ArticlePDF AvailableLiterature Review

Abstract

Aging is defined as progressive physiological alterations in an organism that lead to senescence. In response to stress, when proliferative-competent cells undergo permanent, irreversible growth arrest (like replicative dividing limit, oncogene activation, oxidative stress, or deoxyribonucleic acid (DNA) damage), it is termed as cellular senescence. Biomarkers p53, telomerase, and other inflammatory cytokines have a vital link with senescence, and directed use of these markers might be useful in manipulating cancer and the aging process. We included studies related to topics ' accelerated aging due to cancer', telomerase's relation to Aging and Cancer, p53's relation to Aging and Cancer, Atherosclerosis and Cancer from Search databases like PubMed and Google Scholar. We relied on peer-reviewed articles and included literature from the last 10 years written in the English language. Degenerative diseases in humans are usually linked to atherosclerosis, and atherosclerosis is associated with short leukocyte telomere length. Cancer itself and its treatment are linked with accelerated aging by causing progressive shortening of telomeres during cell replication, resulting in cell death. Gene p53 is known to have a dual effect that works as a tumor suppressor and has pro-aging side effects. In experimental studies, when p53 overcomes multiple regulatory mechanisms controlling its activity, then only the pro-aging side effects of p53 manifested. This might be a potential key for treating cancer without causing the side-effects of aging. In this review, we aim to explain and summarize the interdependent nature of p53, telomeres, and other conventional mechanisms of aging and cancer like inflammation, oxidative stress, uncontrolled proliferation, angiogenesis, micro ribonucleic acids (RNAs), and apoptosis, with a more synergistic approach that can help in developing new therapeutics and play a potential role in shaping modern human lifespan and revolutionize cancer treatment.
Received 08/30/2020
Review began 09/16/2020
Review ended 10/01/2020
Published 10/04/2020
© Copyright 2020
Dhalla et al. This is an open access article
distributed under the terms of the
Creative Commons Attribution License
CC-BY 4.0., which permits unrestricted
use, distribution, and reproduction in any
medium, provided the original author and
source are credited.
Comparing the Role of the p53 Gene and
Telomerase Enzyme in ‘Accelerated Aging Due to
Cancer’: A Literature Review
Paramvijay Singh Dhalla , Arunima Kaul , Jian Garcia , Anusha Bapatla , Raheela Khalid , Ana S.
Armenta-Quiroga , Safeera Khan
1. Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA 2. Internal Medicine,
California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
Corresponding author: Paramvijay Singh Dhalla, dhallaparamvijay@gmail.com
Abstract
Aging is defined as progressive physiological alterations in an organism that lead to senescence. In response
to stress, when proliferative-competent cells undergo permanent, irreversible growth arrest (like replicative
dividing limit, oncogene activation, oxidative stress, or deoxyribonucleic acid (DNA) damage), it is termed as
cellular senescence. Biomarkers p53, telomerase, and other inflammatory cytokines have a vital link with
senescence, and directed use of these markers might be useful in manipulating cancer and the aging process.
We included studies related to topics ' accelerated aging due to cancer', telomerase's relation to Aging and
Cancer, p53's relation to Aging and Cancer, Atherosclerosis and Cancer from Search databases like PubMed
and Google Scholar. We relied on peer-reviewed articles and included literature from the last 10 years
written in the English language. Degenerative diseases in humans are usually linked to atherosclerosis, and
atherosclerosis is associated with short leukocyte telomere length. Cancer itself and its treatment are linked
with accelerated aging by causing progressive shortening of telomeres during cell replication, resulting in
cell death. Gene p53 is known to have a dual effect that works as a tumor suppressor and has pro-aging side
effects. In experimental studies, when p53 overcomes multiple regulatory mechanisms controlling its
activity, then only the pro-aging side effects of p53 manifested. This might be a potential key for treating
cancer without causing the side-effects of aging. In this review, we aim to explain and summarize the
interdependent nature of p53, telomeres, and other conventional mechanisms of aging and cancer like
inflammation, oxidative stress, uncontrolled proliferation, angiogenesis, micro ribonucleic acids (RNAs),
and apoptosis, with a more synergistic approach that can help in developing new therapeutics and play a
potential role in shaping modern human lifespan and revolutionize cancer treatment.
Categories: Internal Medicine, Oncology
Keywords: p53, telomerase, aging, cancer, metformin, statin, p16ink4a, progeria, atherosclerosis, sasp
Introduction And Background
Aging is defined as the constant physiological changes in an organism leading to senescence, biological
function deterioration, and the capacity to adapt to metabolic stress. 'Senescence' can refer to cellular
senescence or whole organism's senescence. Organismal senescence causes an increase in death rates and/or
a decrease in fecundity associated with increasing age. In other words, aging applies to degenerative
alterations that happen in every organism without any death reference. In contrast, senescence applies to
the developmental stage at which 'close to death' manifestations become evident. Manifestations of the
aging process include graying of hair, increased susceptibility to infection, higher risk of heatstroke or
hypothermia, and musculoskeletal changes. The nine hallmarks of aging are mentioned in Figure 1.
1 2 2 2 2
2 2
Open Access Review
Article DOI: 10.7759/cureus.10794
How to cite this article
Dhalla P, Kaul A, Garcia J, et al. (October 04, 2020) Comparing the Role of the p53 Gene and Telomerase Enzyme in ‘Accelerated Aging Due to
Cancer’: A Literature Review. Cureus 12(10): e10794. DOI 10.7759/cureus.10794
FIGURE 1: Nine Hallmarks of Aging
Accelerated aging in cancer
Some studies have investigated accelerated aging in cancer survivors and concluded that accelerated aging
manifests as frailty, which is a clinical syndrome in which an individual cannot revert to baseline functional
status after a physical insult [1]. Accelerated frailty has been linked to a more striking comorbidity burden in
childhood cancer survivors, which was observed in the brains of adult survivors of pediatric lymphoid
malignancies and adult breast cancer survivors, than in non-cancer control groups. Compiling evidence
supports the hypothesis that cancer itself and its treatment are associated with accelerated aging [1-3].
Aging and cancer have many facets; hence, there are numerous theories, each of which may reveal one or
more aging perspectives. One of the notable theories of aging, which we will review in detail, pivots around
telomeres, which are repeated segments of deoxyribonucleic acid (DNA) seen at the ends of chromosomes.
