You are on page 1of 6

REVIEW

Review

Telomere maintenance and disease

Judy M Y Wong, Kathleen Collins

The proliferative capacity of human cells is regulated by telomerase, an enzyme uniquely specialised for telomeric DNA
synthesis. The critical role of telomerase activation in tumour progression and tumour maintenance has been well
established in studies of cancer and of oncogenic transformation in cell culture. New evidence suggests that
telomerase activation has an important role in normal somatic cells, and that failure to activate sufficient telomerase
also promotes disease. We review the evidence for premature telomere attrition in proliferative deficiencies of the
human haemopoietic system, and discuss the potential use of telomerase activation in telomere-restorative gene
therapy.

Telomere biology Short telomeres


Telomeres are found at the ends of linear chromosomes.
In most eukaryotes, functional telomeres are constituted
by short, tandem DNA repeats and a multitude of Checkpoint Checkpoint bypassed:
associated proteins.1,2 The presence of telomeres intact additional telomere shortening
distinguishes the natural ends of chromosomes from
random DNA breaks, thereby preventing unwanted end-
Proliferative Chromosome Non-reciprocal
to-end fusion or nucleolytic degradation. In addition to Apoptosis
arrest fusion recombination
this physical protection of chromosome ends, eukaryotic
telomeres have important roles in cellular processes
including chromatin organisation and control of cell Rampant genomic
proliferation.3,4 instability (crisis)
DNA-dependent DNA polymerases fail to replicate
linear chromosome ends completely. Consequently, up to
Reactivation of telomerase:
a few hundred basepairs of telomeric DNA can be lost Cell death
genome stabilisation
with each mammalian cell division. Telomere erosion is
cumulative. It eventually produces an altered chromatin
structure, which either activates a damage-sensing Cancer
checkpoint to halt cell growth, or escapes checkpoint
control and undergoes repair-mediated rearrangement.2,5-7
Cultured normal human fibroblasts with critically short Figure 1: Cellular responses to short telomeres
telomere length are restrained from cell division in a Telomeres shorten with cell proliferation when not balanced by telomere
quiescent state termed replicative senescence. In other cell synthesis. In healthy somatic cells, critically short telomeres activate a
checkpoint that induces either apoptosis or the proliferative arrest of
types, short telomeres can instead provoke apoptosis. In replicative senescence. In the absence of checkpoint function, telomeres
all cells with an intact telomere checkpoint, telomere erode until they become substrates for aberrant DNA repair. Infrequently,
attrition can serve as a mitotic clock that safeguards spontaneous activation of telomerase during the crisis phase of genomic
normal somatic cells from deregulated proliferation by instability stabilises and allows maintenance of the rearranged genome,
conferring indefinite renewal capacity.
counting down the lineage allotment of cell divisions
(figure 1).
If the cellular pathways monitoring and responding to cell death. Rarely, tumorigenic cells that have activated a
short telomeres are inactivated, continued proliferation mechanism for stable maintenance of telomere length can
will erode telomeres enough to prevent their end- survive (figure 1).
protective function. Telomeres that become uncovered as
double-stranded breaks are subject to DNA repair, Telomere maintenance by telomerase
creating chromosome fusions or translocations. Aberrant Several distinct strategies for telomere length maintenance
chromosomes are further damaged by rounds of anaphase have evolved in eukaryotes. The physiological pathway for
bridge, breakage, and fusion. Cell cultures with genomic telomere maintenance in human cells involves the
instability caused by unstable telomeres are said to be in ribonucleoprotein enzyme telomerase. This enzyme is a
crisis phase, which is characterised by a high probability of specialised reverse transcriptase that copies a region
Lancet 2003; 362: 983–88. Published online May 13, 2003
Search strategy
http://image.thelancet.com/extras/02art7027web.pdf
Department of Molecular and Cell Biology, University of California We searched PubMed with keywords including telomerase,
at Berkeley, 401 Barker Hall, Berkeley, CA 94720-3204, USA telomeres, proliferative senescence, diseases of the
(J M Y Wong PhD, K Collins PhD) haemopoietic system, gene therapy, and others. We aimed to
cite recent studies directly; helpful review articles were cited
Correspondence to: Dr Kathleen Collins
(e-mail: kcollins@socrates.berkeley.edu)
for greater coverage of articles published before 2002.

