Preprint
Review

This version is not peer-reviewed.

What Is Aging, and How Can We Defeat It?

Submitted:

14 November 2025

Posted:

18 November 2025

You are already at the latest version

Abstract
Aging is not an immutable fate but a malleable biological process driven by interconnected molecular, cellular, and systemic failures. While modern medicine excels at managing acute conditions, it largely fails to address the root cause of most chronic diseases – aging itself. In this review, I synthesize current evidence that aging arises from the interplay of stochastic damage and evolutionarily shaped regulatory programs (exemplified by conserved parameters such as AROCM) and manifests through the hallmarks of aging, including genomic instability, proteostatic collapse, mitochondrial dysfunction, cellular senescence, and inflammaging, ultimately driving chronic disease across organ systems. I argue that defeating aging requires a strategic shift from symptom palliation to restoration of youthful function, achieved through three synergistic pillars: (1) elimination of damage (e.g., senolytics, monoclonal antibodies against pathogenic immune clones or DAMPs), (2) reactivation of endogenous repair mechanisms (e.g., caloric restriction, metformin, GLP-1 agonists, transient OSK expression), and (3) cell, tissue, and organ replacement (e.g., stem cell–derived islets, FOXO3-enhanced MSCs). I highlight evolutionary insights from long-lived species, the centrality of chronic inflammation and fibrosis in age-related disease, and the transformative role of AI: from multi-omic aging clocks to agentic systems for target discovery and personalized longevity medicine. The tools to initiate this paradigm shift are already emerging; what is needed now is scientific rigor, interdisciplinary integration, and a collective commitment to treating aging as the fundamental driver of human morbidity and mortality.
Keywords: 
;  ;  ;  ;  ;  ;  ;  ;  ;  

Why Aging is a Fundamental Driver of Human Suffering and Why it is so Hard to Deal With

Today, representatives of our species live longer and healthier than at any time in history. Thanks to vaccination, the later introduction of antibiotics, and now the emerging victories over cancer, aging may become the next frontier our minds are poised to overcome [1,2].
Yet progress in this field seems stalled, and not only due to lack of funding or understanding, but because aging is fundamentally different from infectious diseases or cancer. By its nature, aging is a systemic degenerative process, not caused by a single hostile agent, but by a progressive, body-wide loss of function – observable in the brain, immune system, skeletal muscles, cardiovascular system, and virtually every organ [3,4].
The bad news is that modern medicine remains poorly equipped to address such degenerative and chronic conditions. It often focuses on masking symptoms rather than achieving true recovery or eradication of the underlying pathology. Thus, addressing aging requires answering two intertwined questions: how to genuinely cure chronic disease – and how to defeat aging itself [5,6].
How difficult it will be to defeat aging is, in fact, inscribed in the very architecture of our gene regulatory networks. A recent large-scale epigenomic atlas across 17 human tissues demonstrated that perturbing the majority of age-associated co-methylation modules, representing core gene networks, exacerbates rather than ameliorates aging signatures. Only a handful of modules, such as those linked to NAD⁺ salvage metabolism, showed resilience to intervention. This underscores a sobering truth: effective anti-aging strategies must be exquisitely precise, because in most cases, blunt manipulation does more harm than good [7].
Why aging is harmful is becoming increasingly obvious, yet human psychology remains reluctant to acknowledge it as a treatable condition. Common objections come from various domains: religious beliefs (“this is how God made it”), demographic fears (“the planet is overpopulated!”), or concerns about stagnation (“we’ll run out of new ideas”).
In reality, many polytheistic and especially Abrahamic religions developed the concept of “life after death” as a psychological coping mechanism for mortality – anxiety so profound it echoes in the existential verses of Omar Khayyam [8,9]. Not all traditions reject longevity: in Daoism, for example, longevity and harmony with nature are central virtues, giving rise to practices like Qigong [10].
The demographic argument now seems increasingly outdated: more and more countries face severe economic strain due to aging populations, with no viable solutions in sight [11,12]. Similarly, the fear of intellectual stagnation ignores a key fact: cognitive rigidity is itself a hallmark of brain aging [13].
Most striking, however, is this contradiction: people universally condemn cancer, Alzheimer’s, and heart attacks as evils, and wish them on no one – yet normalize the very process that causes them all: aging, often even refusing to recognize it as a disease [14]. Beneath all objections to fighting aging lies a single, deep-seated belief: “It’s impossible: no one has succeeded before.”
Meanwhile, aging imposes a substantial burden on individuals, healthcare systems, and economies worldwide. But today, for the first time in history, we possess unprecedented data, tools, and biological insight. The question is no longer whether we can defeat aging, but when.
And so, evidence supports the view that: targeting the biological mechanisms of aging represents a viable and urgent strategy to extend healthspan, and potentially lifespan, with growing evidence supporting its feasibility [15].

What Causes Aging: Stochasticity or Programme?

If aging exerts profoundly detrimental effects across biological systems, what is its fundamental cause? This question remains one of the hottest debates in biogerontology, and to date, no single consensus theory of aging exists.
What is widely accepted, however, is the “Hallmarks of Aging” framework, though it is more phenomenological than causal. In brief, aging is characterized by a set of interconnected changes, grouped into three categories. Primary hallmarks, which arise intrinsically and may act as initial drivers: (1) genomic instability, (2) telomere attrition, (3) epigenetic alterations, (4) loss of proteostasis, and (5) disabled macroautophagy. Antagonistic hallmarks, which emerge as compensatory responses to primary damage but become harmful over time: (6) cellular senescence, (7) mitochondrial dysfunction, and (8) deregulated nutrient-sensing. Integrative hallmarks, which result from the interplay of the above and reflect systemic collapse: (9) chronic inflammation, also referred as inflammaging, (10) stem cell exhaustion, (11) altered intercellular communication, and (12) microbiome dysbiosis [16].
I will explore these mechanisms in detail later. For now, I ask: what is the root cause of aging? Two opposing views dominate the field: purely stochastic damage versus an evolutionarily shaped program. Increasingly, however, scientists converge on a middle ground: aging is quasi-stochastic: a blend of random molecular decay and regulated biological responses.
Indeed, aging can be seen as a biological manifestation of the second law of thermodynamics: complex living systems maintain order by exporting entropy to their environment, but over time, they accumulate internal entropy, gradually losing the ability to preserve structural and functional integrity.
This manifests at multiple levels. Somatic mutations accumulate stochastically across the genome [17]. Chromatin architecture, exquisitely organized in youth, becomes increasingly disordered with age: global loss of DNA methylation and histone marks coexists with focal heterochromatinization, such as senescence-associated heterochromatin foci (SAHF) [18]. This epigenetic drift disrupts transcriptional fidelity, derepresses endogenous retroviruses and transposable elements, and fuels genomic instability, inflammation, and cancer risk [19,20].
At the tissue level, aged cells lose their differentiated identity. Meanwhile, the extracellular matrix stiffens due to random non-enzymatic cross-links (e.g., advanced glycation end-products, or AGEs), which not only reduce tissue elasticity but also mechanically induce senescence-like phenotypes in neighboring cells – this all gives rise to the extracellular matrix (ECM) theory of aging [21,22].
At first glance, these processes appear purely stochastic, yet biological programs actively modulate them. For instance, FOXO3, a key regulator of chromatin stability and stress resistance, declines with age but functions as a potent anti-aging safeguard, as demonstrated in arteries, lungs, and skeletal muscle [23,24,25]. SIRT6, an NAD⁺-dependent deacylase, orchestrates genomic integrity, suppresses LINE1 retrotransposons, and enhances DNA repair; its overexpression extends lifespan in mice and it is evolutionarily more efficient in long-lived species, while some rare SIRT6 variants exist in human centenarians [26,27,28,29,30]. Klotho, a circulating anti-aging hormone, represses insulin/IGF-1 and Wnt signaling, protects against fibrosis and vascular calcification, and its deficiency recapitulates accelerated aging [31,32,33]. Polycomb complexes (PRC1/2), by maintaining repression of developmental genes, buffer epigenetic drift; their age-related dysfunction underlies the loss of cellular identity [34,35,36,37]. This suggests that numerous regulatory networks embedded in our biology may shape the trajectory of aging.
Supporting this view, a recent study from Vadim Gladyshev’s lab disentangled stochastic and programmed components of aging: while some age-related gene expression changes are tissue-specific and random, others are highly concordant across cell types and organs, implying systemic regulation [38].
Likewise, work from Andrew Teschendorff’s group showed that, despite the presence of stochastic noise, the core signature of biological aging is largely non-stochastic – with the notable exception of pre-cancerous and cancerous tissues, where randomness dominates [39].

Evolutionary Regulators of Aging

If a biological programme of aging exists, what is it, and who are its main players? As in the previous section, such regulation likely operates at multiple levels, beginning with chromatin integrity.
For mammals, the AROCM (Average Rate of Change in Methylation), calculated across a conserved set of 552 CpG sites located in polycomb-repressed bivalent promoter regions (BivProm2+), robustly predicts species maximum lifespan, according to work by Steve Horvath and the Mammalian Methylation Consortium. These CpGs serve as a biomarker of epigenetic drift: they are hypomethylated in youth and gain methylation with age. Crucially, a lower AROCM (reflecting slower age-related methylation gain and better maintenance of the youthful epigenetic state) correlates strongly with greater longevity, spanning from short-lived rodents to exceptionally long-lived whales and bats. In essence, species with the slowest epigenetic “drift” at these developmentally silenced loci tend to live the longest [40].
Beyond epigenetics, other phenomenological patterns of animal longevity reveal evolutionary adaptations. For example, elephants and whales, despite their vast cell numbers (and thus high cancer risk under Peto’s paradox), rarely develop tumors.
Elephants resolve Peto’s paradox through a remarkable expansion of the TP53 locus, harboring one canonical copy and 19 transcribed retrogenes (TP53RTGs). Although these retrogenes lack DNA-binding domains and do not function as classical transcription factors, they enhance p53 signaling and promote a hypersensitive apoptotic response to DNA damage, effectively eliminating potentially malignant cells before they can proliferate [41,42].
In contrast, cetaceans such as bowhead and other long-lived whales lack TP53 amplification but instead rely on a multi-layered tumor suppression strategy. This includes positive selection and lineage-specific duplications in key apoptosis regulators (e.g., CASP3, APAF1, TP73) and potent inhibitors of angiogenesis (e.g., SERPINE1, CD82, TSC2) [43]. Critically, bowhead whale cells also exhibit uniquely high-fidelity DNA repair: they resolve double-strand breaks and mismatch lesions with exceptional efficiency and accuracy, mediated in part by elevated levels of CIRBP and its downstream effector RPA2, which together enhance genomic stability without eliminating damaged cells via apoptosis [44].
According to the research in the Gorbunova and Seluanov laboratory, he naked mole-rat, a longevity icon, possesses a unique hyaluronan-rich extracellular matrix that confers cancer resistance [45] – and even mice expressing naked mole-rat hyaluronic acid synthase 2 gene (nmrHas2) had lower tissue inflammation and cancer incidence [46]. Moreover, an evolutionarily altered cGAS protein in this species lacks the suppressive function seen in humans and mice, instead enhancing homologous recombination repair and delaying aging through prolonged chromatin retention after DNA damage [47].
Spalax galili (the blind mole rat), another long-lived rodent, does not accumulate clonal expansions of T- and B-lymphocytes with age – unlike humans, whose immune senescence drives chronic “inflammaging” [48].
Early in their evolution, Neoaves acquired a loss-of-function KEAP1 mutation that constitutively activates NRF2, enhancing antioxidant defense and neutralizing the high ROS from their elevated metabolism, decoupling intense metabolic activity from oxidative damage and enabling unusually long lifespans [49].
Longevity is also shaped at the physiological level. Neoteny, the retention of juvenile traits into adulthood, appears tightly linked to extended lifespan [50]. The axolotl, a neotenic salamander, remains in a larval-like state (with external gills and delayed metamorphosis due to impaired thyroxine signaling) and shows negligible senescence for most of its 13-year lifespan [51,52,53]. Naked mole-rats resemble hairless, earless newborn mice (a neotenic phenotype), and their mitochondria retain biophysical properties typical of neonatal, not adult, rodents [54,55]. Even humans are neotenic: compared to other great apes, we have flatter faces, smaller canines, reduced sexual dimorphism, and weaker musculature – traits that emerged alongside extended lifespans and complex social structures (including pair bonding) [54,56]. Indeed, among hominids, humans are among the longest-lived, despite not being the largest.
Conversely, lifespan can also shorten during evolution, often due to relaxed selection or inbreeding. In domestic dogs, large breeds live significantly shorter lives than small ones, primarily due to body size–related aging acceleration. However, within breeds, higher individual inbreeding coefficients are associated with reduced lifespan, and purebred status itself (vs. mixed ancestry) explains 46% of lifespan variation after accounting for body size, highlighting the fitness cost of recent inbreeding and loss of heterozygosity [57,58]. Even more extreme is the case of annual killifish (Nothobranchius spp.), which inhabit temporary African pools. Their life history is compressed into a few months: they mature rapidly, reproduce, and die, while their embryos enter diapause to survive the dry season. Even in captivity, they rarely exceed 12–14 months. This short lifespan stems from evolutionary trade-offs: massive expansion of transposable elements, degradation of DNA repair pathways, and loss of genomic maintenance systems – essentially, they’ve “given up” on longevity [59,60,61].
What can we conclude? Evolution has already shaped Homo sapiens into one of Earth’s longest-lived species. But nature has also equipped other organisms with extraordinary longevity mechanisms, from epigenetic stability to cancer resistance and neotenic physiology. Rather than developing entirely novel paradigms, current efforts can build upon evolutionarily conserved longevity mechanisms observed across species. If we combine them with radical technologies, from epigenetic reprogramming to organ replacement, to extend healthspan beyond evolutionary limits.

Aging Cell Physiology

During aging, profound alterations occur within every cell, many of which are captured by the “Hallmarks of Aging” framework. While a full discussion would require a dedicated review, I focus here on several key cellular processes.

Nuclear Dysfunction

The nucleus is among the first compartments affected. DNA damage, from both endogenous sources (e.g., replication errors, ROS) or exogenous insults (UV, alkylating agents), triggers the DNA damage response (DDR). Initially protective (activating repair and halting cell cycle to prevent oncogenesis), DDR becomes chronically activated in aged cells, driving a senescent phenotype characterized by sustained expression of p21 and other cell cycle inhibitors [62].
A major endogenous trigger of DDR is telomere attrition. Because eukaryotic chromosomes are linear, their ends are capped by telomeres – nucleoprotein structures that prevent recognition as double-strand breaks (DSBs). With each cell division, telomeres shorten; once critically eroded, they mimic DSBs, leading to persistent DDR activation and irreversible cell cycle arrest [63].
Beyond mutations, epigenomic drift is a hallmark of aging nuclei. Global loss of DNA methylation and histone marks reduces chromatin compaction, leading to: (1) increased transcriptional noise (leaky expression of silenced genes), (2) derepression of transposable elements, fueling inflammation and genomic instability, (3) loss of cellular identity, as lineage-specific gene expression programs blur [64,65].
Paradoxically, focal hypermethylation also occurs, often at promoters of tumor suppressor or developmental genes, which forms senescence-associated heterochromatin foci (SAHF), which may be adaptive (e.g., silencing oncogenes) or maladaptive [18,66].

Cytoplasmic and Proteostatic Collapse

Nuclear dysfunction propagates to the cytoplasm. Mutated or truncated mRNAs (from genomic or splicing errors) produce misfolded proteins. Simultaneously, altered gene expression disrupts stoichiometry of multi-subunit complexes (e.g., ribosomes, proteasomes), further impairing protein homeostasis [67,68].
In healthy young cells, misfolded proteins are either refolded by chaperones (foldases) like HSP70/HSP90, or degraded via the ubiquitin–proteasome system (UPS) or autophagy–lysosome pathway (ALP). All these systems (foldases, UPS, ALP) are highly energy-consuming and thus rely on ATP [69].
But in aging, ATP becomes scarce (see below), crippling these systems. Cells resort to ATP-independent “holdases” (e.g., small HSPs, like HSP27) that sequester aggregates, leading to lipofuscin accumulation in long-lived post-mitotic cells (e.g., cardiomyocytes, retinal pigment epithelium) [69].
Critically, impaired macroautophagy affects not only proteins but also organelles. Damaged mitochondria, which produce excess ROS and generate little ATP, are normally cleared by mitophagy, but this process falters with age, creating a vicious cycle [70].
Autophagy is regulated by several nutrient-sensing pathways, such as mTOR (activated by nutrients/energy), which suppresses autophagy, and AMPK (activated by starvation), which promotes it and antagonizes mTOR. This explains why caloric restriction, rapamycin (mTOR inhibitor), and metformin (AMPK activator) enhance proteostasis and extend healthspan [71].

Mitochondrial Decline

Mitochondria suffer a double vulnerability: their DNA lacks protective histones and is located in close proximity to the electron transport chain (ETC), where reactive oxygen species (ROS) production is highest, making mtDNA highly susceptible to mutations. These mutations impair the function of ETC proteins, rendering them even less efficient. As a result, mitochondria produce more ROS and less ATP, creating a vicious cycle. The situation is further aggravated by ROS-mediated oxidation of membrane lipids, which compromises mitochondrial integrity [70].
Additionally, NAD⁺ levels decline with age due to reduced synthesis and increased consumption, particularly by PARP during the DNA damage response (DDR), leading to reduced sirtuin activity and further impairment of ETC function [72].
In fact, some mitochondria remain relatively intact in aged cells. However, because autophagy, especially mitophagy, is impaired with age, a large fraction of mitochondria accumulate damage and become dysfunctional [70].
The result is a triad of hallmarks in aged cells: reduced ATP output, increased ROS leakage, and oxidized membrane lipids [70].

Senescence and Inflammaging

Faced with cumulative stress – from genome, mitochondria, proteome, and extracellular matrix – cells often enter senescence, which is characterized by irreversible cell cycle arrest (via p16/Rb and p53/p21), but at the same time resistance to apoptosis (via upregulation of BCL-2 family anti-apoptotic proteins) [73].
Their true danger lies in the senescence-associated secretory phenotype (SASP): a cocktail of pro-inflammatory cytokines (IL-6, IL-8, TNF-α), chemokines, and matrix metalloproteinases. SASP acts as a damage-associated molecular pattern (DAMP), inducing paracrine senescence in neighboring cells – hence the “zombie cell” analogy [73].
Over time, SASP drives chronic, low-grade inflammation (“inflammaging”), which impairs stem cell function, disrupts tissue regeneration, alters host–microbiome crosstalk, which finally integrates all hallmarks of aging into a systemic collapse [73].
Normally, immune cells clear senescent cells, but immunosenescence with age cripples this surveillance, allowing senescent cells to accumulate and amplify the damage [74,75]…

How Aging Affects Major Organ Systems

Taking into consideration all general cell aging mechanisms listed in the section above, I will briefly look at how this all ends with the well-known age-associated diseases of particular organ systems that we all know, and why aging is indeed their fundamental reason [76].
Just before delving into the details, a recent perspective introduced a quantitative framework linking age-related diseases to the hallmarks of aging and found the strongest alignment for type 2 diabetes, followed by idiopathic pulmonary fibrosis, Parkinson’s disease, and rheumatoid arthritis [77].

Brain

The brain is probably the most complicated organ in our body, constituting as many as 86 billion cells [78]. Main brain cells, neurons, are long-living with very low neurogenesis in some zones in adults, but their function becomes impaired in the elderly. Two main reasons in their biology underlie this: first, neurons rely very heavily on energy due to the need for membrane potential maintenance, and energy becomes scarce because of mitochondrial dysfunction [79]. Second, neurodegenerations are a dramatic illustration of how harmful proteostasis stress may be: beta-amyloid plaques and tau protein neurofibrillary tangles, alpha-synuclein, and TDP-43 and SOD1 are thought to be the most important players in Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), respectively [80].
What may be even more important is chronic inflammation in the neural tissue. The key players here are astrocytes (going into the A1 state), microglia (M1-like), and T-lymphocytes, particularly Th1 and Th17 types [81,82,83]. T-lymphocytes, according to recent research from the Brunet group, are particularly deleterious, as they foster brain aging the most – namely via IFN-γ signaling [84].
A crucial and often underestimated factor in age-related cognitive decline and neurodegenerative diseases is the increasing permeability of blood vessels and the blood-brain barrier (BBB). This occurs for several reasons, including a decrease in the expression of genes encoding tight junction proteins (such as ZO-1, occludin, claudin) [85] and a selective death of M-pericytes, which are responsible for supporting the extracellular matrix [86].
What drives this inflammation, which is so wicked and exacerbates the effects of protein aggregates? The factors include the inflamed state of the whole body (including the brain itself), maladaptive responses of glial cells to protein aggregates, increased blood–brain barrier permeability (so that T cells may enter, while brain antigens can leak out and trigger immunity), impaired gut–brain axis signaling, and the debated role of viral infections such as HSV-1 [87,88,89].