These decide the maximum life span of a cell because each time a cell divides, many repeats are lost, which
shortens telomere, resulting in cell death. Thus, progressive chromosome shortening occurs during cell
replication and is observed with aging [4]. Various studies have shown that most degenerative and
inflammatory diseases, and many cancers, characterized by oxidative stress, contribute to accelerated
telomere shortening [5-6]. As a prerequisite to cellular senescence, a transcriptional regulator called gene
p53 causes apoptosis or cell cycle arrest, represses stem and progenitor cell populations' mobilization,
resulting in an accelerated aging process. Moreover, p53 also represses unregulated proliferation pathways,
leading to senescence-associated secretory phenotype (SASP) and cellular senescence. This generates
degenerative and pro-inflammatory tissue milieu resulting in aging process suppression. Henceforth, p53
has the potential to both hinder and expedite cellular aging processes [1,7]. In this review, we will discuss
the quantitative expression p16ink4a as a biomarker of aging, used to predict radiation toxicity due to
cancer treatment. We aim to explain and summarize the interdependent nature of p53, telomeres, and other
conventional mechanisms of aging and cancer with a more collaborative approach, which might aid in
understanding these mechanisms from a different perspective.
Review
Method
2020 Dhalla et al. Cureus 12(10): e10794. DOI 10.7759/cureus.10794 2 of 11
We collected free full-text articles as data using PubMed and Google Scholar as our primary databases. We
included studies related to the topics 'accelerated aging due to cancer', telomerase's relation to aging and
cancer, p53's relation to aging and cancer, atherosclerosis, and cancer. We relied on peer-reviewed articles
and included literature from the last 10 years written in the English language. We included all possible types
of studies - mixed studies that covered all kinds of ethnicities from around the world. Participants were
cancer patients of all ages and gender with signs of accelerated aging like atherosclerosis, gray hair,
osteoporosis, and frailty, with biomarkers of aging and cancer like p53 and telomerase enzyme. We excluded
non-peer-reviewed articles, literature before 10 years, and articles written in any other language except
English. Non-cancer patients with frailty or cancer patients without signs of frailty were also excluded.
Discussion
One of the leading causes of death in both developing and developed countries being cancer. There is a
robust biological association between the conventional mechanisms of aging and cancer occurrence. In this
review article, we will explain different theories that discuss the evidence on the vital link between aging
mechanisms and carcinogenesis. These theories are worth considering, which can help understand the
relationship between cancer and the aging process in the general population [8].
Telomerase Enzyme
Telomerase enzyme is a vital enzyme for cell survival that prevents telomere shortening; consequently,
cellular senescence is observed after many cell division rounds. By increasing the expression and
reactivation of the telomerase complex, cancer cells bypass cellular senescence. This is one of the processes
that is required for tumor transformation and progression [9]. Multiple studies have concentrated on
identifying strategies or compounds to inhibit telomerase activity in cancer cells with induction of
senescence and subsequent loss of telomere integrity [9-11].
Mechanisms of Oxidative Stress Accelerating Telomeric Shortening
To demonstrate how oxidative stress accelerates telomere shortening, multiple mechanistic theories have
been suggested. One of the theories proposes that oxidative stress triggers cell death or senescence, and to
compensate that, the survivors undergo more increased cell divisions, resulting in increased telomere
shortening. One of the widely mentioned theories proposes that reactive oxygen species (ROS) causes
single-strand breaks (SSB) at telomeres directly or as intermediates during lesion repair, resulting in
replication fork collapse and telomere loss (Figure 2). Alternatively, lesions that impede telomere replication
can cause an accumulation of unreplicated single-stranded DNA (ssDNA) and manifest as multi-telomeric
foci at chromatid ends termed fragile telomeres (Figure 2) [12-14].
FIGURE 2: Stalling and Blocking of Telomere's Replication Fork
Stalling and Blocking of the Telomeres Replication Fork
The schematic exhibits a description of how telomere losses or telomere fragility arises from DNA lesions
that collapse or stall replication fork progression, respectively. When the DNA replication fork faces SSBs,
it can cause the collapse of the fork, resulting in a double-strand break. Due to uncondensed regions that
2020 Dhalla et al. Cureus 12(10): e10794. DOI 10.7759/cureus.10794 3 of 11
result from accumulated unreplicated ssDNA, fragile telomeres manifest as multi-telomeric foci at a
chromatid end. For detection with a telomeric probe, telomere losses exhibit as chromatid ends lacking
sufficient telomeric DNA. By extending a prematurely truncated telomere, telomerase can suppress
telomere losses. Hence, telomerase's concept of regeneration can be used for the development of
therapeutics (Figure 2).
The Inverse Relation Between Cancer and Atherosclerosis Concerning Telomere Length
Degenerative diseases in humans are mostly linked to atherosclerosis, and atherosclerosis is associated with
short leukocyte telomere length (LTL) [15-17]. Short telomerase also gives protection against cancer
development by diminishing the proliferative activity of stem cells and limiting its regenerative capacity,
which gives rise to age-dependent degenerative conditions like atherosclerosis. To create 'genetic risk
scores' for cancers and atherosclerosis, which is expressed as coronary heart disease, recent studies have
used LTL-GWAS (genome-wide association) study findings [18]. As investigations showing, African
Americans have longer telomere length than in European ethnicity individuals; it is interesting to contrast
and compare the incidence of notable cancers and atherosclerosis between these two ethnicities. The
percentage of cancers like lung (after adjustment for smoking), prostate, pancreas, triple-negative breast
cancer, and its incipience at a younger age were greater in African Americans as compared to European
ethnicity [19-21]. On a contrary note, African Americans exhibited a lower incidence of atherosclerosis.
Hence, emphasizing on the role of telomere length in developing cancers and atherosclerosis more than risk
factors itself [22]. In these studies, the LTL-associated alleles (ZNF208, TERC, TERT, OBFC1, ACTP2, RTEL1,
and NAF1) seen are a risk sign for melanoma, lung cancer, and coronary heart disease. Therefore, when the
alleles' collective effect results in a relatively longer LTL, lung cancer and melanoma risk have risen opposite
to the risk of coronary heart disease, which was reduced. Evidencing the fact that increasing telomere length
increases cancer risk and decreases atherosclerosis, which is shown in Figure 3. The limitations of these
studies are that they don't include other common cancers like breast cancer and colorectal cancer, which
might be linked to LTL-associated alleles, and, if not, which other alleles they are linked to should also be
researched thoroughly (Figure 3) [23].
