You are on page 1of 24

Critical Reviews in Clinical Laboratory Sciences

ISSN: 1040-8363 (Print) 1549-781X (Online) Journal homepage: http://www.tandfonline.com/loi/ilab20

Telomere length measurement as a clinical


biomarker of aging and disease

Clare L. Fasching

To cite this article: Clare L. Fasching (2018): Telomere length measurement as a clinical
biomarker of aging and disease, Critical Reviews in Clinical Laboratory Sciences, DOI:
10.1080/10408363.2018.1504274

To link to this article: https://doi.org/10.1080/10408363.2018.1504274

Published online: 28 Sep 2018.

Submit your article to this journal

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ilab20
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES
https://doi.org/10.1080/10408363.2018.1504274

REVIEW ARTICLE

Telomere length measurement as a clinical biomarker of aging and disease


Clare L. Fasching
Telomere Diagnostics, Inc., Menlo Park, CA, USA

ABSTRACT ARTICLE HISTORY


Telomere length measurement is increasingly recognized as a clinical gauge for age-related dis- Received 30 March 2018
ease risk. There are several methods for studying blood telomere length (BTL) as a clinical bio- Revised 11 July 2018
marker. The first is an observational study approach, which directly measures telomere lengths Accepted 22 July 2018
using either cross-sectional or longitudinal patient cohorts and compares them to a population Published online 2 2 2
of age- and sex-matched individuals. These direct traceable measurements can be considered KEYWORDS
reflective of an individual’s current health or disease state. Escalating interest in personalized Leukocyte; telomere length;
medicine, access to high-throughput genotyping and resulting acquisition of large volumes of cardiovascular disease; all-
genetic data corroborates the second method, Mendelian randomization (MR). MR employs telo- cause mortality; age-related
mere length-associated genetic variants to indicate predisposition to disease risk based on the disease; observational
genomic composition of the individual. When assessed from cells in the bloodstream, telomeres studies; Mendelian
can show variation from their genetically predisposed lengths due to environmental-induced randomization
changes. These alterations in telomere length act as an indicator of cellular health, which, in
turn, can provide disease risk status. Overall, BTL measurement is a dynamic marker of biological
health and well-being that together with genetically defined telomere lengths can provide
insights into improved healthcare for the individual.

Abbreviations: apoAI: apolipoprotein AI; BMI: body mass index; BP: blood pressure; BTL: blood
telomere length; CAC: coronary artery calcification; CAD: coronary artery disease; CHD: coronary
heart disease; CLL: chronic lymphocytic leukemia; CMV: cytomegalovirus; CRP: C-reactive protein;
CST: CTC1-STN1-TEN1; CT: computed tomography; CVD: cardiovascular disease; DNA: deoxyribo-
nucleic acid; DC: Dyskeratosis congenita; FISH: fluorescence in-situ hybridization; GWAS: genome-
wide association study; HbA1c: glycated hemoglobin; HDL-C: high-density lipoprotein cholesterol;
HH: Hoyeraal–Hreidarsson syndrome; HSC: hematopoietic stem cell; IGF1: insulin-like growth fac-
tor-1; IHD: ischemic heart disease; IL-6: interleukin-6; LDL-C: low-density lipoprotein cholesterol;
NT-proBNP: N-terminal pro B-type natriuretic peptide; MI: myocardial infarction; MR: Mendelian
randomization; PBMC: peripheral blood mononuclear cell; PC: progenitor cell; PCR: polymerase
chain reaction; qPCR: quantitative polymerase chain reaction; ROS: reactive oxygen species; SCD:
sudden cardiac death; SNP: single nucleotide polymorphism; ssDNA: single-stranded deoxyribo-
nucleic acid; STELA: single telomere length analysis; T2DM: type 2 diabetes mellitus; TC: total
cholesterol; TRF: terminal restriction fragment; TRF1: Terminal repeat Factor 1; TRF2: Terminal
repeat Factor 2; UCP2: uncoupling protein 2; US: United States; WBCs: white blood cells

1. Introduction predisposition. The number and breadth of studies


Telomeres are nucleoprotein structures found at the associating blood telomere length (BTL) with lifetime
ends of linear chromosomes, comprised of repetitive disease risks continues to increase, including reported
hexameric deoxyribonucleic acid (DNA) sequence (50 - associations with autoimmune disorders [5–7] and, in
TTAGGG-30 ) and specific associated proteins [1]. In add- particular, age-related conditions that include cardio-
ition to providing a mechanism for the aging cell to vascular disease (CVD, e.g. coronary heart disease
determine its replicative lifespan [2], telomeres serve as (CHD), atherosclerosis), type 2 diabetes mellitus (T2DM),
molecular beacons for DNA damage that signal the cell stroke, and cancer (Table 1). In a similar fashion, gen-
to activate repair mechanisms [3] or, in the case of ome-wide association studies (GWAS) have found telo-
extreme damage, to signal senescence or cell death mere length-associated genetic variants that associate
pathways [4]. Considerable interest has developed with predisposition to disease risk (Table 1). At face
around the effectiveness and utility of using telomere value, these studies suggest that telomere length meas-
length as a clinical biomarker for disease and/or disease urement and/or specific genetic variants could have

CONTACT Clare L. Fasching cfasching@telomeredx.com Telomere Diagnostics, Inc., 3603 Haven Ave., Suite A Menlo Park, CA 94025, USA
ß 2018 Informa UK Limited, trading as Taylor & Francis Group
2 C. L. FASCHING

Table 1. Summary of studies and cohorts by indication.


TL measurement
Indication All-cause mortality cohorts Sample size Study type method Reference
All-cause mortality Leiden Longevity Study 3359 Observational/MR qPCR [64]
All-cause mortality Copenhagen City Heart Study and 64367 Observational/MR qPCR [66]
Copenhagen General Population Study
All-cause mortality Epidemiological Study on the Chances of 12199 Observational qPCR [68]
Prevention, Early Recognition, and
Optimized Treatment of Chronic Diseases
in the Older Population, Nurse’s
Health Study
All-cause mortality Copenhagen City Heart Study 4576 Observational qPCR [67]
All-cause mortality National Health and Nutrition Examination 3091 Observational qPCR [69]
Survey, 1999–2002
All-cause mortality Zutphen Elderly Study 203 Observational qPCR [72]
All-cause mortality Steno Diabetes Center patients 273 Observational Southern blot [101]
All-cause mortality Italian National Research Center on Aging 568 Observational qPCR [100]
recruitment
All-cause mortality Osteoporotic Fractures in Men Study 2744 Observational qPCR [73]
All-cause mortality Tokyo Oldest Old Survey on Total Health, 1554 Observational Southern blot [15]
Tokyo Centenarians Study, Japanese
Semi-Supercentenarians Study
General Health Genetic Epidemiology Research on Adult 110266 Observational qPCR [65]
Health and Aging
All-cause mortality Lothian Birth cohort (1921) 550 Observational qPCR [74]
All-cause mortality Heart and Soul Study 780 Observational qPCR [71]
All-cause mortality Heart and Soul Study 608 Observational qPCR [70]
TL measurement
Indication Cardiovascular Disease Cohorts Sample Size Study Type method Reference
CVD Copenhagen General Populations Study, 64367 Observational/MR qPCR [66]
Copenhagen City Heart Study
MI INTERHEART study 3972 cases/ Observational qPCR [86]
4321 controls
CHD Mental Stress Ischemia Prognosis Study 566 Observational qPCR [84]
CAD West of Scotland Primary Prevention Study 484 Observational Southern Blot [82]
IHD Copenhagen General Populations Study, 290022 Observational/MR qPCR [87]
Copenhagen City Heart Study,
Copenhagen Ischemic Heart Disease
Study, CARDIoGRAM consortium,
CARDIoGRAMplusC4D consortium
MI/stroke Cardiovascular Health Study 419 Observational Southern Blot [85]
CVD Cardiovascular Disease in Intermittent 241 cases/ Observational qPCR [90]
Claudication study 249 controls
CHD Recruited Healthy people 203 Observational Southern Blot [81]
CVD Bruneck Study 88 Observational Southern Blot [88]
CAC Healthy individuals recruited in 325 Observational qPCR [91]
South Carolina
CAC National Heart, Lung and Blood Institute 3169 Observational Southern Blot [92]
Family Heart Study
CHD Hypercoagulability and Impaired Fibrinolytic 1517 cases/ MR N/A [111]
function MECHanisms, Coronary Artery 1601 controls
Bypass Graft, Simon Broome Familial
Hypercholesterolemia study, University
College London Diabetes and
Cardiovascular Disease Study, European
Atherosclerosis Research Study II
CAD CARDIoGRAM consortium 22233 cases/ MR N/A [105]
64762 controls
CVD UK BioBank 134773 MR N/A [133]
CHD ENGAGE Telomere Consortium, 22233 cases/ MR N/A [134]
CARDIoGRAM consortium, MAGIC consor- 64762 controls
tium, DIAGRAM consortium, GIANT
Consortium, Global Lipids Genetics con-
sortium, ENGAGE 1000 Genome
Consortium, 1000 Genome Consortium
for CHD
TL measurement
Indication Metabolic Disorder Cohorts Sample Size Study Type method Reference
T2DM Recruited urban Chinese population 930 cases/ Observational/MR qPCR [98]
867 controls
T2DM Strong Heart Family Study 2328 Observational qPCR [97]
(continued)
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 3

T2DM Bruneck Study 606 Observational qPCR [96]


Insulin resistance Framingham Heart Study-men 174 cases/ Observational Southern Blot [48]
156 controls
T2DM National Health and Nutrition Examination 3921 Observational qPCR [99]
Survey, 1999-2002
Oxidative stress Recruited rural Australian of 950 MR N/A [137]
European descent
T2DM University College London Diabetes and 569 MR N/A [136]
Cardiovascular Disease Study
TL measurement
Indication Cancer Cohorts Sample Size Study Type method Reference
Cancer Copenhagen City Heart Study, Copenhagen 95568 Observational/MR qPCR [140]
General Population Study
Cancer Copenhagen City Heart Study, Copenhagen 47102 Observational qPCR [139]
General Population Study
Lung Cancer Recruited from University of Texas MD 1385 cases/ Observational qPCR [142]
Anderson Cancer Center 1385 controls
Cancer Bruneck Study 787 Observational qPCR [141]
Pancreatic Cancer European Prospective Investigation into 331 cases/ Observational qPCR [143]
Cancer and Nutrition 331 controls
Chronic myelogenous UK Medical Research Council CMLIII Trial 59 cases/ Observational Southern Blot [145]
Leukemia 75 controls
Chronic myelogenous Recruited from Terry Fox Laboratory 174 cases/ Observational qPCR, Southern [144]
Leukemia 301 controls Blot, Flow FISH
Chronic Lymphocytic Recruited from University Hospital, 152 cases Observational qPCR [146]
Leukemia Ulm, Germany
Chronic Lymphocytic Recruited from University of Turin, Amedeo 401 cases/ Observational Southern Blot [147]
Leukemia Avogadro University of Eastern Piedmont 30 controls
Chronic Lymphocytic Recruited from Department of Clinical and 173 cases Observational qPCR [148]
Leukemia Experimental Medicine, Hematology
Section, Padova and Department of
Hematology, Vicenza
Chronic Lymphocytic UK Lloyd’s Register Foundation CLL4 trial 384 cases Observational qPCR, STELA [149]
Leukemia
Chronic Lymphocytic Recruited from Cardiff University 41 cases Observational STELA [150]
Leukemia
Chronic Lymphocytic Recruited from University Hospitals Umeå, 310 cases Observational qPCR, Southern [151]
Leukemia Uppsala and Link€ oping Blot, Flow FISH
Breast and Ovarian SEARCH study, Sisters in Breast Screening 103991 breast MR N/A [40]
Cancer (SIBS) study, EPIC-Norfolk study, cases, 39774 ovar-
Copenhagen City Heart Study, ian cases, 11705
Copenhagen General Population study. BRCA1 mutation
carrier cases, 53724
leukocyte telomere
length analysis
Colorectal Cancer Wellcome Trust case-control consortium 2157 cases/ MR N/A [155]
National Blood Service cohort, Scottish 3912 controls
population for the COGS, UK CORGI study
controls, VICTOR study, Scottish cases
from COGS, colorectal cancer cases
from CORGI
Glioma UCSF Adult Glioma Study, Wellcome Trust 1644 cases/ MR N/A [41]
Case-Control Consortium, The Mayo 7736 controls
Clinic, Illumina iControls, The Cancer
Genome Atlas
Cancer Genetic Associations and Mechanisms in 51725 cases/ MR N/A [152]
Oncology consortium 62035 controls
Chronic lymphocytic Eastern Cooperative Oncology Group (Mayo 273 cases/ MR N/A [153]
leukemia Clinic), Recruited at the Dana-Farber 5725 controls
Cancer Institute
Non-Hodgkin Meta-analysis of 9 prospective cohort stud- 10102 cases/ MR N/A [154]
lymphoma ies, 8 population based case/control stud- 9562 controls
ies, 5 clinic based case/control studies
Cancer and Non- Meta-analysis compiled by The Telomere 420081 cases MR N/A [157]
Neoplastic Disease Mendelian Randomization Collaboration (median 2526/dis-
ease/ 1093105 con-
trols (median
6789/disease)
COGS: consortium on the genetics of schizophrenia; CORGI: colorectal tumor gene identification; ENGAGE: European network for genetic and genomic epi-
demiology; FISH: fluorescence in-situ hybridization; CARDIoGRAM: coronary artery disease genome-wide replication and meta-analysis;
CARDIoGRAMplusC4D: coronary artery disease genome-wide replication and meta-analysis plus coronary artery disease; DIAGRAM: diabetes genetic repli-
cation and meta-analysis; GIANT: genetic investigation of anthropometric traits; MAGIC: meta-analysis of glucose and insulin-related traits consortium; MR:
Mendelian randomization; TL: telomere length; UCSF: University of California, San Francisco; UK: United Kingdom.
4 C. L. FASCHING