THE LANCET • Vol 362 • September 20, 2003 • www.thelancet.com 983

For personal use. Only reproduce with permission from The Lancet.
REVIEW

Telomerase inactivation in the human somatic tissues


has exceptions, in a highly restricted subset of adult
tissues that are undergoing rapid proliferation.22
Telomerase is known to be active in some germline,
epithelial, and haemopoietic cells. In telomerase-positive
stages of these cell lineages, telomerase activation
correlates with induction of hTERT gene transcription. In
some cases, additional stimulation is provided by an
increase in the amount of ribonucleoprotein, post-
translational modification of hTERT, or subnuclear
shuttling of catalytically active ribonuclearprotein.22,24,25
Figure 2: The human telomerase ribonucleoprotein complex Telomerase in somatic stem cells does not compensate for
Telomerase RNA (red) must interact with H/ACA proteins (including
dyskerin, green) and with other unknown proteins, to accumulate in cells proliferation-dependent telomere loss; it does so only in a
as stable, fully processed functional RNA. A stable but inactive restricted phase of progenitor cell proliferation, and is
telomerase ribonucleoprotein is present in most human somatic cells. To subsequently downregulated by terminal differentiation.
become catalytically active, TERT (blue) and probably other proteins must Intuitively, telomerase activation could be thought to
join the ribonucleoprotein complex. Additional subunits may be required
to mediate interaction of telomerase with the telomeric repeats at a replenish every telomeric repeat lost during multiple
chromosome 3 end (black). preceding rounds of DNA replication. Such telomere
length homoeostasis has been extensively characterised in
single-celled organisms such as yeasts1 and is being
within its integral RNA component to extend intensively studied in long-term cultures of mammalian
chromosome 3 ends by synthesis of telomeric simple cells.26 In cancer cells, the balance between telomere loss
sequence repeats.8 An alternative mechanism for and telomere addition can maintain stable telomere
lengthening of telomeres, which is dependent on lengths over an extended period of proliferation. In
homologous recombination, can arise in human cells normal human somatic tissues, however, telomerase
under selective pressure. This second pathway is activation does not equate with the acquisition or
detectable in some transformed fibroblast cultures and a maintenance of stable telomere lengths. The transient
small minority of cancers.9 nature of telomerase activation in normal somatic cell
Telomerase ribonucleoproteins are only partly lineages differs strikingly from the constitutive activation
characterised in subunit composition. Our knowledge of of telomerase in tumour cells. The amount of telomere
human telomerase-associated proteins has been gained shortening offset by somatic cell telomerase activation
from candidate approaches (figure 2). The telomerase depends on telomere length before activation, rate of
RNA component is ubiquitously transcribed and telomere shortening with each round of cell division,
assembled into a stable but catalytically inactive amount of telomerase activity, and cellular regulation of
ribonucleo protein.10 Precursor processing and stability of telomere-telomerase interaction. Much remains unknown
human telomerase RNA (hTR) require binding of small about mechanisms that establish telomere length to
nucleolar ribonucleoproteins such as dyskerin, NHP2, account for findings such as proliferation-linked activation
NOP10, and GAR1 to the H/ACA motif of hTR.11-15 of telomerase in cells that nonetheless undergo rapid
Catalytic activity is acquired by binding of the telomerase erosion of telomeres.27
reverse transcriptase subunit (TERT)16 and is modulated
by additional factors including chaperones and nucleic The haemopoietic system: telomerase and
acid binding proteins.17 Finally, unknown additional telomere dynamics
conformational changes or interaction partners are The haemopoietic system provides an excellent model for
necessary to recruit telomerase to telomeres during the the study of human telomerase and telomere dynamics in
DNA synthesis phase of the cell cycle. a physiological context.28,29 Haemopoietic stem cells have
Telomerase activity is absent from most human somatic longer telomeres than descendant lineages. The low
cells beyond the early stages of fetal development,18 telomerase activity in stem cells does not seem to
although telomerase-positive cells are more generally compensate for proliferation, resulting in progressive
present in mouse tissues.19 Telomerase inactivation in telomere loss with age. In the differentiation of myeloid
human somatic cells is mainly due to transcriptional cells, including granulocytes and monocytes, telomere
repression of TERT, and can be overcome by constitutive length in progenitor cells sets an upper limit on the
expression of a human TERT (hTERT) transgene.20 proliferative capacity of lineage descendants. Telomere
Constitutive expression of hTERT in presenescent lengths in differentiated myeloid populations correlate
fibroblasts, lymphocytes, epithelial cells, and other cell with their anticipated replicative history.30 By contrast,
types can extend their replicative lifespan.4 Other lymphoid cell lineages have a complicated telomerase and
mechanisms of telomerase inactivation, including telomere length dynamic. Naive and memory T cells and
alternative mRNA splicing to express a catalytically B cells from peripheral blood undergo an age-progressive
inactive form of hTERT, have been noted in early erosion of telomere length.31 However, antigen challenge
development.21,22 The general somatic inhibition of can induce telomerase activation and telomere elongation
telomerase activity may function as a tumour suppression in these cells.
mechanism, which is perhaps most relevant in organisms Germinal centre B cells provide the most striking
with long lives. Because telomere erosion in telomerase- example of telomerase-dependent telomere elongation in
negative cells limits proliferative capacity, the likelihood of the normal human soma.32 Strong telomerase activation in
accumulation of the multiple spontaneous DNA centroblasts and centrocytes leads to a net gain in
mutations essential for carcinogenesis should be telomere length of 3–4 Kb for germinal centre B cells
diminished. This hypothesis is consistent with data from relative to naive B cells. Memory B cells downregulate
some mouse models, although the short telomeres telomerase, returning the lineage to proliferation-
resulting from telomerase disruption can also enhance dependent telomere loss. Although CD4+ memory T cells
tumorigenesis by promoting genomic instability.23 possess shorter telomeres than their naive counterparts,33