FIGURE 3: The Inverse Relation Between Cancer and Atherosclerosis
Concerning Telomere Length
p16ink4a Expression Due to Cancer Treatment Influencing Telomere Length
Radiation and chemotherapy can cause progressive and long-term tissue damage by the formation of pro-
inflammatory cytokines [24]. The accelerated development of second malignancies and other comorbidities
was also seen in survivors of childhood cancer treated with radiations. Chemotherapy has a significant effect
on telomere length. Repetitive standard-dose chemotherapy given for solid tumor patients was associated
with telomere shortening in hematopoietic stem cells and peripheral blood mononuclear cells [25]. Between
the ages of 20 and 80, a more robust marker for predicting molecular aging is the dynamic range of p16ink4a
expression, which increases approximately 10-fold. In one study of early-stage breast cancer, in which
females treated with adjuvant chemotherapy had their p16ink4a expression estimated in peripheral blood T
cells immediately after treatment and was raised by approximately one log2 order of magnitude and
continued to be present for one year following treatment [26], which corresponded to approximately a 15-
year increase in chronologic age. Undoubtedly, chemotherapy and radiation therapy accelerate aging;
hence, patients who are most vulnerable to cancer treatment can be identified by several biomarkers of
aging, which are now available and which might minimize treatment-related toxicity by allowing the earlier
testing of the interventions. To predict radiation toxicity in individual cases, studies testing how
quantitative expression biomarkers like p16ink4a might be utilized for developing possible interventions to
2020 Dhalla et al. Cureus 12(10): e10794. DOI 10.7759/cureus.10794 4 of 11
ameliorate radiation effects on patients; therefore, they are required to be researched in more detail [27].
p53 Association With Aging
A rare genetic disorder Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disorder that
causes de novo point mutation inside exon 11 of the LMNA gene, leading to the accumulation of progerin.
HGPS fibroblasts, which are near-senescent, show reduced levels of Δ133p53α and increased levels of p53β.
Moreover, due to progerin-induced faulty DNA repair and genomic instability, there is an accumulation of
unrepaired DNA and double-strand breaks, which induces cellular senescence [28]. Surprisingly, they found
that due to a tumor protection mechanism controlled by BRD4 (Bromodomain-containing protein 4), cells
from HGPS patients typically do not develop cancer. Hence, research into the unexplored mechanism of
BRD4 and the renewal of Δ133p53α expression can be a crucial link between aging and cancer, which might
aid in cancer prevention [29-31].
p53 Isoforms in Cancer
The role of p53 in cancer formation is paradoxical. The sudden p53 induction in sarcomas and hepatocellular
carcinomas provokes senescence followed by tumor elimination. On the contrary note, cellular senescence
has an ability for tumor promotion, which is presumably related to senescence-associated secretory
phenotype (SASP) factors. Tumors are arrested at the pre-malignant stage due to cellular senescence. SASP
factors are secreted by senescent cells, which promote senescence induction in a paracrine mode (relating to
or denoting a hormone that affects only in the vicinity of the gland secreting it) and further reinforces the
senescence in an autocrine mode (signifying or relating to a cell-produced substance that affects the cell by
which it is secreted) [32]. Through full-length p53 and p53 isoforms, the subsequent model explains the
regulation of aging, age-related disorders, and cellular senescence. Depending on the p53 status and cell
type, multiple stresses stimulate full-length p53 and change various p53 isoforms expressions. The cell-
autonomous effects, like loss of functional cells and regenerative capacity, induce cellular senescence by
changing p53 status like decreased D133p53a and increased p53b. The non-cell-autonomous effect, mainly
SASP, is also seen in senescent cells. Senescence can be reinforced by autocrine SASP; in turn, the induction
of senescence in neighboring cells is achieved by the impact of paracrine SASP and stimulate an immune
response, leading to tumor suppression and aging. Concurrently, tumor progression is caused by SASP,
which also supports fibrosis, angiogenesis, the proliferation of the cells, and tumor invasiveness. Full-length
p53 expressions and various p53 isoforms also regulate this dual outcome through cell-autonomous and
non-cell-autonomous functions. Besides, senescence and age-related outcomes require various p53 isoform
expression for its occurrence. The full-length p53 desirable outcomes are suppressing pre-malignant tumor
cells resulting in tumor suppression, getting rid of damaged cells by stimulating the immune system, and
supporting reparative mechanisms like tissue restoration and wound healing [33-35]. However, the
deleterious effects of full-length p53 comprise prolonged inflammatory reactions, promoting stem cell-like
phenotypes in malignant cells, immune evasion by the tumor, and tumor promotion by inducing
angiogenesis [30,32]. The integration of p53 with anti-growth interventions that enhance survival is shown
in Figure 4.
2020 Dhalla et al. Cureus 12(10): e10794. DOI 10.7759/cureus.10794 5 of 11
FIGURE 4: p53 Isoforms in Cancer
The Integration of p53 With Anti-Growth Interventions That Improve Survival
P53 induces G1 arrest as a part of a stress response, which inhibits mTORC1. G1-arrested cells are enabled
by the pro-growth pathway by introducing these mitogenic signals that change into senescent cells and
manifest as SASP. Cellular senescence represses the cell from developing into cancer; however, the SASP can
trigger cancer development from non-senescent cells by changing the tissue microenvironment.
Furthermore, it also decreases longevity and stimulates tissue degeneration. While p53-deletion promotes
cancer, simple p53 overexpression should reduce cancer. However, a mice experiment revealed that mice
carrying an additional p53 gene within a bacterial artificial chromosome (BAC) exhibited a reduced cancer
incidence, with surprisingly no distinctive exaggerated signs of aging. As ARF elevated p53 levels by
repressing MDM2 and increased gene dosage of p53 collectively with Arf, it resulted in a decline in cancer
rate and improved overall survival. Similarly, increased p53 levels were seen in mice with a hypomorphic
MDM2 allele, showing decreased cancer incidence without adverse side effects [7]. These effects are shown
in Figure 5.
2020 Dhalla et al. Cureus 12(10): e10794. DOI 10.7759/cureus.10794 6 of 11
FIGURE 5: Mitogenic Signals
Therefore, it concludes that enhanced p53-mediated cancer suppression was not toxic to adult mice, and
whenever p53 overwhelms various regulatory mechanisms that modulate its activity, only then pro-aging
side effects of p53 are exhibited. The limitation of the study mentioned above was that it was not conducted
on humans. A broad-based human study on identifying and manipulating these undiscovered regulatory
mechanisms should be done, which might be a potential key to treating cancer without the side effects of
aging [7]. On the other hand, another study done on mice with a reduced level of mdm2 showed a lack of
premature aging phenotype. This suggests that p53 function enhancement is not sufficient to provoke
aging. Hence, more study needs to be conducted to understand the underlying physiology to link p53
expression and aging [36]. Various p53 isoforms abnormal expression was also evidenced in breast cancer,
ovarian cancer, colon carcinoma, head and neck tumors, glioblastoma, melanoma, renal cell carcinoma, lung
cancer, and acute myeloid leukemia, hepatic cholangiocarcinoma, and acute myeloid leukemia [37-39].