clinical utility in evaluating increased disease risk and/ feature of all DC patients is abnormally short telomeres
or as sentinels to monitor health status and disease [9], which fall below the first percentile, that is, telomere
progression. lengths 99% of the population [10]. Phenotypes associ-
The aim of this review is to assess the mounting evi- ated with DC, include idiopathic pulmonary fibrosis,
dence for a relationship between telomere length meas- aplastic anemia, and cancer [11]. Lineages with the auto-
urement and disease risk and to discuss limitations somal-dominant form of DC display genetic anticipation,
surrounding its use as a biomarker for age-related dis- in which telomere lengths shorten across generations
ease risk. As background to genetic predisposition of an and the disease presents earlier and with more severity
individual’s telomere length and factors that can directly in offspring relative to the previous generation [11].
influence measured telomere length, an overview of Studies of DC families strongly support telomere length
studies on telomere length inheritance as well as factors heritability. Studies assessing telomere lengths in healthy
that modulate telomere length during the course of a individuals and their offspring also support high herit-
lifetime are presented. Two primary classes of telomere ability, with some identifying maternal [8] or paternal
length studies, observational and Mendelian randomiza- paths [12–15]. In one study of DC families with individu-
tion (MR), provide the data under review. Using disease- als haplo-insufficient for TERT [16], affected individuals
specific clinical cohorts, observational studies directly (n ¼ 15) had telomere lengths below the 10th percentile,
monitor telomere length from the case population and with 13/15 below the first percentile [16]. Seven of nine
compare them to those from a control population. of their genotypically normal offspring fell below the
Repeat sampling can detect changes in the BTLs attrib- 10th percentile in telomere lengths, suggesting a genetic
uted to fluctuations in the microenvironment from life- inheritance of the parental telomere length [16]. The
style, health, or disease status of an individual. These remaining two genotypically normal offspring had lon-
changes present as a dynamic, measurable feature that ger telomere lengths than their parents, illustrating the
can provide insight into the current disease risk status genetic complexity of telomere length inheritance [16].
of an individual. In MR studies, genomic DNA polymor- A large meta-analysis of six cohorts (n ¼ 19,713) illus-
phisms, single nucleotide polymorphisms (SNPs) associ- trated a strong association between parental and off-
ated with short or long telomere lengths, highlight spring telomere lengths, with a slightly greater maternal
genetic predisposition to disease risk in case-control association [8]. This study also found telomere lengths in
clinical studies. The genetic nature of the MR studies monozygotic twins were more similar than in dizygotic
avoids the dynamic aspect of direct telomere length twins [8]. The telomere lengths of the dizygotic twins
measurement and focuses primarily on personalized and their non-twin siblings were similar [8]. Heritable fac-
telomere length with regard to its disease risk predis- tors are not the only drivers of telomere length. An asso-
position. Benefits and caveats of these two potentially ciation between the telomere lengths of spouses,
useful approaches will be discussed in the context of especially among older couples, suggests there may also
age-related diseases. be environmental components influencing length [8].
Evaluation of parental age and offspring telomere
lengths reveals an association between increased pater-
2. Determinants of telomere length: genetics, nal age at conception and longer telomere lengths in
environmental factors, and their offspring [17–20]. Inquiry into telomere length
molecular pathways inheritance and associated diseases reveals the genetic
2.1. Telomere length inheritance complexity of the telomere length set-point. Extrinsic
conditions such as socioeconomic status and psycho-
Innate telomere lengths, defined as average basal telo- logical stress can affect maternal telomere lengths [21]
mere length distributions characteristic of an individual’s and may also affect the newborn [22]. Given that an indi-
stem cells, vary modestly across populations and are vidual’s telomere length set-point is genetically inherited
determined by inherited genetic components. High her- and is also influenced by age, sex, environment, stress,
itability has been shown in twin and sibling studies [8], and health status, what mechanisms control and desta-
and is dramatically illustrated in a telomere-associated bilize telomere length?
genetic syndrome with polygenic X-linked, autosomal-
recessive and autosomal-dominant variations,
2.2. Telomere length homeostasis:
Dyskeratosis congenita (DC). DC is a genetic disorder
molecular mechanisms
with most known mutations residing within genes
involved in telomere biology, including DKC1, TERC, An individual’s telomere length set-point derives from
TERT, TIN2, NOLA2, and NOLA3 [9]. A common molecular the actions of a combination of heritable pathways,
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 5

including factors such as telomerase [1] (i.e. the ribonu- active hepatitis C virus [30], and persistent cytomegalo-
cleoprotein complex that actively lengthens telomeres virus [31] have been shown to associate with
during development), a telomere trimming mechanism short BTLs.
that prevents over-lengthening [23], and the protein Telomerase and telomere trimming. Telomerase
complexes that protect the telomeric DNA, shelterin extends telomeres using an RNA moiety, hTR. This moi-
[24,25], and CTC1-STN1-TEN1(CST) [26,27]. Regardless of ety partially anneals to a single-stranded DNA 30 over-
the initial set-point, telomere lengths decrease with hang structure of the telomere providing a template for
every cell division in an individual due to the “end-repli- hTERT, the catalytic subunit of telomerase (Figure 1(A))
cation problem” [1]. Both oxidative and replication [32–34]. Telomerase is active during embryonic devel-
stress can induce rapid telomere shortening due to opment to lengthen telomeres during cellular expan-
DNA damage [28]. Any one or all of these mechanisms sion and remains active postnatally in tissues that self-
could be active on blood telomeres at a given time, renew, such as stem cells [1]. This complex can be epi-
influencing its length distribution. In addition to these sodically activated in tissues that require further cell
intrinsic factors, viral infections including human expansion, for instance the hematopoietic system [1].
immunodeficiency virus showing a viral load [29,30], Excessive telomere lengths are prone to replication fork

(A) (B)

hTERT
Nuclease

TTAGGGTTAGGG ECTR
AAUCCC

Shelterin

hTR
Dyskerin NHP2 NOP10 GAR1 NAF1 RAP1 TRF2 TRF1 TIN2 TPP1 POT1

(C) (D)
5’ 3’
3’ 5’ Reactive Oxygen Species
(H2O2)

5’ 3’
3’
Telomere length

5’
Cell divisions

Replication stress

5’ 3’
3’ 5’

Base modification
5’ 3’
3’ 5’ (MNNG)

5’ 3’
3’ 5’

Figure 1. Processes involved in telomere length homeostasis. (A) Telomerase is a ribonucleoprotein that uses an RNA moiety,
hTR, as a template for adding sequence to the leading strand of the telomere. Several proteins, Dyskerin, NHP2, NOP10, and
NAF1 are associated with hTR during its maturation. GAR1 replaces NAF1 in the mature holoenzyme. (B) Telomere trimming, his-
torically known as telomere rapid deletion, involves cleavage of the terminal portion of the telomere, which is thought to involve
the t-loop, and results in the release of linear and circular extra-chromosomal telomeric DNA (ECTR). (C) Telomere attrition, also
known as the “end replication problem”, occurs during every cell division in the absence of any telomere lengthening mechan-
ism. This process results in the gradual shortening of telomere length and provides the means for the characteristic “cellular life-
span.” (D) DNA damage can affect telomere length by introducing single-strand breaks and gaps (H2O2), stalled replication forks
(replication stress), or bulky adducts on the DNA (MNNG), all of which require dissolution or resolution and often result in dra-
matic shortening of the affected telomere.
6 C. L. FASCHING

stalling or arrest due to slippage, or secondary structure (TRF1) and 2 (TRF2), to the telomeric DNA [45]. These
formations (e.g. G-quadruplex), both of which require genetically distinct pathways contribute to setting an
resolution [35]. Telomere trimming is a mechanism that individual’s general telomere length and provide the
rapidly reduces telomere length by removing part of complexity that contributes to changes in telomere
the end structure (Figure 1(B)) [23]. This mechanism has length depending on age and health status.
been demonstrated in cancer cells [36] that have been
modified to have very long telomeres, in leukocytes
3. Telomere lengths in the clinic: sampling a
that have been stimulated with a mitogen [37], and in
potential biomarker
human stem cells [38]. Together, telomerase and telo-
mere trimming provide cells with a homeostasis path- 3.1. White blood cells: a DNA source for dynamic
way for telomere length regulation [23]. and genetic telomere length measurement
Telomere length, DNA replication, and replicative cap- Complete blood counts and leukocyte counts/analysis
acity. Recently, Gadalla et al. [39] showed improved sur- have been recognized for years as useful and viable
vival among recipients of bone marrow transplants for markers of an individual’s health [46] and white blood
treatment of aplastic anemia when donor tissues had cells (WBCs) are the sample of choice for telomere
longer telomere lengths [39]. This increase in survival length studies. This is due, in part, to the critical role
was attributed to an increase in telomere-mediated rep- they perform in immunity, inflammation, and cellular
licative capacity that helped reestablish a productive clearance as well as the fact that WBCs are exposed to
hematopoietic system. Because post-replicative removal the metabolic by-products of other tissues, including
of the Okazaki fragment generates a gap and a single- ROS [47]. Chronic inflammation and ROS, indicative of
stranded 30 DNA overhang, telomeres shorten after adverse health conditions, are observed to shorten BTLs
every cell division in a process known as “the end-repli- [48] and therefore can provide an indicator of disease
cation problem” [1] (Figure 1(C)). Eventually, telomeres or disease risk. In itself, blood is a minimally invasive
reach a point of critical shortness, after which cells source for genomic biomarker analysis and is amenable
enter a senescent state of permanent growth arrest [1]. to repeated sampling over time. Telomere length-asso-
Telomere attrition has long been associated with cellu- ciated genetic variants inform an individual as to their
lar aging and provides proliferating cells with a means predisposition to disease risk. WBC progenitor cells
to determine cellular lifespan [2] or replicative capacity. (PCs), hematopoietic stem cells (HSCs), are capable of
Genetic predisposition for longer telomere lengths has self-renewal [49], using telomerase to delay telomere
been associated with increased cancer risk [34,40,41]. shortening [1]. It is estimated that approximately 1012
Simply, longer telomere lengths promote more cell divi- new WBCs enter the blood every day in healthy adults
sions prior to senescence or cell death [1]. As bone mar- [50]. The telomere lengths of these newly circulating
row transplant recipients exemplify [39], repopulating a cells are reflective of the unadulterated stem cell popu-
cellular niche necessitates sufficient telomere-mediated lation, as the fresh cohort has genetically defined HSC
replicative capacity for success. telomere lengths and thus are subject to telomere
Telomere length, DNA damage, and oxidative stress. In shortening influenced by lifestyle and disease status.
addition to gradual telomere length attrition, DNA dam- Similar to measures of blood pressure (BP), heart rate,
age or resolution of replication stress at the telomere and glucose levels, these changes create a dynamic
can cause rapid shortening (Figure 1(D)). A major indicator of health status.
source of DNA damage is oxidative stress resulting from
reactive oxygen species (ROS), due to inefficient
removal or depletion of antioxidants [28]. ROS directly
3.2. Molecular methods to measure
damages DNA in the form of single-stranded DNA
telomere length
breaks or indirectly through generation of aldehyde Several broadly established methods are available to
metabolites that induce formation of DNA adducts, measure telomere length, each with application-
leading to damage during the replication process [42]. dependent benefits and caveats. The most frequently
The formation of single-stranded DNA gaps and breaks reported method relies on quantitative PCR (qPCR) to
within the telomere can collapse into double-stranded calculate relative average telomere length. qPCR pro-
breaks during replication, shortening the telomere vides rapid, high-throughput analysis for large-scale
[43,44]. Oxidative stress has also been proposed to con- clinical studies [51,52]. Fluorescence-in situ-hybridiza-
tribute to telomere shortening by altering the binding tion (FISH) [53] and Flow-FISH [54] provide the means
of telomere-specific proteins, terminal repeat factor 1 to estimate telomere length within a single cell or
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 7