984 THE LANCET • Vol 362 • September 20, 2003 • www.thelancet.com

For personal use. Only reproduce with permission from The Lancet.
REVIEW

memory and naive B cells can have similar telomere transcription limits expression of hTR, steady state
lengths.32 In culture, mitogenic stimulation of B and amounts of RNA would be reduced by 50%. Additional
T cells induces telomerase activation in a wide range of telomerase RNA gene mutations were identified in
conditions.29 However, telomerase activation in cultured individual patients with autosomal dominant dyskeratosis
lymphocytes does not increase telomere length; it can congenita.43 These mutations would be predicted to have
offset the rate of telomere loss for only a restricted number a range of different effects on telomerase function,
of cell cycles after the initial stimulus to proliferation.34 because some mutations arise within hTR regions that are
important for RNA accumulation, whereas others occur
Haemopoietic proliferative failure induced by within hTR regions important for catalytic activity but
telomerase deficiency dispensable for accumulation.13
Mice without genes for telomerase survive for several Defects in different structural parts of the telomerase
generations despite progressive telomere attrition, since enzyme can lead to different phenotypes. The large degree
some inbred strains have especially long telomeres.35 After of cell type-specific telomerase enzyme regulation means
several generations of knockout breeding, with successive that a particular mechanism of inhibition could markedly
telomere shortening transmitted through the germline, affect enzyme function in one tissue but not in another.22
knockout mice develop haemopoietic deficiencies. These Disease onset is generally later in patients with autosomal
problems include small spleen size, decreased follicle dominant dyskeratosis congenita than in those with
numbers, reduced lymphocyte counts, and impaired X-linked disease; this difference might result from a less
lymphocyte proliferation.36 These findings could not severe telomerase deficiency in autosomal dominant disease
predict whether telomerase function would be required inheritance. Different penetrance of dermatological
for somatic cell renewal within a person’s lifespan. phenotypes relative to bone marrow deficiency is also noted
Dyskeratosis congenita was the first primary telomere among patients with dyskeratosis congenita,37 suggesting
maintenance disorder to be identified in man. The major that different amounts of telomerase ribonucleoprotein loss
cause of death is progressive bone marrow failure. Data can have differential effects on epithelial and haemopoietic
from studies of this disease suggest that the restricted tissue renewal.22 The enhanced risk of carcinoma
amount of telomerase-dependent maintenance of development in some patients probably indicates that short
telomeres in human somatic cells is essential for health telomeres in human epithelial cells have a role in promotion
and viability, serving to boost cellular renewal in highly of tumorigenic genomic instability.44,45
proliferative epithelial and haemopoietic tissues.22 Dyskerin gene mutations have also been noted in
Dyskeratosis congenita is mainly inherited as an association with X-linked Hoyeraal-Hreidarsson
X-linked disorder, and is characterised by reticulate skin syndrome.46-48 Patients with this disorder have prenatal-
pigmentation, nail dystrophy, mucosal leucoplakia, and onset growth retardation, cerebellar hypoplasia,
pancytopenia.37 Genetic linkage analysis in many families microcephaly, and immunodeficiency in the first few years
with X-linked disease showed that a mutant gene termed of life. However, they do not present with the set of
dyskerin might cause the disorder.38 Dyskerin has epithelial features that is typically diagnostic of dyskeratosis
homologues in all eukaryotic organisms. The highly congenita. Additionally, telomerase RNA gene mutations
related protein Cbf5p in budding yeast catalyses the post- have been described in a subset of patients with aplastic
transcriptional conversion of uridine to pseudouridine at anaemia.49 Consistent with a primary defect in telomerase
target RNA sites, which are specified by hybridisation function, peripheral blood leukocytes in these patients can
with tightly bound H/ACA small nucleolar RNAs.39 In have substantially shorter telomeres than age-matched
vertebrate cells, human dyskerin is associated with controls.50-52 Possibly, a large range of proliferative
H/ACA small nucleolar RNAs and also with hTR12 via the deficiency phenotypes could result from primary
hTR H/ACA motif (figure 2).11,13 Most mutations in the deficiencies of telomerase function, with variations
dyskerin gene result in substitutions of a single dependent on genetic background, environmental
aminoacid.40 Since the null allele is lethal for all organisms challenge, and the mechanism of telomerase inhibition.
investigated (including mice41) alleles of dyskerin in
dyskeratosis congenita must alter but not eliminate the Disease with premature telomere loss due to
protein’s function. Dyskerin gene mutations noted in increased proliferative demand
patients with the disease do not seem to affect amounts of Defects in genes other than those encoding telomerase
H/ACA small nucleolar RNA, pseudouridine synthesis in subunits themselves can result in premature telomere loss.
ribosomal RNA, or any other aspect of ribosome For example, an increased burden of proliferative demand
biogenesis tested to date.12,42 Consistent with a partial loss can drive chronologically premature telomere shortening,
of function in disease-associated isoforms of dyskerin, even with an unaltered rate of telomere loss per cell
telomerase RNA is decreased but not absent in cells from division. Phenotypes distinct from dyskeratosis congenita
X-linked dyskeratosis congenita patients with dyskerin would arise dependent on which tissues are subject to the
gene mutations,12 with a 3–5 fold reduction in steady-state highest rates of turnover and which cell types have the least
RNA (unpublished). A reduced concentration of telomerase-dependent compensation for proliferation.
telomerase ribonucleoprotein restricts maximum catalytic Some examples relevant to the human haemopoietic system
activity of telomerase, inducing premature telomere are described below.
shortening that varies in extent according to the type of
cell and its proliferative history. Chronic infection
Mutations in the telomerase RNA gene are a genetic The chronic phase of HIV infection is associated with
basis for autosomal dominant dyskeratosis congenita. In a substantial loss of telomere length in the CD8+ subset of T
large family with autosomal dominant inheritance of this lymphocytes.53 The reason why telomere loss is specific to
disorder, a heterozygous deletion of the 3 region of the this group of T cells is unknown. Excessive telomere
telomerase RNA gene cosegregated with disease.43 This shortening has also been shown in patients with chronic
deletion removes important regions of the H/ACA motif hepatitis or liver cirrhosis,54,55 consistent with accelerated
that are essential for RNA accumulation, and such a liver cirrhosis in telomerase knockout mice.56 In liver
mutation should therefore create a null allele. If disease, and probably in other chronic infections, the rate of