Therapeutics Targeting p53 and Myc
A potential approach in pro-senescence therapy is targeting p53 either indirectly or directly and impacting
p53 in the senescence process. Hence, trials involving therapeutics activating p53 and/or its pathway are
currently under work. One of the strategies tested in tumors that retain wild-type p53 is hindering the
MDM2/p53 interaction and improving p53 function. Through the restoration of the p53-mediated tumor
suppression pathway, cancer cell growth arrest and apoptosis were induced while generating minimal
cytotoxicity and side effects, which has led to the discovery of Nutlin, a precise inhibitor of the p53/MDM2
interaction. Moreover, the effectiveness and side effects of Nutlin-3, such as increases in atherosclerosis, are
yet to be established. The signaling pathways involved in the synergistic effects of combining other drugs
could also be used to identify additional targets [9]. 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside
(AICAR) and metformin are compounds that induce adenosine monophosphate-activated protein kinase
(AMPK), resulting in the induction of p53. Additionally, metformin inhibits mTORC1 via AMPK, which
activates the potential insulin- and IGF-signaling pathways, consequently decreasing the harmful impacts of
diabetes mellitus type 2 (including increased risk of cancer). The activation of AMPK and p53 by metformin
has shown to inhibit melanoma invasion (but not migration or proliferation), inhibits growth, and improves
radiation response for non-small cell lung cancer. Like metformin, AICAR also induces AMPK, resulting in
enhancing p53 phosphorylation and promoting p21 to arrest endothelial cells in G0 or G1. Therefore, AMPK-
activating agents can potentially induce p53 and decrease mTORC1 to suppress cancer and possibly other
diseases [40]. As AMPK-activating agents like metformin have shown to inhibit melanoma invasion, it is a
possibility that the p53 gene may have an indirect effect on cancer through the direct effect of IGF-signaling
pathways. Hence, this association should be thoroughly researched upon which might help formulate new
cancer regimes. Other common causes of aging and cancer and their possible therapeutics are mentioned in
Table 1.
2020 Dhalla et al. Cureus 12(10): e10794. DOI 10.7759/cureus.10794 7 of 11
Mechanisms Mediators Effect Relation to Ageing & Cancers Possible Therapeutic interventions
Inflammation
Atherosclerosis antioxidants –
Phospholipase A2- Leukotrienes
pathway- Cyclooxygenase - TNF-α -
Cancer/atherosclerosis IL-6 - TGF-β
A cascade of biochemical events is
triggered due to the harmful stimuli that
induce the migration of leukocytes from
the blood to damaged tissue, resulting in
an inflammatory response that causes
the growth of the atherosclerotic lesion.
In cancer, macrophages and T cells are the predominant inflammato ry cells
since they are accountable for the secretion into the microenvironm ent of
massive amounts of inflammatory cytokines, proangiogenic factors, a nd reactive
oxygen species [41].
Interventions involving anti-
inflammatory molecule's
upregulation like TNF and
interleukin-1 receptor antagonist.
Oxidative
Stress
ROS's primary endogenous source is
the mitochondrial respiratory chain,
associated with enzymatic reactions
catalyzed by the xanthine oxidase,
nitric oxide synthase, and
NADH/NADPH oxidase.
Oxidative stress associated with local
inflammation, tissue remodeling,
endothelial dysfunction, smooth muscle
growth, and plaque formation has also
been associated with the production of
growth factors and mitogens that may
stimulate cell proliferation in early
atheromatous lesion sites.
As a consequence of the failure of the antioxidant systems, which is r esponsible
for their neutralization, promoting the development of inflammatory pro cesses,
there is an imbalance between RNS and ROS contributing to oxidat ive stress.
Which further causes the development of both atherosclerosis and cancer [42].
Interventions involving cell surface
receptors, such as CD44 and
several integrins that interact with
activated macrophages at sites of
inflammation, stimulates cell
adhesion and migration,
consequently, manipulating cancer
and the process of metastasis.
Uncontrolled
Proliferation
Involves regulatory proteins such as
cyclins and CDKs
Macrophages promote the development
of atherosclerotic plaque and, in various
tissues, stimulate the development of
different types of cancer.
Tissue homeostasis is regulated by two of the predominant physiologic al
processes like cell division and programmed cell death, where deregulat ion of
one of them provokes the development of several diseases, including cancer and
atherosclerosis [43].
Interventions target deregulation in
several control points like the G1-S
transition. It is one of the leading
causes of accelerated cell growth
and accumulated mutations.
Angiogenesis
Activators' growth factor - VEGF. FGF,
Cytokines – IL-1, IL-6, IL-8, cathepsin,
copper, oncogenes- c-Myc, r
Endothelin, erythropoietin, nitric oxide
synthase inhibitors cytokines- IL-10,
IL- 12, metalloproteinase, inhibitor,
zinc, oncogenes - p53, Rb endostatin,
interferon-a.
Formation of micro-vessels in an
atherosclerotic lesion contributes to the
development of plaque, the formation of
micro-vessels stimulated by hypoxia, HIF,
and ROS have a role in atherogenesis.
The progression of the primary atherosclerotic lesion requires angiogen esis; it is
known that the expansion of plaque and its risk complications such as rupture or
vascular thrombosis depends on this mechanism. Concerning cance r, tumor
vascular development is also essential for proliferation in processes suc h as
metastatic expansion since cancer cells depend on an adequate su pply of
oxygen and nutrients for this phenomenon to occur, where new bloo d and
lymphatic vessels are formed through angiogenesis and lymphangiog enesis [44].
An example of a medication that
blocks VEGF is bevacizumab
(Avastin), a monoclonal antibody
used in various cancers like colon
cancer. It blocks VEGF from binding
to the receptors on the cells that line
the blood vessels and stop
angiogenesis.
MicroRNAs in
atherosclerosis
and cancer
Both cancer and atherosclerosis
proangiogenic - Let7-f, miR-27b, and
miR-130a Inhibition of cellular
migration, endothelial proliferation,
and angiogenesis - miR-221, miR-222.
miRNAs are a group of highly conserved,
non-coding small RNAs, which play a
crucial role in gene regulation by acting
as repressors or activators. Since a
single miRNA may have various genes as
targets, and several miRNAs may share
the same target [45].
In atherosclerosis and cancer, the presence and regulation of severa l miRNAs
have also been associated with the control of cell proliferation, differen tiation,
and genomic stability, among other functions.
Since miRNAs have been found to
regulate atherosclerosis and Cancer
development, it becomes crucial to
investigate if similar therapeutic
strategies involving miRNAs could
apply to both diseases.
Apoptosis
The extrinsic or death receptor
pathway is triggered by the binding of
Fas and TNFR1 to Fas-L. The intrinsic
pathway is directed by two groups of
molecules, Bcl-2 and Bax.
Apoptosis is characterized by the
morphological and molecular changes in
cells, including cell shrinking, membrane
vesicle formation, and loss of adhesion to
neighboring cells.
The process of apoptosis is considered a determining factor in the reg ression or
progression of atherosclerosis by intervening in the stability of the plaqu e. On the
other hand, apoptosis has been recognized as an important player in c ancer
development since it functions as a molecular tool that cells employ to avoid the
proliferation of damaged cells, hence inhibiting tumor growth [46].