across a specific population of cells when combined Although telomere length predisposition is heritable,
with specific antibody staining. Terminal restriction dynamic variation of telomere length in an individual’s
fragment (TRF) analysis provides the ability to estimate WBCs is influenced by lifestyle, health, and disease state
absolute telomere length in DNA base pairs [55]. Single [21]. Genetic telomere length has been shown to con-
telomere length analysis (STELA) allows measurement tribute a relatively low percentage to the overall vari-
of a near-exact length of chromosome-specific telo- ability of telomere length [62], potentially justifying
meres [56]. When executed with standard reference direct measurement as a biomarker for non-neoplastic
materials, controls and validated methodologies, any of age-related diseases, especially in individuals between
these methods can provide useful information about the ages of 50 and 75 years (Table 2). As aging popula-
telomere length. This reviewer recommends to the tions adopt habits to improve health and are living lon-
interested reader four in-depth reviews that discuss ger, definitions of age groups tend to evolve. For the
these methodologies, their benefits, and cav- 2010 census, the US Census Bureau divided age groups
eats [57–60]. into: <18 years, 18–44 years, 45–64 years, and 65þ years
Although each of these methodologies provides [63]. For the purpose of this review, middle-age is
valuable data, it is important to note that their assay- defined as 45–64 years, older adults as 65–74 years, and
specific definitions of telomere length typically do not 75þ as elderly. For studies that explored telomere
allow telomere length comparisons across methods. In lengths in nonagenarians and above, those age groups
addition, similar techniques performed in different labo- will be identified as ultra-elderly. As this review dis-
ratories may utilize different quality control compo- cusses the studies, it will start with those of the ultra-
nents, resulting in different absolute values [61]. For elderly, as this population is quite informative when
example, relative average telomere length values as compared to control groups with shorter life spans.
defined by qPCR are based on the source DNA used for All-cause mortality and telomere length. Several
the standard curve, which is not standardized across observational studies have associated direct BTL meas-
laboratories. TRF protocols vary and are not standar- urement with all-cause mortality risk (Table 1), a classifi-
dized to one enzyme cocktail. Absolute or relative aver- cation that encompasses death from any cause. Several
age telomere length data produced across separate, studies evaluated the relationship between BTL and all-
non-standardized studies cannot be easily compared or cause mortality in the ultra-elderly and their offspring.
The inverse association between all-cause mortality and
aggregated, as variations in methodologies may con-
BTL in both middle-aged and ultra-elderly individuals
found analyses. To bypass these limitations, this review
supports the monitoring of telomere lengths to collect
will focus on study data that have been categorized as
additional information for healthcare providers and
percentile divisions of the study population, allowing
their long-lived patients. The Leiden Longevity Study
comparison of population data independent of telo-
(LLS) cohort was used to evaluate the association of
mere measurement method.
telomere length to aging and age-related diseases in a
population of long-lived people, in which nonagenar-
4. Telomere length and disease: is telomere ians (i.e. 90 years), represented the long-lived
length a reliable clinical marker for non- (n ¼ 944; mean age: 93; age range: 89–104 years), their
neoplastic age-related disease risk? offspring represented the genetically predisposed to be
long-lived, (n ¼ 1,671; mean age: 61; age range:
4.1. Observational studies: BTLs as indicators of
39–81 years), and their offspring’s spouses represented
health or disease
the control population, (n ¼ 744; mean age: 60; age
The prevalence of high-powered clinical studies pro- range: 36–79 years) [64]. These studies show an inverse
vides strong evidence of an association between telo- association with BTL and all-cause mortality in the mid-
mere length measurement and several age-related dle-aged groups, a combination of the LLS offspring
diseases. From a clinical perspective, telomere lengths and their spouses, as well as in the nonagenarians,
that fall within the lowest age- and sex-matched per- quantified as a 25% decrease in mortality risk/unit BTL
centiles (shorter telomeres) increase that individual’s [64]. After adjusting for covariates including immune-
disease risk for all-cause mortality, CVD and age-related related markers (i.e. insulin-like growth factor-1 (IGF-1),
metabolic disorders (Table 1). The observational studies C-reactive protein (CRP), interleukin-6 (IL-6), cyto-
that link disease and/or mortality risk to short telomeres megalovirus (CMV) infection and lymphocyte counts),
are agnostic to the mechanism by which these telo- using a Cox proportional hazard model, the association
mere lengths are acquired. of short BTL with all-cause mortality was found to be
8 C. L. FASCHING

Table 2. Association of direct telomere length measurement and age-related diseases.


Middle-aged Older adult Ultra-elderly
Disease Associated BTL Adult 18–44 (years) 45–64 (years) 65–74 (years) Elderly 75–89 (years) 90 þ (years)
All-cause mortality Short Weischer et al. [67], Deelen et al. [64], Farzaneh-Far et al. Deelen et al. [64] Deelen et al. [64]
Astrup et al. [101] Rode et al. [66], [71], Bonfigli
Weischer et al. [67], et al. [100]
Mons et al. [68],
Goglin et al. [70],
Astrup et al. [101]
None N/A N/A N/A Houben et al. [72], N/A
Svensson et al. [73],
Marioni et al. [74]
CVD Short N/A Brouilette et al. [81], Fitzpatrick et al. [85], N/A N/A
Brouilette et al. [82], D’Mello et al. [86],
Hammadah et al. Willeit et al. [88]
[84], D’Mello et al.
[86], Scheller Madrid
et al. [87], Willeit
et al. [88],
Raschenberger et al.
[90], Mainous et al.
[91], Hunt et al. [92]
None N/A N/A Svensson et al. [73], N/A
Willeit et al. [88]
T2DM Short N/A Demissie et al. [48], N/A N/A N/A
Willeit et al. [96],
Zhao et al. [97],
Xiao et al. [98]
None Menke et al. [99] Menke et al. [99] N/A N/A N/A
Cancer Long N/A Sanchez-Espiridion N/A N/A N/A
et al. [142]
Short N/A Willeit et al. [141], Willeit et al. [141] N/A N/A
Sanchez-Espiridion
et al. [142]
None Campa et al. [143] Weischer et al. [139], Weischer et al. [139], Svensson et al. [73] N/A
Rode et al. [140], Rode et al. [140],
Campa et al. [143] Campa et al. [143]

independent of these covariates in the offspring/ The following studies, when taken together, illustrate
spouses and the nonagenarians [64]. No difference in an inverse association between telomere length and all-
BTL or in rate of telomere attrition was found between cause mortality for middle-aged individuals (Table 2),
the long-lived offspring and the controls, suggesting suggesting that telomere length measurement could
that BTL in the middle-aged does not necessarily provide additional information to healthcare manage-
explain the long-lived characteristic of these families. ment when used as a biomarker for mortality risk. A
This similarity in telomere characteristics between the study using the Copenhagen City Heart Study (CCHS)
long-lived offspring and their spouses implicates con- cohort and the Copenhagen General Population Study
tribution from other (i.e. non-genetic) factors affecting (CGPS) cohort (n ¼ 64,367) evaluated the association
telomere lengths. In a similar study, Arai et al. [15] between telomere lengths and all-cause mortality in
examined the telomere lengths in centenarians and the middle-aged [66]. At the time of follow-up (median:
their offspring and found that telomere lengths 7 years; range: 0–22 years), 12% of individuals had died
decreased up to the age of 100 years, after which (n ¼ 7,607). The telomere length data was subdivided
they began lengthening [15]. These long-lived popu- into deciles, highlighting an inverse association
lations and their offspring had longer telomeres than between the shortest telomere length decile compared
expected for their age group [15]. Similarly, a large to the longest and all-cause mortality [66]. Increased
study using the Genetic Epidemiology Research on age, sex, and lifestyle factors including current smoking,
Adult Health and Aging (GERA) cohort (n ¼ 110,266) increased body mass index (BMI), physical inactivity,
confirmed the age-related decline in telomere lengths and increased alcohol intake were the focus of a smaller
up to 75 years of age while the population older study using the CCHS cohort (n ¼ 4576), in which the
than 75 years showed increasing telomere length [65]. cross-sectional data showed a modest association
This association of longer telomere lengths and between short telomere length and all-cause mortality
increased survival in these elderly populations is con- at both baseline and the 10-year follow-up [67]. After
sistent with a survivor effect. adjusting for age and sex, if necessary, smoking, BMI,
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 9

alcohol consumption, and physical activity, Mons et al. multivariate adjusted analysis [73]. Longitudinal data
[68] found the lowest quintile BTL associated with all- from the Lothian Birth cohorts in 1921 (n ¼ 550) and
cause mortality in the Epidemiological Study on the 1936 (n ¼ 1091) showed a modest association of
Chances of Prevention, Early Recognition, and increased BTL with decreased mortality risk in the
Optimized Treatment of Chronic Diseases in the Older 1921 cohort. However, when evaluating the 1936
Population (ESTHER; n ¼ 3,566), and the Nurse’s Health cohort, independently or combined with the 1921
Study (NHS; (n ¼ 8,633), cohorts [68]. Similarly, in the cohort, no significant association between telomere
National Health and Nutrition Examination Survey length and all-cause mortality was identified [74].
(NHANES), 1999–2002 cohort, the two shortest telomere These studies fail to signify an association between
length quartiles were modestly associated with all- telomere length and all-cause mortality in elderly
cause mortality, when compared to the longest [69]. In men. The propensity of the telomeres of some eld-
one of two studies in individuals with stable CHD using erly to increase in length [15,65] could diminish the
the Hearts and Soul cohort [70,71], Goglin et al. [70] usefulness of BTL measurement as a biomarker for
found telomere shortening was associated with this age group.
increased mortality in individuals with stable coronary CVD and telomere length. CVD in the United
artery disease (CAD) over a 5-year period (n ¼ 608) [70]. States is comprised of 45.1% CHD, 16.5% stroke,
In the second study, Farzaneh-Far et al. [71] sought an 8.5% heart failure, 9.1% high BP, 3.2% artery disease,
association between telomere length and other clinical, and 17.6% other [75]. Underlying CAD can precipi-
inflammatory and echocardiographic variables that may tate into sudden cardiac death (SCD), which is
be used as prognostic determinants in individuals with defined as death from an unexpected cardiac event
stable CAD (n ¼ 780) [71]. After adjusting for clinical fac- [76,77]. SCD primarily occurs in adults 35þ years
tors, those in the shortest quartile compared to those in and accounts for approximately 20% of all deaths
the longest were found to have an increased risk of all- due, in part, to undiagnosed CHD [78,79]. SCD
cause mortality [71]. Collectively, these studies promote occurs in 44–52% of men and 59–69% of women
monitoring of BTL as a clinical biomarker for all-cause without previous history of CVD [79]. Current meth-
mortality risk in the middle-aged. ods of identifying individuals at risk for SCD include
In contrast to studies of middle-aged and long- family history of CVD or sudden cardiac arrest, bio-
lived populations, several independent studies focus- chemical and genetic markers for atherosclerosis,
ing on the elderly age group failed to show an asso- coagulation, endothelial function, inflammation, oxi-
ciation between telomere length and all-cause dative stress, renal function, euro-humoral status,
mortality (Tables 1 and 2). Several explanations could and cardiac pump functionality [78]. When account-
play a role, including the advanced mean age of the ing for age, sex, and ethnicity, a CVD risk score such
cohort participants, the relatively narrow age ranges as the Framingham or cardiovascular mortality score
of the participants in these studies, and the tendency can be generated based on BP, total cholesterol
for an increase in telomere lengths in this age (TC), low-density lipoprotein cholesterol (LDL-C), and
group. A group of men from the Zutphen Elderly high-density lipoprotein cholesterol (HDL-C) values
Study (n ¼ 203) were evaluated for association of BTL [78,80]. According to the American Heart Association,
and all-cause and cause-specific mortality. The subse- several lifestyle factors (e.g. smoking, physical activ-
quent analysis showed no association between all- ity, diet, and weight) and health factors (e.g. TC, BP,
cause mortality and telomere length, socioeconomic, and glycemic control) are indicative of CHD risk [75].
or lifestyle factors [72]. Paired samples from the sur- Together, these risk factors and risk scores are the
vivors (n ¼ 75), collected in 1993 (mean age: 78 years) current measure by which CVD is identified
and 2000 (mean age: 83 years), were analyzed for and treated.
BTL; no association with telomere length and all- As early as 2003, short BTLs have been associated
cause mortality, CVD mortality or cancer mortality in with increased risk of myocardial infarction (MI) and
this longitudinal study was found [72]. A larger study CHD (Table 1). Numerous subsequent studies uphold
in the MrOS-Sweden study cohort (n ¼ 2,744), of eld- the utility of BTL as a clinical biomarker for CVD/CHD
erly men (mean age: 75.5; age range: 68–81 years; risk in individuals not otherwise identified by standard
average follow-up: 6 years) where approximately one- cardiac risk factors, including BMI, smoking, and TC
fifth of the participants had died (n ¼ 556), showed (Table 1). Included in the studies showing an inverse
no association between baseline BTL and all-cause relationship between telomere length and CVD/CHD
mortality, CVD mortality, or cancer mortality after risk are as follows: CHD/CAD/ischemic heart disease
10 C. L. FASCHING

