Unlock 15% Off Your First Order

Your Cart

Congrats! You're saving

Your cart is empty. Explore our shop to add products and get started.

The future of healthy aging

Science-backed tools designed to improve your healthspan and longevity.

Shop Now
Diagram illustrating the 12 interconnected hallmarks of aging arranged in a circular layout around a central label.

The 12 Hallmarks of Aging: A Scientific Guide to How—and Why—We Age

Published:

Why do we age? For decades, aging researchers have explored this question using animal and cellular models, uncovering the complex cellular and molecular hallmarks that drive both normal aging and accelerated aging syndromes. Today, scientists have aligned on at least 12 hallmarks of aging—interconnected biological processes that contribute to age-related decline, chronic inflammation, reduced mitochondrial function, stem cell exhaustion, and the onset of age-related diseases like cardiovascular disorders and Alzheimer’s disease. 


Understanding these hallmarks doesn’t necessarily clarify how aging works. It does, however, reveal where interventions—nutrition, lifestyle, targeted supplements, and emerging longevity tools—can help support better healthy aging and potentially delay normal aging.


This is exactly what the Tally Health membership is designed to do: help you understand what’s driving how you’re aging, then support measurable improvement with science-backed, personalized guidance.


Let’s break down the 12 hallmarks of aging, how they interact, and why they matter for long-term health.

1. Genomic Instability

Slide titled “Genomic Instability” featuring a cracked glass visual symbolizing DNA damage and genetic breakdown over time.
Image from Tally Health

Throughout life, our DNA experiences constant damage—from radiation, pollutants, metabolic byproducts, and normal cellular processes. When genome stability declines, DNA repair mechanisms can’t keep up. This accumulation of damage influences aging by:


  • Affecting essential genes

  • Compromising cellular function

  • Increasing susceptibility to age-associated diseases

Genomic instability is a primary driver of both normal human aging and numerous premature aging diseases.

2. Telomere Attrition (Telomere Shortening)

Slide titled “Genomic Instability” featuring a cracked glass visual symbolizing DNA damage and genetic breakdown over time.
Image from Tally Health

Telomeres are protective caps at the ends of chromosomes. Over time, with repeated cell division, they shorten—a process known as telomere attrition.


Shortened telomeres:


  • Increase cell senescence

  • Reduce regenerative capacity

  • Contribute to accelerated aging

In telomerase-deficient mice, lengthened telomeres exhibit decreased markers of aging, highlighting their importance in the aging process.

3. Epigenetic Alterations

Slide titled “Epigenetic Alterations” showing a stylized DNA strand with markers indicating methylated and unmethylated regions.
Image from Tally Health

Epigenetics controls how genes are switched on or off. With age, DNA methylation patterns, histone modifications, and chromatin remodeling factors shift, leading to transcriptional alterations affecting metabolic pathways, cell growth, and repair.


Because epigenetic shifts strongly correlate with chronological age, this hallmark is a major focus of modern aging research.

4. Loss of Proteostasis (Impaired Protein Homeostasis)

Slide titled “Loss of Proteostasis” featuring an older adult holding their head, representing protein misfolding and cellular stress.
Image from Tally Health

Healthy cells rely on tightly regulated protein folding, recycling, and clearance. Over time:


  • Proteins misfold

  • Cleanup systems weaken

  • Toxic aggregates accumulate

These changes are linked to neurodegenerative diseases such as Alzheimer’s and Parkinson’s.

5. Deregulated Nutrient Sensing

Slide titled “Deregulated Nutrient Sensing” with fruits and vegetables symbolizing metabolism and nutrient signaling pathways.
Image from Tally Health

Cells rely on metabolic and signaling pathways (like mTOR, AMPK, and insulin signaling) to sense nutrients. With age, these pathways become dysregulated.


Examples include:

  • Anabolic signaling accelerates aging via mTOR overactivation

  • Dietary restriction may delay normal aging by restoring balance

  • Changes in insulin/IGF-1 signaling influence longevity in both young and old tissues

This hallmark reveals how fasting, nutrient timing, and metabolic health influence aging biology.

6. Mitochondrial Dysfunction

Slide titled “Mitochondrial Dysfunction” showing a close-up illustration of mitochondria associated with reduced cellular energy.
Image from Tally Health

Mitochondria are the cell’s powerhouses—and their decline is a defining feature of organismal aging.


Aging mitochondria produce fewer beneficial mitochondrial proteins and more reactive oxygen species (ROS), contributing to:


  • Lower energy production

  • Higher oxidative stress

  • Increased risk of age-related disease

Mitochondrial DNA mutations and nutrient-sensing changes further accelerate dysfunction.

7. Cellular Senescence

Slide titled “Cellular Senescence” featuring microscopic imagery of aging cells that have stopped dividing.
Image from Tally Health

Cells don’t divide indefinitely. When they accumulate enough damage, they enter cellular senescence—a state of permanent cell cycle arrest.


