence and dissemination of T-cell lymphoma in Lck-Tert mice. Mol Cell Biol. 2004;24:4275–4293.
    1. Castelhano-Carlos MJ, Sousa N, Ohl F, Baumans V. Identification methods in newborn C57BL/6 mice: a developmental and behavioural evaluation. Lab Anim. 2010;44:88–103.
    1. Collado M, Blasco MA, Serrano M. Cellular senescence in cancer and aging. Cell. 2007;130:223–233.
    1. Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman IL, Rando TA. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature. 2005;433:760–764.
    1. de Jesus BB, Schneeberger K, Vera E, Tejera A, Harley CB, Blasco MA. The telomerase activator TA-65 elongates short telomeres and increases health span of adult/old mice without increasing cancer incidence. Aging Cell. 2011;10:604–621.
    1. Doyon J. Skill learning. Int Rev Neurobiol. 1997;41:273–294.
    1. Duque S, Joussemet B, Riviere C, Marais T, Dubreil L, Douar AM, Fyfe J, Moullier P, Colle MA, Barkats M. Intravenous administration of self-complementary AAV9 enables transgene delivery to adult motor neurons. Mol Ther. 2009;17:1187–1196.
    1. Fauce SR, Jamieson BD, Chin AC, Mitsuyasu RT, Parish ST, Ng HL, Kitchen CM, Yang OO, Harley CB, Effros RB. Telomerase-based pharmacologic enhancement of antiviral function of human CD8+ T lymphocytes. J Immunol. 2008;181:7400–7406.
    1. Ferguson VL, Ayers RA, Bateman TA, Simske SJ. Bone development and age-related bone loss in male C57BL/6J mice. Bone. 2003;33:387–398.
    1. Flores I, Canela A, Vera E, Tejera A, Cotsarelis G, Blasco MA. The longest telomeres: a general signature of adult stem cell compartments. Genes Dev. 2008;22:654–667.
    1. Flores I, Cayuela ML, Blasco MA. Effects of telomerase and telomere length on epidermal stem cell behavior. Science. 2005;309:1253–1256.
    1. Flores I, Evan G, Blasco MA. Genetic analysis of myc and telomerase interactions in vivo. Mol Cell Biol. 2006;26:6130–6138.
    1. Flurkey K, Papaconstantinou J, Miller RA, Harrison DE. Lifespan extension and delayed immune and collagen aging in mutant mice with defects in growth hormone production. Proc Natl Acad Sci USA. 2001;98:6736–6741.
    1. Foust KD, Nurre E, Montgomery CL, Hernandez A, Chan CM, Kaspar BK. Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes. Nat Biotechnol. 2009;27:59–65.
    1. Gao GP, Alvira MR, Wang L, Calcedo R, Johnston J, Wilson JM. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc Natl Acad Sci USA. 2002;99:11854–11859.
    1. Garcia-Cao I, Garcia-Cao M, Tomas-Loba A, Martin-Caballero J, Flores JM, Klatt P, Blasco MA, Serrano M. Increased p53 activity does not accelerate telomere-driven ageing. EMBO Rep. 2006;7:546–552.
    1. Gonzalez-Suarez E, Flores JM, Blasco MA. Cooperation between p53 mutation and high telomerase transgenic expression in spontaneous cancer development. Mol Cell Biol. 2002;22:7291–7301.
    1. Gonzalez-Suarez E, Samper E, Ramirez A, Flores JM, Martin-Caballero J, Jorcano JL, Blasco MA. Increased epidermal tumors and increased skin wound healing in transgenic mice overexpressing the catalytic subunit of telomerase, mTERT, in basal keratinocytes. EMBO J. 2001;20:2619–2630.
    1. Guarente L. Sirtuins as potential targets for metabolic syndrome. Nature. 2006;444:868–874.
    1. Hammerman MR. Insulin-like growth factors and aging. Endocrinol Metab Clin North Am. 1987;16:995–1011.
    1. Hao LY, Armanios M, Strong MA, Karim B, Feldser DM, Huso D, Greider CW. Short telomeres, even in the presence of telomerase, limit tissue renewal capacity. Cell. 2005;123:1121–1131.
    1. Harley CB, Liu W, Blasco M, Vera E, Andrews WH, Briggs LA, Raffaele JM. A natural product telomerase activator as part of a health maintenance program. Rejuvenation Res. 2011;14:45–56.
    1. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–395.
    1. Heikkinen S, Argmann CA, Champy MF, Auwerx J. Evaluation of glucose homeostasis. Curr Protoc Mol Biol Chapter. 2007;29 Unit 29B 23.
    1. Hemann MT, Strong MA, Hao LY, Greider CW. The shortest telomere, not average telomere length, is critical for cell viability and chromosome stability. Cell. 2001;107:67–77.
    1. Herrera E, Samper E, Martin-Caballero J, Flores JM, Lee HW, Blasco MA. Disease states associated with telomerase deficiency appear earlier in mice with short telomeres. EMBO J. 1999;18:2950–2960.
    1. Hikosaka O, Nakamura K, Sakai K, Nakahara H. Central mechanisms of motor skill learning. Curr Opin Neurobiol. 2002;12:217–222.