(IHD), MI, coronary artery calcification (CAC), and ath- shorter telomere lengths; after adjusting for age and
erosclerosis (Tables 1 and 2). Brouilette and colleagues sex, BTL was associated with HDL-C and apolipoprotein
[81] evaluated 203 individuals who had experienced an AI (apoAI) and inversely associated with diabetes, CRP
MI by 50 years of age and compared their BTLs to an and CVD in multivariable adjusted models [88]. Analysis
age-, sex-, and smoking status-matched control group of the CVD group based on age showed an inverse rela-
(n ¼ 180) with no history of CHD. The two shortest telo- tionship between baseline telomere lengths and dis-
mere length quartiles showed an inverse association ease presentation of MI or stroke, in the first three age
with CHD [81]. In a study of the West of Scotland groups, 45–54, 55–64, and 65–74 years [88]. Similar
Primary Prevention Study (WOSCOPS) cohort (n ¼ 484), to studies of all-cause mortality, individuals in the
Brouilette and colleagues [82] again assessed the asso- 75–84 year age range did not show an association
ciation of telomere length with prediction of CAD [82]. between telomere length and CVD [88]. These data fur-
A higher risk of CAD was associated with telomere ther strengthen the evidence that telomere length
lengths in the middle and shortest tertiles [82]. When measurement could be an additional predictive bio-
these groups were given statins, their CAD risk lowered, marker for the development of CVD between the ages
while those in the middle and shortest tertiles of the of 45–74 years (Table 2).
placebo group remained at risk for CAD [82]. As statin Atherosclerosis, a chronic disease in which choles-
treatment is the standard of care to reduce CVD risk terol-rich deposits, scar tissue, inflammatory factors,
[83], the association with longer telomere length may and cells build up on the artery walls, contributes to
provide useful data on the use of telomere length as a underlying CVD [75,89]. Different methods of measuring
biomarker to monitor statin efficacy and CVD risk. atherosclerosis-mediated CVD risk include ultrasound to
Hammadah et al. [84] hypothesized that since reduced determine brachial artery flow mediated dilution, intima
numbers of circulating PC had been linked to CAD, and media thickness, and carotid plaques, as well as CT
telomere length could directly impact the replicative scans to evaluate CAC [89]. Atherosclerosis is another
capacity of PCs, PCs would mitigate the association facet of CVD that may use BTL as a biomarker for dis-
between telomere length and adverse CVD outcomes. ease risk. Willeit et al. [88] evaluated telomere lengths
However, in a population of stable CAD patients using in the development of atherosclerosis in the Bruneck
the Mental Stress Ischemia Prognosis study (MIPS) cohort (n ¼ 623), a third of which had developed ath-
cohort (n ¼ 566; average age 63 ± 9 years), they found erosclerosis (n ¼ 294) [88]. Short telomere lengths asso-
history of MI, but not CAD severity associated with ciated with advanced, but not early atherosclerosis in
shorter telomere lengths after multivariate analysis [84]. this cohort [88], suggesting their diagnostic, but not
The lowest quartile of BTL and PCs were each associ- prognostic potential for this indication. Peripheral artery
ated with increased risk of CAD; however, no significant disease is caused by atherosclerosis and increases the
interaction was found between PCs and telomere risk of CVD or stroke [90]. An inverse association
lengths [84]. These data indicate an independent asso- between telomere length and peripheral artery disease
ciation of short telomere length and decreased number risk was identified in Caucasian males, which remained
of PCs with future CVD events. Similar to data for all- after multivariate adjustment of age, log-CRP, HDL-C,
cause mortality, individuals under the age of 73 years current smoking, and log-N-terminal B-type natriuretic
from the Cardiovascular Health Study (CHS) cohort peptide (NT-proBNP) [91]. Growing evidence suggests
(n ¼ 419) showed an association between short telo- that assessing progression of CAC may provide a posi-
mere length and MI, as well as stroke [85]. After adjust- tive cost–benefit ratio and treatment strategies for CHD
ing for diabetes status, physical activity, hypertensive [75]. The following studies explored the association of
status, smoking status, age or ethnicity, an inverse asso- CAC with BTL. Mainous and colleagues [91] hypothe-
ciation of telomere lengths with MI risk was also seen sized that based on the association of atherosclerosis
in the INTERHEART cohort (n ¼ 3972 cases; n ¼ 4321 with increasing age and telomere length with cellular
controls) [86]. An inverse association of telomere age [2] and CAD, telomere length may be associated
lengths with IHD was found in each of the three short- with CAC. They evaluated the association between BTL
est telomere length quartiles when compared with the and CAC in a group of healthy individuals (n ¼ 235; age
longest, after multifactorial adjustment in two Danish range: 40–64 years), with no history of CVD and found
cohorts (CCHS and CGPS; n ¼ 62996) [87]. Willeit et al. telomere lengths in the shortest tertile were associated
[88] studied the association between telomere length with greater atherosclerosis in a multivariate adjusted
and CVD in the Bruneck cohort. Individuals that devel- model of age, sex, race, metabolic syndrome, and smok-
oped CVD at follow-up (n ¼ 88) were positive for ing, when compared to the longest telomere length
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 11

tertile, even after including Framingham Risk Score [91]. the hypertensive group had shorter BTL that could be
Using CAC as a metric of CAD risk and atherosclerosis attributed to its association with insulin resistance [48].
burden, a study using the National Heart, Lung and In another study, Xiao and colleagues [98] explored the
Blood Institute Family Heart Study cohort (n ¼ 3169) association of BTL and T2DM in a Chinese population
found the shortest telomere length tertile was associ- (n ¼ 930 cases; n ¼ 867 controls). They found shorter
ated with increased CAC category [92]. These associa- BTL in the T2DM population compared to controls even
tions support measuring BTL as a disease indicator for after multivariate adjustment [98]. BTL was inversely
atherosclerosis as well as CAD risk (Tables 1 and 2). The associated with age and sex (male) in the control popu-
scale of studies associating telomere length with CVD lation, but not in the T2DM population [98]. In contrast
risk heightens the utility of telomere length measure- a study using the NHANES (1999–2002) cohort
ment as a clinical biomarker for disease risk. (n ¼ 3,921), found no association between telomere
Age-related metabolic disorders. Metabolic syndrome length and T2DM status or duration after adjusting for
is characterized by central obesity, impaired glucose age, ethnicity, sex, education, income, smoking, pack-
tolerance, hypertension, dyslipidemia, high triglyceride years smoked, alcohol consumption, BMI, and CRP [99].
concentration, and low HDL-C concentration, which Bonfigli et al. [100] explored the predictive value of
collectively increase risk of developing T2DM [93]. BTL measurement for all-cause mortality risk in a
T2DM, a prominent metabolic disease, occurs primar- T2DM population [100]. BTL was predictive for all-
ily in middle-aged individuals between 60 and cause mortality, even when adjusted for T2DM com-
74 years [94]. T2DM is associated with an increase in plications, age, sex, and disease duration [100]. In a
insulin resistance and is a risk factor for other age- Kaplan–Meier comparison of T2DM duration and BTL,
related diseases, such as atherosclerosis and CVD [94]. the first 76 months showed the highest mortality risk
Insulin resistance is induced by a sustained inflamma- was conferred by the group that had longer T2DM
tory response and leads with T2DM, hypertension, duration and shorter telomere lengths at baseline
atherosclerosis, and metabolic syndrome [95]. Studies [100]. A study in type I diabetic patients (n ¼ 273)
on the relationship of telomere length with T2DM also showed shorter telomere lengths associated with
and related metabolic disorders are few (Tables 1 and all-cause mortality, when adjusted for age, sex, smok-
2), but present a compelling picture. ing, systolic BP, TC, glycated hemoglobin (HbAlc), log
Diabetes has been associated with oxidative stress, urinary albumin excretion rate, previous MI, or apo-
which in turn affects BTLs. In a prospective study using plexia [101]. Collectively, these studies show a modest
the Bruneck cohort (n ¼ 606) to determine the associ- association of BTL, insulin resistance, and T2DM.
ation of BTL with risk of new-onset T2DM, Willeit et al. Together, these studies present compelling data asso-
[96] measured BTL prior to the clinical detection of ciating telomere lengths, diabetes disease status, and
T2DM [96]. Although no association between BTL and all-cause mortality risk. More studies are needed to
oxidative stress, inflammation or hyperglycemia was elucidate the association of metabolic disorders and
detected, the lowest quartile of BTL was inversely asso- telomere lengths.
ciated with T2DM risk [96]. Another prospective study
used the Strong Heart Family Study (n ¼ 2328) to asso-
4.2. MR: genetic variants as predictors of disease
ciate BTL and T2DM risk [97]. Patient samples with
risk attributable to telomere length
prevalent T2DM or CHD at baseline were excluded and
followed up after 5.5 years [97]. The 292 individuals that Many studies, including those described above in
developed T2DM had shorter baseline BTL. After multi- Observational Studies, report on the association
variate adjustment for sex, age, BMI, fasting glucose, between disease risk and telomere length that have
and total triglycerides, the shortest quartile of BTL been directly measured for each subject; one of the
showed a higher risk of developing T2DM compared to core strengths of the observational data. Direct obser-
the longest [97]. Insulin resistance and oxidative stress vation of telomere length as a phenotype, however,
are associated with hypertension leading to CVD [48]. can be influenced by confounding factors including
Based on its association with oxidative stress, BTL was lifestyle, socioeconomic status, health status, reverse
assessed for an association with hypertension using causation, and selection bias [102]. Increasingly, MR
Framingham Heart Study men (n ¼ 156 controls; methods are being used to model the contribution of
n ¼ 174 hypertensive) [48]. Demissie et al. [48] found telomere length to disease risk to evaluate its potential
BTL was negatively associated with insulin resistance as a biomarker for predisposition to disease risk
and systemic oxidative stress. After adjusting for age, or state, and in some cases, to assign a causal agent.
12 C. L. FASCHING

Table 3. ENGAGE Telomere consortium single nucleotide polymorphisms.


SNP Tel-related gene Pathway Locus Chromosome Reference
rs10936599 TERC Telomerase MYNN 3 [105]
rs2736100 TERT Telomerase TERT 5 [105]
rs7675998 NAF1 Telomerase NAF1 4 [105]
rs9420907 OBFC1/STN1 Telomere end protection OBFC1 10 [105]
rs755017 RTEL1 Telomere replication ZBTB46 20 [105]
rs8105767 Unknown Unknown ZNF208 19 [105]
rs11125529 Unknown Muscle differentiation ACYP2 2 [105]

This method is used extensively in epidemiological Table 4. Complete list of IVs used in Mendelian randomiza-
studies where a genotype closely associated with a tion studies.
phenotype is used as a proxy for the phenotype, most Association with No association
often in GWAS analyses for disease risk [102–104]. leukocyte telomere with telomere
Gene Chromosome length lengths
The rationale for MR methods is rooted in Mendel’s CXCR4 2 [108] Unknown
law of independent assortment, where alleles inherited ACYP2 2 [105,106] Unknown
TERC 3 [62,106–110,155,159] Unknown
from both parents are randomly distributed to off- MNNY 3 [105,110] Unknown
spring. SNPs associated with a particular telomere PXK 3 [106] Unknown
length are used as a proxy for telomere length in stud- NAF1 4 [105,106] Unknown
TERT 5 [40,105,106,159] Unknown
ies for disease risk and should be randomly distributed ZNF311 6 [106] Unknown
throughout an unbiased general population. These TNKS 8 [156] Unknown
OBFC1 10 [62,106,108,111] Unknown
SNPs are assumed to have no direct association with MEN1 11 [156] Unknown
the disease of interest, but can, however, potentially Mre11 11 [156] Unknown
UCP2 11 [136,137] [98]
provide assignment of a causal influence by the pheno- DDX11 12 [160] Unknown
type on the disease risk [104]. Telomere length-associ- BICD1 12 [161] Unknown
CEP95 17 [110] Unknown
ated SNPs have been identified using GWAS by SMURF2 17 [110] Unknown
analyzing the allelic association to specific telomere CTC1 17 [62] Unknown
RECQL5 17 [156] Unknown
lengths (Tables 3 and 4). In addition to determining the ZNF676 19 [62,108] Unknown
telomere length associated with a particular SNP, there ZNF208 19 [105] Unknown
are several assumptions that must hold true for the SNP BCL2L1 20 [106] Unknown
DHX35 20 [62] Unknown
to be considered appropriate for use in MR studies. The RTEL1 20 [105,106] [156]
first assumption is the SNP must only affect the disease ZBTB46 20 [105] Unknown
through its effect on telomere length. To this end, there
should be some understanding of the functional or bio- associate with telomere length. Replication/validation
logical effect of the SNP on the telomere length. In studies in six additional cohorts supported the associ-
other words, the genotype must be strongly associated ation of seven SNPs with mean BTL even after adjust-
with the phenotype it represents and not be associated ment for age, sex, study-specific covariates, genomic
with confounding factors commonly seen in observa- inflation, and other control factors (Table 3) [105]. Five
tional studies. The SNP must be related to the disease of the genetic variants were near genes associated with
risk only through the phenotype it represents [102]. telomere biology: TERC, TERT, OBFC1, NAF1, and RTEL1
Finally, the SNPs should be well-established genotypes [105]. The connection of the two remaining loci,
that have been confirmed across several studies and ZNF208 and ACYP2, to telomere biology remains unclear
laboratories. Several limitations complicate the use of [105]. Several additional screens have confirmed TERT
MR, including linkage disequilibrium, pleiotropy, gen- [106], TERC [62,106–110], OBFC1 [62,106,108,111], NAF1
etic heterogeneity, and population stratification [102]. It [106], and ACYP2 [106] are associated with telomere
may be advantageous to use a set of SNPs instead of length. Many other loci have been associated with telo-
one or a few to assess the genetic relationship to dis- mere length (Table 3); however, most have not been
ease risk. A number of alleles have been identified and replicated across laboratories (Table 4). The most con-
are being used in MR studies to assess the nature of sistent associations between telomere length and dis-
telomere length involvement in age-related disease risk ease risk are found when using a combination of alleles
(Tables 3 and 4). rather than an individual SNP. This corroborates the
Genetic variants associated with telomere length. In requisite use of only those variants that have been
the most extensive GWAS to date, 15 cohorts replicated across laboratories and have a strong associ-
(n ¼ 37,684) were used to identify genetic variants that ation with telomere biology.
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 13

TPP1

POT1

Shelterin CST

STN1/
RAP1 TRF2 TRF1 TIN2 TPP1 POT1 TEN1 CTC1
OBFC1

Figure 2. Telomere-associated protein complexes. Shelterin is a six subunit protein complex, which includes TRF1, TRF2, RAP1,
TIN2, TPP1, and POT1, all of which serve to protect the telomere end. CST is a trimeric protein complex, which includes CTC1,
STN1 (OBFC1), and TEN1, that participates in lagging strand replication and has been implicated in telomerase regulation at
the telomere.