Studies investigating LDLR-
knockout mice have shown the
inactivation of an apoptosis
inhibitor expressed by macrophages
(Spα/Api6), increasing macrophage
apoptosis, and, therefore, inhibiting
atherosclerosis.
TABLE 1: Common Causes of Both Aging and Cancer Formation
TNF-α: tumor necrosis factor-alpha; IL: interleukin; TGF-β: transforming g rowth factor-beta; ROS: reactive oxygen species; CDK: cyclin-dependent
kinases; NADH: nicotinamide adenine dinucleotide + hydrogen; NADPH: nicotinamide adenine dinucleotide phosphate; RNS: reactive nitrogen
species; VEGF: vascular endothelial growth factor; FGF: fibroblast growth factor; RNA: ribonucleic acid; TNFR1: tumor necrosis factor receptor
1; LDLR: low-density lipoprotein receptor; miRNAs: microRNAs; HIF: hypoxia-inducible factor
Source: [45]
Therapeutics Targeting Atherosclerosis
Atherosclerosis has been considered a predisposing risk factor for several malignancies, especially of the
epithelial type (e.g., prostate, colon, ovary, lung), and a marker of aging. For example, tumors treated
with oxazaphosphorines and pyrimidine antagonists decreased the incidence and presence of
atherosclerotic lesions. The probable mechanism behind these effects was the changes in the concentration
of free cholesterol in the plasma membrane of cells affecting the formation of lipid rafts and caveolae. These
2020 Dhalla et al. Cureus 12(10): e10794. DOI 10.7759/cureus.10794 8 of 11
can be directly correlated to the function of crucial receptors, such as the epidermal growth factor receptor
(EGFR), members of the TNF receptor family, and the tumor necrosis factor-related apoptosis-inducing
ligand (TRAIL_ receptor that corresponds well in both cancer development and atherogenesis [47-48].
Combined immunotherapies employing gefitinib or trastuzumab or chemotherapies employing cisplatin or
doxorubicin, when coupled with the use of statins, surprisingly resulted in a higher therapeutic response in
gastrointestinal and lung cancers. Hence, it was concluded that the synergistic induction of cytotoxicity
employing immune- or chemotherapy in conjunction with statins improves the survival rate in patients with
ovarian cancer receiving only statins as a way to prevent an adverse cardiovascular event [47,49], as
atherosclerosis is the leading risk in aging due to cancer and its treatment which increases morbidity and
mortality. A novel therapeutic approach, which should be researched widely, would be the early treatment of
atherosclerosis with statins to prevent future risk of cardiovascular events due to cancer and its treatment.
As statins are readily available, it would be a game-changing strategy and can be manipulated after more in-
depth knowledge of these pathways is available in the future.
Conclusions
In this article, we have discussed various mechanisms such as telomerase enzyme, p53, and other common
mechanisms that indicate that 'accelerated aging' can be caused by cancer and its treatment. The 'double-
edged sword' effects of p53 are tumor suppression with pro-aging side effects and tumor promotion with
anti-aging effects. However, the pro-aging side effects of p53 usually manifest when p53 overwhelms the
many regulatory mechanisms that control its activity. As African American ethnicities have longer telomere
length than European ethnicities, African Americans developed more cancer and less atherosclerosis despite
having more risk factors for atherosclerosis. This could challenge 'the inverse relation theory between cancer
and atherosclerosis concerning telomere length.' Chemotherapy and radiation therapy cause accelerated
aging by affecting telomere length; it has been linked with telomere shortening in hematopoietic stem cells.
Thus, earlier testing of interventions with quantitative measurement of p16ink4a expression might benefit
in minimizing treatment-related toxicity. A new therapeutic strategy researched widely should be the early
treatment of atherosclerosis with statins to prevent future cardiovascular events due to cancer and its
treatment. This paper's limitation was that there was evidence from several experimental mouse studies,
whereas very few human trials have been conducted yet. Finally, we propose platforms for future research
on human telomere and p53 genetics due to its potential role in manipulating the human lifespan, which
might be a possible solution for treating cancer without the side effects of aging.
Additional Information
Disclosures
Conflicts of interest: In compliance with the ICMJE uniform disclosure form, all authors declare the
following: Payment/services info: All authors have declared that no financial support was received from
any organization for the submitted work. Financial relationships: All authors have declared that they have
no financial relationships at present or within the previous three years with any organizations that might
have an interest in the submitted work. Other relationships: All authors have declared that there are no
other relationships or activities that could appear to have influenced the submitted work.
References
1. Alfano CM, Peng J, Andridge RR, et al.: Inflammatory cytokines and comorbidity development in breast
cancer survivors versus non-cancer controls: evidence for accelerated aging?. J Clin Oncol. 2017, 35:149-156.
10.1200/JCO.2016.67.1883
2. Ness KK, Krull KR, Jones KE, et al.: Physiologic frailty as a sign of accelerated aging among adult survivors of
childhood cancer: a report from the St Jude Lifetime Cohort Study. J Clin Oncol. 2013, 31:4496-4503.
10.1200/JCO.2013.52.2268
3. Hurria A, Jones L, Hyman BM: Cancer treatment as an accelerated aging process: assessment, biomarkers,
and interventions. Am Soc Clin Oncol Educ Book. 2016, 35:516-522. 10.1200/EDBK_156160
4. Harley CB, Futcher AB, Greider CW: Telomeres shorten during aging of human fibroblasts . Nature. 1990,
345:458-460. 10.1038/345458a0
5. Sanders JL, Iannaccone A, Boudreau RM, et al.: The association of cataract with leukocyte telomere length
in older adults: defining a new marker of aging. J Gerontol A Biol Sci Med Sci. 2011, 66:639-645.
10.1093/gerona/glr034
6. Zhang J, Rane G, Dai X, et al.: Aging and the telomere connection: an intimate relationship with
inflammation. Ageing Res Rev. 2016, 25:55-69. 10.1016/j.arr.2015.11.006
7. Hasty P, Campisi J, Sharp ZD: Do p53 stress responses impact organismal aging? . Transl Cancer Res. 2016,
5:685-691. 10.21037/tcr.2016.12.02
8. Armenian SH, Gibson CJ, Rockne RC, Ness KK: Premature aging in young cancer survivors . J Natl Cancer
Inst. 2019, 111:226-232. 10.1093/jnci/djy229
9. Calcinotto A, Kohli J, Zagato E, Pellegrini L, Demaria M, Alimonti A: Cellular senescence: aging, cancer, and
injury. Psychol Rev. 2019, 99:1047-1078. 10.1152/physrev.00020.2018
10. Ait-Aissa K, Ebben JD, Kadlec AO, Beyer AM: Friend or foe? Telomerase as a pharmacological target in
cancer and cardiovascular disease. Pharmacol Res. 2016, 111:422-433. 10.1016/j.phrs.2016.07.003
11. Gomez DL, Armando RG, Cerrudo CS, Ghiringhelli PD, Gomez DE: Telomerase as a cancer target.
Development of new molecules. Curr Top Med Chem. 2016, 16:2432-2440.
