Cellular rejuvenation and tissue repair
Cellular ageing is not a single event but a convergence of gradual molecular failures that accumulate over time. The recognised hallmarks include telomere attrition, genomic instability from DNA damage, epigenetic alterations, mitochondrial dysfunction, loss of proteostasis, and the build-up of senescent cells. These mechanisms together erode the regenerative capacity of tissues and increase susceptibility to chronic disease.
Work by Nobel laureates Tomas Lindahl, Paul Modrich, and Aziz Sancar demonstrated that DNA is under constant attack from oxidative stress and replication errors, with thousands of lesions occurring daily in each cell. Survival depends on DNA repair pathways such as base excision repair, mismatch repair, and nucleotide excision repair. Without these processes, cellular identity and function would collapse within days. This recognition reframes ageing not as an inevitable decline but as the progressive failure of repair systems that can, in principle, be restored or enhanced.
Cellular senescence represents another duality. On one hand, it prevents damaged cells from dividing and transforming into malignancies. On the other hand, senescent cells persist in a metabolically active state, secreting pro-inflammatory cytokines, chemokines, and proteases known collectively as the senescence-associated secretory phenotype (SASP). The SASP disrupts the tissue microenvironment and drives systemic inflammation, contributing to frailty, metabolic disease, and neurodegeneration. Removing or modulating senescent cells is therefore a cornerstone of rejuvenation research.
Epigenetic clocks as clinical biomarkers
The ability to measure biological age has transformed research. Chronological age is a poor predictor of disease risk, while epigenetic clocks based on DNA methylation patterns provide a quantifiable marker of cellular ageing. Steve Horvath’s pan-tissue clock, published in 2013, showed that methylation patterns across the genome can accurately estimate age in multiple tissues.
Second-generation clocks such as PhenoAge and GrimAge extend predictive power, correlating with morbidity, mortality, and functional decline. Their development has moved the concept from descriptive biology into clinical research, where methylation patterns are now used to stratify patients in trials and evaluate treatment response.
Recent advances allow cell-type-specific clocks, capable of identifying accelerated ageing within discrete cell populations. For example, glial cells in the brain may show faster epigenetic ageing in individuals with Alzheimer’s disease compared with other tissues. This shift makes the tool diagnostically actionable, guiding therapeutic targeting and providing precise endpoints for clinical studies.
Stem cell and exosome therapies in regenerative medicine
Stem cells remain central to regenerative medicine due to their capacity for self-renewal and differentiation. Three main types dominate clinical research: mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and haematopoietic stem cells (HSCs). These cells contribute to repair either by direct differentiation or by paracrine signalling that recruits native repair pathways.
Exosomes, extracellular vesicles secreted by stem cells, are increasingly recognised as therapeutic in their own right. They carry proteins, RNA, and microRNA that influence cell behaviour, reduce inflammation, and promote repair. Exosomes have the added advantage of crossing the blood–brain barrier, making them a candidate for treating neurodegenerative disease.
Clinical applications now extend to musculoskeletal conditions such as osteoarthritis and tendon injury, cardiovascular disease, and neurodegeneration. A notable advance is the discovery of Prg4+ progenitor cells within skeletal muscle, which can differentiate into bone cells. This has implications for both traumatic fracture repair and the more subtle tissue decline associated with ageing.
The field is undergoing a decisive transition from donor-derived MSCs, which vary in quality and pose ethical challenges, to iPSC-derived MSCs (iMSCs). iMSCs provide unlimited expansion from a standardised source, reducing variability and enabling scalable production. This marks the movement from bespoke therapies toward reproducible, off-the-shelf products.
| Feature | Primary MSCs | iPSC-derived MSCs (iMSCs) |
| Source | Bone marrow, adipose, umbilical cord | Reprogrammed adult cells |
| Scalability | Donor-limited | Unlimited, from master banks |
| Consistency | High donor variability | Homogeneous, reproducible |
| Differentiation | Multipotent | Directed differentiation possible |
| Tumorigenicity risk | Low | Higher if reprogramming incomplete |
| Ethical issues | Invasive harvest, donor consent | Avoids embryo controversy |
Pharmacological strategies targeting senescence
Pharmacological interventions are designed to modulate or eliminate senescent cells. Two categories dominate: senolytics, which selectively induce the death of senescent cells, and senomorphics, which suppress the SASP without cell clearance.
The most studied senolytic combination is dasatinib with quercetin. In animal studies, this duo improved bone density and cognitive function. Human trials have been more complex. A Phase 2 study in post-menopausal women with osteoporosis did not show population-wide benefit, but a subgroup with higher senescent burden responded more strongly. Similarly, early studies in older adults with mild cognitive impairment suggest benefit only in those with low baseline cognitive scores.
These results highlight that senolytics are not universally effective but instead may work best as precision medicine targeted at individuals with a demonstrable senescent cell burden. Future development is likely to focus on diagnostics that can identify responsive patients and on compounds that target specific senescent subtypes.
Epigenetic reprogramming is another strategy. By transiently activating Yamanaka factors or using small-molecule cocktails, cells can be partially reprogrammed to a younger state without losing identity. This avoids the risk of tumour formation associated with complete reprogramming, offering a safer path to age reversal.