Two protein complexes involved in telomere length Pot1a [127] result in embryonic lethality. These studies
protection and regulation at each chromosome end are illustrate the importance of shelterin complex members
shelterin and CST. Shelterin is composed of six proteins in vivo, which may in part explain their absence as use-
including TRF1, TRF2, TIN2, TPP1, POT1, and RAP1 ful proxies for genetic telomere length in MR studies.
(Figure 2), which together preserve the telomere struc- There are reports of genetic variants in POT1 associat-
ture and protect the telomere ends in part by prevent- ing with cancer, but not telomere length [128,129]. The
ing DNA repair mechanisms from identifying them as fact that so few SNPs associated with shelterin have
double-strand DNA breaks [24,25]. The double-strand been identified as candidate genetic variants for MR
telomere repeat sequence-binding proteins, TRF1 and studies suggests that an additional level of regulation
TRF2, act as a scaffold for the shelterin complex mem- on genetic telomere length outside the known path-
bers (Figure 2) [24]. While depletion of TRF1 results in ways may play an important role.
cellular senescence [25], overexpression of TRF1 [112], While shelterin has not been associated with genetic
or TRF2 [113] causes a decrease in telomere length. telomere length, two members of CST have. CST, a tri-
TIN2 interacts with both TRF1 and TRF2 (Figure 2) and meric protein complex comprised of CTC1, STN1, and
has been shown to destabilize the shelterin complex TEN1, was identified and is thought to play a role in
when depleted [114]. It provides the bridge to TPP1, telomere length homeostasis [26,27]. STN1, also known
which binds and recruits POT1, the single-strand telo- as OBFC1, binds nonspecifically to single-stranded DNA
mere binding protein (Figure 2) [25,115]. In addition, [130,131]. Genetic variants in OBFC1 (STN1) associated
TPP1 interacts with telomerase [24] and is thought to with telomere length have been identified by multiple
recruit and regulate it [116,117]. Decreased expression laboratories [62,106,108,111], making them useful for
of POT1 or TPP1 induces telomere lengthening, sug- MR studies. Driven by CTC1, the CST complex localizes
gesting that POT1 is involved in telomere length regula- to the single-stranded portion of the telomere (Figure
tion [25]. RAP1 relies on binding to TRF2 for its 2) [26,131]. Although TEN1 is necessary for the telomere
telomeric localization [24] and is thought to participate capping function of the CST complex, its exact telo-
in repression of rapid telomere deletion and repression meric function remains a mystery [131]. Its depletion,
of telomere fusion [118]. Given the intimate association however, results in telomere dysfunction [132].
of the shelterin complex to telomere length, how have Decreasing the expression of any of the three CST com-
no genetic variants been associated with variation in plex members results in an increase in telomere length
telomere length? Two shelterin members, TIN2 in telomerase-positive cells, suggesting a role together
[119,120] and TPP1 [121,122], are implicated in clinical with telomerase in telomere length homeostasis [27]. It
syndromes characterized by extremely short telomere has been proposed that the TPP1/POT1 complex
length, DC and the more severe form, recruits and promotes telomerase activity at the telo-
Hoyeraal–Hreidarsson syndrome (HH), respectively. meres (Figure 2) and the CST complex reduces recruit-
Deficiency of several shelterin members in mice includ- ment and subsequent telomerase activity and
ing Trf1 [123], Trf2 [124], Tin2 [125], Tpp1 [126], and promotes lagging strand synthesis to complete
14 C. L. FASCHING

telomere elongation (Figure 2) [27,34,131]. In contrast using populations of general health and others includ-
to the shelterin members, genetic variants from OBFC1, ing case-control populations (Tables 1 and 6). Studies
and less frequently CTC1, have been associated with using sufficient numbers of cases yield some associ-
short/long telomere length, substantiating their use in ation between disease risk and genetic telomere length
MR studies. The number of proteins and complexity of (Table 6). As an example, using the most vetted SNPs
the mechanisms associated with telomere length for genetic telomere lengths (i.e. OBFC1, TERC, and
homeostasis and telomere protection illustrate a ration- TERT), two of the strongest studies found different
ale for the broad range of telomere lengths at a given results [66,87]. The first study used the CCHS cohort
age and sex. Further research will help resolve the con- (n ¼ 64,367) and found no association of genetic telo-
undrum of genetic variants with unknown biological mere length and all-cause or cardiovascular mortality
relationships to telomere length and pathways within [66]. Using the combined CCHS data and the CGPS
telomere biology that do not seem to significantly asso- data, the second study began by characterizing genetic
ciate with genetic telomere length. telomere length and found a 67 bp decrease in telo-
Non-neoplastic disease associated with genetic telo- mere length/allele, which accounted for 0.7% of the
mere lengths. Identification of telomere length-associ- telomere length variation [87]. A modest association
ated predisposition to disease risk using genetic between genetic telomere length and IHD risk was
methodologies is ever more popular. The studies below seen when the SNPs were applied as an allele sum on
explore this avenue within several non-neoplastic dis- the combined Danish cohorts, Copenhagen Ischemic
ease categories (Tables 2 and 5). In a first glance at the Heart Disease Study (CIHDS), CGPS, and CCHS
association of genetic telomere length and disease risk, (n ¼ 105,000 total; n ¼ 17,000 IHD), which was improved
a common theme appears: the power of numbers. by including data from the CARDIoGRAMplusC4D con-
Using a small population (n ¼ 3118; 1601 controls; 1517 sortium [87]. These differences in association between
cases) [111], or a general healthy population [64,66], genetic telomere length and disease risk, when includ-
several studies were unable to find an association ing the CIHDS and CARDIoGRAMplusC4D cohorts, sug-
between genetic telomere lengths and CHD [111], gest the association may be masked in the general
T2DM [111], all-cause mortality [64,66], and CVD [64,66]. population and becomes more apparent with inclusion
Many studies have the means to use cohorts with of a sufficient disease population. This illustrates the
greatly increased overall population numbers, some importance of not only increased study power in

Table 5. Association of direct telomere length measurement in cancer types and subtypes.
Observational studies
Cancer subtypes Direct association Inverse association No association
Lung cancer N/A Rode et al. [140] N/A
Squamous cell carcinoma of the lung N/A Sanchez-Espiridion et al. [142] N/A
Adenocarcinoma of the lung Sanchez-Espiridion N/A N/A
et al. [142]
Gastric cancer N/A Willeit et al. [141] N/A
Ovarian cancer N/A Willeit et al. [141] N/A
CLL N/A Roos et al. [146], Rossi et al. [147], Rampazzo et al. [148], N/A
Strefford et al. [149], Grabowski et al. [151]
CML N/A Brummendorf et al. [144], Boultwood et al. [145] N/A
Melanoma N/A Rode et al. [140] N/A
Leukemia N/A Rode et al. [140] N/A
Pancreatic cancer N/A N/A Campa et al. [143]

Table 6. Association of genetic telomere length and age-related diseases.


Disease Genetic telomere length Associated studies
All-cause mortality Longer allele N/A
Shorter allele Deelen et al. [64], Rode et al. [66], Mauberet et al. [111]
No association Said et al. [133]
CVD Longer allele N/A
Shorter allele Deelen et al. [64], Rode et al. [66], Scheller Madrid et al. [87], Codd et al.
[105], Mauberet et al. [111], Said et al. [133], Zhan et al. [134]
Cancer Longer allele Bojesen et al. [40], Walsh et al. [41], Rode et al. [66], Rode et al. [140],
Zhang et al. [152], Ojha et al. [153], Machiela et al. [154], Jones et al.
[155], Haycock et al. [157]
Shorter allele Said et al. [133]
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 15

general, but especially of the cases. The value of BTL/allele and the UCP2-A55V-CC haplotype is associ-
increased “case” study power is further illustrated by ated with shorter telomere lengths than the TT and CT
Codd et al. [105], who, using ENGAGE SNPs (Table 3), haplotypes [137]. These data highlight the complexity
explored the association of genetic telomere length of using genetic variants as proxies for telomere length
with CAD in the CARDIoGRAM cohort (n ¼ 22,233 cases; with regard to disease risk and illustrate the rationale
n ¼ 64,762 controls) [105]. The genetic risk score of the for replication across several studies, populations, and
set of all seven SNPs showed an association between laboratories. There are a number of messages that can
genetically shorter telomere length and higher risk of be taken from these studies. First, association of genetic
CAD [105]. These results were similar to a study using telomere length and non-neoplastic disease risk may
the Danish cohorts combined with the CARDIoGRAM not be empowered using all of the SNPs. Second, a suf-
cohort, which evaluated three of the telomere associ- ficiently high number of cases may be required for
ated SNPs, suggesting that OBFC1, TERC, and TERT are identification of an association between genetic telo-
sufficient to determine CHD disease risk. The first study, mere length and disease risk. This may be due to subtle
using a UK Biobank population (n ¼ 134,773), reported differences in genomic backgrounds of study partici-
an association between genetically short telomeres and pants that could confer compensatory or enhancing
increased hypertension, CVD, and cancer, but not dia- functional effects. Finally, the majority of these studies
betes or all-cause mortality after adjusting for age, sex, used the CARDIoGRAM consortium or
and genotyping arrays [133]. These data illustrate the CARDIoGRAMplusC4D consortium for CHD, resulting in
potential of using significantly powered studies, specif- similar outcomes. Additional studies in a variety of pop-
ically demonstrating the number of cases with hyper- ulations and laboratories will strengthen or challenge
tension (n ¼ 41.847), CVD (n ¼ 46,979), and cancer the validity of these associations.
(n ¼ 22,448), compared to T2DM (n ¼ 7,969). Similar to
previous studies [87,105], analyses of the CARDIoGRAM
5. Telomere length and disease: is telomere
consortium data and the ENGAGE Telomere consortium
length a reliable clinical marker for assessing
SNPs (Table 3) showed genetically longer telomere
cancer risk?
lengths associated with decreased risk of CHD [134].
Zhan et al. [134] also examined the association of gen- The investigations exploring a link between cancer and
etic telomere lengths to several metabolic markers and telomeres have been long and multifaceted. The pro-
found an association with fasting insulin after replica- gression of non-neoplastic cells to a neoplastic state
tion in an independent cohort [134]. Increased fasting includes activation of a telomere maintenance mechan-
insulin was also associated with higher CHD risk, sug- ism [138]. Eighty-five to 90% of cancers activate tel-
gesting that it acts as a mediator between genetic telo- omerase; 10%–15% use a recombination-based
mere length and CHD [134]. Insulin resistance and mechanism, alternative lengthening of telomeres
T2DM are associated with uncoupling protein 2 (UCP2), [1,138]. While directly measuring telomere length in
an inner mitochondrial membrane protein [135]. tumors may provide useful information, it is unclear
Genetic variants in UCP2 have been found to associate how measurement of BTL strengthens the understand-
with T2DM [135] and oxidative stress levels [98]. Several ing of cancer or cancer risk (Tables 2 and 5). There may
studies have explored the association of telomere be systemic changes, such as activation of cancer asso-
length to two UCP2 SNPs. For example, in a population ciated with inflammatory pathways that can affect BTL,
of urban Chinese, Xiao et al. [98] found the UCP2–886A but the impact of these changes has not proven to be
allele to be a risk factor for T2DM, but neither the A nor as promising as compared to other diseases, rendering
G allele was associated with BTL [98]. In a population of the clinical utility of BTL as a biomarker for cancer risk
Caucasians with T2DM, the UCP-866A allele is associated unsubstantiated. The following observational studies
with shorter BTL than the UCP-866G allele [136]; neither show that BTL is directly, inversely, or not associated
of the UCP-A55V alleles were associated with BTL [136]. with cancer risk, illustrating this challenge (Tables 2
These first two studies show conflicting results with and 5).
regard to BTL and UCP2 variants, which could be attrib- BTL measurement for cancer risk analysis. A large
uted to underlying population genomic differences. population (n ¼ 47,102) of the CCHS and the CGPS
Based on the association of UCP2 and oxidative stress, cohorts was evaluated for years of survival after cancer
Zhou et al. [137] explored the association of UCP2 with and cancer risk [139]. Compared to individuals in the
telomere length in a control population lacking T2DM. first quartile, representing the longest telomere lengths,
They found the UCP2–866A allele confers an increase in those in the fourth quartile had reduced survival time
16 C. L. FASCHING