12. Zglinicki VT: Oxidative stress shortens telomeres . Trends Biochem Sci. 2002, 27:339-344. 10.1016/S0968-
2020 Dhalla et al. Cureus 12(10): e10794. DOI 10.7759/cureus.10794 9 of 11
0004(02)02110-2
13. Sfeir A, Kosiyatrakul ST, Hockemeyer D, MacRae SL, Karlseder J, Schildkraut CL, de Lange T: Mammalian
telomeres resemble fragile sites and require TRF1 for efficient replication . Cell. 2009, 138:90-103.
10.1016/j.cell.2009.06.021
14. Barnes RP, Fouquerel E, Opresko PL: The impact of oxidative DNA damage and stress on telomere
homeostasis. Mech Ageing Dev. 2019, 177:37-45. 10.1016/j.mad.2018.03.013
15. Hunt SC, Kark JD, Aviv A: Association between shortened leukocyte telomere length and cardio-metabolic
outcomes. Circ Cardiovasc Genet. 2015, 8:4-7. 10.1161/CIRCGENETICS.114.000964
16. D'Mello MJ, Ross SA, Briel M, Anand SS, Gerstein H, Pare G: Association between shortened leukocyte
telomere length and cardiometabolic outcomes. Systematic review and meta-analysis. Circ Cardiovasc
Genet. 2015, 8:82-90. 10.1161/CIRCGENETICS.113.000485
17. Haycock PC, Heydon EE, Kaptoge S, Butterworth AS, Thompson A, Willeit P: Leucocyte telomere length and
risk of cardiovascular disease: systematic review and meta-analysis. BMJ. 2014, 349:g4227.
10.1136/bmj.g4227
18. Williams GC: Pleiotropy, natural selection, and the evolution of senescence . Evolution. 1957, 11:398-411.
10.2307/2406060
19. El-Telbany A, Ma PC: Cancer genes in lung cancer: racial disparities: are there any? . Genes Cancer. 2012,
3:467-480. 10.1177/1947601912465177
20. Olson SH, Kurtz RC: Epidemiology of pancreatic cancer and the role of family history . J Surg Oncol. 2013,
107:1-7. 10.1002/jso.23149
21. Dietze EC, Sistrunk C, Miranda-Carboni G, O'Regan R, Seewaldt VL: Triple-negative breast cancer in
African-American women: disparities versus biology. Nat Rev Cancer. 2015, 15:248-254. 10.1038/nrc3896
22. Nault JC, Ningarhari M, Rebouissou S, Rossi JZ: The role of telomeres and in cirrhosis and liver cancer . Nat
Rev Gastroenterol Hepatol. 2019, 16:544-558. 10.1038/s41575-019-0165-3
23. Stone RC, Horvath K, Kark JD, Susser E, Tishkoff SA, Aviv A: Telomere length and the cancer-atherosclerosis
trade-Off. PLoS Genet. 2016, 12:e1006144. 10.1371/journal.pgen.1006144
24. Kim JH, Jenrow KA, Brown SL: Mechanisms of radiation-induced normal tissue toxicity and implications for
future clinical trials. Radiat Oncol J. 2014, 32:103-115. 10.3857/roj.2014.32.3.103
25. Diker-Cohen T, Uziel O, Szyper-Kravitz M, Shapira H, Natur A, Lahav M: The effect of chemotherapy on
telomere dynamics: clinical results and possible mechanisms. Leuk Lymphoma. 2013, 54:2023-2029.
10.3109/10428194.2012.757765
26. Sanoff HK, Deal AM, Krishnamurthy J, et al.: Effect of cytotoxic chemotherapy on markers of molecular age
in patients with breast cancer . J Natl Cancer Inst. 2014, 106:dju057. 10.1093/jnci/dju057
27. Hurria A, Jones L, Hyman BM, Muss MD: Cancer treatment as an accelerated aging process: assessment,
biomarkers, and interventions. Am Soc Clin Oncol Educ Book. 2018, 36:e516-e522. 10.1200/EDBK_156160
28. Musich PR, Zou Y: DNA-damage accumulation and replicative arrest in Hutchinson-Gilford progeria
syndrome. Biochem Soc Trans. 2011, 39:1764-1769. 10.1042/BST20110687
29. Muhlinen VN, Horikawa I, Alam F, et al.: p53 isoforms regulate premature aging in human cells . Oncogene.
2018, 37:2379-2393. 10.1038/s41388-017-0101-3
30. Fujita K: p53 isoforms in cellular senescence- and aging-associated biological and physiological functions .
Int J Mol Sci. 2019, 20:6023. 10.3390/ijms20236023
31. Fernandez P, Scaffidi P, Markert E, Lee JH, Rane S, Misteli T: Transformation resistance in a premature aging
disorder identifies a tumor-protective function of BRD4. Cell Rep. 2014, 9:248-260.
10.1016/j.celrep.2014.08.069
32. Coppe JP, Patil CK, Rodier F, Et al.: A human-like senescence-associated secretory phenotype is conserved
in mouse cells dependent on physiological oxygen. PLoS One. 2010, 5:e9188. 10.1371/journal.pone.0009188
33. Kang T.W, Yevsa T, Woller N, et al.: Senescence surveillance of pre-malignant hepatocytes limits liver
cancer development. Nature. 2011, 479:547-551. 10.1038/nature10599
34. Lujambio A, Akkari L, Simon J, et al.: Non-cell-autonomous tumor suppression by p53 . Cell. 2013, 153:449-
460. 10.1016/j.cell.2013.03.020
35. Acosta JC, Banito A, Wuestefeld T, et al.: A complex secretory program orchestrated by the inflammasome
controls paracrine senescence . Nat Cell Biol. 2013, 15:978-990. 10.1038/ncb2784
36. Mendrysa SM, O'Leary KA, McElwee MK, Michalowski J, Eisenman RN, Powell DA, Perry ME: Tumor
suppression and normal aging in mice with constitutively high p53 activity . Genes Dev. 2006, 20:16-21.