Cardiovascular System, Lungs, and Skeletal Muscles

Cardiovascular diseases (CVD) remain the leading cause of death worldwide. This group includes atherosclerosis-related conditions such as hypertension, myocardial infarction, stroke, and acute coronary syndrome, primarily affecting blood vessels, as well as heart-specific disorders like cardiac insufficiency, arrhythmias, and bradycardia [90].
The key deleterious processes in blood vessels involve cholesterol deposition in the vessel wall, followed by maladaptive responses of macrophages that infiltrate to clear it but instead become pro-inflammatory foam cells. Additional contributors include vascular calcification and extracellular matrix (ECM) stiffening [91]. Among ECM-related pathways, SPP1–CD44 signaling is particularly harmful, promoting inflammation and fibrosis [92]. On the protective side, FOXO3 (a stress-resistance transcription factor) declines in aged endothelial cells, as does nitric oxide (NO) production, impairing vasodilation and vascular homeostasis [23].
As for the heart, major age-related changes include fibrosis (driven by factors such as WWP2) and chronic inflammation [93,94]. Cardiomyocytes become hypertrophied yet decrease in number due to limited regenerative capacity. The gene encoding FOXP1, a transcriptional regulator that maintains cardiac identity and function, shows reduced expression with age, contributing to functional decline [95].
Lungs are tightly interconnected with the cardiovascular system, and their aging contributes to conditions such as chronic obstructive pulmonary disease (COPD), one of the leading causes of death worldwide, as well as idiopathic pulmonary fibrosis (IPF) and increased susceptibility to severe pneumonias. The underlying mechanisms include enlargement of alveoli, loss of elastic recoil, and impaired mucociliary clearance [96,97].
As for skeletal muscles, they share several physiological features with the heart. Inflammation and fibrosis also occur in aged muscle tissue, and together with impaired regenerative capacity and intracellular lipid accumulation, they drive sarcopenia, a condition characterized by reduced muscle mass, declining physical strength, and increased risk of disability [98]. Interestingly, FOXO3 in myotubes also serves as a key homeostasis regulator, whose expression is gradually lost during aging [25].

Immune System

Immune system aging is probably one of the most important aspects of organismal aging, because it dramatically affects all other organ systems and impairs the body’s ability to eliminate precancerous, cancerous, and senescent cells [99,100].
At the population level, a shift toward a higher myeloid-to-lymphoid cell ratio occurs. This results from chronic inflammatory signaling, which biases hematopoietic stem cells toward myeloid lineage commitment. Myeloid cells belong to the innate immune system, which is less “clever” than adaptive immunity and more prone to drive nonspecific inflammation rather than precise responses, further fueling the vicious cycle of inflammaging [101,102].
Within the lymphoid lineage, profound changes also take place. Thymic involution with age drastically reduces the production of new naïve T cells in elderly individuals [103]. Meanwhile, memory T cells specific to a very limited set of antigens – often from chronic viral infections like cytomegalovirus or Epstein-Barr virus – become dominant. In some cases, individual clones expand to represent more than 1% of all T lymphocytes, which is excessive given the normal diversity of the T-cell repertoire. These expanded clones not only pour fuel on the fire of chronic inflammation but also suppress the maturation of new naïve T cells and occupy their niches [104,105,106,107]. Notably, as has been discussed earlier, the long-lived rodent Spalax does not exhibit this pathological clonal expansion [48].
With age, the immune system also accumulates errors and begins to attack self-antigens as tolerance mechanisms break down. This is why immune aging shares more features with autoimmune disorders and hypersensitivity reactions than is commonly appreciated [108,109].
Yet paradoxically, this same chronically activated and dysregulated immune system becomes largely ineffective in the elderly, against both infectious agents and cancers. This state is known as immunosenescence, and it explains why older adults face significantly higher risks from infections such as COVID-19 [110].

Stomach, Intestine, Microbiome, Liver, and Pancreas

Some of the most important age-related changes occur in the gastrointestinal (GI) system and associated organs, disrupting interactions with the gut microbiome and triggering systemic effects in blood and distant tissues [111].
In the aged stomach, hypoacidity (reduced HCl secretion) develops alongside decreased pepsin synthesis. This is often linked to Helicobacter pylori infection and gastric mucosal atrophy, leading to impaired protein digestion, a factor that exacerbates sarcopenia [112,113].
In the intestine, the mucosa also deteriorates, and, most critically, synthesis of tight junction proteins (e.g., occludin) declines, resulting in “leaky gut” (increased intestinal permeability) [114,115,116]. This coincides with dramatic shifts in the gut microbiome: overall diversity drops, with a notable loss of Akkermansia muciniphila (a key symbiont that strengthens the epithelial barrier) and SCFA-producing genera such as Faecalibacterium, Roseburia, and Eubacterium. Simultaneously, potentially pathogenic Proteobacteria expand [117,118,119]. Combined with weakened local immune surveillance and tolerance mechanisms, this dysbiosis fuels chronic inflammation, both locally in the gut and systemically across other organs [120].
The aging liver undergoes progressive structural and functional decline. Hepatocytes become enlarged and increasingly polyploid, while liver sinusoidal endothelial cells lose their fenestrations, a process known as pseudocapillarization, which impairs the exchange of metabolites, lipoproteins, and insulin between blood and hepatocytes. This directly contributes to hepatic insulin resistance and age-related type 2 diabetes. Concurrently, mitochondrial dysfunction, reduced autophagy, and accumulation of lipofuscin and fat (even in non-obese individuals) lead to steatosis and diminished detoxification capacity [121,122,123]. A recent study in aged monkeys further showed that SREBP2 (a master regulator of cholesterol biosynthesis) is upregulated in the aging liver, driving hypercholesterolemia and accelerating liver aging itself [124].
The exocrine pancreas also experiences significant age-related remodeling. Acinar cells atrophy, and fatty infiltration (pancreatic lipomatosis) increases, reducing secretion of digestive enzymes such as amylase, lipase, and trypsin. This exocrine pancreatic insufficiency further compromises nutrient absorption, especially of proteins and lipids, amplifying the risk of malnutrition and sarcopenia [125,126]. In the endocrine compartment, aging drives degeneration of pancreatic β-cells, marked by a heightened unfolded protein response, accumulation of protein aggregates, and β-cell-specific upregulation of the ER chaperone HSP90B1, which impairs glucose-stimulated insulin secretion. Together, these changes disrupt proteostasis, cause functional decline, and ultimately contribute to impaired glucose tolerance and type 2 diabetes [127,128].

Other Organs

Kidneys. With age, nephron mass declines, accompanied by glomerulosclerosis and interstitial fibrosis. Impaired autophagy in podocytes and tubular cells leads to accumulation of damaged DNA and ROS, resulting in reduced glomerular filtration rate, predisposing to chronic kidney disease and heightened vulnerability to acute kidney injury [129,130].
Bones. Osteoblast activity wanes due to attenuated Wnt/β-catenin signaling and AGE-mediated collagen cross-linking, driving osteoporosis. In postmenopausal women, estrogen loss unleashes RANKL, causing a surge in osteoclast-mediated resorption, culminating in hip and vertebral fractures [131,132].
Male reproductive system. Leydig cells show reduced expression of INSL3 and LHCGR, leading to age-related testosterone decline (late-onset hypogonadism), which exacerbates sarcopenia and osteoporosis. Concurrently, senescent cell accumulation in prostatic stroma and chronic inflammation fuel benign prostatic hyperplasia and increase prostate adenocarcinoma risk [133,134].
Female reproductive system. Ovarian reserve depletion stems from accumulated double-strand DNA breaks and mitochondrial dysfunction in oocytes, culminating in menopause. The resulting estradiol drop disrupts the RANKL/OPG balance, accelerating bone loss, while also causing vaginal epithelial atrophy and genitourinary syndrome of menopause [135,136].
Skin. Dermal thinning and wrinkling arise from suppressed TGF-β/Smad signaling and senescent fibroblasts, reducing COL1A1 and elastin production. Combined with DNA damage in keratinocytes and NAD⁺ decline, this impairs wound healing and elevates squamous cell carcinoma risk [137,138].

Cancer Risks and Beyond

Aging is the major risk factor for nearly all cancers, probably excluding only juvenile forms. This occurs due to a combination of factors. Most obviously, cells accumulate mutations over time, and if such a cell evades death, it may eventually give rise to cancer. As I have already discussed, the immune system, whose primary role includes cancer surveillance, becomes increasingly ineffective with age. Moreover, a chronically inflamed environment provides an ideal niche for pre-cancerous and cancerous cells to thrive. Additional contributors include VEGF secretion by senescent cells, extracellular matrix (ECM) remodeling, and age-related microbiome alterations, all of which further fuel tumor initiation and progression [100,139].
Thus, as we can see, aging lies at the core of almost every major human disease. If we truly want to develop effective treatments, we must adopt a holistic perspective – and place aging itself at the center of our therapeutic strategy.

What Technologies Could be Used to Fight Aging

Given the deep interconnections between aging, chronic inflammation, fibrosis, and age-related diseases, the next critical question is: what tools do we already have, or will soon have, to intervene?
Today, the most effective lifespan-extending interventions remain deceptively simple: caloric restriction (CR) and regular exercise act as powerful rejuvenating therapies, alongside sufficient sleep, stress reduction, sunlight exposure, and a nutrient-rich diet [140]. Yet, as powerful as they are, they are not enough to halt aging. Here, I focus on the concrete therapeutic tools already in our arsenal, and those emerging on the near horizon.

Small Molecules

Small molecules represent the oldest and still dominant class of therapeutics, accounting for over half of all approved drugs. Despite the rise of biologics, gene therapies, and cell-based approaches, big pharma is now returning to small molecules – a trend highlighted by Alex Zhavoronkov in recent LinkedIn posts. Their advantages are clear: (1) the most easy tissue delivery, especially compared to gene therapies, (2) often orally bioavailable (no injections needed) (3) relatively inexpensive to manufacture, (4) target and off-target effects can be predicted with increasing accuracy using molecular docking and machine learning [141,142,143,144].
This category includes several well-known “pro-longevity” candidates: rapamycin, an mTOR inhibitor that extends lifespan and healthspan in model organisms, but in humans, its immunosuppressive side effects limit chronic use [145]. Skulachev ions (SkQ1), mitochondria-targeted antioxidants designed to neutralize ROS; however, their systemic efficacy in humans remains modest [146,147].
Metformin and NMN represent particularly promising candidates for near-term clinical translation, given their safety profiles, mechanistic rationale, and ongoing trials. Metformin, initially an anti-diabetic drug, activates AMPK, enhances autophagy, and antagonizes mTOR signaling, while NMN serves as a precursor to NAD⁺, a coenzyme that declines sharply with age and is essential for mitochondrial and DNA repair functions. Both are already in clinical trials for aging-related outcomes (e.g., TAME and MILES trials for metformin) [148,149,150,151,152,153].
But the real game-changers may come from precision small molecules that block specific pathological drivers. For example, Insilico Medicine identified TNIK as a master regulator of fibrosis and developed INS018_055 (Rentositib), a TNIK inhibitor now in trials for idiopathic pulmonary fibrosis and kidney fibrosis [154,155], while the Petretto lab has pinpointed WWP2 as another promising target in heart and kidney fibrosis [94,156]. At the same time Insilico has also unveiled ISM8969, a potent NLRP3 inflammasome inhibitor, designed for CNS inflammatory disorders like Parkinson’s disease [157]. Evidence supports that NLRP3 inhibitors could have a far broader impact, since the NLRP3 inflammasome is a central hub of inflammaging, one of the core pillars of aging itself [158].
Ultimately, no single molecule will be a silver bullet. But a rational combination, boosting autophagy, restoring NAD⁺, suppressing chronic inflammation, halting fibrosis, already offers a credible, near-term path toward significantly extended healthspan.

Antibodies

This class of therapeutics emerged relatively recently but has already proven highly effective in treating many chronic diseases, often in combination with other agents.
The best-known examples are immune checkpoint inhibitors used in cancer immunotherapy, such as pembrolizumab and nivolumab. These antibodies block the interaction between PD-L1 (expressed on cancer cells as a “don’t eat me” signal) and PD-1 (its receptor on T cells), thereby restoring anti-tumor immunity [159].
Beyond oncology, monoclonal antibodies have achieved remarkable success in autoimmune disorders. For instance, Tribuvia (senprutug), developed by Russian scientists Sergey Lukyanov and Dmitry Chudakov in collaboration with BIOCAD, targets the TRBV9+ T-cell receptor of a pathogenic T-cell clone responsible for ankylosing spondylitis (Bekhterev’s disease), leading to its selective elimination. This breakthrough was made possible by years of high-throughput TCR sequencing, which identified the disease-driving clone. This approach could be expanded in the future towards other autoimmune disorders [160,161,162]. Notably, Chudakov has proposed in his interview that similar approaches could be used to eliminate hyperexpanded, pro-inflammatory T-cell clones that accumulate with age, a key driver of inflammaging [163]. If so, antibodies may become a precision tool against immune aging itself.
Antibodies are also highly effective in allergic diseases. A combination of omalizumab (anti-IgE), dupilumab (anti-IL-4Rα), and mepolizumab (anti-IL-5) acts at multiple levels: omalizumab reduces cell-bound IgE, dupilumab blocks Th2 signaling to B cells via the IL4/13-IL-4Rα axis, while mepolizumab inhibits eosinophil and basophil activation via IL-5. Together, they disrupt the allergic cascade more comprehensively than any single agent. However, in clinics dual combinations are more widespread [164,165,166].
In cardiovascular disease (CVD), Repatha (evolocumab), an antibody against PCSK9, enhances hepatic clearance of LDL cholesterol by preventing PCSK9-mediated degradation of LDL receptors, significantly lowering circulating LDL levels [167].
Most recently, Lecanemab (Leqembi) and Donanemab (Kisunla) received FDA approval for Alzheimer’s disease. Both target amyloid-β (Aβ) aggregates in the brain and promote their clearance by microglia. While effective in slowing cognitive decline, they carry risks – most notably amyloid-related imaging abnormalities (ARIA), including brain edema [168,169,170,171].
Overall, monoclonal antibodies excel when the goal is to eliminate a specific pathological cell population, block extracellular signaling, or neutralize a harmful soluble or membrane-bound molecule.
They are highly potent and generally well-tolerated, though limitations remain: (1) targets must be extracellular or cell-surface exposed, (2) manufacturing is complex and costly, (3) administration typically requires injections or infusions, (4) and there’s a risk of anti-drug immune responses [172,173].
Looking ahead, antibodies could be engineered to target: (1) SASP (senescence-associated secretory phenotype) factors, (2) potential pro-aging proteins identified in plasma proteomic studies (e.g., by Tony Wyss-Coray [174], or (3) detrimental inter-tissue signaling pathways that drive systemic aging; (4) pathogenic immune clones, in particular, represent a promising frontier.
Regardless of the specific approach, monoclonal antibodies are already among our most powerful weapons, not just against disease, but against aging itself.

mRNA, Oligonucleotides, and siRNA

mRNA vaccines entered mainstream medicine during the COVID-19 pandemic, thanks to Pfizer–BioNTech’s Comirnaty and Moderna’s Spikevax. They elicit potent and durable immune responses – a breakthrough that earned the 2023 Nobel Prize in Physiology or Medicine [175,176,177]. Beyond infectious disease, this platform is now being applied to cancer immunotherapy, with mRNA vaccines in development for melanoma and non-small cell lung cancer [178,179,180,181].
In the context of aging, tolerogenic vaccines (both protein- and mRNA-based), designed not to activate, but to silence or reprogram the immune system, hold particular promise for autoimmune and chronic inflammatory conditions, which are central to inflammaging [182,183,184].
Oligonucleotides and siRNA represent another powerful class of gene-targeting therapeutics. They work by binding to specific mRNA transcripts, thereby blocking translation or triggering mRNA degradation. Among the most compelling examples are pelacarsen (an antisense oligonucleotide, Novartis), and lepodisiran (an siRNA, Eli Lilly), both currently in Phase 3 clinical trials [185,186,187,188]. They target the LPA gene, whose product, lipoprotein(a), is a major independent genetic risk factor for atherosclerosis and thrombosis. Elevated Lp(a) affects ~20% of the population, yet no effective therapies existed until now, largely because Lp(a) is structurally similar to plasminogen, with predictable consequences [189,190,191].
In brief,If we want to temporarily express a gene (e.g., a pro-longevity factor like FOXO3) without altering the genome, mRNA delivery is a viable option. If we aim to knock down a harmful gene, antisense oligonucleotides or siRNA offer precise, reversible control. The main challenge for both approaches remains efficient intracellular delivery, since these molecules must reach the cytoplasm to function [192].

Genomic and Epigenomic Editors

Genome and epigenome editing are among the most transformative technologies in modern biomedicine. Early tools like zinc finger nucleases (ZFNs) and TALENs used engineered protein domains to recognize and cleave specific DNA sequences. Today, however, CRISPR/Cas systems dominate the field, leveraging guide RNA (gRNA) for programmable targeting [193,194].
Beyond standard CRISPR nucleases, its daughter technologies offer greater precision and safety: base editors fuse a nickase Cas9 (D10A or H840A mutant) to a deaminase enzyme, enabling direct C•G→T•A or A•T→G•C conversions without double-strand breaks [195,196]. Epigenome editors use catalytically dead Cas9 (dCas9) or dCas12 fused to epigenetic effectors (e.g., DNMT3A, TET1, p300), allowing reversible, tunable control of gene expression, which is ideal for modulating aging pathways like FOXO3, SIRT1, or NF-κB [197,198].
While CRISPR’s applications are vast, I focus here on its potential against aging. For blood vessels, key genetic risk factors, such as PCSK9 (which regulates LDL receptor degradation) and LPA (encoding lipoprotein(a), a major independent risk factor for atherosclerosis), can be permanently inactivated, not just transiently suppressed by antibodies or RNAi. Companies are already advancing this vision: Verve Therapeutics is developing VERVE-102, a base editor that introduces a premature stop codon in PCSK9, leading to lifelong LDL reduction [199,200]. Scribe Therapeutics is advancing STX-1200 to knockout LPA [201] and STX-1150 to epigenetically silence PCSK9 [202]. Critically, the liver, the primary target for these therapies, is among the most accessible organs for in vivo gene editing, supporting optimistic forecasts for clinical translation [203].
Despite their promise, all nuclease-based platforms face major hurdles. One is about delivery: only adeno-associated viruses (AAVs) offer reasonable tissue specificity and safety, but their cargo capacity is limited, and pre-existing immunity is common. The blood–brain barrier (BBB) makes CNS targeting especially difficult; even successful delivery risks neuroinflammation or edema. Additional challenges include off-target edits, reduced on-target efficiency (especially in high-fidelity variants), PAM sequence constraints, and immune responses to bacterial Cas proteins [204,205].
A potential paradigm shift may come from de novo protein design. Recently, David Baker’s lab used AI-driven protein folding to generate ultra-compact DNA-binding proteins (~55–60 amino acids vs. 1,368 in SpCas9). These "mini-Cas" analogs are: small enough for non-viral delivery (e.g., LNPs), non-immunogenic (human-like sequence), and highly specific, with tunable binding affinity [206].
In my view, CRISPR/Cas, while revolutionary today, will likely be superseded by this new generation of AI-designed genomic and epigenomic editors. Ultimately, two strategies will be essential in the fight against aging: (1) permanent removal of detrimental alleles (e.g., PCSK9, LPA, APOE4) via precise genome editing, and (2) dynamic, reversible reprogramming of gene networks via epigenome editing – restoring youthful expression patterns without altering the genomic sequence. Together, they form a powerful toolkit for precision geroscience.
We will separately discuss cell therapies and tissue engineering in the next section - probably, some of the most promising approaches, yet in the very beginning of their development.
To summarize, aging is an extremely complex process, and only a multimodal approach, rather than a single therapeutic intervention, can drive a true revolution.