THE LANCET • Vol 362 • September 20, 2003 • www.thelancet.com 985

For personal use. Only reproduce with permission from The Lancet.
REVIEW

Telomere erosion
with proliferation
Normal genome structure
Normal physiology
Telomere length
Extended proliferative lifespan

Early telomerase activation

Genome rearrangements
Transformed phenotypes
Short telomere checkpoint Extended proliferative lifespan

Genomic
Late telomerase instability
activation

Cell divisions Senescence, Crisis


apoptosis Cell death

Figure 3: Cellular responses to telomerase activation


Most human cells do not express telomerase and lose terminal telomeric repeats with each round of cell division (blue line). Short telomeres trigger the
proliferative arrest of replicative senescence or apoptosis. Telomerase activation before this checkpoint allows indefinite proliferation with normal cellular
physiology (green curve). Activation of telomerase after onset of genomic instability promotes acquisition of transformed and tumorigenic phenotypes (red
curve, dashed to denote the small proportion of cells that evade death).

cellular renewal rises to meet higher demand from the DNA damage-triggered signal transduction to p53
increase in cell death. activation and checkpoint arrest. T cells from patients
with ataxia telangiectasia have shorter telomeres than
Bone marrow transplant age-matched controls, consistent with a greater rate of
Bone marrow transplant is currently the only therapeutic cell turnover due to unrepaired genome damage.63
option for some patients with haematological disorders. For Constitutive expression of hTERT in patients’ cells
long-term engraftment after transplantation, an expansion increases telomere length and prevents proliferative
of the transplanted cells must happen to repopulate the senescence, but does not fully rescue telomere
host’s marrow. This unusually high demand for dysfunction; these findings suggest that ATM may have a
haemopoietic system renewal, although transient, could direct role at the telomere as well.64 Similar findings have
substantially deplete the telomere reserves of stem and been reported for cells from patients with Nijmegen
progenitor cells. Shorter telomere lengths have been shown breakage syndrome.65 This disorder derives from
in transplant recipients than in their donors.57-60 Although mutations of an ATM target gene, and shares with ataxia
clinical data is not yet available to implicate telomere telangiectasia the features of radiosensitivity, cancer
shortening as an important factor in the failure of bone predisposition, and immunodeficiency.
marrow transplant, some relevant evidence has been Fanconi anaemia is an autosomal recessive disorder
reported.61 characterised by progressive bone marrow failure and an
increased risk of cancer, most commonly acute myeloid
DNA damage repair syndromes leukaemia.62 Patients with this disease have chrono-
Damaged DNA and aberrant DNA replication logically accelerated telomere shortening.66 Although the
intermediates must be repaired to protect genome spectrum of disease phenotypes has some similarities to
stability. When gene mutations compromise the function dyskeratosis congenita, only cells from patients with
of DNA repair, an increase in cell death occurs and the Fanconi anaemia have increased sensitivity to DNA
proliferative demands for renewal are increased. Cell crosslinking agents. Fanconi anaemia can arise from
lines derived from patients with DNA-repair gene mutations in different loci, including the breast cancer
mutations undergo premature senescence, which is susceptibility gene BRCA2.67 These and other findings
causally linked to chronologically accelerated telomere firmly establish a direct role for Fanconi anaemia group
shortening. proteins in the response to DNA damage.68 In view of the
Ataxia telangiectasia is an autosomal recessive raised oxidative stress sensitivity of Fanconi anaemia
disorder. Patients with this disease have several defects, cells, an increased rate of cell turnover could
including hypersensitivity to ionising radiation. Death is chronologically accelerate telomere shortening.
mostly due to infection, with increased predisposition to Alternatively, a faster rate of telomere shortening with
lymphoid malignant diseases.62 The ataxia telangiectasia each cell division could result from increased telomere
gene locus encodes ATM, a protein kinase involved in damage.69 Cells of patients with ataxia telangiectasia show

