Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 9

“The role of telomeres in aging or anti-aging

programs”

Introduction
Good Morning Dr. LOO and Dr. WU
Today I am going to present the topic about “The role of telomeres in aging or anti-
aging programs.”

Here is the content of today’s presentation.

Telomeres, which are protective caps on our chromosomes, play an important role in
this aging process. Our cells keep dividing and making copies of themselves. However,
a bit of the telomere, gets lost each time this happens. This occurs due to the incomplete
replication of chromosome ends during the DNA copying process.

Consequently, telomeres undergo gradual shortening over time. When telomeres reach a
critical length, cells may either cease dividing or undergo programmed cell death,
known as replicative senescence. Telomere shortening is closely associated with
different aspects of the aging process and is recognized as one of the hallmarks of aging,

This review aims to understand how telomere problems are linked to aging-related
traits, and examine the important roles of telomeres and telomerase in the aging
processand common age-related diseases. Additionally, it will explore the role of
telomeres in aging and discuss the emerging field of anti-aging programs.

Telomere
The concept of telomeres, introduced by Hermann Muller in the early 1930s as
"terminal genes," In 1961, Hayflick and Moorhard demonstrated that human fetal cell
have finite replicative potential, which is 50-60 time and this is the so-called “Hayflick
Limits”
In 1971, Blackburn and Gall's pioneering work on (T. thermophila) uncovered unique,
conserved repetitive DNA sequences at the chromosome ends. These repetitive
sequences were distinct from the genes present in the body of the chromosome.
Building on this initial observation, Blackburn, along with her graduate student Carol
Greider, conducted further investigations in the early 1980s.

Using DNA sequencing techniques, they successfully determined the exact sequence of
repetitive DNA found at the ends of T. thermophila chromosomes, which is known as
Telomere. For their discovery of telomeres’ protection and the enzyme telomerase,
Blackburn, Greider and Szostak were awarded with Nobel Prize in Physiology or
Medicine in 2009

What is Telomere?
The word ‘Telomere’ is derived from the Greek and means “End-part”. It is a region of
repetitive nucleotide sequences at each end of a chromosome. The tandem repeat
sequences are varies from species to species and are “TTAGGG” in vertebrates.
Telomeres are important in preventing chromosome end fusion, maintaining the
stability of the genome and ensuring the accurate replication of DNA during cell
division.

Telomeres have unique features. One of these features is the formation of a T-loop,
where the 3' overhang of telomeric DNA folds back on itself. Additionally, a D-loop is
formed when one strand is displaced. In addition, telomeres are protected by a group of
at least six protein subunits collectively known as Shelterin. Shelterin plays a crucial
role in safeguarding the DNA at the telomeres. It prevents DNA damage signaling from
the ends of telomeres and inhibits DNA repair mechanisms that could lead to fusion of
the chromosome ends. Shelterin also regulates the access and activity of telomerase, the
enzyme responsible for maintaining telomere length.

Telomerase
Telomerase is a ribonucleoprotein that acts on the 3’ overhang of telomeres, it
encompasses a highly conserved reverse transcriptase (TERT) and a template RNA
component (TERC). Using the 3’overhang of telomeres as a primer to align with TERC
sequences, TERT adds telomeric repeats to chromosome ends, thus lengthening the
telomeres. The major function of telomerase is to compensate for the "end replication
problem" of conventional DNA polymerase to replicate chromosome. During DNA
replication, the conventional DNA polymerases are unable to fully replicate the ends of
chromosomes, resulting in the progressive shortening of telomeres with each cell
division. This shortening is a natural consequence of the "end replication problem".

By extending the telomeres, telomerase helps to prevent the loss of genetic information
and allows cells to continue dividing. Telomerase activity is tightly regulated, with most
somatic cells in the body having low or no telomerase expression, which contributes to
the limited replicative lifespan of these cells. On the other hand, telomerase was found
to be highly active in germ cells, stem cells, and most cancer cells. The reactivation of
telomerase in cancer cells allows them to overcome the Hayflick limit and continue
proliferating indefinitely, leading to the immortality of cancer.

Telomere and Aging


As I mentioned before, during each round of DNA replication, telomeres undergo
gradual shortening due to the end replication problem, where the DNA polymerase
cannot fully replicate the very ends of the linear chromosomes. Telomeres, which are
limited in nature, act as a barrier to cellular immortality. However, when telomere
function is compromised, it coincides with a decline in fitness associated with aging. In
fact, telomere attrition is considered one of the hallmarks of aging. These hallmarks of
aging include various factors such as genomic instability, epigenetic alterations, loss of
proteostasis, disrupted nutrient sensing, mitochondrial dysfunction, cellular senescence,
depletion of stem cells, and altered intercellular communication. Alongside telomere
attrition, these factors collectively contribute to the aging process.