    1. Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Even PC, Cervera P, Le Bouc Y. IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature. 2003;421:182–187.
    1. Inagaki K, Fuess S, Storm TA, Gibson GA, McTiernan CF, Kay MA, Nakai H. Robust systemic transduction with AAV9 vectors in mice: efficient global cardiac gene transfer superior to that of AAV8. Mol Ther. 2006;14:45–53.
    1. Ingram DK, Reynolds MA. Assessing the predictive validity of psychomotor tests as measures of biological age in mice. Exp Aging Res. 1986;12:155–162.
    1. Jaskelioff M, Muller FL, Paik JH, Thomas E, Jiang S, Adams AC, Sahin E, Kost-Alimova M, Protopopov A, Cadinanos J, et al. Telomerase reactivation reverses tissue degeneration in aged telomerase-deficient mice. Nature. 2011;469:102–106.
    1. Jiang H, Lillicrap D, Patarroyo-White S, Liu T, Qian X, Scallan CD, Powell S, Keller T, McMurray M, Labelle A, et al. Multiyear therapeutic benefit of AAV serotypes 2, 6, and 8 delivering factor VIII to hemophilia A mice and dogs. Blood. 2006;108:107–115.
    1. Kaplitt MG. Gene therapy clinical trials in the human brain. Protocol development and review of current applications. Front Neurol Neurosci. 2009;25:180–188.
    1. Krishnamurthy J, Ramsey MR, Ligon KL, Torrice C, Koh A, Bonner-Weir S, Sharpless NE. p16INK4a induces an age-dependent decline in islet regenerative potential. Nature. 2006;443:453–457.
    1. Kuhlow D, Florian S, von Figura G, Weimer S, Schulz N, Petzke KJ, Zarse K, Pfeiffer AF, Rudolph KL, Ristow M. Telomerase deficiency impairs glucose metabolism and insulin secretion. Aging (Albany NY) 2010;2:650–658.
    1. Lin J, Epel E, Cheon J, Kroenke C, Sinclair E, Bigos M, Wolkowitz O, Mellon S, Blackburn E. Analyses and comparisons of telomerase activity and telomere length in human T and B cells: insights for epidemiology of telomere maintenance. J Immunol Methods. 2010;352:71–80.
    1. Maguire AM, Simonelli F, Pierce EA, Pugh EN, Jr, Mingozzi F, Bennicelli J, Banfi S, Marshall KA, Testa F, Surace EM, et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N Engl J Med. 2008;358:2240–2248.
    1. Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ, Ozelo MC, Hoots K, Blatt P, Konkle B, et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med. 2006;12:342–347.
    1. Martinez P, Thanasoula M, Carlos AR, Gomez-Lopez G, Tejera AM, Schoeftner S, Dominguez O, Pisano DG, Tarsounas M, Blasco MA. Mammalian Rap1 controls telomere function and gene expression through binding to telomeric and extratelomeric sites. Nat Cell Biol. 2010;12:768–780.
    1. Mas A, Montane J, Anguela XM, Munoz S, Douar AM, Riu E, Otaegui P, Bosch F. Reversal of type 1 diabetes by engineering a glucose sensor in skeletal muscle. Diabetes. 2006;55:1546–1553.
    1. Matheu A, Maraver A, Klatt P, Flores I, Garcia-Cao I, Borras C, Flores JM, Vina J, Blasco MA, Serrano M. Delayed ageing through damage protection by the Arf/p53 pathway. Nature. 2007;448:375–379.
    1. Matsushita T, Elliger S, Elliger C, Podsakoff G, Villarreal L, Kurtzman GJ, Iwaki Y, Colosi P. Adeno-associated virus vectors can be efficiently produced without helper virus. Gene Ther. 1998;5:938–945.
    1. Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 1985;28:412–419.
    1. McKay JD, Hung RJ, Gaborieau V, Boffetta P, Chabrier A, Byrnes G, Zaridze D, Mukeria A, Szeszenia-Dabrowska N, Lissowska J, et al. Lung cancer susceptibility locus at 5p15.33. Nat Genet. 2008;40:1404–1406.
    1. Mitchell JR, Wood E, Collins K. A telomerase component is defective in the human disease dyskeratosis congenita. Nature. 1999;402:551–555.
    1. Molofsky AV, Slutsky SG, Joseph NM, He S, Pardal R, Krishnamurthy J, Sharpless NE, Morrison SJ. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature. 2006;443:448–452.
    1. Moynihan KA, Grimm AA, Plueger MM, Bernal-Mizrachi E, Ford E, Cras-Meneur C, Permutt MA, Imai S. Increased dosage of mammalian Sir2 in pancreatic beta cells enhances glucose-stimulated insulin secretion in mice. Cell Metab. 2005;2:105–117.
    1. Niemeyer GP, Herzog RW, Mount J, Arruda VR, Tillson DM, Hathcock J, van Ginkel FW, High KA, Lothrop CD., Jr Long-term correction of inhibitor-prone hemophilia B dogs treated with liver-directed AAV2-mediated factor IX gene therapy. Blood. 2009;113:797–806.
