Skip to main content
. Author manuscript; available in PMC: 2013 Jun 20.
Published in final edited form as: Nat Rev Cancer. 2008 Jun;8(6):450–458. doi: 10.1038/nrc2393

Table 1.

Dysfunctional telomeres (mTerc−/−) reduce tumor incidence in mouse models of cancer

Genotype/Treatment Tumor phenotypes Effect of dysfunctional telomeres
(phenotype in mTerc−/− background)
Ref.
No mutations Few tumors normally seen Compared to wild-type and early generation
telomerase null mice, aging late generation mice
show an increase in the incidence of cancer.
32, 42
DMBA/TPA
treatment
Treatment with these
carcinogens allow for the
monitoring tumor initiation
(papillomas) and progression to squamous cell carcinomas of the
skin.
Loss of telomerase (G1) resulted in decreased growth
rate and size of papillomas, with a slight decrease in
numbers. G5 mice with dysfunctional telomeres
were almost completely resistant to papilloma
formation.
50
Ink4a/Arf Deletion of this locus results in
loss of both p16Ink4a and p19ARF.
The resulting mice develop
lymphomas and sarcomas.
Late generation double knockouts show decreased
incidence of spontaneous and carcinogen-induced
tumors, and increased tumor latency.
49, 93
APC min 100% of mice with this mutation
develop multiple intestinal
neoplasias (MIN) that progress
from micro- to macro-adenomas.
Short telomeres led to increase in tumor initiation
(microadenomas) but decreased size and number of
macroscopic adenomas
51
Alb-uPA
transgene or
CCl4 or DEN
treatment
This transgenic mouse and the
carcinogenic treatment are both
effective ways of modeling
hepatocellular carcinoma (HCC)
Successive breeding of Alb-uPA onto a late
generation telomerase null background or treatment
of G3/G4 mice with CCl4, or DEN resulted
in decreased number and size of liver nodules.
53
PMS2 Deficiency of this mismatch
repair genes leads to increased
susceptibility for lymphomas,
sarcomas, and colon carcinomas
Progressively shortening telomeres reduced the
incidence of all three tumors types.
94
Eμ-myc Transgenic expression of c-myc
in B-cells leads to potent
formation of lymphoma in this
model for Burkitt’s lymphoma.
Formation of lymphoma was almost completely
suppressed for two years in mice with dysfunctional
telomeres (G5/G6), unlike wildtype and G1 mice that
rapidly developed cancer within 6 months.
57
ATM Thymic lymphoma Delayed onset and decreased incidence of thymic
lymphomas
95, 96
P53 Loss of this important tumor
suppressor leads to rapid
development of mainly
lymphomas and soft tissue
sarcomas.
Combined homozygous loss of p53 and
dysfunctional telomeres led to increased incidence of
lymphomas. In addition, late generation mTerc−/−
combined with heterozygous loss of p53 showed a
shift in tumor spectrum from lymphomas to
epithelial cancers (breast, GI, SCC) with non-reciprocal
translocations.
47
P53 R172P This point mutation commonly
found in human tumors
abolishes the ability of p53 to
induce apoptosis and delays
tumor formation for about 6
months.
Mice from an intergenerational cross with mTerc−/−
animals (iG1) have dysfunctional telomeres and
show almost complete suppression of tumorigenesis.
58
ATM, P53 T-cell lymphoma Loss of p53 accelerated onset of lymphomas in
mTerc−/− ATM−/− mice.
97
K5-Terf2 Specific overexpression of TRF2
in the skin of these mice leads to
development of spontaneous
SCC on the skin.
Increasing generations of telomerase deficient mice
showed accelerated onset of spontaneous and UV-induced
skin neoplasms.
98

Note: APCmin, adenomatous polyposis coli min (multiple intestinal neoplasias) allele; ATM, ataxia telangiectasia mutated; Terc, telomerase RNA component; PMS2, postmeiotic segregation increased 2;