Main Approaches to Fight Aging

Given that we have discussed in the previous section the tools available against age-related dysfunctions, let us now classify them into several logical approaches. While alternative taxonomies exist, we highlight three major strategies: (1) eliminating hostile agents, (2) stimulating the body’s intrinsic rejuvenation capacity, and (3) cell, tissue, and organ replacement.

Eliminating Hostile Agents

When this approach is mentioned, the first targets that come to mind are pre-cancerous/cancerous cells and senescent cells. Leaving oncology to specialists, senescent cells are already primed for death: their survival hinges solely on the hyperexpression of anti-apoptotic proteins, which constitutes their Achilles’ heel.
This insight gave rise to senolytics – compounds designed to selectively eliminate senescent cells while sparing healthy ones. Examples include dasatinib, quercetin, fesitin, and navitoclax. In practice, however, these first-generation agents exhibit significant toxicity: fatigue, gastrointestinal and hematological side effects, and thrombocytopenia, among others. Many scientists rightly criticize these drawbacks, but the approach itself is sound; what deserves scrutiny is the current pharmacological implementation [207,208,209,210].
A ray of hope comes from a recent study by the Rando lab: a combination of ABT-199 (a navitoclax analog with weaker senolytic activity) and birinapant (an anti-cancer drug that mimics the pro-apoptotic protein SMAC) achieved potent senolytic effects without inducing thrombocytopenia [211]. Such rationally designed combinations may pave the way for safer, more effective clearance of senescent cells in aging and chronic disease.
It is also a vital strategy to target specific age-related pathologies driven by senescent cell accumulation, particularly those with a strong inflammatory component – rather than positioning senolytics as broad anti-aging agents. Recently, senolytics had success in treating animal models neurodegenerations, via eliminating senescent-like neurons, astrocytes, and microglia [212,213], while senolytic treatment reduces pain and senescence burden in human intervertebral disc tissue and preclinical models of low back pain [214,215,216]. Senolytics also have high potential in treating lung fibrosis [217,218,219], CVD [220,221], type 2 diabetes [222,223,224], postmenopause bone loss [225], and so on.
Beyond senescent cells, pathogenic immune clones (such as the age-expanded T lymphocytes discussed earlier) also represent valid targets for elimination. In fact, detrimental cell populations have been identified across multiple tissues. For instance, the Deep Dixit group recently described CD38⁺CD169⁻CD11c⁻ age-associated macrophages and dysfunctional adipose ILC2 cells, both pro-inflammatory and tissue-disruptive, making them prime candidates for selective removal [226,227].
Not only cells, but also harmful molecules, can be targeted. Obvious examples include SASP factors that fuel chronic inflammation. A key case in point is HMGB1, a nuclear protein involved in DNA architecture that, when released extracellularly, acts as a damage-associated molecular pattern (DAMP) and induces senescence in neighboring cells. Critically, its activity depends on redox state: the reduced form is potently pro-senescent, whereas the oxidized form is inert. This makes HMGB1 a compelling therapeutic target, either through neutralization or forced oxidation, to disrupt paracrine SASP signaling and halt the spread of senescence [228].
Equally promising are cross-tissue signaling axes, such as the MIF–CD74 pathway uncovered by the Kellis lab: MIF is secreted by fibro-adipogenic progenitors in skeletal muscle, binds to CD74 on myeloid cells in white adipose tissue, and drives adipose inflammation – a process that declines with exercise [229]. Why not pharmacologically block this, and other similarly vicious, circuits?

Stimulating the Body’s Intrinsic Rejuvenation Capacity

Our body possesses more regenerative capacity than it normally uses. Many of the most promising interventions do not invent something entirely new – they reactivate pro-longevity mechanisms already embedded in our biology.
The simplest examples are caloric restriction (CR), rapamycin, and metformin. Our cells can degrade protein aggregates and damaged mitochondria via autophagy, but they rarely operate at full capacity. Physiological challenges (like fasting) or pharmaceuticals can safely enhance this intrinsic cleanup system [230,231]. Similarly, supplementing with NAD⁺ precursors (e.g., NMN), currently in clinical trials, helps restore normal cellular metabolism by boosting sirtuin activity [232,233].
GLP-1 receptor agonists, initially developed for diabetes and obesity, demonstrate broad geroprotective effects by suppressing chronic inflammation, oxidative stress, and cellular senescence – core hallmarks of aging [234]. Preclinical studies show they reverse brain aging signatures and reduce Alzheimer’s-related gene expression [235], while clinical trials are now evaluating their impact on physical function and molecular aging biomarkers in older adults [236]. Their multimodal action across metabolic, cardiovascular, and neurodegenerative pathways positions GLP-1 agonists as leading candidates for healthspan extension [237,238].
The same principle applies to DNA repair, which can be enhanced by lifestyle factors (e.g., high-quality sleep) or pharmacological agents, thereby reducing senescence signaling and cancer risk.
Exercise is another well-established rejuvenating intervention. In skeletal muscle, for instance, it acts through the SPP1–CD44 axis, which modulates the stem cell niche and promotes regeneration, as recently shown by the Rando and Brunet labs [239]. Could we pharmacologically mimic this pathway (specifically in muscle, not systemically) to treat sarcopenia?
One of the most actively debated strategies is transient expression of Yamanaka factors: the cocktail of Oct4, Sox2, c-Myc, and Klf4 (OSKM). Despite concerns about carcinogenicity, recent studies from the Belmonte group demonstrate that short-term, cyclic induction is generally safe, even with long-term use in mice [240]. Even safer is the OSK combination (without c-Myc), which retains efficacy while minimizing tumorigenic risk. Controlled OSK exposure restores chromatin integrity, activates tissue-resident stem cells (e.g., in muscle), and, counterintuitively, triggers apoptosis in cancer cells: once their epigenome is reset, they recognize irreparable genomic damage and self-destruct, according to recent David Sinclair’s and Tao Cheng’s research [241,242,243]. Since OSK delivery currently requires gene therapy (a major hurdle), the Sinclair lab is developing small molecules that mimic OSK activity [244]. While this approach remains high-risk and demands precise control, we believe it holds strong potential for partial, targeted rejuvenation, once proven safe in humans.
Another controversial but intriguing avenue is the “young blood” effect. Heterochronic parabiosis experiments show that joining the circulatory systems of young and old mice partially rejuvenates the old while accelerating aging in the young (compared to isochronic pairs) [245,246]. This inspired practices like young plasma transfusions (e.g., by Bryan Johnson), widely criticized as unscientific and potentially harmful (transfusion itself is a physiological stressor). Moreover, no significant results were achieved in clinical trials by the commercial company Ambrosia [247], which were widely criticized for flawed methodology. This was followed by a strong 2019 FDA warning against such unproven "young blood" practices, leading Ambrosia to cease treatments [248,249]. Furthermore, a trial involving young fresh frozen plasma infusions for Alzheimer's patients showed no significant improvement in cognition [250,251]. On the other hand, a separate study exploring umbilical cord plasma concentrate in elderly individuals showed potential in affecting some biomarkers related to aging, though the researchers emphasized that these results are highly preliminary [252]. And despite all this, the underlying idea may hold merit: what specific factors in young blood drive rejuvenation? If we identify these proteins, metabolites, or extracellular vesicles, they could become components of a rational, cell-free rejuvenation therapy.
In general, the first two strategies – elimination of damage and activation of repair – are deeply intertwined. Take microglia: in response to beta-amyloid, they can launch either a damaging inflammatory program or an adaptive, phagocytic one (driven by GPNMB and lysosomal clearance) [253,254,255]. If we block the harmful pathway, will cells default to the protective one? If we remove chronic stressors like senescent cells, will the body’s innate healing capacity rebound more fully? Likely, yes. And if we carefully combine both strategies, we may shift human healthspan toward its biological ceiling of 120–150 years [256].
But what if we want more?

Cell, Tissue, and Organ Replacement

Cell, tissue, and organ replacement holds near-unlimited potential for extending both healthspan and lifespan, potentially enabling biological immortality. The main limitation is that most approaches remain preclinical or early clinical, with only a few validated applications.
Probably the most widely used cell therapy today is CAR-T cells, which fuse an antibody-derived targeting domain with T-cell receptor signaling components to selectively eliminate tumor cells [257,258]. Beyond oncology, CAR-T and TCR-T cells (T cells engineered with antigen-specific receptors) are now being explored for autoimmune disorders [259]. Importantly, it is not only conventional CD8⁺ cytotoxic and CD4⁺ helper T cells that can be engineered: CAR/TCR-Tregs (regulatory T cells) can be designed to induce antigen-specific immune tolerance [260,261]. Such cells could potentially suppress autoimmune-like processes in aging, including chronic inflammation driven by self-reactive clones.
Equally promising is immune system replacement. As discussed above, aging drastically reduces naïve T-cell production, partly due to age-associated clonal expansions and inflammaging, and partly due to thymic involution [103,262]. Recent studies show that FOXN1 overexpression (in mice) [263,264] or IL-22 administration can partially restore thymic architecture and naïve T-cell output [265]; bioengineered thymic organoids are also emerging [266]. Replacement is not limited to adaptive immunity: innate immune cells, such as ILC2s, can also be replenished [227]. Critically, this should be coupled with the clearance of old, senescent, and chronically inflamed immune cells.
What is already used in clinics is skin and corneal epithelia – autologous or allogeneic sheets derived from stem or progenitor cells. These are standard care for severe burns, chronic ulcers, and corneal injuries, with decades of safety data [267,268,269,270]. In the context of aging, they hold clear potential for age-impaired wound healing, where delayed re-epithelialization, reduced growth factor secretion, and chronic inflammation impede recovery [271].
Also in high demand are cartilage and bone grafts. Engineered cartilage, often from chondrocytes or MSCs seeded on biodegradable scaffolds, is used for nasal and auricular reconstruction and is being tested for focal osteoarthritic lesions [272,273]. Bone substitutes, including 3D-printed ceramic or collagen-based matrices loaded with osteoprogenitors, are increasingly deployed in orthopedics and dentistry to treat osteoporotic fractures and non-unions [274,275]. Both tissues are highly relevant to aging: cartilage loss drives osteoarthritis, while age-related declines in osteoblast function and matrix mineralization underlie fragility fractures [276,277].
One of the most encouraging success stories is currently unfolding in diabetes therapy. Clinical trials have demonstrated that stem cell–derived islets (SC-islets) – pancreatic β-like cells differentiated from human pluripotent stem cells – can restore glucose-responsive insulin secretion in both type 1 [278,279,280] and advanced type 2 diabetes [281,282]. In multiple studies, patients achieved insulin independence for over a year, with near-normal HbA1c and time-in-range metrics [283,284]. Both autologous (patient-specific iPSC-derived) and allogeneic approaches are being explored, but the latter are considered more scalable. To overcome the need for systemic immunosuppression, scientists are developing several advanced strategies. These include the use of encapsulation devices (e.g., ViaCyte’s PEC-Direct/Encap, now under Vertex) to protect grafts [285,286]. Another major effort involves engineering hypoimmunogenic SC-islets, for example by knocking out B2M (to eliminate HLA class I) and CIITA (to suppress HLA class II), which have shown promise in preclinical models for evading T-cell and NK-cell rejection [287,288,289,290]. This represents one of the clearest proofs that chronic metabolic disease can be eradicated, not just managed, by fully restoring endogenous insulin regulation.
Very promising is neuronal replacement for neurodegenerative conditions – to my view, perhaps the only way to truly restore function in a dying brain. Antibody-based therapies (e.g., lecanemab, donanemab) and emerging anti-neuroinflammatory strategies slow decline but do not halt or reverse neuronal loss [291]. In contrast, iPSC-derived dopaminergic neurons are already in clinical trials for Parkinson’s disease (e.g., by BlueRock Therapeutics and Kyoto University), with grafts showing survival, integration, and motor improvement at 2 years [292,293,294]. For Alzheimer’s, cortical and cholinergic neurons derived from iPSCs are in preclinical development, aiming to replace lost circuitry in the entorhinal cortex and basal forebrain [295]. Unlike symptomatic approaches, neuronal replacement targets the core pathology: irreversible cell loss.
Very promising, minimally invasive, and still underutilized is microbiome replacement. Fecal microbiota transplantation (FMT) not only cures recurrent C. difficile infection but also reverses age-associated dysbiosis in preclinical models, restoring SCFA production, gut barrier integrity, and systemic immune tone [296]. Engineered consortia (e.g., VE303 by Vedanta Biosciences) containing defined Clostridia strains induce colonic Tregs and suppress inflammaging [297,298,299]. In aged mice, FMT from young donors improves cognitive performance, reduces microglial activation, and extends healthspan – effects linked to bile acid and tryptophan metabolite shifts [300,301]. Human trials are now exploring precision microbiota therapeutics for sarcopenia and frailty treatment [302].
Conceptually intriguing work has emerged from China, where the Liu Guang-Hui lab, in collaboration with Belmonte, leveraged extensive single-cell profiling of aged cynomolgus monkeys (Macaca fascicularis) to pinpoint FOXO3 as a central anti-aging regulator. They engineered mesenchymal stem cells (MSCs) carrying a gain-of-function FOXO3 mutation, which, upon transplantation, induced robust systemic rejuvenation with no significant adverse effects. Wild-type MSCs provided modest benefit, but far less pronounced. The effect was largely mediated by exosome-dependent signaling, not direct engraftment [303].
What’s next? The main bottleneck in engineering complex organs (such as the heart, lungs, or kidneys) is achieving functional vascularization and innervation. In large organoids, cells in the core rapidly undergo necrosis due to hypoxia and poor metabolite exchange [304,305]. Additional hurdles include the trade-off between autologous approaches (patient-derived iPSCs: low immunogenicity but high cost and complex QC) and allogeneic hypoimmunogenic iPSC platforms (e.g., with B2M/CIITA knockout), which are now entering clinical development [306].
Once these barriers are overcome, and we can reliably replace aged or failing organs, we may finally confront the ultimate question: how long can a fully repaired human live?

But What If We are to Act Right Now?

I will end this section with a sobering reality.
Some individuals are, or soon will be, facing imminent death (e.g., from terminal cancer), and one more technology, albeit controversial and unproven, offers a speculative lifeline: cryonics. In this procedure, the body (or brain) is preserved at –196°C in liquid nitrogen after perfusion with cryoprotective agents designed to minimize ice crystal formation and reduce cellular damage [307].
The good news: vitrification (ice-free glass-like solidification) has enabled successful recovery of functionality in small tissues and organisms, including nematodes (Caenorhabditis elegans), mammalian retinas, and even whole pig kidneys, which were later transplanted and functioned ex vivo [308,309,310,311].
The bad news: no complex mammal, let alone a human, has ever been revived after cryopreservation. Current protocols typically begin hours after legal death, during which ischemic damage and imperfect CPA perfusion may already compromise neural integrity [312].
In my view, no known law of physics forbids future revival, if sufficient structural information (especially in the brain’s connectome) is preserved. The real question is whether today’s cryonics protocols preserve enough fine-scale neural architecture to retain identity and memory. To answer this, we need systematic, high-resolution comparisons of brain ultrastructure before and after vitrification – ideally using connectomics [313].
Encouragingly, the FlyWire Consortium and Google recently mapped the full synaptic-resolution connectome of the adult Drosophila melanogaster brain, a milestone that demonstrates our growing capacity to image, store, and analyze neural wiring at scale [314]. A lot of progress is needed to move from the fruit fly brain to the human one, but this is the first step.
While cryonics remains a high-risk, long-shot bet, it may one day serve as a bridge to future rejuvenation technologies, if we can prove that “you” are still there in the frozen brain.

Aging Clocks

How can we quantify aging to assess how biologically “young” or “old” an individual is, irrespective of chronological age, or to measure the efficacy of anti-aging therapies? For this, multiple aging clocks have been developed. They estimate biological age as an integrative metric of organismal state: if it exceeds chronological age, the individual is at higher risk of serious illness or death; if it is lower, the opposite holds true. Today, aging clocks exist for numerous tissues and species, working with data ranging from clinical blood tests to single-cell RNA-seq.
The era of aging clocks began with Steve Horvath, who in 2013 introduced the first multi-tissue DNA methylation clock. Like most clocks today, it uses an elastic net regression model based on methylation levels at 353 CpG sites [315]. This was followed by other first-generation clocks such as Hannum (71 CpGs) [316] and Zhang (514 CpGs) – one of the most accurate models for predicting chronological age to date [317].
However, these first-generation models face the “biomarker paradox”: the more precisely they predict chronological age, the less they reflect health status or disease risk. Many CpG sites in these clocks correlate strongly with age but weakly with pathology. Returning to the stochasticity question, clocks like Horvath’s and Zhang’s are largely driven by stochastically drifting CpGs, unlike second-generation clocks such as PhenoAge [39,318].
Second-generation clocks answer not “How old am I?” but “How much time do I have until death, cancer, or a cardiovascular event?” Many CpGs in these models show modest age correlation but strong association with clinical outcomes. According to the ComputAgeBench study by the Ekaterina Khrameeva’s group, PhenoAgeV2 [319] is currently the strongest predictor, followed by GrimAgeV1 [320], PhenoAgeV1 [318], and GrimAgeV2 [321,322].
Moreover, the Mammalian Methylation Consortium has recently developed pan-tissue, pan-mammalian epigenetic clocks applicable to 185 species. These clocks are built on evolutionarily conserved CpG sites, highly enriched in polycomb-repressed developmental loci, that form the molecular backbone of cross-species aging biomarkers and underpin the AROCM framework [323].
Not all clocks rely on DNA methylation. In fact, they can be built from any biomedical data modality. The Wyss-Coray group recently developed plasma proteome clocks, capturing both systemic aging signals and organ-specific proteins from 11 major organ systems [174]. The Kennedy and Gruber groups created DoliClock, a lipidome-based brain aging clock [324], as well as PCAge and LinAge2, designed for routine clinical data [325,326]. The Anne Brunet lab built cell type–specific clocks for the brain: separate models for neuroblasts, astrocytes, oligodendrocytes, and microglia [327]. And the Vadim Gladyshev lab recently introduced ScAge, an aging clock for single-cell data [328].
We must understand that aging clocks are not just diagnostic tools or risk predictors – they can reveal causal drivers of aging. Genes, proteins, or metabolites that strongly increase predicted age may themselves be active contributors to aging and thus valid therapeutic targets. This insight underpins Insilico Medicine’s Precious1GPT, Precious2GPT, and Precious3GPT clock models, which use transformer-based models to identify such targets [329,330,331].
Finally, we must remember that not all age-related changes are harmful. Some are adaptive responses to damage, and blocking them could worsen outcomes. To distinguish between the two, Gladyshev and Horvath recently developed DamAge (measuring detrimental alterations) and AdaptAge (measuring protective or compensatory changes) – a crucial step toward precision geroscience [332].