    1. Ornish D, Lin J, Daubenmier J, Weidner G, Epel E, Kemp C, Magbanua MJ, Marlin R, Yglecias L, Carroll PR, et al. Increased telomerase activity and comprehensive lifestyle changes: a pilot study. Lancet Oncol. 2008;9:1048–1057.
    1. Park JI, Venteicher AS, Hong JY, Choi J, Jun S, Shkreli M, Chang W, Meng Z, Cheung P, Ji H, et al. Telomerase modulates Wnt signalling by association with target gene chromatin. Nature. 2009;460:66–72.
    1. Rafnar T, Sulem P, Stacey SN, Geller F, Gudmundsson J, Sigurdsson A, Jakobsdottir M, Helgadottir H, Thorlacius S, Aben KK, et al. Sequence variants at the TERT-CLPTM1L locus associate with many cancer types. Nat Genet. 2009;41:221–227.
    1. Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature. 2005;434:843–850.
    1. Sachsinger J, Gonzalez-Suarez E, Samper E, Heicappell R, Muller M, Blasco MA. Telomerase inhibition in RenCa, a murine tumor cell line with short telomeres, by overexpression of a dominant negative mTERT mutant, reveals fundamental differences in telomerase regulation between human and murine cells. Cancer Res. 2001;61:5580–5586.
    1. Sahin E, Colla S, Liesa M, Moslehi J, Muller FL, Guo M, Cooper M, Kotton D, Fabian AJ, Walkey C, et al. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature. 2011;470:359–365.
    1. 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;2:800–807.
    1. Saretzki G. Telomerase, mitochondria and oxidative stress. Exp Gerontol. 2009;44:485–492.
    1. Sarin KY, Cheung P, Gilison D, Lee E, Tennen RI, Wang E, Artandi MK, Oro AE, Artandi SE. Conditional telomerase induction causes proliferation of hair follicle stem cells. Nature. 2005;436:1048–1052.
    1. Schoeftner S, Blanco R, de Silanes IL, Munoz P, Gomez-Lopez G, Flores JM, Blasco MA. Telomere shortening relaxes X chromosome inactivation and forces global transcriptome alterations. Proc Natl Acad Sci USA. 2009;106:19393–19398.
    1. Sharma GG, Gupta A, Wang H, Scherthan H, Dhar S, Gandhi V, Iliakis G, Shay JW, Young CS, Pandita TK. hTERT associates with human telomeres and enhances genomic stability and DNA repair. Oncogene. 2003;22:131–146.
    1. Shimokata H, Tobin JD, Muller DC, Elahi D, Coon PJ, Andres R. Studies in the distribution of body fat: I. Effects of age, sex, and obesity. J Gerontol. 1989;44:M66–M73.
    1. Stroes ES, Nierman MC, Meulenberg JJ, Franssen R, Twisk J, Henny CP, Maas MM, Zwinderman AH, Ross C, Aronica E, et al. Intramuscular administration of AAV1-lipoprotein lipase S447X lowers triglycerides in lipoprotein lipase-deficient patients. Arterioscler Thromb Vasc Biol. 2008;28:2303–2304.
    1. Tafuro S, Ayuso E, Zacchigna S, Zentilin L, Moimas S, Dore F, Giacca M. Inducible adeno-associated virus vectors promote functional angiogenesis in adult organisms via regulated vascular endothelial growth factor expression. Cardiovasc Res. 2009;83:663–671.
    1. Tomas-Loba A, Flores I, Fernandez-Marcos PJ, Cayuela ML, Maraver A, Tejera A, Borras C, Matheu A, Klatt P, Flores JM, et al. Telomerase reverse transcriptase delays aging in cancer-resistant mice. Cell. 2008;135:609–622.
    1. Tsakiri KD, Cronkhite JT, Kuan PJ, Xing C, Raghu G, Weissler JC, Rosenblatt RL, Shay JW, Garcia CK. Adult-onset pulmonary fibrosis caused by mutations in telomerase. Proc Natl Acad Sci USA. 2007;104:7552–7557.
    1. Vulliamy T, Marrone A, Goldman F, Dearlove A, Bessler M, Mason PJ, Dokal I. The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita. Nature. 2001;413:432–435.
    1. Wang DB, Dayton RD, Henning PP, Cain CD, Zhao LR, Schrott LM, Orchard EA, Knight DS, Klein RL. Expansive gene transfer in the rat CNS rapidly produces amyotrophic lateral sclerosis relevant sequelae when TDP-43 is overexpressed. Mol Ther. 2010;18:2064–2074.
    1. Yamaguchi H, Calado RT, Ly H, Kajigaya S, Baerlocher GM, Chanock SJ, Lansdorp PM, Young NS. Mutations in TERT, the gene for telomerase reverse transcriptase, in aplastic anemia. N Engl J Med. 2005;352:1413–1424.
    1. Zincarelli C, Soltys S, Rengo G, Rabinowitz JE. Analysis of AAV serotypes 1–9 mediated gene expression and tropism in mice after systemic injection. Mol Ther. 2008;16:1073–1080.

Source: PubMed

Подписаться