Table 7. Association of genetic telomere length in cancer the accelerated phase more quickly [145]. Shorter telo-
types and subtypes. mere length was also associated with Philadelphia
Mendelian randomization studies chromosome-positive cells [144]. In chronic lymphocytic
Cancer subtypes Longer allele Shorter allele leukemia (CLL), BTL is a predictor of treatment-free sur-
Lung cancer Rode et al. [40] N/A vival and overall survival [146–149]. B cells purified
Adenocarcinoma of the lung Zhang et al. [152], N/A
Haycock et al. [157] from CLL patients had higher genomic rearrangements
Ovarian cancer Bojesen et al. [40], N/A in the population with the shortest telomeres [150].
Haycock et al. [157]
Breast cancer Bojesen et al. [40] N/A Roos et al. [146] found similar genomic aberrations
Chronic Lymphocytic Leukemia Ojha et al. [153], N/A independent of telomere length, but short telomere
Machiela et al. [154]
Melanoma Rode et al. [140], N/A length was associated with an unfavorable clinical out-
Haycock et al. [157] come, thus, could serve as an independent prognostic
Endometrial cancer Haycock et al. [157] N/A
Kidney cancer Haycock et al. [157] N/A
indicator for treatment-free survival [146]. B-CLL
Colorectal carcinoma Jones et al. [155] N/A patients with mutated VH genes were able to be further
Colorectal adenoma Haycock et al. [157] N/A
Bladder cancer Haycock et al. [157] N/A
subcategorized based on telomere lengths with shorter
Testicular germ-cell cancer Haycock et al. [157] N/A telomere length lending a worse prognosis, similar to
Glioma Walsh et al. [4], N/A the unmutated VH genes [151]. There is a high clinical
Haycock et al. [157]
Neuroblastoma Haycock et al. [157] N/A utility for telomere length measurement in hemato-
logical cancers, which in addition to disease phase and
disease survival, in CLL may provide useful independent
after cancer. Stratification of the cohort based on length prognostic information [151].
of survival/early death and for cancer type, including Genetic telomere length association with cancer risk
lung cancer, melanoma, leukemia, and esophageal can- analysis. Similar to CVD, the utilization of genetic telo-
cer, showed an association between telomere length mere length studies using MR is becoming common for
and early death, but no significant association with can- surveying cancer risk (Tables 6 and 7). The ENGAGE
cer risk [139]. These results were corroborated in an Telomere consortium SNPs (Table 3) were originally
additional study using these same cohorts [140]. Willeit characterized with regard to their association to telo-
and colleagues evaluated the Bruneck Study cohort mere length [105]. One study assessing genetic telo-
(n ¼ 787; age range: 40–79 years) and found an associ- mere length and cancer risk or cancer mortality found
ation between short telomere length and gastric, lung lower cancer mortality significantly associated with the
and ovarian cancer prognosis as well as cancer mortal- shortest genetic telomere length from the allele sum of
ity, when the longest telomere lengths were compared TERC, TERT, and OBFC1 [66]. In a subsequent study, gen-
with the middle and the shortest [141]. In a case-control etically increased telomere length was associated with
study, Sanchez-Espiridion and colleagues [142] eval- increased cancer risk individually and as an allele sum
uated histological subtypes of lung cancer and found of TERT, TERC, and OBFC1 alleles [140]. When Rode et al.
adenocarcinomas were associated with longer BTL, [140] evaluated specific cancer types, longer per allele
independent of sex, age, and smoking. In contrast, genetic telomere length was associated with increased
squamous cell carcinomas of the lung were associated risk in melanomas and lung cancer [140]. Using the
with short telomere length [142]. A study on pancreatic GAME-ON consortium data, another MR study eval-
cancer using the European Prospective Investigation uated the association between cancer risk from five
into Cancer and Nutrition (EPIC) cohort, (n ¼ 331 cases; common cancer types (i.e. breast, lung, colorectal, ovar-
n ¼ 331 controls), found no association, indicating that ian, and prostate) and telomere-associated genetic var-
BTL is not a strong predictor of pancreatic cancer risk iants corresponding to the TERC, TERT, NAF1, OBFC1,
[143]. These observational studies indicate that BTL PXK, ZNF208, RTEL1, ZNF676, CTC1, and ACPY2 loci [152].
measurement as a global biomarker for cancer risk Zhang et al. [152] found NAF1, OBFC1, TERT, and ZNF208
remains inconclusive (Table 2) and the clinical utility as associated with adenocarcinoma of the lung. Ojha and
a biomarker for specific cancer subtypes (Table 5) is at colleagues [153] studied the association of genetic telo-
best limited. Hematological cancers seem to be the mere length with risk of a hematopoietic malignancy,
exception. Compared to a normal population, average CLL, using the ENGAGE SNPs (Table 3) plus CTC1. The
BTL in chronic myelogenous leukemia patients was TERC, TERT, and OBFC1 alleles corresponding to longer
shorter [144] and associated with time from diagnosis telomere length were associated with increased CLL
to entering the accelerated and blast phase [144,145]; risk [153]. A study on non-Hodgkin lymphoma found
those with shorter BTL at the time of diagnosis entered that in 3104 patients, genetically longer telomere
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 17

length was associated with increased CLL and small in which two of the most common SNPs associated
lymphocytic lymphoma risk [154]. SNPs showing the with cancer risk are found at loci, TERC and TERT, which
strongest association were TERC, TERT, and OBFC1 [154]. encode macromolecules essential for telomerase func-
Five SNPs in the region of TERC were identified after tion [1]. This relationship between telomerase and can-
GWAS analyses of telomere biology associated gene cer may present a more relevant association with these
pathway SNPs by BTL [155]. Jones et al. [155] went on SNPs than telomere length in the neoplastic studies.
to assess these SNPs for colorectal carcinoma risk using The Telomeres Mendelian Randomization Collaboration
a GWAS approach and found one SNP, rs10936599, pre- undertook a large meta-analysis of 22 different cancer
viously associated with longer telomeres [156], to be types [157]. Using 16 different SNPs that represented 10
associated with colorectal carcinoma and adenomas different regions, longer genetic telomere length was
[155]. This study highlights the association of cancer associated with glioma, endometrial cancer, kidney can-
risk with telomere maintenance as an alternative cause cer, testicular germ-cell cancer, melanoma, bladder can-
for the association with genetic telomere length. A cer, neuroblastoma, lung adenocarcinoma, and serous
meta-analysis identified an additional SNP near TERC low malignancy potential ovarian cancer [157].
that is associated with glioma risk [41]. Previously iden- Collectively, these observational and MR studies under-
tified alleles near TERC and TERT [105], representing lon- score the early nature of BTL associations in the elucida-
ger BTL, were also associated with increased glioma risk tion of cancer risk and the need for more research.
[41]. On the other hand, RTEL1 SNPs associated with
increased glioma risk showed no significant association
6. Summary and perspectives
with regard to telomere length [41]. This study indicates
two loci, TERC and TERT may provide helpful informa- The importance of telomere length measurement as a
tion in glioma-specific studies. In an extensive analysis clinical biomarker is gaining acceptance. There is
of the TERT locus for SNPs associated with mean telo- increasing evidence from large cohort studies demon-
mere length, breast, and ovarian cancer, Bojesen and strating that direct measurement of telomere length is
colleagues [40] found telomere length-dependent as relevant for all-cause mortality and CVD risk assessment
well as telomere length-independent SNPs associated in individuals under the age of 75 years. Some age-
with increased cancer risk across three regions. The related metabolic disorders also indicate the potential
minor alleles of two BTL- and cancer-associated SNPs in clinical usefulness of BTL measurement that will likely
the first two regions were associated with long telo- become clearer with additional studies including more
mere length. A common theme seems to be emerging, diverse populations. The relationship between

Disease
Disease risk
states
(e.g. CVD)
(e.g. T2DM)

Leukocyte Telomere
Lengths
(Dynamic)

Genetic
Other
Telomere
Genetic
Length
(Set point)
Variants

Environmental
Factors
(e.g. inflammation,
chronic infection)

Figure 3. Contributions to telomere length as a clinical biomarker. Many factors play a role in the dynamic nature of measured
telomere length. Leukocyte telomere length can be affected by disease risk, as well as disease state driven by environmental fac-
tors, such as inflammation and insulin resistance. Genetic telomere length provides an inherited telomere length set point predis-
position to disease risk. Environmental factors, such as chronic viral infection or other causes of inflammation as well as
additional contributing factors such as disease state (e.g. type 2 diabetes mellitus (T2DM), atherosclerosis), can create shortened
telomere length in adulthood that increases disease risk (e.g. cardiovascular disease (CVD)). Genetic variants (e.g. UCP2) associ-
ated with environmental factors, such as oxidative stress, may affect telomere length and disease risk directly.
18 C. L. FASCHING

neoplastic diseases and telomeres, however, is compli- this methodology, given the potential different genetic
cated, overshadowing BTLs as a clinical indication of clades in an individual.
cancer. Studies evaluating predisposition to disease risk The collection of studies presented in this review
based on genetic telomere length are also gaining provides an interwoven picture of BTL measurement,
momentum. Direct and genetic telomere length meas- disease risk, and genetic predisposition (Figure 3). BTLs
urement approaches each provide interrelated comple- are dynamic, reflecting the body’s current physiological
mentary data. One provides a glimpse into an state. As new WBCs emerge into circulation, their telo-
individual’s genetic disease risk and the other a glimpse meres are subjected to environmental conditions such
into the effects of current health and lifestyle choices. as chronic viral infection, to which lifestyle and disease
Knowledge from each provides valuable information for states, such as inflammation and T2DM, may contribute
downstream health and lifestyle decisions. (Figure 3). These environmental conditions and molecu-
A number of different methodologies are currently lar disruptions can exacerbate disease risk, which are
being explored to measure telomere lengths in a clin- reflected in the BTLs (Figure 3). Observational studies
ical setting, as well as being used in the basic and clin- monitoring dynamic telomere length provide strong
evidence for telomere length measurement as a viable
ical research environment. A study comparing
clinical biomarker in non-neoplastic diseases, especially
methodologies resulted in large coefficients of variation
for the middle-aged. Genetic telomere length analysis
that would be unacceptable in a clinical laboratory
contributes an additional level of understanding to per-
[158]. Challenges for implementation of direct telomere
sonalized disease risk predisposition (Figure 3).
length measurement in a clinical setting include devel-
oping standard methods, reagents, and protocols for
proficiency testing across methodologies, as well as Acknowledgements
standardizing the tissue analyzed across laboratories. I would like to thank Dr. Lyndal Hesterberg, Dr. Kirk Ehmsen,
Studies have used whole blood, isolated peripheral Dr. Janet Warrington, Dr. Sara Cole, and Dr. Jessica Lao for
blood mononuclear cells (PBMCs), fractionated subsets critical review and comments on this manuscript.
of WBCs or saliva as the tissue for telomere length ana-
lysis, each of which may provide subtle, but measurable Disclosure statement
differences. To minimize laboratory-induced variation in
This author alone is responsible for the content and writing
telomere length, storage and handling of the samples,
of this article. The author is an employee and has received
both prior to and after DNA extraction, should also be stock options from Telomere Diagnostics, Inc.
standardized. Regardless of these challenges, many
studies report acceptable variation and present their
References
data in percentile format, which allows the data to
be reconciled. [1] Reddel RR. Telomere maintenance mechanisms in
Genomic analysis to determine genetic telomere cancer: clinical implications. CPD. 2014;20:6361–6374.
[2] Allsopp RC, Vaziri H, Patterson C, et al. Telomere
length is not subject to the same challenges that direct
length predicts replicative capacity of human fibro-
telomere length measurement incurs. Interpretation of blasts. Proc Natl Acad Sci USA. 1992;89:10114–10118.
genetic telomere length is, however, not without a few [3] Verdun RE, Crabbe L, Haggblom C, et al. Functional
challenges of its own. The number of individuals for human telomeres are recognized as DNA damage in
which genetic telomere length confers increased dis- G2 of the cell cycle. Mol Cell. 2005;20:551–561.
[4] Ramirez R, Carracedo J, Jimenez R, et al. Massive
ease risk is representative of only a small percentage of telomere loss is an early event of DNA damage-
the population. Caution in choosing genetic variants induced apoptosis. J Biol Chem. 2003;278:836–842.
that have demonstrably been associated with telomere [5] Andrews NP, Fujii H, Goronzy JJ, et al. Telomeres and
length seems paramount to the success of this strategy. immunological diseases of aging. Gerontology.
2010;56:390–403.
A larger challenge toward implementation of genetic
[6] Costenbader KH, Prescott J, Zee RY, et al.
telomere length analysis as a personalized screen in the Immunosenescence and rheumatoid arthritis: does
clinic is to increase the diversity in the genomic back- telomere shortening predict impending disease?
grounds necessary to apply these data broadly across Autoimmun Rev. 2011;10:569–573.
populations. Currently, the majority of studies have [7] Kordinas V, Ioannidis A, Chatzipanagiotou S. The
telomere/telomerase system in chronic inflammatory
been performed using northern European populations. diseases. Cause or effect? Genes (Basel). 2016;7:60.
Understanding common risk alleles across genomic [8] Broer L, Codd V, Nyholt DR, et al. Meta-analysis of
backgrounds seems key to clinical implementation of telomere length in 19,713 subjects reveals high
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 19