To study the roles of telomeres and telomerase in the aging process, TERC and TERT
knockout mouse models has been used extensively. The TERC and TERT knockout
mouse models have confirmed the crucial role of telomeres in the aging process.
Through these models, it has been demonstrated that telomere dysfunction accelerates
various signs of aging, including shortened lifespan, aged appearance, reduced tissue
stem cells, and etc.

Mitochondrial dysfunction
On of the mechanism leading to aging is Mitochondrial dysfunction. It revealed that the
telomere dysfunction in late-generation RNA Component knockout actives the p53-
PGC pathway of aging. The p53 protein is widely recognized as a tumor suppressor
that has a critical function in controlling the cell cycle, DNA repair, and apoptosis in
response to stress.

It has been found that telomere dysfunction activate the p53 pathway. When telomeres
become critically short or dysfunctional, it triggers a DNA damage response, leading to
the activation of p53 and suppression of peroxisome proliferator-activated receptor
gamma coactivator 1-alpha and beta (PGC-1α and PGC-1β), leading to a decrease in
mitochondrial biogenesis and function, as well as metabolic alterations. These changes
resemble the effects observed when PGC-1α and PGC-1β are reduced or eliminated.
The repression of the PGC network, along with the resulting mitochondrial dysfunction
and metabolic changes, combined with telomere-induced apoptotic and proliferative
checkpoints, collectively contribute to the decline in multiple systems' functionality in
the presence of telomere dysfunction.

Stem cell exhaustion


Another mechanism leading to aging by Telomeres dysfunction is stem cell exhaustion.
The shortage of telomeres eventually triggers replicative senescence, leading to
irreversible growth arrest and preventing cells from further dividing and replenishing
tissues. Consequently, senescent cells accumulate in aging tissues, contributing to the
impairment of replicative potential in tissue stem cells and immune cells, which is
known as immunosenescence.

During the process of forming blood cells, aged mice have demonstrated a decline in the
cell-cycle activity of hematopoietic stem cells. This leads to a decline in hematopoiesis,
resulting in a decreased production of immune cells. Consequently, the immune system
becomes less efficient and making individuals more susceptible to infections and
diseases. Additionally, the decrease in hematopoietic stem cells with age is associated
with an increased incidence of anemia and certain types of leukemia.

Stem cell exhaustion and telomere attrition also impair tissue regeneration in various
organs, including the skin, muscles, and nervous system and leads to functional decline
and age-related diseases in these organs.

Inflammation
• Additionally, Senescent cells in aging tissues acquire a senescence-associated
secretory phenotype (SASP) and release pro-inflammatory substances, leading to
chronic inflammation.
• Chronic inflammation, known as inflammaging, creates an unfavorable
microenvironment for stem cells and impairs their regenerative potential.
• It contributes to tissue damage, impairs tissue repair and regeneration, and plays a
role in the onset and progression of age-related diseases.
• Cardiovascular diseases and hypertension are closely linked to chronic
inflammation, promoting plaque formation and increasing the risk of heart attacks
and strokes.

Moreover, neurodegenerative disorders, such as Alzheimer's disease and Parkinson's


disease, have also been associated with chronic inflammation and leading to progressive
neuronal damage and cognitive decline.

Moreover, chronic inflammation has implications in the onset of metabolic syndrome


such as obesity, hypertension etc. It can disrupt insulin signaling, leading to insulin
resistance and type 2 diabetes.

Loss of proteostasis
Loss of proteostasis, another mechanism of aging, it means loss of maintenance of
proper protein folding and functionality within cells, is linked to telomere dysfunction
and the repression of SIRT1 expression, mediated by the p53 pathway.

Proteostasis refers to the maintenance of proper protein folding and functionality within
cells. The impairment of chaperone proteins, which play a crucial role in aiding proper
protein folding, can result in the disruption of proteostasis.
Proper protein folding is crucial for neuronal homeostasis, as neurons cannot dilute
misfolded proteins through cell division.
Neurological diseases associated with aging, such as Alzheimer's disease, involve the
accumulation of misfolded proteins and result in neuronal loss and cognitive decline.