Gene therapy and tissue engineering innovations
Gene therapy provides tools to repair or enhance cellular function. Viral and non-viral vectors are used to deliver corrective genes or therapeutic proteins. CRISPR-Cas9 technology enables precise gene editing, allowing mutations to be corrected or pathogenic sequences to be removed.
Tissue engineering, often combined with gene therapy, integrates cells with scaffolds and bioactive factors to build functional tissues. The most visible innovation is 3D bioprinting, where living cells act as bio-inks to fabricate structures layer by layer. The challenge has been vascularisation: without blood vessels, printed tissues cannot survive at scale. Recent breakthroughs in creating vascularised constructs represent significant progress towards functional organs suitable for transplantation.
These technologies together address one of healthcare’s most significant crises: the shortage of donor organs. If scalable, engineered tissues could eventually replace current transplant systems.


Clinical trials and regulatory frameworks in 2025
As of 2025, the clinical pipeline for rejuvenation and repair spans musculoskeletal disease, spinal cord injury, cardiovascular repair, and neurodegenerative disorders. For example, Regeneron’s COURAGE trial has shown promise for muscle-preserving antibodies in patients losing weight on GLP-1 therapies. Another trial, OST-HER2 immunotherapy, has demonstrated benefit in delaying osteosarcoma recurrence, achieving RMAT designation from the US Food and Drug Administration (FDA).
Despite these signals, approval rates for regenerative products remain low. The FDA’s Regenerative Medicine Advanced Therapy (RMAT) designation and the European Medicines Agency (EMA) Advanced Therapy Medicinal Products (ATMP) framework provide expedited routes. However, sponsors often struggle to meet the evidentiary standards. Manufacturing variability, difficulty in developing potency assays, and reliance on small, open-label studies reduce regulatory confidence. This paradox means the very innovation that qualifies therapies for accelerated review often creates the barriers that prevent approval.
Safety remains the overriding concern. Pluripotent cells carry the risk of tumour formation if differentiation is incomplete. Immunogenicity and unpredictable behaviour of infused cells also present hazards. Regulatory bodies mandate Good Manufacturing Practice (GMP) and rigorous quality control to address these issues, but reproducibility is still a significant challenge.
Ethical considerations and equitable access
The debate over embryonic stem cells once dominated discussion. While induced pluripotent stem cells largely sidestep this issue, other ethical concerns remain. Donor exploitation, particularly in egg donation, and inadequate informed consent in commercial stem cell clinics are persistent problems. The marketing of unproven therapies to vulnerable patients undermines trust and has triggered regulatory enforcement actions worldwide.
Germline editing raises more profound questions. Changes introduced at the embryo stage would be heritable. The consensus across leading scientific bodies is that germline modification should not proceed outside tightly controlled basic research, as the risks are poorly understood and the societal consequences irreversible.
Equity is a defining challenge. The high cost of regenerative therapies raises the prospect of restricted access, with treatment available only to the wealthy. In parallel, research in epigenetics shows that poverty, pollution, and malnutrition can leave heritable marks on DNA methylation, influencing disease risk in subsequent generations. If therapies to reverse this damage are only available to privileged groups, a biologically enforced class divide could emerge. The issue extends beyond economics to social justice, demanding policy action to ensure accessibility.
Leading researchers and institutions in cellular rejuvenation
Progress in the field reflects the work of pioneering scientists. Steve Horvath’s development of epigenetic clocks has reshaped how biological age is quantified. Elaine Fuchs at Rockefeller University has mapped the behaviour of epithelial stem cells in skin repair, demonstrating how epigenetic memory can either promote regeneration or perpetuate inflammation. Gordon Keller received the 2025 ISSCR Achievement Award for advancing directed differentiation of human pluripotent stem cells into cardiovascular and blood lineages.
Industrial investment is shaping research strategy. Altos Labs has attracted over $3 billion in funding to pursue large-scale investigations into cellular programming. Mesoblast has focused on allogeneic stem cell therapies that do not require donor matching. Pharmaceutical leaders such as Pfizer and Regeneron have expanded their pipelines to include regenerative approaches.
Academic institutions remain vital, with the Icahn School of Medicine at Mount Sinai and the Cedars-Sinai Regenerative Medicine Institute both driving translational research and training the next generation of scientists.
Future directions for cellular rejuvenation and tissue repair
The next phase will integrate artificial intelligence to accelerate discovery and personalisation. Machine learning can mine genomic and epigenetic data to predict treatment response, optimise trial design, and discover new pharmacological agents.
The central aim is to extend healthspan, not merely lifespan. By delaying frailty, dementia, and multimorbidity, rejuvenation therapies could reduce healthcare burden while improving quality of life. For this potential to be realised, regulatory clarity, rigorous long-term safety data, and equitable access must align.
Real Fact: The number of people aged over 65 worldwide is projected to double by 2050, creating unprecedented demand for therapies that extend healthy years rather than prolong life.
The field has matured from speculative science into a data-driven endeavour. However, its success will not be measured only in laboratory milestones. It will be judged by whether therapies become safe, affordable, and accessible to patients across all societies. Restoring cellular health offers a profound opportunity: to reframe medicine from treating disease towards maintaining vitality at its most fundamental level.