986 THE LANCET • Vol 362 • September 20, 2003 • www.thelancet.com

For personal use. Only reproduce with permission from The Lancet.
REVIEW

increased telomere damage,70 but immortal ataxia 4 Harley CB. Telomerase is not an oncogene. Oncogene 2002; 21:
telangiectasia cell lines do not differ from controls in any 494–502.
measured variable of telomere length maintenance.71 5 Goytisolo FA, Blasco MA. Many ways to telomere dysfunction: in vivo
studies using mouse models. Oncogene 2002; 21: 584–91.
Therefore, rates of telomere shortening will have to be 6 Harrington L, Robinson MO. Telomere dysfunction: multiple paths to
measured directly in different cell types from individuals the same end. Oncogene 2002; 21: 592–97.
with Fanconi anaemia and from healthy people, to 7 Hackett JA, Greider CW. Balancing instability: dual roles for
establish a direct effect of disease on rate of telomere loss. telomerase and telomere dysfunction in tumorigenesis. Oncogene 2002;
21: 619–26.
8 Blackburn EH. The end of the (DNA) line. Nat Struct Biol 2000; 7:
Clinical applications 847–50.
Expanded proliferative capacity would reverse or prevent 9 Henson JD, Neumann AA, Yeager TR, Reddel RR. Alternative
many adverse symptoms of haemopoietic deficiencies, lengthening of telomeres in mammalian cells. Oncogene 2002; 21:
regardless of their precise molecular cause. Patients with 598–610.
primary genetic defects in telomere maintenance 10 Feng J, Funk WD, Wang S, et al. The RNA component of human
telomerase. Science 1995; 269: 1236–41.
pathways or with a higher burden of demand for cellular
11 Mitchell JR, Cheng J, Collins K. A box H/ACA small nucleolar RNA-
renewal would both be served by boosting proliferative like domain at the human telomerase RNA 3 end. Mol Cell Biol 1999;
capacity. Patients who might be helped by a telomere- 19: 567–76.
based treatment could be identified by use of genetic tests 12 Mitchell JR, Wood E, Collins K. A telomerase component is defective
or telomere length measurements, well before the onset in the human disease dyskeratosis congenita. Nature 1999; 402:
551–55.
of severe haemopoietic failure. Constitutive activation of
13 Mitchell JR, Collins K. Human telomerase activation requires two
telomerase before activation of the short-telomere independent interactions between telomerase RNA and telomerase
checkpoint (figure 3, green curve), instead of as a reverse transcriptase in vivo and in vitro. Mol Cell 2000; 6: 361–71.
selection in the crisis phase of telomere dysfunction (red 14 Pogacic V, Dragon F, Filipowicz W. Human H/ACA small nucleolar
curve), has been shown to leave typical cellular physiology RNPs and telomerase share evolutionarily conserved proteins NHP2
unperturbed.4 and NOP10. Mol Cell Biol 2000; 20: 9028–40.
15 Dragon F, Pogacic V, Filipowicz W. In vitro assembly of human
Several features of hTERT transgene expression as a H/ACA small nucleolar RNPs reveals unique features of U17 and
gene therapy protocol to allow telomerase activation in telomerase RNAs. Mol Cell Biol 2000; 20: 3037–48.
haemopoietic cells seem favourable. First, constitutive 16 Nakamura TM, Cech TR. Reversing time: origin of telomerase. Cell
hTERT expression with a standard retroviral vector is 1998; 92: 587–90.
sufficient for telomere length maintenance or gain in most 17 Ford LP, Wright WE, Shay JW. A model for heterogeneous nuclear
ribonucleoproteins in telomere and telomerase regulation. Oncogene
cell types, including T cells72,73 and fibroblasts from 2002; 21: 580–83.
patients with dyskeratosis congenita (unpublished). 18 Wright WE, Piatyszek MA, Rainey WE, Byrd W, Shay JW.
Second, hTERT expression can become downregulated Telomerase activity in human germline and embryonic tissues and
after restoration of telomere reserves and still provide an cells. Dev Genet 1996; 18: 173–79.
adequate expansion of renewal capacity. Third, because 19 Prowse KR, Greider CW. Developmental and tissue-specific
regulation of mouse telomerase and telomere length.