Telomerase and cancer


Extensive research has been conducted on telomerase and its involvement in cancer.
Telomeres, which are protective structures found at the ends of chromosomes, play a
crucial role in preventing the activation of a DNA damage response (DDR). When
telomeres become critically short, cells typically enter a state of replicative senescence
or undergo programmed cell death as a mechanism to halt the proliferation of damaged
cells. Nevertheless, specific cancer cells possess the capability to surpass this constraint
through the activation of telomerase. This reactivation of telomerase contributes to the
immortalization of cancer cells and facilitates their unrestrained proliferation.

Cancer cells can activate telomerase through several mechanisms, such as oncogenes
acting as transcriptional regulators of telomerase, alternative splicing, and epigenetic
modifications and upregulation of TERT expression

Targeting telomerase has emerged as a promising strategy for cancer therapy. Several
telomerase inhibitors and telomerase-targeted immunotherapies are being investigated
in preclinical and clinical studies as potential anti-cancer treatments. It is worth noting
that although telomerase activation is a prevalent mechanism for maintaining telomeres
in cancer cells, not all cancer cells depend on telomerase. Some cancer cells can employ
an alternative pathway called the alternative lengthening of telomeres (ALT) to elongate
their telomeres.

Telomere and anti-aging interventions


In fact, the potential of telomerase as an anti-aging therapy has been demonstrated in
various studies. An important study utilized a mouse model lacking telomerase have
demonstrated the capability of telomerase activation to reverse age-related phenotypes.
The researchers utilized an inducible system to reactivate telomerase in adult mice with
critically short telomeres. The outcomes demonstrated significant reductions in DNA
damage signaling and the reversal of degenerative characteristics in variousorgans.
These findings strongly support the potential of telomerase activation as a promising
intervention against aging.

Besides, there is evidence indicating that oxidative stress can accelerate telomere
shortening and contribute to the development of metabolic syndrome, such as obesity,
high blood pressure. Oxidative stress can accelerate telomere shortening by causing
damage to telomeric DNA. The relationship between telomere length, oxidative stress,
and metabolic syndrome appears to be bidirectional. On one hand, oxidative stress can
accelerate telomere shortening, potentially contributing to the development or
progression of metabolic syndrome. On the other hand, metabolic syndrome
components can increase oxidative stress levels, leading to further telomere attrition.
Implementing strategies to decrease oxidative stress, such as adopting a healthy
lifestyle, effectively addressing inflammation, and enhancing antioxidant defenses, have
the potential to alleviate telomere shortening and potentially decelerate the aging
process.

Telomerase therapy and future perspectives


Several compounds have been discovered as potential activators of telomerase. One of
which being extensively studied is TA-65, it is a natural compound derived from an
extract of a plant commonly used in traditional Chinese medicine, Astragalus
membranaceous. Studies have shown that the compound TA-65 has the ability to
increase the levels of mouse telomerase reverse transcriptase (mTERT) in specific
tissues and lengthen telomeres that have become critically short. These findings suggest
that TA-65 has the potential to enhance telomerase activity and elongate telomeres in
mice. It is noteworthy that the use of TA-65 did not significantly raise the risk of cancer
in these experimental mice.
Furthermore, the effectiveness of hormonal agents, such as danazol. In a study
involving patients with telomere diseases, it was found that treatment with danazol not
only prevented telomere loss but also led to a significant increase in telomeric repeats

In addition to small molecules and hormonal agents, recent advancements have been
made in targeting TERC stability, providing new therapeutic options for
telomeropathies. Specifically, the enzyme PAPD5 has been found to play a role in the
degradation of TERC, the RNA component of telomerase. In recent studies, researchers
have explored the potential of a small-molecule inhibitor of PAPD5. By inhibiting
PAPD5, the inhibitor shows promise in increasing telomere length, suggesting its
potential as a therapeutic approach for telomere-related disorders.

These findings provide new insights into compounds and therapeutic approaches for
modulating telomerase activity and telomere length, paving the way for novel
treatments for telomeropathies and age-related diseases.

Conclusion
In conclusion, telomeres and telomerase have been found to play crucial roles in cellular
senescence and the aging process through various mechanisms

Numerous studies have demonstrated the close relationship between telomere


shortening and age-related decline, dysfunction, and reduced lifespan. Additionally,
telomere shortening has been linked to the development of cancer.

The importance of telomeres tells us that more research is needed on aging and diseases
related to getting older. More studies on telomeres and telomerase has the potential to
improve our understanding of aging and develop strategies to combat it.

You might also like