10.1101/gad.1378506
37. Bourdon JC, Khoury MP, Diot A, et al.: p53 mutant breast cancer patients expressing p53γ have as good a
prognosis as wild-type p53 breast cancer patients. Breast Cancer Res. 2011, 13:7. 10.1186/bcr2811
38. Nutthasirikul N, Limpaiboon T, Leelayuwat C, Patrakitkomjorn S, Jearanaikoon P: Ratio disruption of the
133p53 and TAp53 isoform equilibrium correlates with poor clinical outcome in intrahepatic
cholangiocarcinoma. Int J Oncol. 2013, 42:1181-1188. 10.3892/ijo.2013.1818
39. Takahashi R, Markovic SN, Scrable HJ: Dominant effects of Δ40p53 on p53 function and melanoma cell fate .
J Invest Dermatol. 2014, 134:791-800. 10.1038/jid.2013.391
40. Hasty P, Christy BA: p53 as an intervention target for cancer and aging . Pathobiol Aging Age Relat Dis.
2013, 3:e22702. 10.3402/pba.v3i0.22702
41. Vanaclocha FV: Inflammation in the molecular pathogenesis of cancer and atherosclerosis . Reumatol Clin.
2009, 5:40-43. 10.1016/j.reuma.2008.12.008
42. Zhivotovsky B, Orrenius S: Cell cycle and cell death in disease: past present and future . J Intern Med. 2010,
268:395-409. 10.1111/j.1365-2796.2010.02282.x
43. Chang BD, Watanabe K, Broude EV, Fang J, Poole JC, Kalinichenko TV, Roninson IB: Effects of
p21Waf1/Cip1/Sdi1on cellular gene expression: implications for carcinogenesis, senescence, and age-related
diseases. Proc Natl Acad Sci USA. 2000, 97:4291-4296. 10.1073/pnas.97.8.4291
44. Folkman J: Tumor angiogenesis therapeutic implications’. Engl J Med. 1971, 285:1182-1186.
10.1056/NEJM197111182852108
45. Olivieri F, Rippo MR, Procopio AD, et al.: Circulating inflamma-miRs in aging and age-related diseases .
2020 Dhalla et al. Cureus 12(10): e10794. DOI 10.7759/cureus.10794 10 of 11
Front Genet. 2013, 4:121. 10.3389/fgene.2013.00121
46. Green DR, Llambi F: Cell death signaling cold spring . Harb Perspect Biol. 2015, 7:1-24.
47. Virginia J, Vieyra T, Coello BD, Oliva JM: Atherosclerosis and cancer; a resemblance with far-reaching
implications. Arch Med Res. 2017, 48:12-26. 10.1016/j.arcmed.2017.03.005
48. Staubach S, Hanisch FG: Lipid rafts: signaling and sorting platforms of cells and their roles in cancer . Expert
Rev Proteomics. 2011, 8:263-277. 10.1586/epr.11.2
49. Kato S, Smalley S, Sadarangani A, et al.: Lipophilic but not hydrophilic statins selectively induce cell death
in gynecological cancers expressing high levels of HMGCoA reductase. J Cell Mol Med. 2010, 14:1180-1193.
10.1111/j.1582-4934.2009.00771.x
2020 Dhalla et al. Cureus 12(10): e10794. DOI 10.7759/cureus.10794 11 of 11
Article
Background Hidradenitis suppurativa (HS) is a chronic debilitating disease with a significant burden of both organic and psychological comorbidities. It has been shown that certain telomere‐related genes (TRGs) affect a wide range of diseases, including HS and its associated comorbidities, but their exact role in HS pathogenesis is still unknown. Objectives To determine whether TRG methylomes can be used as biomarkers in HS. Methods Using the Illumina HumanMethylation450 BeadChip array, we examined methylation variations associated with TRGs in HS cases and age‐, sex‐, and ethnicity‐matched healthy controls . The study utilized integrated bioinformatics statistical methods, such as a false discovery rate (FDR), the area under the receiver operating characteristic curve (AUC), and principal component analysis. Results There were a total of 585 different differentially methylated CpG sites identified in 585 TRGs associated with HS (474 hypomethylated and 111 hypermethylated) (FDR p‐value <0.05). A number of these CpGs have been identified as being involved in increased pain sensitivity including EPAS1, AHR, CSNK1D, DNMT1, IKBKAP, NOS3 , PLCB1, and PRDM16 genes; GABRB3 as a potential alcohol addiction marker; DDB1, NSMCE2, and HNRNPA2B1 associated with cancers. Pathway analysis identified 67 statistically significant pathways, including DNA repair, telomere maintenance, mismatch repair, and cell cycle control. (p<0.001). Conclusion The disruption of TRGs leads to the shortening of telomeres, which is associated with HS progression, aging, cellular senescence, and an increased risk of various diseases, including cancer and associated comorbidities, such as metabolic syndrome, cardiovascular disease, and inflammatory disorders. Further research is necessary to better understand the underlying mechanisms and establish causal links between TRGs and HS. The present study is the first effort to comprehend potential pathomechanisms of sporadic HS cases concentrating on PBMC methylome since ours.
Article
Full-text available
Cellular senescence, a term originally used to define the characteristics of normal human fibroblasts that reached their replicative limit, is an important factor for ageing, age-related diseases including cancer, and cell reprogramming. These outcomes are mediated by senescence-associated changes in gene expressions, which sometimes lead to the secretion of pro-inflammatory factors, or senescence-associated secretory phenotype (SASP) that contribute to paradoxical pro-tumorigenic effects. p53 functions as a transcription factor in cell-autonomous responses such as cell-cycle control, DNA repair, apoptosis, and cellular senescence, and also non-cell-autonomous responses to DNA damage by mediating the SASP function of immune system activation. The human TP53 gene encodes twelve protein isoforms, which provides an explanation for the pleiotropic p53 function on cellular senescence. Recent reports suggest that some short isoforms of p53 may modulate gene expressions in a full-length p53-dependent and -independent manner, in other words, some p53 isoforms cooperate with full-length p53, whereas others operate independently. This review summarizes our current knowledge about the biological activities and functions of p53 isoforms, especially Δ40p53, Δ133p53α, and p53β, on cellular senescence, ageing, age-related disorder, reprogramming, and cancer. Numerous cellular and animal model studies indicate that an unbalance in p53 isoform expression in specific cell types causes age-related disorders such as cancer, premature ageing, and degenerative diseases.
Article
Full-text available
Cellular senescence is a hallmark of normal aging and aging-related syndromes, including the premature aging disorder Hutchinson-Gilford Progeria Syndrome (HGPS), a rare genetic disorder caused by a single mutation in the LMNA gene that results in the constitutive expression of a truncated splicing mutant of lamin A known as progerin. Progerin accumulation leads to increased cellular stresses including unrepaired DNA damage, activation of the p53 signaling pathway and accelerated senescence. We previously established that the p53 isoforms ∆133p53 and p53β regulate senescence in normal human cells. However, their role in premature aging is unknown. Here we report that p53 isoforms are expressed in primary fibroblasts derived from HGPS patients, are associated with their accelerated senescence and that their manipulation can restore the replication capacity of HGPS fibroblasts. We found that in near-senescent HGPS fibroblasts, which exhibit low levels of ∆133p53 and high levels of p53β, restoration of Δ133p53 expression was sufficient to extend replicative lifespan and delay senescence, despite progerin levels and abnormal nuclear morphology remaining unchanged. Conversely, Δ133p53 depletion or p53β overexpression accelerated the onset of senescence in otherwise proliferative HGPS fibroblasts. Our data indicate that Δ133p53 exerts its role by modulating full-length p53 (FLp53) signaling to extend the replicative lifespan and promotes the repair of spontaneous progerin-induced DNA double-strand breaks (DSBs). We showed that Δ133p53 dominant-negative inhibition of FLp53 occurs directly at the p21/CDKN1A and miR-34a promoters, two p53 senescence-associated genes. In addition, Δ133p53 expression increased the expression of DNA repair RAD51, likely through upregulation of E2F1, a transcription factor that activates RAD51, to promote repair of DSBs. In summary, our data indicate that Δ133p53 modulates p53 signaling to repress progerin-induced early onset of senescence in HGPS cells. Therefore, restoration of ∆133p53 expression may be a novel therapeutic strategy to treat aging-associated phenotypes of HGPS in vivo.