cells that regain telomere length have a proliferative Proc Natl Acad Sci USA 1995; 92: 4818–22.
advantage, they should compete well after engraftment. 20 Bodnar AG, Ouellette M, Frolkis M, et al. Extension of life-span by
Finally, trangene-mediated expression of hTERT protein introduction of telomerase into normal human cells. Science 1998;
should not provoke rejection because this molecule is 279: 349–52.
expressed widely in development and in some adult 21 Ulaner GA, Hu J, Vu TH, Giudice LC, Hoffman AR. Telomerase
activity in human development is regulated by human telomerase
somatic cells as well. Because many cell types in the reverse transcriptase (hTERT) transcription and by alternate splicing
human haemopoietic system can activate telomerase of hTERT transcripts. Cancer Res 1998; 58: 4168–72.
endogenously, constitutive activation of the enzyme might 22 Collins K, Mitchell JR. Telomerase in the human organism.
not increase cancer risk, and could even decrease risk by Oncogene 2002; 21: 564–79.
reduction of the genomic instability caused by short 23 Artandi SE, DePinho RA. A critical role for telomeres in suppressing
telomeres.4,22 These favourable features should provide a and facilitating carcinogenesis. Curr Opin Genet Dev 2000; 10: 39–46.
24 Aisner DL, Wright WE, Shay JW. Telomerase regulation: not just
stimulus for clinical trials. However, putative telomere- flipping the switch. Curr Opin Genet Dev 2002; 12: 80–85.
independent roles of telomerase in the stimulation of cell 25 Wong JM, Kusdra L, Collins K. Subnuclear shuttling of human
growth and the prevention of apoptosis will also need to be telomerase induced by transformation and DNA damage. Nat Cell Biol
considered.74 Experimental analysis will be needed to show 2002; 4: 731–36.
whether manipulation of telomere length by telomerase 26 Smogorzewska A, van Steensel B, Bianchi A, et al. Control of human
telomere length by TRF1 and TRF2. Mol Cell Biol 2000; 20: 1659–68.
activation is clinically useful.
27 Chiu C-P, Dragowska W, Kim KW. Differential expression of
telomerase activity in hematopoietic progenitors from adult human
Conflict of interest statement bone marrow. Stem Cells 1996; 14: 239–48.
None declared. 28 Weng N-P, Hathcock KS, Hodes RJ. Regulation of telomere length
and telomerase in T and B cells: a mechanism for maintaining
Acknowledgments replicative potential. Immunity 1998; 9: 151–57.
We thank staff of the Collins Laboratory for comments about the 29 Weng N-P. Regulation of telomerase expression in human
manuscript, and many colleagues for general discussion. Both authors lymphocytes. Springer Semin Immunopathol 2002; 24: 23–33.
contributed to all aspects of manuscript production. JMYW was funded in 30 Rufer N, Brümmendorf TH, Kolvraa S, et al. Telomere fluorescence
part by a postdoctoral fellowship from the National Cancer Institute of measurements in granulocytes and T lymphocyte subsets point to a
Canada, with additional funding from a grant of the American Cancer high turnover of hematopoietic stem cells and memory T cells in early
Society to the laboratory of KC. This review does not reflects the interests childhood. J Exp Med 1999; 190: 157–67.
of any funding agency. 31 Son NH, Murray S, Yanovski J, Hodes RJ, Weng N-P. Lineage-
specific telomere shortening and unaltered capacity for telomerase
expression in human T and B lymphocytes with age. J Immunol 2000;
References 165: 1191–96.
1 McEachern MJ, Krauskopf A, Blackburn EH. Telomeres and their 32 Weng N-P, Granger L, Hodes RJ. Telomere lengthening and
control. Annu Rev Genet 2000; 34: 331–58. telomerase activation during human B cell differentiation.
2 de Lange T. Protection of mammalian telomeres. Oncogene 2002; 21: Proc Natl Acad Sci USA 1997; 94: 10827–32.
532–40. 33 Weng N-P, Levine BL, June CH, Hodes RJ. Human naive and
3 Price CM. Telomeres and telomerase: broad effects on cell growth. memory T lymphocytes differ in telomeric length and replicative
Curr Opin Genet Dev 1999; 9: 218–24. potential. Proc Natl Acad Sci USA 1995; 92: 11091–94.