The Power of AI in Aging Research and Drug Development

To comprehend and ultimately defeat a complex process like biological aging, we don’t need to be alchemists, but our science must be data-driven. Fortunately, thanks to trends like Moore’s Law, more data is generated every year. AI tools can now ingest numerous datasets and read millions of scientific papers to uncover patterns invisible to humans, and thus drive real breakthroughs.
One of the brightest examples of AI in biopharmaceutical innovation is Insilico Medicine, led by Alex Zhavoronkov. They’ve built a complete end-to-end pipeline that has already yielded a strong portfolio of novel small molecules. In brief, PandaOmics identifies therapeutic targets by integrating multi-omics and biomedical literature [333], Chemistry42 designs small-molecule inhibitors and predicts their properties (e.g., target binding, toxicity) [334], and inClinico forecasts clinical trial outcomes to avoid costly failures [335].
Equally transformative is the work of the Manolis Kellis lab, which harnesses machine learning to comprehend the regulation of the human genome in aging and disease. Just some examples of their work includes Hydra, an interpretable deep generative model for robust annotation of single-cell omics data [336]; scCLIP, a multimodal contrastive learning framework that aligns chromatin accessibility and gene expression across tissues [337]; SPATIA, a spatially aware foundation model that unifies cellular morphology, transcriptomics, and tissue architecture [338]; Procyon, a multimodal protein language model that predicts functional phenotypes across the proteome [339]; and Epilogos, an information-theoretic browser for navigating epigenomic landscapes across thousands of samples [340].
Such AI solutions will be transformative across virtually every area of research and development. They are not replacements for scientists – they are force multipliers, accelerating discovery and enabling us to tackle the systemic complexity of aging with unprecedented precision.

AI Tools as Assistants in Personalized Medicine

AI tools must also be integrated into clinical medicine, and for compelling reasons [341].
First, a typical doctor–patient consultation lasts only 15 to 30 minutes – far too little time to thoroughly assess an individual’s full medical history, lifestyle, and risk profile. In some settings, such as pre-shift medical checks for miners, it can be even shorter [342,343].
Second, no single physician or scientist can keep up with the entire body of biomedical literature, especially when dealing with rare or complex cases. An AI agent, however, can instantly access and synthesize millions of publications, ensuring no relevant evidence is overlooked [344,345].
Third, computer vision algorithms can outperform humans in specific diagnostic tasks, serving as powerful assistants that reduce medical error. A prime example is the use of AI in dermatology, where algorithms now routinely distinguish benign nevi from melanomas with high accuracy [346,347]. Another one is IRA Labs, a Skolkovo-based startup that analyzes CT scans to detect pathologies, automating routine radiological workflows with precision [348].
AI also excels at interpreting genomic data. By analyzing a patient’s sequencing results, it can highlight clinically relevant variants and guide decision-making – a major step toward precision medicine. In some cases, AI even helps stratify patients for targeted therapies. For instance, Neuron23, in collaboration with QIAGEN, developed NEU-411, a small-molecule inhibitor of LRRK2, a kinase linked to vesicle trafficking, inflammation, and increased Parkinson’s disease risk in certain genetic variants. Since NEU-411 is effective only against specific LRRK2 forms, an ML-based tool is used to identify likely responders based on the patient’s LRRK2 gene sequence [349,350].
Moreover, omics-based diagnostics are enabling ultra-early disease detection. For example, aberrant methylation of the SEPT9 gene is already used as a blood-based biomarker for colorectal cancer screening [351,352]. In neurodegeneration, although not “pure” omics, companies like ToxGenSolutions and DiamiR, along with the NeuroMiR project, are developing miRNA-based panels that can detect Alzheimer’s disease 5–6 years before symptom onset – far earlier than current blood tests for Aβ40/42 (which offer only 1–2 years of lead time) [353,354,355,356,357]. As such tools enter clinical practice, AI will become indispensable for interpreting their complex outputs.
Finally, AI agents can serve as personal health companions, acting as psychotherapists (in high demand today), nutritionists, fitness coaches, and even life mentors, all tailored to an individual’s biology and behavior [358].
The future belongs to integrated health ecosystems that fuse scientific, genomic, clinical, and psychological data, empowering both doctors and patients to understand, prevent, and ultimately cure disease. Some great examples of such complex ecosystems include Tempus [359], Luria Health [360], and numerous other platforms.

Conclusions

In 2025, we are witnessing a tectonic shift in how society perceives aging, and not as an immutable fate, but as a modifiable biological process. Yet biogerontology remains marginalized, even ridiculed, by parts of the scientific establishment. This is not merely due to inertia or dogma; it is also a consequence of overhyped claims that outpace evidence. Premature promises of “curing aging tomorrow” erode public trust – a tragedy for those of us committed to rigorous, data-driven science.
The pragmatic path forward begins not with abstract longevity fantasies, but with concrete, high-burden chronic diseases, particularly those rooted in inflammaging and fibrosis: atherosclerosis, type 2 diabetes, Alzheimer’s disease, and autoimmune conditions. We must move beyond the palliative mindset of “managing symptoms” and instead aim to restore tissues to a functional, youthful reference state, whether we call it rejuvenation, repair, or simply cure.
Critically, the same interventions can serve dual roles: prevention and reversal. Caloric restriction and exercise lay the foundation; next-generation tools: senolytics, metformin, GLP-1 agonists, and emerging gene and cell therapies like anti-LPA editing or FOXO3-augmented MSCs – offer far greater precision. But their true power emerges only when we reframe our intent: not just “treating disease X,” but “delaying systemic aging” or “rejuvenating organ system Y.” This conceptual shift is essential: it aligns clinical practice with geroscience and unlocks regulatory, funding, and public support pathways.
As AI and machine learning mature, they will increasingly guide this effort: identifying causal drivers of aging, predicting individual responses to interventions, and integrating multi-omics data into personalized health strategies. The future belongs not to miracle cures, but to precision restoration, grounded in biology and scaled by technology.
If our generations commit to this mission enough, not with hype, but with humility, rigor, and vision, we may not only extend healthspan for future generations, but ourselves live as long as biology and the laws of physics allow.