Article
Full-text available
Figure 1. Factors involved in the early stages and development of atherosclerosis and cancer. Among the important similarities found in both multifactorial diseases, the identification of changes in the DNA sequence in close correlation to epigenetic modifications, can be transmitted across generations in relationship with key environmental factors. These external factors can by themselves start or potentiate anomalous processes evidenced in the long-term as oxidative stress, inflammation, aberrant apoptosis, uncontrolled cell proliferation and angiogenesis.
Article
Telomerase is a key enzyme for cell survival that prevents telomere shortening and the subsequent cellular senescence that is observed after many rounds of cell division. In contrast, inactivation of telomerase is observed in most cells of the adult liver. Absence of telomerase activity and shortening of telomeres has been implicated in hepatocyte senescence and the development of cirrhosis, a chronic liver disease that can lead to hepatocellular carcinoma (HCC) development. During hepatocarcinogenesis, telomerase reactivation is required to enable the uncontrolled cell proliferation that leads to malignant transformation and HCC development. Part of the telomerase complex, telomerase reverse transcriptase, is encoded by TERT, and several mechanisms of telomerase reactivation have been described in HCC that include somatic TERT promoter mutations, TERT amplification, TERT translocation and viral insertion into the TERT gene. An understanding of the role of telomeres and telomerase in HCC development is important to develop future targeted therapies and improve survival of this disease. In this Review, the roles of telomeres and telomerase in liver carcinogenesis are discussed, in addition to their potential translation to clinical practice as biomarkers and therapeutic targets.
Article
Advances in early detection, treatment, and supportive care have resulted in an estimated 16 million cancer survivors who are alive in the United States today. Outcomes have notably improved for children with cancer as well as young adults with hematologic malignancies due, in part, to the intensification of cancer treatment, including the use of hematopoietic cell transplantation. Emerging evidence suggests that these cancer survivors are at risk for premature aging, manifesting as early onset of chronic health conditions and a higher risk of mortality compared with the general population. Although the pathophysiology of premature aging in these survivors has not been fully elucidated, emerging concepts in aging research could help shed light on this phenomenon. Longitudinal studies are needed to better characterize aging in these survivors, setting the stage for much-needed interventions to halt the trajectory of accelerated aging. These efforts will be enhanced through collaborations between translational researchers, clinical oncologists, primary care providers, geriatricians, patient caretakers, and other stakeholders committed to improving the lives of the growing population of survivors. © The Author(s) 2019. Published by Oxford University Press. All rights reserved. For permissions, please email: [email protected]
Article
Cellular senescence is a permanent state of cell cycle arrest that occurs in proliferating cells subjected to different stresses. Senescence is, therefore, a cellular defense mechanism that prevents the cells to acquire an unnecessary damage. The senescent state is accompanied by a failure to re-enter the cell cycle in response to mitogenic stimuli, an enhanced secretory phenotype and resistance to cell death. Senescence takes place in several tissues during different physiological and pathological processes such as tissue remodeling, injury, cancer, and aging. Although senescence is one of the causative processes of aging and it is responsible of aging-related disorders, senescent cells can also play a positive role. In embryogenesis and tissue remodeling, senescent cells are required for the proper development of the embryo and tissue repair. In cancer, senescence works as a potent barrier to prevent tumorigenesis. Therefore, the identification and characterization of key features of senescence, the induction of senescence in cancer cells, or the elimination of senescent cells by pharmacological interventions in aging tissues is gaining consideration in several fields of research. Here, we describe the known key features of senescence, the cell-autonomous, and noncell-autonomous regulators of senescence, and we attempt to discuss the functional role of this fundamental process in different contexts in light of the development of novel therapeutic targets.
Article
An accumulating body of evidence supports the hypothesis that cancer and/or cancer treatment is associated with accelerated aging. The majority of these data come from the pediatric literature; however, a smaller yet growing body of literature points toward similar findings in the geriatric population. This is a key survivorship issue the growing number of older adults with cancer face, along with the short- and long-term impact of cancer therapy on the aging process. This article will review clinical and biologic markers of aging in older adults with cancer, use cardiovascular disease as a model of accelerated aging, and discuss potential interventions to decrease the risk.
Article
Telomeres are dynamic nucleoprotein-DNA structures that cap and protect linear chromosome ends. Because telomeres shorten progressively with each replication, they impose a functional limit on the number of times a cell can divide. Critically short telomeres trigger cellular senescence in normal cells, or genomic instability in pre-malignant cells, which contribute to numerous degenerative and aging-related diseases including cancer. Therefore, a detailed understanding of the mechanisms of telomere loss and preservation is important for human health. Numerous studies have shown that oxidative stress is associated with accelerated telomere shortening and dysfunction. Oxidative stress caused by inflammation, intrinsic cell factors or environmental exposures, contributes to the pathogenesis of many degenerative diseases and cancer. Here we review the studies demonstrating associations between oxidative stress and accelerated telomere attrition in human tissue, mice and cell culture, and discuss possible mechanisms and cellular pathways that protect telomeres from oxidative damage.
Article
p53 is a transcriptional regulator that responds to cellular stresses to suppress oncogenesis, but some of these responses can have unintended consequences that influence non-cancer-related aging processes. The impact of these consequences is not well understood-partly due to the many complex processes that influence p53 function and partly due to the vast array of processes that p53 affects. p53 has the potential to both accelerate and hinder cellular aging processes, which would likely have antithetical biological outcomes with regard to organismal aging. To accelerate aging, p53 induces apoptosis or cell cycle arrest as a prerequisite to cellular senescence; both can impair the mobilization of stem and progenitor cell populations. To suppress aging, p53 inhibits unregulated proliferation pathways that could lead to cellular senescence and a senescence-associated secretory phenotype (SASP), which creates a pro-inflammatory and degenerative tissue milieu. A review of mouse models supports both possibilities, highlighting the complexity of the p53 influence over organismal aging. A deeper knowledge of how p53 integrates and is integrated with various biological processes will improve our understanding of its influence over the aging process.