THE LANCET • Vol 362 • September 20, 2003 • www.thelancet.com 987

For personal use. Only reproduce with permission from The Lancet.
REVIEW

34 Bodnar AG, Kim NW, Effros RB, Chiu C-P. Mechanism of human liver tissues with age and chronic inflammation. Exp Cell Res
telomerase induction during T cell activation. Exp Cell Res 1996; 228: 2000; 256: 578–82.
58–64. 56 Rudolph KL, Chang S, Millard M, Schreiber-Agus N, DePinho RA.
35 Blasco MA, Lee H, Hande MP, et al. Telomere shortening and tumor Inhibition of experimental liver cirrhosis in mice by telomerase gene
formation by mouse cells lacking telomerase RNA. Cell 1997; 91: delivery. Science 2000; 287: 1253–58.
25–34. 57 Notaro R, Cimmino A, Tabarini D, Rotoli B, Luzzatto L. In vivo
36 Blasco MA. Immunosenescence phenotypes in the telomerase telomere dynamics of human hematopoietic stem cells.
knockout mouse. Springer Semin Immunopathol 2002; 24: 75–85. Proc Natl Acad Sci USA 1997; 94: 13782–85.
37 Dokal I. Dyskeratosis congenita in all its forms. Br J Haematol 2000; 58 Wynn RF, Cross MA, Hatton C, et al. Accelerated telomere
110: 768–79. shortening in young recipients of allogeneic bone-marrow transplants.
38 Heiss NS, Knight SW, Vulliamy TJ, et al. X-linked dyskeratosis Lancet 1998; 351: 178–81.
congenita is caused by mutations in a highly conserved gene with 59 Akiyama M, Hoshi Y, Sakurai S, Yamada H, Yamada O,
putative nucleolar functions. Nat Genet 1998; 19: 32–38. Mizoguchi H. Changes of telomere length in children after
39 Kiss T. Small nucleolar RNA-guided post-transcriptional modification hematopoietic stem cell transplantation. Bone Marrow Transplant
of cellular RNAs. EMBO J 2001; 20: 3617–22. 1998; 21: 167–71.
40 Knight SW, Heiss NS, Vulliamy TJ, et al. X-linked dyskeratosis 60 Lee JJ, Kook H, Chung IJ, et al. Telomere length changes in patients
congenita is predominantly caused by missense mutations in the undergoing hematopoietic stem cell transplantation.
DKC1 gene. Am J Hum Genet 1999; 65: 50–58. Bone Marrow Transplant 1999; 24: 411–15.
41 He J, Navarrete S, Jasinski M, et al. Targeted disruption of Dkc1, the 61 Akiyama M, Asai O, Kuraishi Y, et al. Shortening of telomeres in
gene mutated in X-linked dyskeratosis congenita, causes embryonic recipients of both autologous and allogeneic hematopoietic stem cell
lethality in mice. Oncogene 2002; 21: 7740–44. transplantation. Bone Marrow Transplant 2000; 25: 441–47.
42 Montanaro L, Chilla A, Trere D, et al. Increased mortality rate and not 62 Vessey CJ, Norbury CJ, Hickson ID. Genetic disorders associated
impaired ribosomal biogenesis is responsible for proliferative defect in with cancer predisposition and genomic instability.
dyskeratosis congenita cell lines. J Invest Dermatol 2002; 118: 193–98. Prog Nucl Acids Res Mol Biol 2000; 63: 189–221.
43 Vulliamy T, Marrone A, Goldman F, et al. The RNA component of 63 Metcalfe JA, Parkhill J, Campbell L, et al. Accelerated telomere
telomerase is mutated in autosomal dominant dyskeratosis congenita. shortening in ataxia telangiectasia. Nat Genet 1996; 13: 350–53.
Nature 2001; 413: 432–35. 64 Wood LD, Halvorsen TL, Dhar S, et al. Characterization of ataxia
44 Maser RS, DePinho RA. Connecting chromosomes, crisis and cancer. telangiectasia fibroblasts with extended life-span through telomerase
Science 2002; 297: 565–69. expression. Oncogene 2001; 20: 278–88.
45 Tlsty TD, Romanov SR, Kozakiewicz BK, Holst CR, Haupt LM, 65 Ranganathan V, Heine WF, Ciccone DN, et al. Rescue of a telomere