References

  1. "A Brief History of Vaccination." World Health Organization, 2023. Accessed October 27, 2025. https://www.who.int/news-room/spotlight/history-of-vaccination/a-brief-history-of-vaccination#:~:text=A%20plasma-derived%20inactivated%20vaccine,Smallpox%20Eradication%2C%20declares%20smallpox%20eradicated.
  2. World Health Organization. "Milestones: 75 Years of Improving Public Health." World Health Organization. Accessed October 27, 2025. https://www.who.int/campaigns/75-years-of-improving-public-health/milestones#year-1988.
  3. Li, Yumeng, Xutong Tian, Juyue Luo, Tongtong Bao, Shujin Wang, and Xin Wu. "Molecular mechanisms of aging and anti-aging strategies." Cell Communication and Signaling 22, no. 1 (2024): 285. [CrossRef]
  4. Kennedy, Brian K., Shelley L. Berger, Anne Brunet, Judith Campisi, Ana Maria Cuervo, Elissa S. Epel, Claudio Franceschi et al. "Aging: a common driver of chronic diseases and a target for novel interventions." Cell 159, no. 4 (2014): 709.
  5. Brunner-La Rocca, Hans-Peter, Lutz Fleischhacker, Olga Golubnitschaja, Frank Heemskerk, Thomas Helms, Thom Hoedemakers, Sandra Huygen Allianses et al. "Challenges in personalised management of chronic diseases—heart failure as prominent example to advance the care process." EPMA Journal 7, no. 1 (2016): 2. [CrossRef]
  6. Thomas, Shane A., Colette J. Browning, Fadi J. Charchar, Britt Klein, Marcia G. Ory, Henrietta Bowden-Jones, and Samuel R. Chamberlain. "Transforming global approaches to chronic disease prevention and management across the lifespan: integrating genomics, behavior change, and digital health solutions." Frontiers in public health 11 (2023): 1248254. [CrossRef]
  7. Jacques, Macsue, Kirsten Seale, Sarah Voisin, Anna Lysenko, Robin Grolaux, Bernadette Jones-Freeman, Severine Lamon et al. "DNA Methylation Ageing Atlas Across 17 Human Tissues." bioRxiv (2025): 2025-07.
  8. Alsuhibani, Azzam, Mark Shevlin, and Richard P. Bentall. "Atheism is not the absence of religion: Development of the monotheist and atheist belief scales and associations with death anxiety and analytic thinking." Psychology of Religion and Spirituality 14, no. 3 (2022): 362. [CrossRef]
  9. Routledge, Clay, Andrew A. Abeyta, and Christina Roylance. "Death and end times: The effects of religious fundamentalism and mortality salience on apocalyptic beliefs." Religion, brain & behavior 8, no. 1 (2018): 21-30. [CrossRef]
  10. Ai, Amy L. "Daoist spirituality and philosophy: Implications for holistic health, aging, and longevity." Holistic approaches to health aging: Complementary and alternative medicine for older adults. New York, NY: Springer (2006): 149-160.
  11. Chen, Jie, Meizhen Zhao, Renyi Zhou, Wenjing Ou, and Pin Yao. "How heavy is the medical expense burden among the older adults and what are the contributing factors? A literature review and problem-based analysis." Frontiers in public health 11 (2023): 1165381. [CrossRef]
  12. Bodnár, Katalin, Carolin Nerlich, Alexander Al-Haschimi, and Ana Isabel Lima. "The macroeconomic and fiscal impact of population ageing." ECB Occasional Paper Series, no. 296 (June 2022).
  13. Xia, Haishuo, Yongqing Hou, Qing Li, and Antao Chen. "A meta-analysis of cognitive flexibility in aging: Perspective from functional network and lateralization." Human brain mapping 45, no. 14 (2024): e70031. [CrossRef]
  14. Mendoza-Núñez, Víctor Manuel, and Ana Belén Mendoza-Soto. "Is aging a disease? A critical review within the framework of ageism." Cureus 16, no. 2 (2024).
  15. Lyu, Yu-Xuan, Qiang Fu, Dominika Wilczok, Kejun Ying, Aaron King, Adam Antebi, Aleksandar Vojta et al. "Longevity biotechnology: bridging AI, biomarkers, geroscience and clinical applications for healthy longevity." Aging (Albany NY) 16, no. 20 (2024): 12955.
  16. López-Otín, Carlos, Maria A. Blasco, Linda Partridge, Manuel Serrano, and Guido Kroemer. "Hallmarks of aging: An expanding universe." Cell 186, no. 2 (2023): 243-278.
  17. Vijg, Jan, and Xiao Dong. "Pathogenic mechanisms of somatic mutation and genome mosaicism in aging." Cell 182, no. 1 (2020): 12-23.
  18. Aird, Katherine M., and Rugang Zhang. "Detection of senescence-associated heterochromatin foci (SAHF)." In Cell Senescence: Methods and Protocols, pp. 185-196. Totowa, NJ: Humana Press, 2012.
  19. la Torre, Annamaria, Filomena Lo Vecchio, and Antonio Greco. "Epigenetic mechanisms of aging and aging-associated diseases." Cells 12, no. 8 (2023): 1163.
  20. Yu, Chenglong, Ee Ming Wong, Jihoon Eric Joo, Allison M. Hodge, Enes Makalic, Daniel Schmidt, Daniel D. Buchanan et al. "Epigenetic drift association with cancer risk and survival, and modification by sex." Cancers 13, no. 8 (2021): 1881. [CrossRef]
  21. Birch, Helen L. "Extracellular matrix and ageing." Biochemistry and cell biology of ageing: part I biomedical science (2019): 169-190.
  22. Statzer, Cyril, Ji Young Cecilia Park, and Collin Y. Ewald. "Extracellular matrix dynamics as an emerging yet understudied hallmark of aging and longevity." Aging and Disease 14, no. 3 (2023): 670. [CrossRef]
  23. Zhang, Weiqi, Shu Zhang, Pengze Yan, Jie Ren, Moshi Song, Jingyi Li, Jinghui Lei et al. "A single-cell transcriptomic landscape of primate arterial aging." Nature communications 11, no. 1 (2020): 2202. [CrossRef]
  24. Sun, Guoqiang, Kuan Li, Jiale Ping, Liyun Zhao, Chao Cui, Junping Wu, Lixin Xie et al. "A single-cell transcriptomic atlas reveals senescence and inflammation in the post-tuberculosis human lung." Nature Microbiology (2025): 1-19.
  25. Jing, Ying, Yuesheng Zuo, Yang Yu, Liang Sun, Zhengrong Yu, Shuai Ma, Qian Zhao et al. "Single-nucleus profiling unveils a geroprotective role of the FOXO3 in primate skeletal muscle aging." Protein & Cell 14, no. 7 (2023): 499-514.
  26. Van Meter, Michael, Mehr Kashyap, Sarallah Rezazadeh, Anthony J. Geneva, Timothy D. Morello, Andrei Seluanov, and Vera Gorbunova. "SIRT6 represses LINE1 retrotransposons by ribosylating KAP1 but this repression fails with stress and age." Nature communications 5, no. 1 (2014): 5011.
  27. Roichman, A., S. Elhanati, M. A. Aon, I. Abramovich, A. Di Francesco, Y. Shahar, M. Y. Avivi et al. "Restoration of energy homeostasis by SIRT6 extends healthy lifespan." Nature communications 12, no. 1 (2021): 3208.
  28. Tian, Xiao, Denis Firsanov, Zhihui Zhang, Yang Cheng, Lingfeng Luo, Gregory Tombline, Ruiyue Tan et al. "SIRT6 is responsible for more efficient DNA double-strand break repair in long-lived species." Cell 177, no. 3 (2019): 622-638.
  29. Simon, Matthew, Jiping Yang, Jonathan Gigas, Eric J. Earley, Eric Hillpot, Lei Zhang, Maria Zagorulya et al. "A rare human centenarian variant of SIRT6 enhances genome stability and interaction with Lamin A." The EMBO Journal 42, no. 3 (2023): e113326.
  30. Yang, Jiping, HyeRim Han, Xifan Wang, Yizhou Zhu, Haiqi Chen, and Yousin Suh. "Rare centenarian SIRT6 coding variants elevate SIRT6 protein levels and resist cellular senescence." bioRxiv (2025): 2025-09.
  31. Kim, Ji-Hee, Kyu-Hee Hwang, Kyu-Sang Park, In Deok Kong, and Seung-Kuy Cha. "Biological role of anti-aging protein Klotho." Journal of lifestyle medicine 5, no. 1 (2015): 1. [CrossRef]
  32. Bi, Xianjin, Ke Yang, Bo Zhang, and Jinghong Zhao. "The protective role of klotho in CKD-associated cardiovascular disease." Kidney Diseases 6, no. 6 (2020): 395-406. [CrossRef]
  33. Zhou, Lili, Yingjian Li, Dong Zhou, Roderick J. Tan, and Youhua Liu. "Loss of Klotho contributes to kidney injury by derepression of Wnt/β-catenin signaling." Journal of the American Society of Nephrology 24, no. 5 (2013): 771-785. [CrossRef]
  34. Tauc, Helen M., Imilce A. Rodriguez-Fernandez, Jason A. Hackney, Michal Pawlak, Tal Ronnen Oron, Jerome Korzelius, Hagar F. Moussa et al. "Age-related changes in polycomb gene regulation disrupt lineage fidelity in intestinal stem cells." Elife 10 (2021): e62250.
  35. Blackledge, Neil P., and Robert J. Klose. "The molecular principles of gene regulation by Polycomb repressive complexes." Nature reviews Molecular cell biology 22, no. 12 (2021): 815-833.
  36. Li, Yan, Yanxiang Mo, Chen Chen, Jin He, and Zhiheng Guo. "Research advances of polycomb group proteins in regulating mammalian development." Frontiers in Cell and Developmental Biology 12 (2024): 1383200.
  37. Much, Christian, Sandy M. Rajkumar, Liming Chen, John M. Cohen, Aravind R. Gade, Geoffrey S. Pitt, and Yicheng Long. "Macromolecular interactions dictate Polycomb-mediated epigenetic repression." bioRxiv (2025): 2025-05.
  38. Tarkhov, Andrei E., Thomas Lindstrom-Vautrin, Sirui Zhang, Kejun Ying, Mahdi Moqri, Bohan Zhang, Alexander Tyshkovskiy, Orr Levy, and Vadim N. Gladyshev. "Nature of epigenetic aging from a single-cell perspective." Nature Aging 4, no. 6 (2024): 854-870.
  39. Tong, Huige, Varun B. Dwaraka, Qingwen Chen, Qi Luo, Jessica A. Lasky-Su, Ryan Smith, and Andrew E. Teschendorff. "Quantifying the stochastic component of epigenetic aging." Nature Aging 4, no. 6 (2024): 886-901.
  40. Horvath, Steve, Joshua Zhang, Amin Haghani, Ake T. Lu, and Zhe Fei. "Fundamental equations linking methylation dynamics to maximum lifespan in mammals." Nature Communications 15, no. 1 (2024): 8093. [CrossRef]
  41. Abegglen, Lisa M., Aleah F. Caulin, Ashley Chan, Kristy Lee, Rosann Robinson, Michael S. Campbell, Wendy K. Kiso et al. "Potential mechanisms for cancer resistance in elephants and comparative cellular response to DNA damage in humans." Jama 314, no. 17 (2015): 1850-1860. [CrossRef]
  42. Sulak, Michael, Lindsey Fong, Katelyn Mika, Sravanthi Chigurupati, Lisa Yon, Nigel P. Mongan, Richard D. Emes, and Vincent J. Lynch. "TP53 copy number expansion is associated with the evolution of increased body size and an enhanced DNA damage response in elephants." elife 5 (2016): e11994. [CrossRef]
  43. Liu, Xing, Fei Yang, Yi Li, Zhen-Peng Yu, Xin Huang, Lin-Xia Sun, Wen-Hua Ren, Guang Yang, and Shi-Xia Xu. "Evolution of p53 pathway-related genes provides insights into anticancer mechanisms of natural longevity in cetaceans." Zoological Research 44, no. 5 (2023): 947. [CrossRef]
  44. Firsanov, Denis, Max Zacher, Xiao Tian, Todd L. Sformo, Yang Zhao, Greg Tombline, J. Yuyang Lu et al. "DNA repair and anti-cancer mechanisms in the long-lived bowhead whale." BioRxiv (2024): 2023-05.
  45. Tian, Xiao, Jorge Azpurua, Christopher Hine, Amita Vaidya, Max Myakishev-Rempel, Julia Ablaeva, Zhiyong Mao, Eviatar Nevo, Vera Gorbunova, and Andrei Seluanov. "High-molecular-mass hyaluronan mediates the cancer resistance of the naked mole rat." Nature 499, no. 7458 (2013): 346-349.
  46. Zhang, Zhihui, Xiao Tian, J. Yuyang Lu, Kathryn Boit, Julia Ablaeva, Frances Tolibzoda Zakusilo, Stephan Emmrich et al. "Increased hyaluronan by naked mole-rat Has2 improves healthspan in mice." Nature 621, no. 7977 (2023): 196-205.
  47. Chen, Yu, Zhixi Chen, Hao Wang, Zhen Cui, Kai-Le Li, Zhiwei Song, Lingjiang Chen et al. "A cGAS-mediated mechanism in naked mole-rats potentiates DNA repair and delays aging." Science 390, no. 6769 (2025): eadp5056.
  48. Izraelson, M., M. Metsger, A. N. Davydov, I. A. Shagina, M. A. Dronina, A. S. Obraztsova, D. A. Miskevich et al. "Distinct organization of adaptive immunity in the long-lived rodent Spalax galili." Nature Aging 1, no. 2 (2021): 179-189.
  49. Castiglione, Gianni M., Zhenhua Xu, Lingli Zhou, and Elia J. Duh. "Adaptation of the master antioxidant response connects metabolism, lifespan and feather development pathways in birds." Nature Communications 11, no. 1 (2020): 2476.
  50. Skulachev, Vladimir P., Susanne Holtze, Mikhail Y. Vyssokikh, Lora E. Bakeeva, Maxim V. Skulachev, Alexander V. Markov, Thomas B. Hildebrandt, and Viktor A. Sadovnichii. "Neoteny, prolongation of youth: from naked mole rats to “naked apes”(humans)." Physiological reviews 97, no. 2 (2017): 699-720.
  51. Lazcano, I., A. Olvera, S. M. Pech-Pool, L. Sachs, N. Buisine, and A. Orozco. "Differential effects of 3, 5-T2 and T3 on the gill regeneration and metamorphosis of the Ambystoma mexicanum (axolotl)." Frontiers in Endocrinology 14 (2023): 1208182. [CrossRef]
  52. Haluza, Yuliia, Joseph A. Zoller, Ake T. Lu, Hannah E. Walters, Martina Lachnit, Robert Lowe, Amin Haghani et al. "Axolotl epigenetic clocks offer insights into the nature of negligible senescence." BioRxiv (2024): 2024-09.
  53. Yun, Maximina H. "Salamander insights into ageing and rejuvenation." Frontiers in cell and developmental biology 9 (2021): 689062. [CrossRef]
  54. Popov, N. A., and V. P. Skulachev. "Neotenic Traits in Heterocephalus glaber and Homo sapiens." Biochemistry (Moscow) 84, no. 12 (2019): 1484-1489.
  55. Munro, Daniel, Cécile Baldy, Matthew E. Pamenter, and Jason R. Treberg. "The exceptional longevity of the naked mole-rat may be explained by mitochondrial antioxidant defenses." Aging Cell 18, no. 3 (2019): e12916.
  56. Somel, Mehmet, Lin Tang, and Philipp Khaitovich. "The role of neoteny in human evolution: from genes to the phenotype." In Post-genome biology of primates, pp. 23-41. Tokyo: Springer Tokyo, 2011.
  57. Yordy, Jennifer, Cornelia Kraus, Jessica J. Hayward, Michelle E. White, Laura M. Shannon, Kate E. Creevy, Daniel EL Promislow, and Adam R. Boyko. "Body size, inbreeding, and lifespan in domestic dogs." Conservation genetics 21, no. 1 (2020): 137-148.
  58. Mata, Fernando, and Andreia Mata. "Investigating the relationship between inbreeding and life expectancy in dogs: mongrels live longer than pure breeds." PeerJ 11 (2023): e15718.
  59. Platzer, Matthias, and Christoph Englert. "Nothobranchius furzeri: a model for aging research and more." Trends in Genetics 32, no. 9 (2016): 543-552.
  60. Cui, Rongfeng, David Willemsen, and Dario Riccardo Valenzano. "Nothobranchius furzeri (African turquoise killifish)." Trends in Genetics 36, no. 7 (2020): 540-541. [CrossRef]
  61. Bulavkina, Elizaveta V., Alexander A. Kudryavtsev, Margarita A. Goncharova, Margarita S. Lantsova, Anastasija I. Shuvalova, Maxim A. Kovalev, and Anna V. Kudryavtseva. "Multifaceted Nothobranchius." Biochemistry (Moscow) 87, no. 12 (2022): 1563-1578. [CrossRef]
  62. Shreeya, Tejal, Mohd Saifullah Ansari, Prabhat Kumar, Muskan Saifi, Ali A. Shati, Mohammad Y. Alfaifi, and Serag Eldin I. Elbehairi. "Senescence: A DNA damage response and its role in aging and Neurodegenerative Diseases." Frontiers in Aging 4 (2024): 1292053. [CrossRef]
  63. Victorelli, Stella, and João F. Passos. "Telomeres and cell senescence-size matters not." EBioMedicine 21 (2017): 14-20. [CrossRef]
  64. An, Yongpan, Qian Wang, Ke Gao, Chi Zhang, Yanan Ouyang, Ruixiao Li, Zhou Ma et al. "Epigenetic Regulation of Aging and its Rejuvenation." MedComm 6, no. 9 (2025): e70369.
  65. Senapati, Parijat, Masaru Miyano, Rosalyn W. Sayaman, Mudaser Basam, Amy Leung, Mark A. LaBarge, and Dustin E. Schones. "Loss of epigenetic suppression of retrotransposons with oncogenic potential in aging mammary luminal epithelial cells." Genome Research 33, no. 8 (2023): 1229-1241.
  66. Olan, Ioana, Tetsuya Handa, and Masashi Narita. "Beyond SAHF: An integrative view of chromatin compartmentalization during senescence." Current Opinion in Cell Biology 83 (2023): 102206.
  67. Ishikawa, Koji. "Multilayered regulation of proteome stoichiometry." Current Genetics 67, no. 6 (2021): 883-890.
  68. Ding, Baojin, and Masood Sepehrimanesh. "Nucleocytoplasmic transport: regulatory mechanisms and the implications in neurodegeneration." International journal of molecular sciences 22, no. 8 (2021): 4165.
  69. Al-Muftia, Yasmeen, Stephen Cranwella, and Rahul S. Samant. "Dysregulated proteostasis: mechanisms and links to aging." Molecular, Cellular, and Metabolic Fundamentals of Human Aging (2022): 55.
  70. Zhang, Jianying, He-Ling Wang, Evandro Fei Fang, E. F. Fang, L. H. Bergersen, and B. C. Gilmour. Autophagy and bioenergetics in aging. Elsevier Inc./Academic Press, 2022.
  71. Yang, Lili. "Nutrient sensing and aging." Molecular, Cellular, and Metabolic Fundamentals of Human Aging (2022): 41.
  72. Poljšak, Borut, Vito Kovač, Stjepan Špalj, and Irina Milisav. "The central role of the NAD+ molecule in the development of aging and the prevention of chronic age-related diseases: strategies for NAD+ modulation." International journal of molecular sciences 24, no. 3 (2023): 2959. [CrossRef]
  73. Lautrup, Sofie, Alexander Anisimov, Maria Jose Lagartos-Donate, and Evandro Fei Fang. "Senescence in aging." Molecular, Cellular, and Metabolic Fundamentals of Human Aging 149 (2022).
  74. Tizazu, Anteneh Mehari, Eyerusalem Amossa Tessema, and Olivier NF Cexus. "Targeting immunosenesence promotes clearance of senescent cells." Immunity & Ageing 22, no. 1 (2025): 35.
  75. Wang, Shuaiqi, Tong Huo, Mingyang Lu, Yueqi Zhao, Jianmin Zhang, Wei He, and Hui Chen. "Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies." Cells 14, no. 7 (2025): 499.
  76. Aging Biomarker Consortium, Hainan Bao, Jiani Cao, Mengting Chen, Min Chen, Wei Chen, Xiao Chen et al. "Biomarkers of aging." Science China Life Sciences 66, no. 5 (2023): 893-1066.
  77. Zhavoronkov, Alex, Dominika Wilczok, Feng Ren, and Fedor Galkin. "Age-related diseases as a testbed for anti-aging therapeutics: the case of idiopathic pulmonary fibrosis." Aging (Albany NY) 17, no. 8 (2025): 1911.
  78. Goriely, Alain. "Eighty-six billion and counting: do we know the number of neurons in the human brain?." Brain 148, no. 3 (2025): 689-691.
  79. Bartman, Sydney, Giuseppe Coppotelli, and Jaime M. Ross. "Mitochondrial dysfunction: a key player in brain aging and diseases." Current issues in molecular biology 46, no. 3 (2024): 1987-2026.
  80. Lim, Su Min, Minyeop Nahm, and Seung Hyun Kim. "Proteostasis and ribostasis impairment as common cell death mechanisms in neurodegenerative diseases." Journal of Clinical Neurology (Seoul, Korea) 19, no. 2 (2023): 101. [CrossRef]
  81. Murdock, Mitchell H., and Li-Huei Tsai. "Insights into Alzheimer’s disease from single-cell genomic approaches." Nature neuroscience 26, no. 2 (2023): 181-195. [CrossRef]
  82. Li, Ke, Rongsha Chen, Ruohua Wang, Wenhui Fan, Ninghui Zhao, Zhongshan Yang, and Jinyuan Yan. "Neuroinflammation in neurodegenerative diseases: Focusing on the mediation of T lymphocytes." Neural Regeneration Research 21, no. 5 (2026): 1864-1889. [CrossRef]
  83. Fu, Jiajia, Yan Huang, Ting Bao, Chengcheng Liu, Xi Liu, and Xueping Chen. "The role of Th17 cells/IL-17A in AD, PD, ALS and the strategic therapy targeting on IL-17A." Journal of Neuroinflammation 19, no. 1 (2022): 98. [CrossRef]
  84. Sun, Eric D., Olivia Y. Zhou, Max Hauptschein, Nimrod Rappoport, Lucy Xu, Paloma Navarro Negredo, Ling Liu, Thomas A. Rando, James Zou, and Anne Brunet. "Spatial transcriptomic clocks reveal cell proximity effects in brain ageing." Nature 638, no. 8049 (2025): 160-171.
  85. Knox, Emily G., Maria R. Aburto, Gerard Clarke, John F. Cryan, and Caitriona M. O’Driscoll. "The blood-brain barrier in aging and neurodegeneration." Molecular psychiatry 27, no. 6 (2022): 2659-2673.
  86. Yang, Andrew C., Ryan T. Vest, Fabian Kern, Davis P. Lee, Maayan Agam, Christina A. Maat, Patricia M. Losada et al. "A human brain vascular atlas reveals diverse mediators of Alzheimer’s risk." Nature 603, no. 7903 (2022): 885-892.
  87. Solanki, Riddhi, Anjali Karande, and Prathibha Ranganathan. "Emerging role of gut microbiota dysbiosis in neuroinflammation and neurodegeneration." Frontiers in Neurology 14 (2023): 1149618.
  88. Koumpouli, Despoina, Varvara Koumpouli, and Antonios E. Koutelidakis. "The Gut–Brain Axis and Neurodegenerative Diseases: The Role of Nutritional Interventions Targeting the Gut Microbiome—A Systematic Review." Applied Sciences 15, no. 10 (2025): 5558.