Over time, senescent cells build up in tissues and secrete inflammatory molecules known as the senescence-associated secretory phenotype (SASP), which drives:


  • Tissue degeneration

  • Chronic inflammation

  • Premature aging phenotypes

Although senescent cells play important and beneficial roles in the body, reducing the burden of senescent cells is an emerging longevity strategy.

8. Stem Cell Exhaustion

Slide titled “Stem Cell Exhaustion” showing an older adult’s face in profile, representing reduced regenerative capacity with age.
Image from Tally Health

Stem cells replenish tissues—from your skin to your gut to your immune system. But with age:


  • Their regenerative capacity declines

  • DNA repair weakens

  • Chronic inflammation disrupts their environment

  • Senescent cells interfere with regulating stem cell behavior

Stem cell exhaustion reduces the body’s ability to heal, repair, and maintain organ function.

9. Altered Intercellular Communication

Slide titled “Altered Intercellular Communication” with neuron-like cells illustrating disrupted signaling between cells.
Image from Tally Health

Cells talk to each other through chemical signals. With age, this communication shifts due to:


  • Increased inflammatory signaling

  • Diminished immune surveillance

  • Hormonal changes

  • Senescent cell activity

This contributes to systemic issues like chronic inflammation and impaired tissue function.

10. Dysbiosis of the Gut Microbiome

Slide titled “Dysbiosis of the Gut Microbiome” showing magnified bacteria-like shapes representing imbalances in gut microbes.
Image from Tally Health

Only recently added as a critical hallmark, gut microbiome imbalance can:


  • Promote chronic inflammation

  • Influence immune function

  • Affect nutrient absorption

  • Accelerate aging

Microbial shifts are seen in both normal physiological aging and premature aging phenotypes.

11. Compromised Autophagy

Slide titled “Compromised Autophagy” featuring a person holding a recycling bin, symbolizing reduced cellular cleanup and recycling processes.
Image from Tally Health

Autophagy is the cell’s recycling and cleanup process. With age, autophagy slows, allowing:


  • Damaged proteins

  • Dysfunctional mitochondria

  • Toxic metabolites

…to accumulate—contributing to accelerated aging and metabolic dysfunction.

12. Chronic Inflammation (“Inflammaging”)

Slide titled “Chronic Inflammation” showing a simplified human silhouette highlighted against a warm background, representing widespread inflammation in the body.
Image from Tally Health

Low-grade, persistent inflammation is a primary risk factor for many age-related diseases. This inflammation results from:


  • Senescent cells

  • Mitochondrial dysfunction

  • Altered intercellular communication

  • Immune system decline

Inflammaging disrupts tissue integrity and amplifies many other hallmarks.

Why These Hallmarks Matter

The hallmarks don’t act in isolation—they form a network. Genomic instability accelerates telomere shortening; mitochondrial dysfunction increases senescence; deregulated nutrient sensing fuels chronic inflammation. When combined, these hallmarks explain why aging is complex but also why targeted interventions can help.


And that’s where the Tally Health membership comes in. By measuring an indicator of biological age and offering evidence-based recommendations tailored to these hallmarks, Tally Health helps you take real, measurable steps toward an extended healthy lifespan.

The Bottom Line

Photo of an older couple smiling outdoors in warm light, representing healthy aging and quality of life over time.
Image from Tally Health

The 12 hallmarks of aging—spanning DNA damage, mitochondrial decline, senescent cell buildup, and nutrient-sensing changes—represent the most comprehensive scientific model for understanding the aging process. They reveal both the complexity of normal human aging and the opportunities to influence it through lifestyle, nutrition, and targeted interventions.


While aging remains inevitable, its pace is not fixed. By addressing these hallmarks proactively, it’s possible to support healthier tissues, stronger mitochondrial function, better stem cell health, and an overall more resilient aging cell.

References

  1. Carlos López-Otín et al. Hallmarks of aging: An expanding universe. Cell 2023; https://doi.org/10.1016/j.cell.2022.11.001

  2. Carlos López-Otín. The hallmarks of aging. Cell 2013; https://doi.org/10.1016/j.cell.2013.05.039

What are the 12 hallmarks of aging?

The 12 hallmarks of aging are interconnected cellular and molecular processes—such as genomic instability, telomere shortening, mitochondrial dysfunction, cellular senescence, and chronic inflammation—that drive age-related decline, increase disease risk, and influence how the body ages over time.

Why do telomeres shorten as we age?

Telomeres shorten with repeated cell division over time, a process known as telomere attrition. Shortened telomeres increase cellular senescence, reduce regenerative capacity, and contribute to accelerated aging.

What is chronic inflammation in aging (“inflammaging”)?

Inflammaging is low-grade, persistent inflammation that increases with age. It results from factors like senescent cells, mitochondrial dysfunction, altered intercellular communication, and immune system decline, and it raises the risk of many age-related diseases.

Recommended Supplements