heritability, stronger maternal inheritance and a stranded DNA and protects telomeres independently
paternal age effect. Eur J Hum Genet. of the Pot1 pathway. Mol Cell. 2009;36:193–206.
2013;21:1163–1168. [27] Chen LY, Redon S, Lingner J. The human CST com-
[9] Savage SA, Alter BP. Dyskeratosis congenita. Hematol plex is a terminator of telomerase activity. Nature.
Oncol Clin North Am. 2009;23:215–231. 2012;488:540–544.
[10] Alter BP, Baerlocher GM, Savage SA, et al. Very short [28] Halliwell B, Aruoma OI. DNA damage by oxygen-
telomere length by flow fluorescence in situ hybrid- derived species. Its mechanism and measurement in
ization identifies patients with dyskeratosis conge- mammalian systems. FEBS Lett. 1991;281:9–19.
nita. Blood. 2007;110:1439–1447. [29] Cote HC, Soudeyns H, Thorne A, et al. Leukocyte
[11] Armanios M, Blackburn EH. The telomere syndromes. telomere length in HIV-infected and HIV-exposed
Nat Rev Genet. 2012;13:693–704. uninfected children: shorter telomeres with uncon-
[12] Nordfjall K, Larefalk A, Lindgren P, et al. Telomere trolled HIV viremia. PLoS One. 2012;7:e39266.
length and heredity: indications of paternal inherit- [30] Zanet DL, Thorne A, Singer J, et al. Association
ance. Proc Natl Acad Sci USA. 2005;102:16374–16378. between short leukocyte telomere length and HIV
[13] Nordfjall K, Svenson U, Norrback KF, et al. Large-scale infection in a cohort study: no evidence of a relation-
parent-child comparison confirms a strong paternal ship with antiretroviral therapy. Clin Infect Dis.
influence on telomere length. Eur J Hum Genet. 2014;58:1322–1332.
2010;18:385–389. [31] van de Berg PJ, Griffiths SJ, Yong SL, et al.
[14] Njajou OT, Cawthon RM, Damcott CM, et al. Cytomegalovirus infection reduces telomere length
Telomere length is paternally inherited and is associ- of the circulating T cell pool. J Immunol. 2010;184:
ated with parental lifespan. Proc Natl Acad Sci USA. 3417–3423.
2007;104:12135–12139. [32] Greider CW, Blackburn EH. A telomeric sequence in
[15] Arai Y, Martin-Ruiz CM, Takayama M, et al. the RNA of Tetrahymena telomerase required for
Inflammation, but not telomere length, predicts suc- telomere repeat synthesis. Nature. 1989;337:331–337.
cessful ageing at extreme old age: a longitudinal [33] Schmidt JC, Cech TR. Human telomerase: biogenesis,
study of semi-supercentenarians. EBioMedicine. trafficking, recruitment, and activation. Genes Dev.
2015;2:1549–1558.
2015;29:1095–1105.
[16] Chiang YJ, Calado RT, Hathcock KS, et al. Telomere
[34] Blackburn EH, Epel ES, Lin J. Human telomere biol-
length is inherited with resetting of the telomere
ogy: a contributory and interactive factor in aging,
set-point. Proc Natl Acad Sci USA. 2010;107:
disease risks, and protection. Science. 2015;350:
10148–10153.
1193–1198.
[17] De Meyer T, Rietzschel ER, De Buyzere ML, et al.
[35] Vannier JB, Pavicic-Kaltenbrunner V, Petalcorin MI,
Paternal age at birth is an important determinant of
et al. RTEL1 dismantles T loops and counteracts telo-
offspring telomere length. Hum Mol Genet.
meric G4-DNA to maintain telomere integrity. Cell.
2007;16:3097–3102.
2012;149:795–806.
[18] Kimura M, Cherkas LF, Kato BS, et al. Offspring’s
[36] Pickett HA, Cesare AJ, Johnston RL, et al. Control of
leukocyte telomere length, paternal age, and telo-
telomere length by a trimming mechanism that
mere elongation in sperm. PLoS Genet. 2008;4:e37
[19] Hjelmborg JB, Dalgard C, Mangino M, et al. Paternal involves generation of t-circles. Embo J.
age and telomere length in twins: the germ stem 2009;28:799–809.
cell selection paradigm. Aging Cell. 2015;14:701–703. [37] Pickett HA, Henson JD, Au AY, et al. Normal mamma-
[20] Factor-Litvak P, Susser E, Kezios K, et al. Leukocyte lian cells negatively regulate telomere length by
telomere length in newborns: implications for the telomere trimming. Hum Mol Genet. 2011;20:
role of telomeres in human disease. Pediatrics. 4684–4692.
2016;137:e20153927. [38] Rivera T, Haggblom C, Cosconati S, et al. A balance
[21] Lin J, Epel E, Blackburn E. Telomeres and lifestyle between elongation and trimming regulates telo-
factors: roles in cellular aging. Mutat Res. 2012; mere stability in stem cells. Nat Struct Mol Biol.
730:85–89. 2017;24:30–39.
[22] Entringer S, Epel ES, Kumsta R, et al. Stress exposure [39] Gadalla SM, Wang T, Haagenson M, et al. Association
in intrauterine life is associated with shorter telomere between donor leukocyte telomere length and sur-
length in young adulthood. Proc Natl Acad Sci USA. vival after unrelated allogeneic hematopoietic cell
2011;108:E513–E518. transplantation for severe aplastic anemia. JAMA.
[23] Pickett HA, Reddel RR. The role of telomere trimming 2015;313:594–602.
in normal telomere length dynamics. Cell Cycle. [40] Bojesen SE, Pooley KA, Johnatty SE, et al. Multiple
2012;11:1309–1315. independent variants at the TERT locus are associ-
[24] Palm W, de Lange T. How shelterin protects mamma- ated with telomere length and risks of breast and
lian telomeres. Annu Rev Genet. 2008;42:301–334. ovarian cancer. Nat Genet. 2013;45:371–384.
[25] Chan SS, Chang S. Defending the end zone: studying 384e371-372.
the players involved in protecting chromosome [41] Walsh KM, Codd V, Smirnov IV, et al. Variants near
ends. FEBS Lett. 2010;584:3773–3778. TERT and TERC influencing telomere length are asso-
[26] Miyake Y, Nakamura M, Nabetani A, et al. RPA-like ciated with high-grade glioma risk. Nat Genet.
mammalian Ctc1-Stn1-Ten1 complex binds to single- 2014;46:731–735.
20 C. L. FASCHING

[42] Marnett LJ. Oxyradicals and DNA damage. [60] Bojesen SE. Telomeres and human health. J Intern
Carcinogenesis. 2000;21:361–370. Med. 2013;274:399–413.
[43] Petersen S, Saretzki G, von Zglinicki T. Preferential [61] Martin-Ruiz CM, Baird D, Roger L, et al.
accumulation of single-stranded regions in telomeres Reproducibility of Telomere Length Assessment–An
of human fibroblasts. Exp Cell Res. 1998;239: International Collaborative Study. Int J Epidemiol.
152–160. 2015;44:1749–1754.
[44] Oikawa S, Kawanishi S. Site-specific DNA damage at [62] Mangino M, Hwang SJ, Spector TD, et al. Genome-
GGG sequence by oxidative stress may accelerate wide meta-analysis points to CTC1 and ZNF676 as
telomere shortening. FEBS Lett. 1999;453:365–368. genes regulating telomere homeostasis in humans.
[45] Opresko PL, Fan J, Danzy S, et al. 3rd, Bohr VA. Hum Mol Genet. 2012;21:5385–5394.
Oxidative damage in telomeric DNA disrupts recogni- [63] Howden LMaJAM. Age and Sex Composition: 2010.
tion by TRF1 and TRF2. Nucleic Acids Res. In: Bureau USC, ed. Washington, D.C.: U. S. Census
2005;33:1230–1239., Bureau; 2011.
[46] Madjid M, Fatemi O. Components of the complete [64] Deelen J, Beekman M, Codd V, et al. Leukocyte telo-
blood count as risk predictors for coronary heart dis- mere length associates with prospective mortality
ease: in-depth review and update. Tex Heart Inst J. independent of immune-related parameters and
2013;40:17–29. known genetic markers. Int J Epidemiol. 2014;43:
[47] Mittal M, Siddiqui MR, Tran K, et al. Reactive oxygen 878–886.
species in inflammation and tissue injury. Antioxid [65] Lapham K, Kvale MN, Lin J, et al. Automated assay of
Redox Signal. 2014;20:1126–1167. telomere length measurement and informatics for
[48] Demissie S, Levy D, Benjamin EJ, et al. Insulin resist- 100,000 subjects in the genetic epidemiology
ance, oxidative stress, hypertension, and leukocyte research on adult health and aging (GERA) cohort.
telomere length in men from the Framingham Heart Genetics. 2015;200:1061–1072.
Study. Aging Cell. 2006;5:325–330. [66] Rode L, Nordestgaard BG, Bojesen SE. Peripheral
[49] Doulatov S, Notta F, Laurenti E, et al. Hematopoiesis: blood leukocyte telomere length and mortality
a human perspective. Cell Stem Cell. 2012;10:
among 64,637 individuals from the general popula-
120–136.
tion. J Natl Cancer Inst. 2015;107:djv074
[50] Rufer N, Brummendorf TH, Kolvraa S, et al. Telomere
[67] Weischer M, Bojesen SE, Nordestgaard BG. Telomere
fluorescence measurements in granulocytes and T
shortening unrelated to smoking, body weight, phys-
lymphocyte subsets point to a high turnover of hem-
ical activity, and alcohol intake: 4,576 general popu-
atopoietic stem cells and memory T cells in early
lation individuals with repeat measurements 10
childhood. J Exp Med. 1999;190:157–167.
years apart. PLoS Genet. 2014;10:e1004191.
[51] Cawthon RM. Telomere measurement by quantitative
[68] Mons U, Muezzinler A, Schottker B, et al. Leukocyte
PCR. Nucleic Acids Res. 2002;30:e47
telomere length and all-cause, cardiovascular disease,
[52] Cawthon RM. Telomere length measurement by a
and cancer mortality: results from individual-partici-
novel monochrome multiplex quantitative PCR
pant-data meta-analysis of 2 large prospective cohort
method. Nucleic Acids Res. 2009;37:e21
[53] Canela A, Vera E, Klatt P, et al. High-throughput telo- studies. Am J Epidemiol. 2017;185:1317–1326.
mere length quantification by FISH and its applica- [69] Needham BL, Rehkopf D, Adler N, et al. Leukocyte
tion to human population studies. Proc Natl Acad Sci telomere length and mortality in the National Health
USA. 2007;104:5300–5305. and Nutrition Examination Survey, 1999-2002.
[54] Poon SS, Martens UM, Ward RK, et al. Telomere Epidemiology. 2015;26:528–535.
length measurements using digital fluorescence [70] Goglin SE, Farzaneh-Far R, Epel ES, et al. Change in
microscopy. Cytometry. 1999;36:267–278. leukocyte telomere length predicts mortality in
[55] Kimura M, Stone RC, Hunt SC, et al. Measurement of patients with stable coronary heart disease from the
telomere length by the Southern blot analysis of ter- Heart and Soul Study. PLoS One. 2016;11:e0168868.
minal restriction fragment lengths. Nat Protoc. [71] Farzaneh-Far R, Cawthon RM, Na B, et al. Prognostic
2010;5:1596–1607. value of leukocyte telomere length in patients with
[56] Baird DM, Rowson J, Wynford-Thomas D, et al. stable coronary artery disease: data from the Heart
Extensive allelic variation and ultrashort telomeres in and Soul Study. Arterioscler Thromb Vasc Biol.
senescent human cells. Nat Genet. 2003;33:203–207. 2008;28:1379–1384.
[57] Aubert G, Hills M, Lansdorp PM. Telomere length [72] Houben JM, Giltay EJ, Rius-Ottenheim N, et al.
measurement-caveats and a critical assessment of Telomere length and mortality in elderly men: the
the available technologies and tools. Mutat Res. Zutphen Elderly Study. J Gerontol A Biol Sci Med Sci.
2012;730:59–67. 2011;66:38–44.
[58] Lai TP, Wright WE, Shay JW. Comparison of telomere [73] Svensson J, Karlsson MK, Ljunggren O, et al.
length measurement methods. Phil Trans R Soc B. Leukocyte telomere length is not associated with
2018;373:20160451. mortality in older men. Exp Gerontol. 2014;57:6–12.
[59] Sanders JL, Newman AB. Telomere length in epi- [74] Marioni RE, Harris SE, Shah S, et al. The epigenetic
demiology: a biomarker of aging, age-related dis- clock and telomere length are independently associ-
ease, both, or neither? Epidemiol Rev. 2013;35: ated with chronological age and mortality. Int J
112–131. Epidemiol. 2016;45:424–432.
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 21

[75] Benjamin EJ, Blaha MJ, Chiuve SE, et al. Heart [91] Mainous AG, 3rd, Codd V, Diaz VA, et al. Leukocyte
Disease and Stroke Statistics-2017 Update: A Report telomere length and coronary artery calcification.
From the American Heart Association. Circulation. Atherosclerosis. 2010;210:262–267.
2017;135:e146–e603. [92] Hunt SC, Kimura M, Hopkins PN, et al. Leukocyte
[76] Bezzina CR, Lahrouchi N, Priori SG. Genetics of sud- telomere length and coronary artery calcium. Am J
den cardiac death. Circ Res. 2015;116:1919–1936. Cardiol. 2015;116:214–218.
[77] Yang KC, Kyle JW, Makielski JC, et al. Mechanisms of [93] Rask-Madsen C, Kahn CR. Tissue-specific insulin
sudden cardiac death: oxidants and metabolism. Circ signaling, metabolic syndrome, and cardiovascular
Res. 2015;116:1937–1955. disease. Arterioscler Thromb Vasc Biol. 2012;32:
[78] Wellens HJ, Schwartz PJ, Lindemans FW, et al. Risk 2052–2059.
stratification for sudden cardiac death: current status [94] Goldsworthy ME, Potter PK. Modelling age-related
and challenges for the future. Eur Heart J. metabolic disorders in the mouse. Mamm Genome.
2014;35:1642–1651. 2014;25:487–496.
[79] Hayashi M, Shimizu W, Albert CM. The spectrum of [95] Abel ED, O’Shea KM, Ramasamy R. Insulin resistance:
epidemiology underlying sudden cardiac death. Circ metabolic mechanisms and consequences in the
Res. 2015;116:1887–1906. heart. Arterioscler Thromb Vasc Biol. 2012;32:
[80] Wilson PW, D’Agostino RB, Levy D, et al. Prediction 2068–2076.
of coronary heart disease using risk factor categories. [96] Willeit P, Raschenberger J, Heydon EE, et al.
Circulation. 1998;97:1837–1847. Leucocyte telomere length and risk of type 2 dia-
[81] Brouilette S, Singh RK, Thompson JR, et al. White cell betes mellitus: new prospective cohort study and lit-
telomere length and risk of premature myocardial erature-based meta-analysis. PLoS One. 2014;9:
infarction. Arterioscler Thromb Vasc Biol. 2003;23: e112483.
842–846. [97] Zhao J, Zhu Y, Lin J, et al. Short leukocyte telomere
[82] Brouilette SW, Moore JS, McMahon AD, et al. length predicts risk of diabetes in American Indians:
Telomere length, risk of coronary heart disease, and the strong heart family study. Diabetes. 2014;63:
statin treatment in the West of Scotland Primary 354–362.
Prevention Study: a nested case-control study. [98] Xiao F, Zheng X, Cui M, et al. Telomere dys-
Lancet. 2007;369:107–114. function-related serological markers are associated
[83] Stone NJ, Robinson JG, Lichtenstein AH, et al. ACC/ with type 2 diabetes. Diabetes Care. 2011;34:
AHA guideline on the treatment of blood cholesterol 2273–2278.
to reduce atherosclerotic cardiovascular risk in [99] Menke A, Casagrande S, Cowie CC. Leukocyte telo-
adults: a report of the American College of mere length and diabetes status, duration, and
Cardiology/American Heart Association Task Force on control: the 1999-2002 National Health and
Practice Guidelines. Circulation. 2013;2014:S1–S45. Nutrition Examination Survey. BMC Endocr Disord.
[84] Hammadah M, Al Mheid I, Wilmot K, et al. Telomere 2015;15:52.
shortening, regenerative capacity, and cardiovascular [100] Bonfigli AR, Spazzafumo L, Prattichizzo F, et al.
outcomes. Circ Res. 2017;120:1130–1138. Leukocyte telomere length and mortality risk in
[85] Fitzpatrick AL, Kronmal RA, Gardner JP, et al. patients with type 2 diabetes. Oncotarget.
Leukocyte telomere length and cardiovascular dis- 2016;7:50835–50844.
ease in the cardiovascular health study. Am J [101] Astrup AS, Tarnow L, Jorsal A, et al. Telomere length
Epidemiol. 2007;165:14–21. predicts all-cause mortality in patients with type 1
[86] D’Mello MJJ, Ross SA, Anand SS, et al. Telomere diabetes. Diabetologia. 2010;53:45–48.
Length and Risk of Myocardial Infarction in a [102] Lawlor DA, Harbord RM, Sterne JA, et al. Mendelian
MultiEthnic Population: The INTERHEART Study. J Am randomization: using genes as instruments for mak-
Coll Cardiol. 2016;67:1863–1865. ing causal inferences in epidemiology. Statist Med.
[87] Scheller Madrid A, Rode L, Nordestgaard BG, et al. 2008;27:1133–1163.
Short Telomere Length and Ischemic Heart Disease: [103] Thomas DC, Conti DV. Commentary: the concept of
Observational and Genetic Studies in 290 022 ’Mendelian Randomization’. Int J Epidemiol. 2004;33:
Individuals. Clin Chem. 2016;62:1140–1149. 21–25.
[88] Willeit P, Willeit J, Mayr A, et al. Telomere length and [104] Didelez V, Sheehan N. Mendelian randomization as
risk of incident cancer and cancer mortality. JAMA. an instrumental variable approach to causal infer-
2010;304:69–75. ence. Stat Methods Med Res. 2007;16:309–330.
[89] Peters SA, den Ruijter HM, Bots ML, et al. [105] Codd V, Nelson CP, Albrecht E, et al. Identification of
Improvements in risk stratification for the occurrence seven loci affecting mean telomere length and their
of cardiovascular disease by imaging subclinical ath- association with disease. Nat Genet. 2013;45:
erosclerosis: a systematic review. Heart. 2012;98: 422–427.
177–184. [106] Pooley KA, Bojesen SE, Weischer M, et al. A genome-
[90] Raschenberger J, Kollerits B, Hammerer-Lercher A, wide association scan (GWAS) for mean telomere
et al. The association of relative telomere length with length within the COGS project: identified loci show
symptomatic peripheral arterial disease: results from little association with hormone-related cancer risk.
the CAVASIC study. Atherosclerosis. 2013;229:469–474. Hum Mol Genet. 2013;22:5056–5064.
22 C. L. FASCHING