  89. Bruno, Francesco, Paolo Abondio, Rossella Bruno, Leognano Ceraudo, Ersilia Paparazzo, Luigi Citrigno, Donata Luiselli et al. "Alzheimer’s disease as a viral disease: Revisiting the infectious hypothesis." Ageing research reviews 91 (2023): 102068.
  90. World Health Organization. "Cardiovascular Diseases." Accessed October 27, 2025. https://www.who.int/health-topics/cardiovascular-diseases.
  91. Mehu, Marcelle, Chandrakala Aluganti Narasimhulu, and Dinender K. Singla. "Inflammatory cells in atherosclerosis." Antioxidants 11, no. 2 (2022): 233.
  92. Cheng, Jun, Hong Wu, Cheng Xie, Yangyan He, Rong Mou, Hongkun Zhang, Yan Yang, and Qingbo Xu. "Single-cell mapping of large and small arteries during hypertensive aging." The Journals of Gerontology: Series A 79, no. 2 (2024): glad188. [CrossRef]
  93. Chen, Huimei, Aida Moreno-Moral, Francesco Pesce, Nithya Devapragash, Massimiliano Mancini, Ee Ling Heng, Maxime Rotival et al. "WWP2 regulates pathological cardiac fibrosis by modulating SMAD2 signaling." Nature communications 10, no. 1 (2019): 3616. [CrossRef]
  94. Chen, Huimei, Gabriel Chew, Nithya Devapragash, Jui Zhi Loh, Kevin Y. Huang, Jing Guo, Shiyang Liu et al. "The E3 ubiquitin ligase WWP2 regulates pro-fibrogenic monocyte infiltration and activity in heart fibrosis." Nature Communications 13, no. 1 (2022): 7375.
  95. Zhang, Yiyuan, Yandong Zheng, Si Wang, Yanling Fan, Yanxia Ye, Yaobin Jing, Zunpeng Liu et al. "Single-nucleus transcriptomics reveals a gatekeeper role for FOXP1 in primate cardiac aging." Protein & Cell 14, no. 4 (2023): 279-293.
  96. Subramaniam, Karthik, Haribalan Kumar, and Merryn H. Tawhai. "Evidence for age-dependent air-space enlargement contributing to loss of lung tissue elastic recoil pressure and increased shear modulus in older age." Journal of Applied Physiology 123, no. 1 (2017): 79-87.
  97. Bailey, Kristina L. "Aging diminishes mucociliary clearance of the lung." Advances in geriatric medicine and research 4, no. 2 (2022): e220005.
  98. Cai, Lubing, Luze Shi, Zhen Peng, Yaying Sun, and Jiwu Chen. "Ageing of skeletal muscle extracellular matrix and mitochondria: finding a potential link." Annals of Medicine 55, no. 2 (2023): 2240707.
  99. Fu, Zhibin, Hailong Xu, Lanping Yue, Weiwei Zheng, Linkang Pan, Fangyi Gao, and Xingshan Liu. "Immunosenescence and cancer: Opportunities and challenges." Medicine 102, no. 47 (2023): e36045.
  100. Wang, Leihan, and Dong Tang. "Immunosenescence promotes cancer development: from mechanisms to treatment strategies." Cell Communication and Signaling 23, no. 1 (2025): 128. [CrossRef]
  101. Lv, Jianjie, Chun Zhang, Xiuxing Liu, Chenyang Gu, Yidan Liu, Yuehan Gao, Zhaohao Huang et al. "An aging-related immune landscape in the hematopoietic immune system." Immunity & Ageing 21, no. 1 (2024): 3. [CrossRef]
  102. Kovtonyuk, Larisa V., Kristin Fritsch, Xiaomin Feng, Markus G. Manz, and Hitoshi Takizawa. "Inflamm-aging of hematopoiesis, hematopoietic stem cells, and the bone marrow microenvironment." Frontiers in immunology 7 (2016): 502. [CrossRef]
  103. Liang, Zhanfeng, Xue Dong, Zhaoqi Zhang, Qian Zhang, and Yong Zhao. "Age-related thymic involution: Mechanisms and functional impact." Aging cell 21, no. 8 (2022): e13671. [CrossRef]
  104. Egorov, Evgeny S., Sofya A. Kasatskaya, Vasiliy N. Zubov, Mark Izraelson, Tatiana O. Nakonechnaya, Dmitriy B. Staroverov, Andrea Angius et al. "The changing landscape of naive T cell receptor repertoire with human aging." Frontiers in Immunology 9 (2018): 1618.
  105. Lanfermeijer, Josien, Peter C. de Greef, Marion Hendriks, Martijn Vos, Josine van Beek, José AM Borghans, and Debbie van Baarle. "Age and CMV-infection jointly affect the EBV-specific CD8+ T-cell repertoire." Frontiers in Aging 2 (2021): 665637.
  106. Hosie, Louise, Annette Pachnio, Jianmin Zuo, Hayden Pearce, Stanley Riddell, and Paul Moss. "Cytomegalovirus-specific T cells restricted by HLA-Cw* 0702 increase markedly with age and dominate the CD8+ T-cell repertoire in older people." Frontiers in immunology 8 (2017): 1776.
  107. Messaoudi, Ilhem, Joël LeMaoult, Jose A. Guevara-Patino, Beatrix M. Metzner, and Janko Nikolich-Žugich. "Age-related CD8 T cell clonal expansions constrict CD8 T cell repertoire and have the potential to impair immune defense." The Journal of experimental medicine 200, no. 10 (2004): 1347-1358.
  108. Yu, Weiru, Yifei Yu, Siyuan Sun, Chenxu Lu, Jianan Zhai, Yumei Lei, Feirong Bai, Ran Wang, and Juan Chen. "Immune alterations with aging: mechanisms and intervention strategies." Nutrients 16, no. 22 (2024): 3830.
  109. Zhao, Tuantuan V., Yuki Sato, Jorg J. Goronzy, and Cornelia M. Weyand. "T-cell aging-associated phenotypes in autoimmune disease." Frontiers in Aging 3 (2022): 867950.
  110. Tizazu, Anteneh Mehari, Hylemariam Mihiretie Mengist, and Gebreselassie Demeke. "Aging, inflammaging and immunosenescence as risk factors of severe COVID-19." Immunity & Ageing 19, no. 1 (2022): 53. [CrossRef]
  111. Yorgancı, Kaya, and Hilmi Anıl Dinçer. "Aging in gastrointestinal system." In Beauty, Aging, and AntiAging, pp. 339-345. Academic Press, 2023.
  112. Skokowski, Jaroslaw, Yogesh Vashist, Sergii Girnyi, Tomasz Cwalinski, Piotr Mocarski, Carmine Antropoli, Antonio Brillantino et al. "The aging stomach: clinical implications of H. pylori infection in older adults—Challenges and strategies for improved management." International Journal of Molecular Sciences 25, no. 23 (2024): 12826. [CrossRef]
  113. Liu, Yiting, and Wen Peng. "Helicobacter pylori Infection and Its Hidden Role in Low Skeletal Muscle Mass: A Comprehensive Review." International Journal of General Medicine (2025): 4659-4670. [CrossRef]
  114. Aleman, Ricardo Santos, Marvin Moncada, and Kayanush J. Aryana. "Leaky gut and the ingredients that help treat it: a review." Molecules 28, no. 2 (2023): 619.
  115. Tran, Lee, and Beverley Greenwood-Van Meerveld. "Age-associated remodeling of the intestinal epithelial barrier." Journals of Gerontology Series A: Biomedical Sciences and Medical Sciences 68, no. 9 (2013): 1045-1056.
  116. Qi, YanFei, Ruby Goel, Seungbum Kim, Elaine M. Richards, Christy S. Carter, Carl J. Pepine, Mohan K. Raizada, and Thomas W. Buford. "Intestinal permeability biomarker zonulin is elevated in healthy aging." Journal of the American Medical Directors Association 18, no. 9 (2017): 810-e1.
  117. Ragonnaud, Emeline, and Arya Biragyn. "Gut microbiota as the key controllers of “healthy” aging of elderly people." Immunity & Ageing 18, no. 1 (2021): 2.
  118. Deng, Feilong, Ying Li, and Jiangchao Zhao. "The gut microbiome of healthy long-living people." Aging (Albany NY) 11, no. 2 (2019): 289.
  119. van der Lugt, Benthe, Adriaan A. Van Beek, Steven Aalvink, Ben Meijer, Bruno Sovran, Wilbert P. Vermeij, Renata MC Brandt et al. "Akkermansia muciniphila ameliorates the age-related decline in colonic mucus thickness and attenuates immune activation in accelerated aging Ercc1−/Δ7 mice." Immunity & Ageing 16, no. 1 (2019): 6.
  120. Conway, Jessica, Erica N. De Jong, Andrea J. White, Ben Dugan, Nia Paddison Rees, Sonia M. Parnell, Lisa E. Lamberte et al. "Age-related loss of intestinal barrier integrity plays an integral role in thymic involution and T cell ageing." Aging Cell 24, no. 3 (2025): e14401. [CrossRef]
  121. Kuchay, Mohammad Shafi, José Ignacio Martínez-Montoro, Narendra Singh Choudhary, José Carlos Fernández-García, and Bruno Ramos-Molina. "Non-alcoholic fatty liver disease in lean and non-obese individuals: current and future challenges." Biomedicines 9, no. 10 (2021): 1346. [CrossRef]
  122. Matsumoto, T., L. Wakefield, and Markus Grompe. "The significance of polyploid hepatocytes during aging process." Cellular and Molecular Gastroenterology and Hepatology 11, no. 5 (2021): 1347-1349. [CrossRef]
  123. Hunt, Nicholas J., Glen P. Lockwood, Alessandra Warren, Hong Mao, Peter AG McCourt, David G. Le Couteur, and Victoria C. Cogger. "Manipulating fenestrations in young and old liver sinusoidal endothelial cells." American Journal of Physiology-Gastrointestinal and Liver Physiology 316, no. 1 (2019): G144-G154.
  124. Yang, Shanshan, Chengyu Liu, Mengmeng Jiang, Xiaoqian Liu, Lingling Geng, Yiyuan Zhang, Shuhui Sun et al. "A single-nucleus transcriptomic atlas of primate liver aging uncovers the pro-senescence role of SREBP2 in hepatocytes." Protein & Cell 15, no. 2 (2024): 98-120.
  125. Möller, Kathleen, Christian Jenssen, Barbara Braden, Michael Hocke, Stephan Hollerbach, André Ignee, Siegbert Faiss et al. "Pancreatic changes with lifestyle and age: What is normal and what is concerning?." Endoscopic ultrasound 12, no. 2 (2023): 213-227.
  126. Löhr, J-M., Nikola Panic, Miroslav Vujasinovic, and Caroline S. Verbeke. "The ageing pancreas: a systematic review of the evidence and analysis of the consequences." Journal of internal medicine 283, no. 5 (2018): 446-460.
  127. Zhu, Min, Xiaohong Liu, Wen Liu, Yanrong Lu, Jingqiu Cheng, and Younan Chen. "β cell aging and age-related diabetes." Aging (Albany NY) 13, no. 5 (2021): 7691.
  128. Li, Jingyi, Yuxuan Zheng, Pengze Yan, Moshi Song, Si Wang, Liang Sun, Zunpeng Liu et al. "A single-cell transcriptomic atlas of primate pancreatic islet aging." National science review 8, no. 2 (2021): nwaa127.
  129. Chou, Yu-Hsiang, and Yung-Ming Chen. "Aging and renal disease: old questions for new challenges." Aging and disease 12, no. 2 (2021): 515.
  130. Zhang, Meiqi, Haifeng Ni, Yumeng Lin, Ke Wang, Tingke He, Lan Yuan, Zhongyu Han, and Xiaohong Zuo. "Renal aging and its consequences: navigating the challenges of an aging population." Frontiers in Pharmacology 16 (2025): 1615681. [CrossRef]
  131. Gao, Yongguang, Na Chen, Zhanda Fu, and Qing Zhang. "Progress of Wnt signaling pathway in osteoporosis." Biomolecules 13, no. 3 (2023): 483. [CrossRef]
  132. Hsu, Shao-Heng, Li-Ru Chen, and Kuo-Hu Chen. "Primary osteoporosis induced by androgen and estrogen deficiency: the molecular and cellular perspective on pathophysiological mechanisms and treatments." International Journal of Molecular Sciences 25, no. 22 (2024): 12139. [CrossRef]
  133. Ivell, Richard, Kee Heng, Katie Severn, Leen Antonio, Gyorgy Bartfai, Felipe F. Casanueva, Ilpo T. Huhtaniemi et al. "The Leydig cell biomarker INSL3 as a predictor of age-related morbidity: Findings from the EMAS cohort." Frontiers in Endocrinology 13 (2022): 1016107. [CrossRef]
  134. Di Carlo, Emma, and Carlo Sorrentino. "The multifaceted role of the stroma in the healthy prostate and prostate cancer." Journal of Translational Medicine 22, no. 1 (2024): 825.
  135. Ju, Wenhan, Yuewen Zhao, Yi Yu, Shuai Zhao, Shan Xiang, and Fang Lian. "Mechanisms of mitochondrial dysfunction in ovarian aging and potential interventions." Frontiers in Endocrinology 15 (2024): 1361289.
  136. Cox, Shanice, Ryan Nasseri, Rachel S. Rubin, and Yahir Santiago-Lastra. "Genitourinary syndrome of menopause." Medical Clinics 107, no. 2 (2023): 357-369.
  137. Wong, Qi Yi Ambrose, and Fook Tim Chew. "Defining skin aging and its risk factors: a systematic review and meta-analysis." Scientific reports 11, no. 1 (2021): 22075.
  138. Wong, Chloe, Jun Yan Ng, Yang Yie Sio, and Fook Tim Chew. "Genetic determinants of skin ageing: a systematic review and meta-analysis of genome-wide association studies and candidate genes." Journal of Physiological Anthropology 44, no. 1 (2025): 4.
  139. Montégut, Léa, Carlos López-Otín, and Guido Kroemer. "Aging and cancer." Molecular cancer 23, no. 1 (2024): 106.
  140. Liu, Ce, Zhaoru Yang, Li He, Ya Xiao, Hao Zhao, Ling Zhang, Tong Liu, Rentong Chen, Kai Zhang, and Bin Luo. "Optimal lifestyle patterns for delaying ageing and reducing all-cause mortality: insights from the UK Biobank." European Review of Aging and Physical Activity 21, no. 1 (2024): 27. [CrossRef]
  141. Zhavoronkov, Alex. "Insilico's bet on small molecules for metabolic, CNS and I&I diseases." LinkedIn. 28 oктября 2025. https://www.linkedin.com/posts/zhavoronkov_in-the-current-environment-this-is-probably-activity-7383253850398363648-tQ4Y.
  142. Howes, Laura. "Why Small-Molecule Drug Discovery Is Having a Moment." Chemical & Engineering News, January 21, 2025. https://cen.acs.org/pharmaceuticals/drug-discovery/small-molecule-drug-discovery-having/101/i36.
  143. Small Molecule Drug Trends | Kyinno Bio. 23 Jan. 2024, https://www.kyinno.com/articles/small-molecule-drugs-new-trends-in-targeting-large-molecules/.
  144. Niazi, Sarfaraz K. "Artificial Intelligence in Small-Molecule Drug Discovery: A Critical Review of Methods, Applications, and Real-World Outcomes." Pharmaceuticals 18, no. 9 (2025): 1271.
  145. Arriola Apelo, Sebastian I., and Dudley W. Lamming. "Rapamycin: an InhibiTOR of aging emerges from the soil of Easter Island." Journals of Gerontology Series A: Biomedical Sciences and Medical Sciences 71, no. 7 (2016): 841-849.
  146. Petrov, Anton, Natalia Perekhvatova, Maxim Skulachev, Linda Stein, and George Ousler. "SkQ1 ophthalmic solution for dry eye treatment: results of a phase 2 safety and efficacy clinical study in the environment and during challenge in the controlled adverse environment model." Advances in therapy 33, no. 1 (2016): 96-115.
  147. Jia, Bo, Jingjing Ye, Lebin Gan, Rui Li, Mengwei Zhang, Diya Sun, Lin Weng et al. "Mitochondrial antioxidant SkQ1 decreases inflammation following hemorrhagic shock by protecting myocardial mitochondria." Frontiers in physiology 13 (2022): 1047909.
  148. Gnesin, Filip, Anne Cathrine Baun Thuesen, Lise Katrine Aronsen Kähler, Sten Madsbad, and Bianca Hemmingsen. "Metformin monotherapy for adults with type 2 diabetes mellitus." Cochrane Database of Systematic Reviews 6 (2020).
  149. American Federation for Aging Research. "TAME - Targeting Aging with Metformin." Accessed October 28, 2025. https://www.afar.org/tame-trial.
  150. Albert Einstein College of Medicine. "Metformin in Longevity Study (MILES)." ClinicalTrials.gov, NCT02432287. Last updated May 19, 2021. https://clinicaltrials.gov/study/NCT02432287.
  151. Song, Qin, Xiaofeng Zhou, Kexin Xu, Sishi Liu, Xinqiang Zhu, and Jun Yang. "The safety and antiaging effects of nicotinamide mononucleotide in human clinical trials: an update." Advances in nutrition 14, no. 6 (2023): 1416-1435. [CrossRef]
  152. Nadeeshani, Harshani, Jinyao Li, Tianlei Ying, Baohong Zhang, and Jun Lu. "Nicotinamide mononucleotide (NMN) as an anti-aging health product–promises and safety concerns." Journal of advanced research 37 (2022): 267-278. [CrossRef]
  153. Huang, Hao. "A multicentre, randomised, double blind, parallel design, placebo controlled study to evaluate the efficacy and safety of uthever (NMN supplement), an orally administered supplementation in middle aged and older adults." Frontiers in aging 3 (2022): 851698. [CrossRef]
  154. Ewald, Collin Y., Fadi E. Pulous, Sarah Wing Yan Lok, Frank W. Pun, Alex Aliper, Feng Ren, and Alex Zhavoronkov. "TNIK’s emerging role in cancer, metabolism, and age-related diseases." Trends in pharmacological sciences 45, no. 6 (2024): 478-489.
  155. Ren, Feng, Alex Aliper, Jian Chen, Heng Zhao, Sujata Rao, Christoph Kuppe, Ivan V. Ozerov et al. "A small-molecule TNIK inhibitor targets fibrosis in preclinical and clinical models." Nature Biotechnology 43, no. 1 (2025): 63-75.
  156. Chen, Huimei, Ran You, Jing Guo, Wei Zhou, Gabriel Chew, Nithya Devapragash, Jui Zhi Loh et al. "WWP2 regulates renal fibrosis and the metabolic reprogramming of profibrotic myofibroblasts." Journal of the American Society of Nephrology 35, no. 6 (2024): 696-718.
  157. Insilico Medicine, "Transforming Parkinson’s Disease Treatment, Insilico Medicine Announces IND-Enabling Completion for AI-Empowered Oral NLRP3 Inhibitor ISM8969," press release, August 14, 2025, https://insilico.com/tpost/fyvt4m40f1-transforming-parkinsons-disease-treatmen.
  158. Liang, Ruikai, Xinrui Qi, Qi Cai, Liyan Niu, Xi Huang, Deju Zhang, Jitao Ling et al. "The role of NLRP3 inflammasome in aging and age-related diseases." Immunity & Ageing 21, no. 1 (2024): 14.
  159. Hossain, Md Arafat. "A comprehensive review of immune checkpoint inhibitors for cancer treatment." International Immunopharmacology 143 (2024): 113365.
  160. Komech, Ekaterina A., Anastasia D. Koltakova, Anna A. Barinova, Anastasia A. Minervina, Maria A. Salnikova, Evgeniya I. Shmidt, Tatiana V. Korotaeva et al. "TCR repertoire profiling revealed antigen-driven CD8+ T cell clonal groups shared in synovial fluid of patients with spondyloarthritis." Frontiers in Immunology 13 (2022): 973243. [CrossRef]
  161. Britanova, Olga V., Kseniia R. Lupyr, Dmitry B. Staroverov, Irina A. Shagina, Alexey A. Aleksandrov, Yakov Y. Ustyugov, Dmitry V. Somov et al. "Targeted depletion of TRBV9+ T cells as immunotherapy in a patient with ankylosing spondylitis." Nature medicine 29, no. 11 (2023): 2731-2736. [CrossRef]
  162. BIOCAD. «Трибувиа®». Accessed October 28, 2025. https://biocad.ru/products/autoimmune/tribuvia.
  163. PCR News, «Сбрoсить Иммунную Память, Прoдлить Жизнь». Accessed October 28, 2025. https://pcr.news/stati/sbrosit-immunnuyu-pamyat-prodlit-zhizn/.
  164. Silva, Isabel C., Rachel Daher, and Saakshi Khattri. "Combined use of Omalizumab and dupilumab: safety and efficacy data from a large academic center." Archives of Dermatological Research 317, no. 1 (2025): 674.
  165. Seyed Jafari, S. Morteza, Laurence Feldmeyer, Simon Bossart, Dagmar Simon, Christoph Schlapbach, and Luca Borradori. "Case report: combination of omalizumab and dupilumab for recalcitrant bullous pemphigoid." Frontiers in immunology 11 (2021): 611549.
  166. Pavord, Ian D., Pascal Chanez, Gerard J. Criner, Huib AM Kerstjens, Stephanie Korn, Njira Lugogo, Jean-Benoit Martinot et al. "Mepolizumab for eosinophilic chronic obstructive pulmonary disease." New England Journal of Medicine 377, no. 17 (2017): 1613-1629.
  167. Abduljabbar, Maram H. "PCSK9 inhibitors: focus on evolocumab and its impact on atherosclerosis progression." Pharmaceuticals 17, no. 12 (2024): 1581.
  168. Chowdhury, Selia, and Nurjahan Shipa Chowdhury. "Novel anti-amyloid-beta (Aβ) monoclonal antibody lecanemab for Alzheimer’s disease: A systematic review." International journal of immunopathology and pharmacology 37 (2023): 03946320231209839.
  169. U.S. Food and Drug Administration, Office of the Commissioner, "FDA Converts Novel Alzheimer’s Disease Treatment to Traditional Approval," press release, August 9, 2024, https://www.fda.gov/news-events/press-announcements/fda-converts-novel-alzheimers-disease-treatment-traditional-approval.
  170. U.S. Food and Drug Administration, Center for Drug Evaluation and Research, "FDA Approves Treatment for Adults with Alzheimer’s Disease," press release, July 2024, https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-treatment-adults-alzheimers-disease.
  171. Wang, Hong, Emel Serap Monkul Nery, Paul Ardayfio, Rashna Khanna, Diana Otero Svaldi, Ivelina Gueorguieva, Sergey Shcherbinin et al. "Modified titration of donanemab reduces ARIA risk and maintains amyloid reduction." Alzheimer's & Dementia 21, no. 4 (2025): e70062. [CrossRef]
  172. Ranbhor, Ranjit. "Advancing Monoclonal Antibody Manufacturing: Process Optimization, Cost Reduction Strategies, and Emerging Technologies." Biologics: Targets and Therapy (2025): 177-187. [CrossRef]
  173. Singh, Rohit, Pankaj Chandley, and Soma Rohatgi. "Recent advances in the development of monoclonal antibodies and next-generation antibodies." Immunohorizons 7, no. 12 (2023): 886-897. [CrossRef]
  174. Oh, Hamilton Se-Hwee, Jarod Rutledge, Daniel Nachun, Róbert Pálovics, Olamide Abiose, Patricia Moran-Losada, Divya Channappa et al. "Organ aging signatures in the plasma proteome track health and disease." Nature 624, no. 7990 (2023): 164-172.