Crawford YG. Loss of chromosomal integrity in human mammary length defect of Nijmegen breakage syndrome cells requires NBS and
epithelial cells subsequent to escape from senescence. J Mammary telomerase catalytic subunit. Curr Biol 2001; 11: 962–66.
Gland Biol Neoplasia 2001; 6: 235–43. 66 Leteurtre F, Li X, Guardiola P, et al. Accelerated telomere shortening
46 Knight SW, Heiss NS, Vulliamy TJ, et al. Unexplained aplastic and telomerase activation in Fanconi’s anaemia. Br J Haematol 1999;
anaemia, immunodeficiency, and cerebellar hypoplasia (Hoyeraal- 105: 883–93.
Hreidarsson syndrome) due to mutations in the dyskeratosis congenita 67 Howlett NG, Taniguchi T, Olson S, et al. Biallelic inactivation of
gene, DKC1. Br J Haematol 1999; 107: 335–39. BRCA2 in Fanconi anemia. Science 2002; 297: 606–09.
47 Yaghmai R, Kimyai-Asadi A, Rostamiani K, et al. Overlap of 68 Bogliolo M, Cabre O, Callen E, et al. The Fanconi anaemia genome
dyskeratosis congenita with the Hoyeraal-Hreidarsson syndrome. stability and tumour suppressor network. Mutagenesis 2002; 17:
J Pediatrics 2000; 136: 390–93. 529–38.
48 Cossu F, Vulliamy TJ, Marrone A, Badiali M, Cao A, Dokal I. A novel 69 Callen E, Samper E, Ramirez JL, et al. Breaks at telomeres and
DKC1 mutation, severe combined immunodeficiency (T+B-NK- TRF2-independent end fusions in Fanconi anemia. Hum Mol Genet
SCID) and bone marrow transplantation in an infant with Hoyeraal- 2002; 11: 439–44.
Hreidarsson syndrome. Br J Haematol 2002; 119: 765–68. 70 Hande MP, Balajee AS, Tchirkov A, Wynshaw-Boris A,
49 Vulliamy TJ, Marrone A, Dokal I, Mason PJ. Association between Lansdorp PM. Extra-chromosomal telomeric DNA in cells from
aplastic anaemia and mutations in telomerase RNA. Lancet 2002; 359: Atm(-/-) mice and patients with ataxia-telangiectasia. Hum Mol Genet
2168–70. 2001; 10: 519–28.
50 Ball SE, Gibson FM, Rizzo S, Tooze JA, Marsh JC, Gordon-Smith 71 Sprung CN, Bryan TM, Reddel RR, Murnane JP. Normal telomere
EC. Progressive telomere shortening in aplastic anemia. Blood 1998; maintenance in immortal ataxia telangiectasia cell lines. Mutat Res
91: 3582–92. 1997; 379: 177–84.
51 Brummendorf TH, Maciejewski JP, Mak J, Young NS, Lansdorp PM. 72 Hooijberg E, Ruizendaal JJ, Snijders PJ, Kueter EW, Walboomers JM,
Telomere length in leukocyte subpopulations of patients with aplastic Spits H. Immortalization of human CD8+ T cell clones by ectopic
anemia. Blood 2001; 97: 895–900. expression of telomerase reverse transcriptase. J Immunol 2000; 165:
52 Lee J, Kook H, Chung I, et al. Telomere length changes in patients 4239–45.
with aplastic anaemia. Br J Haematol 2001; 112: 1025–30. 73 Rufer N, Migliaccio M, Antonchuk J, Humphries RK, Roosnek E,
53 Effros RB. Telomeres and HIV disease. Microbes Infect 2000; 2: 69–76. Lansdorp PM. Transfer of the human telomerase reverse transcriptase
54 Kitada T, Seki S, Kawakita N, Kuroki T, Monna T. Telomere (TERT) gene into T lymphocytes results in extension of replicative
shortening in chronic liver diseases. Biochem Biophys Res Commun potential. Blood 2001; 98: 597–603.
1995; 211: 33–39. 74 Blasco MA. Telomerase beyond telomeres. Nat Rev Cancer 2002; 2:
55 Aikata H, Takaishi H, Kawakami Y, et al. Telomere reduction in 1–6.

988 THE LANCET • Vol 362 • September 20, 2003 • www.thelancet.com

For personal use. Only reproduce with permission from The Lancet.

You might also like