[107] Codd V, Mangino M, van der Harst P, et al. Common [123] Karlseder J, Kachatrian L, Takai H, et al. Targeted
variants near TERC are associated with mean telo- deletion reveals an essential function for the telo-
mere length. Nat Genet. 2010;42:197–199. mere length regulator Trf1. Mol Cell Biol.
[108] Levy D, Neuhausen SL, Hunt SC, et al. Genome-wide 2003;23:6533–6541.
association identifies OBFC1 as a locus involved in [124] Celli GB, de Lange T. DNA processing is not required
human leukocyte telomere biology. Proc Natl Acad for ATM-mediated telomere damage response after
Sci USA. 2010;107:9293–9298. TRF2 deletion. Nat Cell Biol. 2005;7:712–718.
[109] Prescott J, Kraft P, Chasman DI, et al. Genome-wide [125] Chiang YJ, Kim SH, Tessarollo L, et al. Telomere-asso-
association study of relative telomere length. PLoS ciated protein TIN2 is essential for early embryonic
One. 2011;6:e19635 development through a telomerase-independent
[110] Lee JH, Cheng R, Honig LS, et al. Genome wide asso- pathway. Mol Cell Biol. 2004;24:6631–6634.
ciation and linkage analyses identified three loci- [126] Kibe T, Osawa GA, Keegan CE, et al. Telomere protec-
4q25, 17q23.2, and 10q11.21-associated with vari- tion by TPP1 is mediated by POT1a and POT1b. Mol
ation in leukocyte telomere length: the Long Life Cell Biol. 2010;30:1059–1066.
Family Study. Front Genet. 2014;4:310. [127] Hockemeyer D, Daniels JP, Takai H, et al. Recent
[111] Maubaret CG, Salpea KD, Romanoski CE, et al. expansion of the telomeric complex in rodents: two
Association of TERC and OBFC1 haplotypes with distinct POT1 proteins protect mouse telomeres. Cell.
mean leukocyte telomere length and risk for coron- 2006;126:63–77.
ary heart disease. PLoS One. 2013;8:e83122 [128] Robles-Espinoza CD, Harland M, Ramsay AJ, et al.
[112] van Steensel B, de Lange T. Control of telomere POT1 loss-of-function variants predispose to familial
length by the human telomeric protein TRF1. Nature. melanoma. Nat Genet. 2014;46:478–481.
1997;385:740–743. [129] Speedy HE, Kinnersley B, Chubb D, et al. Germ line
[113] Smogorzewska A, van Steensel B, Bianchi A, et al. mutations in shelterin complex genes are associated
Control of human telomere length by TRF1 and with familial chronic lymphocytic leukemia. Blood.
TRF2. Mol Cell Biol. 2000;20:1659–1668. 2016;128:2319–2326.
[114] Ye JZ, Donigian JR, van Overbeek M, et al. TIN2 [130] Wan M, Qin J, Songyang Z, et al. OB fold-containing
binds TRF1 and TRF2 simultaneously and stabilizes protein 1 (OBFC1), a human homolog of yeast Stn1,
the TRF2 complex on telomeres. J Biol Chem. associates with TPP1 and is implicated in telomere
2004;279:47264–47271. length regulation. J Biol Chem. 2009;284:
[115] Takai KK, Kibe T, Donigian JR, et al. Telomere protec- 26725–26731.
tion by TPP1/POT1 requires tethering to TIN2. Mol [131] Rice C, Skordalakes E. Structure and function of the
Cell. 2011;44:647–659. telomeric CST complex. Comput Struct Biotechnol J.
[116] Nandakumar J, Cech TR. Finding the end: recruit- 2016;14:161–167.
ment of telomerase to telomeres. Nat Rev Mol Cell [132] Kasbek C, Wang F, Price CM. Human TEN1 maintains
Biol. 2013;14:69–82. telomere integrity and functions in genome-wide
[117] Sexton AN, Regalado SG, Lai CS, et al. Genetic and replication restart. J Biol Chem. 2013;288:
molecular identification of three human TPP1 func- 30139–30150.
tions in telomerase action: recruitment, activation, [133] Said MA, Eppinga RN, Hagemeijer Y, et al. Telomere
and homeostasis set point regulation. Genes Dev. length and risk of cardiovascular disease and cancer.
2014;28:1885–1899. J Am Coll Cardiol. 2017;70:506–507.
[118] Rai R, Chen Y, Lei M, et al. TRF2-RAP1 is required to [134] Zhan Y, Karlsson IK, Karlsson R, et al. Exploring the
protect telomeres from engaging in homologous Causal Pathway From Telomere Length to Coronary
recombination-mediated deletions and fusions. Nat Heart Disease: A Network Mendelian Randomization
Comms. 2016;7:10881. Study. Circ Res. 2017;121:214–219.
[119] Savage SA, Giri N, Baerlocher GM, et al. TINF2, a [135] Wang H, Chu WS, Lu T, et al. Uncoupling protein-2
component of the shelterin telomere protection polymorphisms in type 2 diabetes, obesity, and insu-
complex, is mutated in dyskeratosis congenita. Am J lin secretion. Am J Physiol Endocrinol Metab.
Hum Genet. 2008;82:501–509. 2004;286:E1–E7.
[120] Walne AJ, Vulliamy T, Beswick R, et al. TINF2 muta- [136] Salpea KD, Talmud PJ, Cooper JA, et al. Association
tions result in very short telomeres: analysis of a of telomere length with type 2 diabetes, oxidative
large cohort of patients with dyskeratosis congenita stress and UCP2 gene variation. Atherosclerosis.
and related bone marrow failure syndromes. Blood. 2010;209:42–50.
2008;112:3594–3600. [137] Zhou Y, Simmons D, Hambly BD, et al. Interactions
[121] Kocak H, Ballew BJ, Bisht K, et al. Hoyeraal- between UCP2 SNPs and telomere length exist in
Hreidarsson syndrome caused by a germline muta- the absence of diabetes or pre-diabetes. Sci Rep.
tion in the TEL patch of the telomere protein TPP1. 2016;6:33147
Genes Dev. 2014;28:2090–2102. [138] Reddel RR. The role of senescence and immortaliza-
[122] Guo Y, Kartawinata M, Li J, et al. Inherited bone mar- tion in carcinogenesis. Carcinogenesis. 2000;21:
row failure associated with germline mutation of 477–484.
ACD, the gene encoding telomere protein TPP1. [139] Weischer M, Nordestgaard BG, Cawthon RM, et al.
Blood. 2014;124:2767–2774. Short telomere length, cancer survival, and cancer
CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES 23

risk in 47102 individuals. J Natl Cancer Inst. [150] Lin TT, Letsolo BT, Jones RE, et al. Telomere dysfunc-
2013;105:459–468. tion and fusion during the progression of chronic
[140] Rode L, Nordestgaard BG, Bojesen SE. Long telo- lymphocytic leukemia: evidence for a telomere crisis.
meres and cancer risk among 95 568 individuals Blood. 2010;116:1899–1907.
from the general population. Int J Epidemiol. [151] Grabowski P, Hultdin M, Karlsson K, et al. Telomere
2016;45:1634–1643. length as a prognostic parameter in chronic lympho-
[141] Willeit P, Willeit J, Kloss-Brandstatter A, et al. Fifteen- cytic leukemia with special reference to VH gene
year follow-up of association between telomere mutation status. Blood. 2005;105:4807–4812.
length and incident cancer and cancer mortality. [152] Zhang C, Doherty JA, Burgess S, et al. Genetic deter-
JAMA. 2011;306:42–44. minants of telomere length and risk of common can-
[142] Sanchez-Espiridion B, Chen M, Chang JY, et al. cers: a Mendelian randomization study. Hum Mol
Telomere length in peripheral blood leukocytes and Genet. 2015;24:5356–5366.
lung cancer risk: a large case-control study in [153] Ojha J, Codd V, Nelson CP, et al. Genetic variation
Caucasians. Cancer Res. 2014;74:2476–2486. associated with longer telomere length increases risk
[143] Campa D, Mergarten B, De Vivo I, et al. Leukocyte of chronic lymphocytic leukemia. Cancer Epidemiol
telomere length in relation to pancreatic cancer risk: Biomarkers Prev. 2016;25:1043–1049.
a prospective study. Cancer Epidemiol Biomarkers [154] Machiela MJ, Lan Q, Slager SL, et al. Genetically pre-
Prev. 2014;23:2447–2454. dicted longer telomere length is associated with
[144] Brummendorf TH, Holyoake TL, Rufer N, et al. increased risk of B-cell lymphoma subtypes. Hum
Prognostic implications of differences in telomere Mol Genet. 2016;25:1663–1676.
[155] Jones AM, Beggs AD, Carvajal-Carmona L, et al. TERC
length between normal and malignant cells from
polymorphisms are associated both with susceptibil-
patients with chronic myeloid leukemia measured by
ity to colorectal cancer and with longer telomeres.
flow cytometry. Blood. 2000;95:1883–1890.
Gut. 2012;61:248–254.
[145] Boultwood J, Peniket A, Watkins F, et al. Telomere
[156] Mirabello L, Yu K, Kraft P, et al. The association of
length shortening in chronic myelogenous leukemia
telomere length and genetic variation in telomere
is associated with reduced time to accelerated
biology genes. Hum Mutat. 2010;31:1050–1058.
phase. Blood. 2000;96:358–361.
[157] Haycock PC, Burgess S, Nounu A, et al. Association
[146] Roos G, Krober A, Grabowski P, et al. Short telomeres
Between Telomere Length and Risk of Cancer and
are associated with genetic complexity, high-risk Non-Neoplastic Diseases: A Mendelian
genomic aberrations, and short survival in chronic Randomization Study. JAMA Oncol. 2017;3:636–651.
lymphocytic leukemia. Blood. 2008;111:2246–2252. [158] Martin-Ruiz CM, Baird D, Roger L, et al.
[147] Rossi D, Lobetti Bodoni C, Genuardi E, et al. Reproducibility of telomere length assessment: an
Telomere length is an independent predictor of sur- international collaborative study. Int J Epidemiol.
vival, treatment requirement and Richter’s syndrome 2015;44:1673–1683.
transformation in chronic lymphocytic leukemia. [159] Melin BS, Nordfjall K, Andersson U, et al. hTERT can-
Leukemia. 2009;23:1062–1072. cer risk genotypes are associated with telomere
[148] Rampazzo E, Bonaldi L, Trentin L, et al. Telomere length. Genet Epidemiol. 2012;36:368–372.
length and telomerase levels delineate subgroups of [160] Vasa-Nicotera M, Brouilette S, Mangino M, et al.
B-cell chronic lymphocytic leukemia with different Mapping of a major locus that determines telomere
biological characteristics and clinical outcomes. length in humans. Am J Hum Genet.
Haematologica. 2012;97:56–63. 2005;76:147–151.
[149] Strefford JC, Kadalayil L, Forster J, et al. Telomere [161] Mangino M, Brouilette S, Braund P, et al. A regula-
length predicts progression and overall survival in tory SNP of the BICD1 gene contributes to telomere
chronic lymphocytic leukemia: data from the UK LRF length variation in humans. Hum Mol Genet.
CLL4 trial. Leukemia. 2015;29:2411–2414. 2008;17:2518–2523.

You might also like