  175. Patel, Rikin, Mohamad Kaki, Venkat S. Potluri, Payal Kahar, and Deepesh Khanna. "A comprehensive review of SARS-CoV-2 vaccines: Pfizer, moderna & Johnson & Johnson." Human vaccines & immunotherapeutics 18, no. 1 (2022): 2002083.
  176. European Medicines Agency, "Comirnaty". Accessed October 28, 2025. https://www.ema.europa.eu/en/medicines/human/EPAR/comirnaty.
  177. European Medicines Agency, "Spikevax (Previously COVID-19 Vaccine Moderna)". Accessed October 28, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/spikevax.
  178. Zoroddu, Stefano, and Luigi Bagella. "Next-Generation mRNA Vaccines in Melanoma: Advances in Delivery and Combination Strategies." Cells 14, no. 18 (2025): 1476.
  179. Kabut, Jacek, Grzegorz J. Stępień, Tomasz Furgoł, Michał Miciak, Natalia Nafalska, Małgorzata Stopyra, Marcin Jezierzański, Krzysztof Feret, and Iwona Gisterek-Grocholska. "mRNA Vaccines in Modern Immunotherapy for Non-Small Cell Lung Cancer (NSCLC)—A Comprehensive Literature Review with Focus on Current Clinical Trials." Biomedicines 13, no. 9 (2025): 2187.
  180. Weber, Jeffrey S., Matteo S. Carlino, Adnan Khattak, Tarek Meniawy, George Ansstas, Matthew H. Taylor, Kevin B. Kim et al. "Individualised neoantigen therapy mRNA-4157 (V940) plus pembrolizumab versus pembrolizumab monotherapy in resected melanoma (KEYNOTE-942): a randomised, phase 2b study." The Lancet 403, no. 10427 (2024): 632-644. [CrossRef]
  181. Merck.com. “Merck and Moderna Initiate Phase 3 Trial Evaluating Adjuvant V940 (mRNA-4157) in Combination with KEYTRUDA® (Pembrolizumab) After Neoadjuvant KEYTRUDA and Chemotherapy in Patients With Certain Types of Non-Small Cell Lung Cancer (NSCLC).” Accessed October 28, 2025. https://www.merck.com/news/merck-and-moderna-initiate-phase-3-trial-evaluating-adjuvant-v940-mrna-4157-in-combination-with-keytruda-pembrolizumab-after-neoadjuvant-keytruda-and-chemotherapy-in-patients-with-certain-ty/.
  182. Geng, Shuang, Huiyuan Zhang, Xian Zhou, Yue He, Xiaoqian Zhang, Xiaoping Xie, Chaofan Li et al. "Diabetes tolerogenic vaccines targeting antigen-specific inflammation." Human vaccines & immunotherapeutics 11, no. 2 (2015): 522-530. [CrossRef]
  183. Mannie, Mark D., Kayla B. DeOca, Alexander G. Bastian, and Cody D. Moorman. "Tolerogenic vaccines: Targeting the antigenic and cytokine niches of FOXP3+ regulatory T cells." Cellular immunology 355 (2020): 104173. [CrossRef]
  184. Wang, Fazhan, Jia Lou, Xiaohan Lou, Fang Wu, Xiaoke Gao, Xiaohan Yao, Jiajia Wan et al. "A Spleen-Targeted Tolerogenic mRNA-LNPs Vaccine for the Treatment of Experimental Asthma." Advanced Science 12, no. 13 (2025): 2412543.
  185. Bhatia, Harpreet S., Archna Bajaj, Sascha N. Goonewardena, and Patrick M. Moriarty. "Pelacarsen: Mechanism of action and Lp (a)-lowering effect." Journal of Clinical Lipidology (2025).
  186. Novartis Pharmaceuticals. “A Randomized Double-Blind, Placebo-Controlled, Multicenter Trial Assessing the Impact of Lipoprotein (a) Lowering With Pelacarsen (TQJ230) on Major Cardiovascular Events in Patients With Established Cardiovascular Disease.” Clinical trial registration. clinicaltrials.gov, July 4, 2025. https://clinicaltrials.gov/study/NCT04023552.
  187. Nissen, Steven E., Helle Linnebjerg, Xi Shen, Kathy Wolski, Xiaosu Ma, Shufen Lim, Laura F. Michael et al. "Lepodisiran, an extended-duration short interfering RNA targeting lipoprotein (a): a randomized dose-ascending clinical trial." Jama 330, no. 21 (2023): 2075-2083.
  188. Eli Lilly and Company. “A Phase 3, Randomized, Double-Blind, Placebo-Controlled Study to Investigate the Effect of Lepodisiran on the Reduction of Major Adverse Cardiovascular Events in Adults With Elevated Lipoprotein(a) Who Have Established Atherosclerotic Cardiovascular Disease or Are at Risk for a First Cardiovascular Event - ACCLAIM-Lp(a).” Clinical trial registration. clinicaltrials.gov, October 15, 2025. https://clinicaltrials.gov/study/NCT06292013.
  189. Lau, Freddy Duarte, and Robert P. Giugliano. "Lipoprotein (a) and its significance in cardiovascular disease: a review." Jama Cardiology 7, no. 7 (2022): 760-769.
  190. Rehberger Likozar, Andreja, Mark Zavrtanik, and Miran Šebeštjen. "Lipoprotein (a) in atherosclerosis: from pathophysiology to clinical relevance and treatment options." Annals of medicine 52, no. 5 (2020): 162-177.
  191. Yao, Matthew, S. Kent Dickeson, Karthik Dhanabalan, Sergey Solomevich, Connor Dennewitz, David Gailani, and Wen-Liang Song. "Investigation of the Influence of Lipoprotein (a) and Oxidized Lipoprotein (a) on Plasminogen Activation and Fibrinolysis." Journal of Lipid and Atherosclerosis 14, no. 2 (2025): 229. [CrossRef]
  192. Pozdniakova, Natalia, Evgenii Generalov, Alexei Shevelev, and Olga Tarasova. "RNA Therapeutics: Delivery Problems and Solutions—A Review." Pharmaceutics 17, no. 10 (2025): 1305. [CrossRef]
  193. Shamshirgaran, Yasaman, Jun Liu, Huseyin Sumer, Paul J. Verma, and Amir Taheri-Ghahfarokhi. "Tools for efficient genome editing; ZFN, TALEN, and CRISPR." Applications of genome modulation and editing (2022): 29-46.
  194. Wani, Atif Khurshid, Nahid Akhtar, Reena Singh, Ajit Prakash, Sayed Haidar Abbas Raza, Simona Cavalu, Chirag Chopra, Mahmoud Madkour, Ahmed Elolimy, and Nesrein M. Hashem. "Genome centric engineering using ZFNs, TALENs and CRISPR-Cas9 systems for trait improvement and disease control in Animals." Veterinary research communications 47, no. 1 (2023): 1-16.
  195. Porto, Elizabeth M., Alexis C. Komor, Ian M. Slaymaker, and Gene W. Yeo. "Base editing: advances and therapeutic opportunities." Nature Reviews Drug Discovery 19, no. 12 (2020): 839-859.
  196. Chen, Liang, Mengjia Hong, Changming Luan, Hongyi Gao, Gaomeng Ru, Xinyuan Guo, Dujuan Zhang et al. "Adenine transversion editors enable precise, efficient A• T-to-C• G base editing in mammalian cells and embryos." Nature biotechnology 42, no. 4 (2024): 638-650.
  197. McCutcheon, Sean R., Dahlia Rohm, Nahid Iglesias, and Charles A. Gersbach. "Epigenome editing technologies for discovery and medicine." Nature Biotechnology 42, no. 8 (2024): 1199-1217.
  198. Kovalev, Maxim A., Naida Yu Mamaeva, Nikolay V. Kristovskiy, Pavel G. Feskin, Renat S. Vinnikov, Pavel D. Oleinikov, Anastasiia O. Sosnovtseva, Valeriy A. Yakovlev, Grigory S. Glukhov, and Alexey K. Shaytan. "Epigenome Engineering Using dCas Systems for Biomedical Applications and Biotechnology: Current Achievements, Opportunities and Challenges." International Journal of Molecular Sciences 26, no. 13 (2025): 6371.
  199. “Verve 102 | Verve Therapeutics.” Accessed October 28, 2025. https://www.vervetx.com/our-programs/verve-102.
  200. “Verve Announces Initial Data from Heart-2 Phase 1b Trial of VERVE-102 - TipRanks.Com.” TipRanks Financial. Accessed October 28, 2025. https://www.tipranks.com/news/the-fly/verve-announces-initial-data-from-heart-2-phase-1b-trial-of-verve-102.
  201. CRISPR Medicine. “Press Release Service: Scribe Therapeutics Highlights Latest Data on Lp(a)-Lowering Therapy and Novel AI Platform for Next-Generation CRISPR-Based Therapeutics at Two Premier Conferences.” Accessed October 28, 2025. https://crisprmedicinenews.com/press-release-service/card/scribe-therapeutics-highlights-latest-data-on-lpa-lowering-therapy-and-novel-ai-platform-for-next/.
  202. “Scribe → Scribe Therapeutics Reports Positive Preclinical Data on Its Novel CRISPR Technologies at the 2025 EAS Congress and ASGCT Annual Meeting.” Accessed October 28, 2025. https://www.scribetx.com/news/scribe-therapeutics-reports-positive-preclinical-data-on-its-novel-crispr-technologies-at-the-2025-eas-congress-and-asgct-annual-meeting.
  203. Maestro, Sheila, Nicholas D. Weber, Nerea Zabaleta, Rafael Aldabe, and Gloria Gonzalez-Aseguinolaza. "Novel vectors and approaches for gene therapy in liver diseases." JHEP Reports 3, no. 4 (2021): 100300. [CrossRef]
  204. Kovalev, Maxim A., Artem I. Davletshin, and Dmitry S. Karpov. "Engineering Cas9: next generation of genomic editors." Applied Microbiology and Biotechnology 108, no. 1 (2024): 209.
  205. Gohil, Nisarg, Gargi Bhattacharjee, Navya Lavina Lam, Samuel D. Perli, and Vijai Singh. "CRISPR-Cas systems: challenges and future prospects." Progress in Molecular Biology and Translational Science 180 (2021): 141-151.
  206. Glasscock, Cameron J., Robert J. Pecoraro, Ryan McHugh, Lindsey A. Doyle, Wei Chen, Olivier Boivin, Beau Lonnquist et al. "Computational design of sequence-specific DNA-binding proteins." Nature Structural & Molecular Biology (2025): 1-10.
  207. Kirkland, J. L., and T. Tchkonia. "Senolytic drugs: from discovery to translation." Journal of internal medicine 288, no. 5 (2020): 518-536.
  208. Saliev, Timur, and Prim B. Singh. "Targeting Senescence: A Review of Senolytics and Senomorphics in Anti-Aging Interventions." Biomolecules 15, no. 6 (2025): 860.
  209. Cheng, Fang, Qiling Xu, Qiang Li, Zheng Cui, Weiming Li, and Fang Zeng. "Adverse reactions after treatment with dasatinib in chronic myeloid leukemia: Characteristics, potential mechanisms, and clinical management strategies." Frontiers in oncology 13 (2023): 1113462.
  210. Wilson, Wyndham H., Owen A. O'Connor, Myron S. Czuczman, Ann S. LaCasce, John F. Gerecitano, John P. Leonard, Anil Tulpule et al. "Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity." The lancet oncology 11, no. 12 (2010): 1149-1159. [CrossRef]
  211. Colville, Alex, Jie-Yu Liu, Cristina Rodriguez-Mateo, Samantha Thomas, Heather D. Ishak, Ronghao Zhou, Julian DD Klein et al. "Death-seq identifies regulators of cell death and senolytic therapies." Cell metabolism 35, no. 10 (2023): 1814-1829. [CrossRef]
  212. Acklin, Scarlett, Manchao Zhang, Wuying Du, Xin Zhao, Matthew Plotkin, Jianhui Chang, Judith Campisi, Daohong Zhou, and Fen Xia. "Depletion of senescent-like neuronal cells alleviates cisplatin-induced peripheral neuropathy in mice." Scientific reports 10, no. 1 (2020): 14170. [CrossRef]
  213. Riordan, Ruben, Wang Rong, Zhen Yu, Grace Ross, Juno Valerio, Jovita Dimas-Muñoz, Valeria Heredia, Kathy Magnusson, Veronica Galvan, and Viviana I. Perez. "Effect of Nrf2 loss on senescence and cognition of tau-based P301S mice." Geroscience 45, no. 3 (2023): 1451-1469. [CrossRef]
  214. Mannarino, Matthew, Hosni Cherif, Saber Ghazizadeh, Oliver Wu Martinez, Kai Sheng, Elsa Cousineau, Seunghwan Lee et al. "Senolytic treatment for low back pain." Science Advances 11, no. 11 (2025): eadr1719.
  215. Silwal, Prashanta, Allison M. Nguyen-Thai, Haneef Ahamed Mohammad, Yanshan Wang, Paul D. Robbins, Joon Y. Lee, and Nam V. Vo. "Cellular senescence in intervertebral disc aging and degeneration: molecular mechanisms and potential therapeutic opportunities." Biomolecules 13, no. 4 (2023): 686.
  216. Wu, Yuhao, Shiwei Shen, Yifeng Shi, Naifeng Tian, Yifei Zhou, and Xiaolei Zhang. "Senolytics: eliminating senescent cells and alleviating intervertebral disc degeneration." Frontiers in bioengineering and biotechnology 10 (2022): 823945.
  217. Zhou, Yueying, Ming Xu, and Sumit Yadav. "Temporomandibular joint aging and potential therapies." Aging (Albany NY) 13, no. 14 (2021): 17955.
  218. Lee, Jin Young, Nabora S. Reyes, Supriya Ravishankar, Minqi Zhou, Maria Krasilnikov, Christian Ringler, Grace Pohan et al. "An in vivo screening platform identifies senolytic compounds that target p16 INK4a+ fibroblasts in lung fibrosis." The Journal of Clinical Investigation 134, no. 9 (2024).
  219. Ozdemir, Said Ali, Md Imam Faizan, Gagandeep Kaur, Sadiya Bi Shaikh, Khursheed Ul Islam, and Irfan Rahman. "Heterogeneity of Cellular Senescence, Senotyping, and Targeting by Senolytics and Senomorphics in Lung Diseases." International Journal of Molecular Sciences (2025).
  220. Dagher, Olina, Pauline Mury, Pierre-Emmanuel Noly, Annik Fortier, Guillaume Lettre, Eric Thorin, and Michel Carrier. "Design of a randomized placebo-controlled trial to evaluate the anti-inflammatory and senolytic effects of quercetin in patients undergoing coronary artery bypass graft surgery." Frontiers in Cardiovascular Medicine 8 (2021): 741542. [CrossRef]
  221. Sweeney, Mark, Stuart A. Cook, and Jesús Gil. "Therapeutic opportunities for senolysis in cardiovascular disease." The FEBS journal 290, no. 5 (2023): 1235-1255. [CrossRef]
  222. Hickson, LaTonya J., Larissa GP Langhi Prata, Shane A. Bobart, Tamara K. Evans, Nino Giorgadze, Shahrukh K. Hashmi, Sandra M. Herrmann et al. "Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease." EBioMedicine 47 (2019): 446-456. [CrossRef]
  223. Klier, Sharon, Jamie Dananberg, Lauren Masaki, Robert B. Bhisitkul, Arshad M. Khanani, Raj Maturi, Hani Salehi-Had et al. "Safety and efficacy of senolytic UBX1325 in diabetic macular edema." NEJM evidence 4, no. 5 (2025): EVIDoa2400009. [CrossRef]
  224. Sodini, Selene, and Milton Fabián Suarez-Ortegón. "Senolytic Interventions for Type 2 Diabetes: Current Evidence and Future Directions." Diabetology 6, no. 6 (2025): 48.
  225. Farr, Joshua N., Elizabeth J. Atkinson, Sara J. Achenbach, Tammie L. Volkman, Amanda J. Tweed, Stephanie J. Vos, Ming Ruan et al. "Effects of intermittent senolytic therapy on bone metabolism in postmenopausal women: a phase 2 randomized controlled trial." Nature Medicine 30, no. 9 (2024): 2605-2612.
  226. Gonzalez-Hurtado, Elsie, Claire Leveau, Keyi Li, Manish Mishra, Rihao Qu, Emily L. Goldberg, Sviatoslav Sidorov et al. "Nerve-associated macrophages control adipose homeostasis across lifespan and restrain age-related inflammation." Nature Aging (2025): 1-16.
  227. Goldberg, Emily L., Irina Shchukina, Yun-Hee Youm, Seungjin Ryu, Takeshi Tsusaka, Kyrlia C. Young, Christina D. Camell, Tamara Dlugos, Maxim N. Artyomov, and Vishwa Deep Dixit. "IL-33 causes thermogenic failure in aging by expanding dysfunctional adipose ILC2." Cell metabolism 33, no. 11 (2021): 2277-2287.
  228. Shin, Ji-Won, Dong-Hyun Jang, So Young Kim, Je-Jung Lee, Tae-Hwan Gil, Eunha Shim, Ji Yeon Kim et al. "Propagation of senescent phenotypes by extracellular HMGB1 is dependent on its redox state." Metabolism 168 (2025): 156259.
  229. Yang, Jiekun, Maria Vamvini, Pasquale Nigro, Li-Lun Ho, Kyriakitsa Galani, Marcus Alvarez, Yosuke Tanigawa et al. "Single-cell dissection of the obesity-exercise axis in adipose-muscle tissues implies a critical role for mesenchymal stem cells." Cell metabolism 34, no. 10 (2022): 1578-1593.
  230. Wolff, Christopher A., Justin J. Reid, Robert V. Musci, Danielle R. Bruns, Melissa A. Linden, Adam R. Konopka, Frederick F. Peelor III, Benjamin F. Miller, and Karyn L. Hamilton. "Differential effects of rapamycin and metformin in combination with rapamycin on mechanisms of proteostasis in cultured skeletal myotubes." The Journals of Gerontology: Series A 75, no. 1 (2020): 32-39. [CrossRef]
  231. Chung, Ki Wung, and Hae Young Chung. "The effects of calorie restriction on autophagy: role on aging intervention." Nutrients 11, no. 12 (2019): 2923. [CrossRef]
  232. Freeberg, Kaitlin A., CeAnn C. Udovich, Christopher R. Martens, Douglas R. Seals, and Daniel H. Craighead. "Dietary supplementation with NAD+-boosting compounds in humans: current knowledge and future directions." The Journals of Gerontology: Series A 78, no. 12 (2023): 2435-2448. [CrossRef]
  233. Liao, Guanyi, Yuchen Xie, Hong Peng, Tianke Li, Xinsen Zou, Faguo Yue, Jinjun Guo, and Li Rong. "Advancements in NMN biotherapy and research updates in the field of digestive system diseases." Journal of Translational Medicine 22, no. 1 (2024): 805. [CrossRef]
  234. Newman, John. "ASSESSING GEROTHERAPEUTIC POTENTIAL OF GLP-1 RECEPTOR AGONISTS." Innovation in Aging 8, no. Suppl 1 (2024): 140.
  235. Li, Zhongqi, Xinyi Chen, Joaquim SL Vong, Lei Zhao, Junzhe Huang, Leo YC Yan, Bonaventure Ip et al. "Systemic GLP-1R agonist treatment reverses mouse glial and neurovascular cell transcriptomic aging signatures in a genome-wide manner." Communications biology 4, no. 1 (2021): 656.
  236. Cortes, Tiffany M., Libia Vasquez, Monica C. Serra, Ronna Robbins, Allison Stepanenko, Kevin Brown, Hannah Barrus, Annalisa Campos, Sara E. Espinoza, and Nicolas Musi. "Effect of semaglutide on physical function, body composition, and biomarkers of aging in older adults with overweight and insulin resistance: protocol for an open-labeled randomized controlled trial." JMIR research protocols 13, no. 1 (2024): e62667.
  237. Kreiner, Frederik Flindt, Bernt Johan von Scholten, Peter Kurtzhals, and Stephen Charles Langford Gough. "Glucagon-like peptide-1 receptor agonists to expand the healthy lifespan: Current and future potentials." Aging Cell 22, no. 5 (2023): e13818.
  238. Peng, Wei, Rui Zhou, Ze-Fang Sun, Jia-Wei Long, and Yong-Qiang Gong. "Novel insights into the roles and mechanisms of GLP-1 receptor agonists against aging-related diseases." Aging and disease 13, no. 2 (2022): 468.
  239. Liu, Ling, Soochi Kim, Matthew T. Buckley, Jaime M. Reyes, Jengmin Kang, Lei Tian, Mingqiang Wang et al. "Exercise reprograms the inflammatory landscape of multiple stem cell compartments during mammalian aging." Cell stem cell 30, no. 5 (2023): 689-705.
  240. Wang, Chao, Ruben Rabadan Ros, Paloma Martinez-Redondo, Zaijun Ma, Lei Shi, Yuan Xue, Isabel Guillen-Guillen et al. "In vivo partial reprogramming of myofibers promotes muscle regeneration by remodeling the stem cell niche." Nature Communications 12, no. 1 (2021): 3094. [CrossRef]
  241. Lu, Yuancheng, Benedikt Brommer, Xiao Tian, Anitha Krishnan, Margarita Meer, Chen Wang, Daniel L. Vera et al. "Reprogramming to recover youthful epigenetic information and restore vision." Nature 588, no. 7836 (2020): 124-129. [CrossRef]
  242. Drake, Sienna S., Abdulshakour Mohammadnia, Aliyah Zaman, Christine Gianfelice, Kali Heale, Adam MR Groh, Elizabeth M-L. Hua et al. "Cellular rejuvenation protects neurons from inflammation-mediated cell death." Cell reports 44, no. 2 (2025). [CrossRef]
  243. Wang, Yajie, Ting Lu, Guohuan Sun, Yawei Zheng, Shangda Yang, Hongyan Zhang, Sha Hao et al. "Targeting of apoptosis gene loci by reprogramming factors leads to selective eradication of leukemia cells." Nature Communications 10, no. 1 (2019): 5594. [CrossRef]
  244. Yang, Jae-Hyun, Christopher A. Petty, Thomas Dixon-McDougall, Maria Vina Lopez, Alexander Tyshkovskiy, Sun Maybury-Lewis, Xiao Tian et al. "Chemically induced reprogramming to reverse cellular aging." Aging (Albany NY) 15, no. 13 (2023): 5966.
  245. Conboy, Irina M., and Thomas A. Rando. "Heterochronic parabiosis for the study of the effects of aging on stem cells and their niches." Cell cycle 11, no. 12 (2012): 2260-2267.
  246. Ximerakis, Methodios, Kristina M. Holton, Richard M. Giadone, Ceren Ozek, Monika Saxena, Samara Santiago, Xian Adiconis et al. "Heterochronic parabiosis reprograms the mouse brain transcriptome by shifting aging signatures in multiple cell types." Nature Aging 3, no. 3 (2023): 327-345.
  247. Ambrosia LLC. “Young Donor Plasma Transfusion and Age-Related Biomarkers.” Clinical trial registration. clinicaltrials.gov, May 10, 2018. https://clinicaltrials.gov/study/NCT02803554.
  248. Research, Center for Biologics Evaluation and. “Important Information about Young Donor Plasma Infusions for Profit.” FDA, December 20, 2019. https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/important-information-about-young-donor-plasma-infusions-profit.
  249. Reporter, Kate Gibson, MoneyWatch Kate Gibson is a reporter for CBS MoneyWatch in New York, Where She Covers Business, and consumer finance Read Full Bio Kate Gibson. “Startup Halts Treatment Using Young People’s Blood after FDA Warning - CBS News,” February 19, 2019. https://www.cbsnews.com/news/startup-halts-treatments-using-young-peoples-blood-after-fda-warning/.
  250. Sha, Sharon. “The PLasma for Alzheimer SymptoM Amelioration (PLASMA) Study: Intravenously-Administered Plasma From Young Donors for Treatment of Mild-To-Moderate Alzheimer's Disease.” Clinical trial registration. clinicaltrials.gov, October 9, 2017. https://clinicaltrials.gov/study/NCT02256306.
  251. Khoury, Rita, and Elias Ghossoub. "Young blood products: emerging treatment for Alzheimer's disease?." Neural Regeneration Research 13, no. 4 (2018): 624-627. [CrossRef]
  252. Clement, James, Qi Yan, Megha Agrawal, Ramon E. Coronado, John A. Sturges, Markus Horvath, Ake T. Lu, Robert T. Brooke, and Steve Horvath. "Umbilical cord plasma concentrate has beneficial effects on DNA methylation GrimAge and human clinical biomarkers." Aging Cell 21, no. 10 (2022): e13696. [CrossRef]
  253. Liu, Mei, Jianping Zhu, Jiawei Zheng, Xuan Han, Lijuan Jiang, Xiangzhen Tong, Yue Ke et al. "GPNMB and ATP6V1A interact to mediate microglia phagocytosis of multiple types of pathological particles." Cell Reports 44, no. 3 (2025). [CrossRef]
  254. Prater, Katherine E., Kevin J. Green, Sainath Mamde, Wei Sun, Alexandra Cochoit, Carole L. Smith, Kenneth L. Chiou et al. "Human microglia show unique transcriptional changes in Alzheimer’s disease." Nature Aging 3, no. 7 (2023): 894-907.
  255. Martins-Ferreira, Ricardo, Josep Calafell-Segura, Bárbara Leal, Javier Rodríguez-Ubreva, Elena Martínez-Saez, Elisabetta Mereu, Paulo Pinho E Costa, Ariadna Laguna, and Esteban Ballestar. "The Human Microglia Atlas (HuMicA) unravels changes in disease-associated microglia subsets across neurodegenerative conditions." Nature Communications 16, no. 1 (2025): 739.
  256. Pyrkov, Timothy V., Konstantin Avchaciov, Andrei E. Tarkhov, Leonid I. Menshikov, Andrei V. Gudkov, and Peter O. Fedichev. "Longitudinal analysis of blood markers reveals progressive loss of resilience and predicts human lifespan limit." Nature communications 12, no. 1 (2021): 2765.
  257. Asmamaw Dejenie, Tadesse, Markeshaw Tiruneh G/Medhin, Gashaw Dessie Terefe, Fitalew Tadele Admasu, Wondwossen Wale Tesega, and Endeshaw Chekol Abebe. "Current updates on generations, approvals, and clinical trials of CAR T-cell therapy." Human vaccines & immunotherapeutics 18, no. 6 (2022): 2114254.
  258. Brudno, Jennifer N., Marcela V. Maus, and Christian S. Hinrichs. "CAR T cells and T-cell therapies for cancer: A translational science review." Jama (2024).
  259. Nie, Wen, Xinyu Yang, Mingrui Ma, Xiaogang Xu, and Jin Hou. "Chimeric antigen receptor T cell therapy for autoimmune diseases." Current Opinion in Immunology 95 (2025): 102596.
  260. Fisher, Marina S., and Sergey V. Sennikov. "T-regulatory cells for the treatment of autoimmune diseases." Frontiers in Immunology 16 (2025): 1511671. [CrossRef]
  261. Riet, Tobias, and Markus Chmielewski. "Regulatory CAR-T cells in autoimmune diseases: progress and current challenges." Frontiers in immunology 13 (2022): 934343. [CrossRef]
  262. Evangelou, Konstantinos, and Vassilis G. Gorgoulis. "Rejuvenating the immune system." Molecular Oncology 19, no. 3 (2025): 584-587. [CrossRef]
  263. Zhao, Jin, Rong Hu, Kuan Chen Lai, Zhenzhen Zhang, and Laijun Lai. "Recombinant FOXN1 fusion protein increases T cell generation in old mice." Frontiers in Immunology 15 (2024): 1423488. [CrossRef]
  264. Li, Jie, Lucas P. Wachsmuth, Shiyun Xiao, Brian G. Condie, and Nancy R. Manley. "Foxn1 overexpression promotes thymic epithelial progenitor cell proliferation and mTEC maintenance, but does not prevent thymic involution." Development 150, no. 8 (2023): dev200995.
  265. Shen, Jingyi, Ying Wang, Fei Zheng, Shuo Cao, Qiu Lan, Kailin Xu, and Bin Pan. "Aryl hydrocarbon receptor regulates IL-22 receptor expression on thymic epithelial cell and accelerates thymus regeneration." npj Regenerative Medicine 8, no. 1 (2023): 64.
  266. Hübscher, Tania, L. Francisco Lorenzo-Martín, Thomas Barthlott, Lucie Tillard, Jakob J. Langer, Paul Rouse, C. Clare Blackburn, Georg Holländer, and Matthias P. Lutolf. "Thymic epithelial organoids mediate T-cell development." development 151, no. 17 (2024): dev202853.
  267. Dean, Jillian, Cosima Hoch, Barbara Wollenberg, Justin Navidzadeh, Bhagvat Maheta, Anisha Mandava, Samuel Knoedler et al. "Advancements in bioengineered and autologous skin grafting techniques for skin reconstruction: a comprehensive review." Frontiers in Bioengineering and Biotechnology 12 (2025): 1461328.
  268. Palomino-de Anda, Luis Felipe, Gustavo Ortiz-Morales, Mauricio Muleiro-Alvarez, Guillermo R. Vera-Duarte, Nicolás Kahuam-López, Alejandro Navas, Arturo Ramirez-Miranda, and Enrique O. Graue-Hernandez. "Clinical outcomes of simple limbal epithelial transplantation for limbal stem cell deficiency in a Mexican population." Indian Journal of Ophthalmology 73, no. 10 (2025): 1508-1512.
  269. Li, Xiaohui, Honglin Wu, Hao Yang, Peng Wang, Xiaoling Cao, Peng Wang, Yongfei Chen et al. "Efficacy and safety of epidermal cell regeneration for postoperative non-healing wounds: a randomized, partially blinded, parallel-controlled trial." Stem Cell Research & Therapy 16, no. 1 (2025): 284.
  270. Prabhasawat, Pinnita, Chareenun Chirapapaisan, Panotsom Ngowyutagon, Pattama Ekpo, Wimolwan Tangpagasit, Kaevalin Lekhanont, Rosanun Sikarinkul et al. "Efficacy and outcome of simple limbal epithelial transplantation for limbal stem cell deficiency verified by epithelial phenotypes integrated with clinical evaluation." The Ocular Surface 22 (2021): 27-37. [CrossRef]
  271. Dube, Christabel Thembela, Yasmin Hui Binn Ong, Kelly Wemyss, Siddharth Krishnan, Tiak Ju Tan, Baptiste Janela, John R. Grainger, Matthew Ronshaugen, Kimberly A. Mace, and Chin Yan Lim. "Age-related alterations in macrophage distribution and function are associated with delayed cutaneous wound healing." Frontiers in immunology 13 (2022): 943159. [CrossRef]
  272. Kaiser, Benedict, Sylvie Miot, Anke Wixmerten, Oliver Pullig, Matthias Eyrich, Ilario Fulco, Josef Vavrina et al. "Engineered autologous nasal cartilage for repair of nasal septal perforations: a case series." International Journal of Surgery 110, no. 10 (2024): 6573-6580. [CrossRef]
  273. Mumme, Marcus, Anke Wixmerten, Alan Ivkovic, Giuseppe M. Peretti, Tayfun Yilmaz, Stephan Reppenhagen, Oliver Pullig et al. "Clinical relevance of engineered cartilage maturation in a randomized multicenter trial for articular cartilage repair." Science Translational Medicine 17, no. 788 (2025): eads0848. [CrossRef]
  274. Yang, Sha, Huapeng Lin, and Cong Luo. "Meta-analysis of 3D printing applications in traumatic fractures." Frontiers in Surgery 8 (2021): 696391.
  275. Cosyn, Jan, Célien Eeckhout, Thomas De Bruyckere, Aryan Eghbali, Stijn Vervaeke, Faris Younes, and Véronique Christiaens. "A multi-centre randomized controlled trial comparing connective tissue graft with collagen matrix to increase soft tissue thickness at the buccal aspect of single implants: 1-year results." Journal of Clinical Periodontology 49, no. 9 (2022): 911-921.
  276. Zhang, Zhengdong, Pan Liu, Yu Song, Liang Ma, Yan Xu, Jie Lei, Bide Tong et al. "Addressing osteoblast senescence: Molecular pathways and the frontier of anti-ageing treatments." Clinical and Translational Medicine 15, no. 7 (2025): e70417.
  277. Zeng, Wenhui, Wei Liu, Lulu Zhang, Yiping Zhang, Yuxiang He, Weijuan Su, Peiying Huang et al. "Trends in osteoporosis assessment, diagnosis after fragility fractures, and treatment for hospitalized patients with osteoporosis or fragility fractures between 2012 and 2021." Archives of Osteoporosis 20, no. 1 (2025): 8.
  278. Reichman, Trevor W., James F. Markmann, Jon Odorico, Piotr Witkowski, John J. Fung, Martin Wijkstrom, Fouad Kandeel et al. "Stem Cell–Derived, Fully Differentiated Islets for Type 1 Diabetes." New England Journal of Medicine (2025).
  279. “Sernova Biotherapeutics Provides Update on Phase 1/2 Clinical Trial of Cell PouchTM Bio-Hybrid Organ for Treatment of Type 1 Diabetes | Sernova.Com,” April 1, 2025. https://sernova.com/press_releases/sernova-biotherapeutics-provides-update-on-phase-1-2-clinical-trial-of-cell-pouch-bio-hybrid-organ-for-treatment-of-type-1-diabetes/.
  280. “Vertex Announces Positive Results From Ongoing Phase 1/2 Study of VX-880 in Patients with Type 1 Diabetes.” News release. June 21, 2024. https://news.vrtx.com/news-releases/news-release-details/vertex-announces-positive-results-ongoing-phase-12-study-vx-880.
  281. Lu, Jiayi, Haojin Cheng, Keyu Chen, and Feng Zhang. "From bench to bedside: future prospects in stem cell therapy for diabetes." Journal of Translational Medicine 23, no. 1 (2025): 72. [CrossRef]
  282. Arte, Priyamvada Amol, Kanchanlata Tungare, Mustansir Bhori, Renitta Jobby, and Jyotirmoi Aich. "Treatment of type 2 diabetes mellitus with stem cells and antidiabetic drugs: a dualistic and future-focused approach." Human Cell 37, no. 1 (2024): 54-84. [CrossRef]
  283. Kumar, Dinesh, Rajni Tanwar, and Vrinda Gupta. "First-ever stem cell therapy restores insulin independence in type 1 diabetes: A medical milestone." World Journal of Stem Cells 17, no. 7 (2025): 106856. [CrossRef]
  284. Wu, Jiaying, Tuo Li, Meng Guo, Junsong Ji, Xiaoxi Meng, Tianlong Fu, Tengfei Nie et al. "Treating a type 2 diabetic patient with impaired pancreatic islet function by personalized endoderm stem cell-derived islet tissue." Cell Discovery 10, no. 1 (2024): 45.
  285. Shapiro, AM James, David Thompson, Thomas W. Donner, Melena D. Bellin, Willa Hsueh, Jeremy Pettus, Jon Wilensky et al. "Insulin expression and C-peptide in type 1 diabetes subjects implanted with stem cell-derived pancreatic endoderm cells in an encapsulation device." Cell Reports Medicine 2, no. 12 (2021).
  286. Keymeulen, Bart, Kaat De Groot, Daniel Jacobs-Tulleneers-Thevissen, David M. Thompson, Melena D. Bellin, Evert J. Kroon, Mark Daniels et al. "Encapsulated stem cell–derived β cells exert glucose control in patients with type 1 diabetes." Nature biotechnology 42, no. 10 (2024): 1507-1514.
  287. Gerace, Dario, Quan Zhou, Jennifer Hyoje-Ryu Kenty, Adrian Veres, Elad Sintov, Xi Wang, Kyle R. Boulanger, Hongfei Li, and Douglas A. Melton. "Engineering human stem cell-derived islets to evade immune rejection and promote localized immune tolerance." Cell Reports Medicine 4, no. 1 (2023).
  288. Thongsin, Nontaphat, Siriwal Suwanpitak, and Methichit Wattanapanitch. "CRISPR-Cas9-mediated disruption of B2M and CIITA genes eliminates HLA class I and II expression in human induced pluripotent stem cells (MUSIi001-A-2)." Stem cell research 71 (2023): 103138.
  289. Karpov, Dmitry S., Anastasiia O. Sosnovtseva, Svetlana V. Pylina, Asya N. Bastrich, Darya A. Petrova, Maxim A. Kovalev, Anastasija I. Shuvalova, Anna K. Eremkina, and Natalia G. Mokrysheva. "Challenges of CRISPR/cas-based cell therapy for type 1 diabetes: how not to engineer a “Trojan horse”." International Journal of Molecular Sciences 24, no. 24 (2023): 17320.
  290. Ph.D, Sandy Vogt. “New Data: Hypoimmune Islets Survive with No Immunosuppression.” Breakthrough T1D, August 8, 2025. https://www.breakthrought1d.org/news-and-updates/new-publication-engineered-islets-making-insulin-and-avoiding-immune-detection-in-first-person-treated/.
  291. Daly, Timothy, Kasper P. Kepp, and Bruno P. Imbimbo. "Are lecanemab and donanemab disease-modifying therapies?." Alzheimer's & Dementia 20, no. 9 (2024): 6659. [CrossRef]
  292. Sawamoto, Nobukatsu, Daisuke Doi, Etsuro Nakanishi, Masanori Sawamura, Takayuki Kikuchi, Hodaka Yamakado, Yosuke Taruno et al. "Phase I/II trial of iPS-cell-derived dopaminergic cells for Parkinson’s disease." Nature (2025): 1-7. [CrossRef]
  293. CiRA | Center for iPS Cell Research and Application, Kyoto University. “Pioneering a cure for Parkinson’s disease: First allogeneic iPS cell-based therapy demonstrates safety and efficacy|News & Events.” Accessed October 28, 2025. http://www.cira.kyoto-u.ac.jp/e.
  294. BlueRock Therapeutics LP. “BlueRock Therapeutics’ Investigational Cell Therapy Bemdaneprocel for Parkinson’s Disease Shows Positive Data at 24-Months.” News release, September 27, 2024. https://www.bluerocktx.com/bluerock-therapeutics-investigational-cell-therapy-bemdaneprocel-for-parkinsons-disease/.
  295. Tao, Ran, Chunmei Yue, Zhijie Guo, Wenke Guo, Yao Yao, Xianfa Yang, Zhen Shao et al. "Subtype-specific neurons from patient iPSCs display distinct neuropathological features of Alzheimer’s disease." Cell Regeneration 13, no. 1 (2024): 21.
  296. Yadegar, Abbas, Sepideh Pakpour, Fathima F. Ibrahim, Ali Nabavi-Rad, Laura Cook, Jens Walter, Anna M. Seekatz, Karen Wong, Tanya M. Monaghan, and Dina Kao. "Beneficial effects of fecal microbiota transplantation in recurrent Clostridioides difficile infection." Cell host & microbe 31, no. 5 (2023): 695-711.
  297. Vedanta Biosciences, Inc. “VE303.” Accessed October 28, 2025. https://www.vedantabio.com/pipeline-programs/ve303/.
  298. Times, Microbiome. “Vedanta Biosciences Publishes Additional Phase 2 VE303 Results in Nature Medicine - Microbiome Times Magazine,” January 27, 2025. https://www.microbiometimes.com/vedanta-biosciences-publishes-additional-phase-2-ve303-results-in-nature-medicine/.
  299. Menon, Rajita, Shakti K. Bhattarai, Emily Crossette, Amanda L. Prince, Bernat Olle, Jeffrey L. Silber, Vanni Bucci, Jeremiah Faith, and Jason M. Norman. "Multi-omic profiling a defined bacterial consortium for treatment of recurrent Clostridioides difficile infection." Nature Medicine 31, no. 1 (2025): 223-234.
  300. Cerna, Camila, Nicole Vidal-Herrera, Francisco Silva-Olivares, Daniela Álvarez, Camila González-Arancibia, Miltha Hidalgo, Pabla Aguirre et al. "Fecal microbiota transplantation from young-trained donors improves cognitive function in old mice through modulation of the gut-brain axis." Aging and disease 16, no. 6 (2024): 3649. [CrossRef]
  301. Conn, Meghan O., Erica N. DeJong, Daniel M. Marko, Russta Fayyazi, Dana Kukje Zada, Kevin P. Foley, Nicole G. Barra, Dawn ME Bowdish, and Jonathan D. Schertzer. "Microbiota protect against frailty and loss of skeletal muscle, and maintain inflammatory tone during aging in mice." American Journal of Physiology-Cell Physiology 328, no. 3 (2025): C887-C894. [CrossRef]
  302. Yang, Bo, Xinhui Li, Jiahui Wang, Yue Xu, Le Wang, Zhifeng Wu, Di Zhao et al. "The efficacy and safety of fecal microbiota transplantation in the treatment of sarcopenia: a retrospective study." Journal of Translational Medicine 23, no. 1 (2025): 645. [CrossRef]
  303. Lei, Jinghui, Zijuan Xin, Ning Liu, Taixin Ning, Ying Jing, Yicheng Qiao, Zan He et al. "Senescence-resistant human mesenchymal progenitor cells counter aging in primates." Cell (2025). [CrossRef]
  304. Liu, Haitao, Xu Zhang, Jiayue Liu, and Jianhua Qin. "Vascularization of engineered organoids." BMEMat 1, no. 3 (2023): e12031.
  305. Das, Suradip, Wisberty J. Gordián-Vélez, Harry C. Ledebur, Foteini Mourkioti, Panteleimon Rompolas, H. Isaac Chen, Mijail D. Serruya, and D. Kacy Cullen. "Innervation: the missing link for biofabricated tissues and organs." NPJ Regenerative medicine 5, no. 1 (2020): 11.
  306. Moy, Alan B., Anant Kamath, Sara Ternes, and Jay Kamath. "The challenges to advancing induced pluripotent stem cell-dependent cell replacement therapy." Medical research archives 11, no. 11 (2023): 4784.
  307. The Cryonics Institute. “Guide to Cryonics Procedures.” Accessed October 28, 2025. https://cryonics.org/guide-to-cryonics-procedures/.
  308. Vita-More, Natasha, and Daniel Barranco. "Persistence of long-term memory in vitrified and revived caenorhabditis elegans." Rejuvenation research 18, no. 5 (2015): 458-463.
  309. Abbas, Fatima, Silke Becker, Bryan W. Jones, Ludovic S. Mure, Satchidananda Panda, Anne Hanneken, and Frans Vinberg. "Revival of light signalling in the postmortem mouse and human retina." Nature 606, no. 7913 (2022): 351-357.
  310. Warner, Ross M., Jun Yang, Andrew Drake, Youngjoo Lee, Sarah Nemanic, David Scott, and Adam Z. Higgins. "Osmotic response during kidney perfusion with cryoprotectant in isotonic or hypotonic vehicle solution." PeerJ 11 (2023): e16323. [CrossRef]
  311. Matsui, Kenji, Yoshitaka Kinoshita, Yuka Inage, Naoto Matsumoto, Keita Morimoto, Yatsumu Saito, Tsuyoshi Takamura et al. "Cryopreservation of fetal porcine kidneys for xenogeneic regenerative medicine." Journal of Clinical Medicine 12, no. 6 (2023): 2293. [CrossRef]
  312. Best, Benjamin P. "Vascular and neuronal ischemic damage in cryonics patients." Rejuvenation research 15, no. 2 (2012): 165-169. [CrossRef]
  313. McKenzie, Andrew T., Ariel Zeleznikow-Johnston, Jordan S. Sparks, Oge Nnadi, John Smart, Keith Wiley, Michael A. Cerullo et al. "Structural brain preservation: a potential bridge to future medical technologies." Frontiers in medical technology 6 (2024): 1400615. [CrossRef]
  314. Dorkenwald, Sven, Arie Matsliah, Amy R. Sterling, Philipp Schlegel, Szi-Chieh Yu, Claire E. McKellar, Albert Lin et al. "Neuronal wiring diagram of an adult brain." Nature 634, no. 8032 (2024): 124-138.
  315. Horvath, Steve. "DNA methylation age of human tissues and cell types." Genome biology 14, no. 10 (2013): 3156.
  316. Hannum, Gregory, Justin Guinney, Ling Zhao, L. I. Zhang, Guy Hughes, SriniVas Sadda, Brandy Klotzle et al. "Genome-wide methylation profiles reveal quantitative views of human aging rates." Molecular cell 49, no. 2 (2013): 359-367.
  317. Zhang, Qian, Costanza L. Vallerga, Rosie M. Walker, Tian Lin, Anjali K. Henders, Grant W. Montgomery, Ji He et al. "Improved precision of epigenetic clock estimates across tissues and its implication for biological ageing." Genome medicine 11, no. 1 (2019): 54.
  318. Levine, Morgan E., Ake T. Lu, Austin Quach, Brian H. Chen, Themistocles L. Assimes, Stefania Bandinelli, Lifang Hou et al. "An epigenetic biomarker of aging for lifespan and healthspan." Aging (albany NY) 10, no. 4 (2018): 573.
  319. Higgins-Chen, Albert T., Kyra L. Thrush, Yunzhang Wang, Christopher J. Minteer, Pei-Lun Kuo, Meng Wang, Peter Niimi et al. "A computational solution for bolstering reliability of epigenetic clocks: implications for clinical trials and longitudinal tracking." Nature aging 2, no. 7 (2022): 644-661.
  320. Lu, Ake T., Austin Quach, James G. Wilson, Alex P. Reiner, Abraham Aviv, Kenneth Raj, Lifang Hou et al. "DNA methylation GrimAge strongly predicts lifespan and healthspan." Aging (albany NY) 11, no. 2 (2019): 303. [CrossRef]
  321. Lu, Ake T., Alexandra M. Binder, Joshua Zhang, Qi Yan, Alex P. Reiner, Simon R. Cox, Janie Corley et al. "DNA methylation GrimAge version 2." Aging (Albany NY) 14, no. 23 (2022): 9484. [CrossRef]
  322. Kriukov, Dmitrii, Evgeniy Efimov, Ekaterina Kuzmina, Anastasiia Dudkovskaia, Ekaterina E. Khrameeva, and Dmitry V. Dylov. "ComputAgeBench: epigenetic aging clocks benchmark." In Proceedings of the 31st ACM SIGKDD Conference on Knowledge Discovery and Data Mining V. 2, pp. 5560-5570. 2025.
  323. Lu, Ake T., Zhe Fei, Amin Haghani, Todd R. Robeck, J. A. Zoller, C. Z. Li, R. Lowe et al. "Universal DNA methylation age across mammalian tissues." Nature aging 3, no. 9 (2023): 1144-1166. [CrossRef]
  324. Latumalea, Djakim, Maximilian Unfried, Diogo Barardo, Jan Gruber, and Brian K. Kennedy. "DoliClock: a lipid-based aging clock reveals accelerated aging in neurological disorders." Aging (Albany NY) 17, no. 6 (2025): 1405.
  325. Fong, Sheng, Kamil Pabis, Djakim Latumalea, Nomuundari Dugersuren, Maximilian Unfried, Nicholas Tolwinski, Brian Kennedy, and Jan Gruber. "Principal component-based clinical aging clocks identify signatures of healthy aging and targets for clinical intervention." Nature Aging 4, no. 8 (2024): 1137-1152.
  326. Fong, Sheng, Kirill A. Denisov, Anastasiia A. Nefedova, Brian K. Kennedy, and Jan Gruber. "LinAge2: providing actionable insights and benchmarking with epigenetic clocks." npj Aging 11, no. 1 (2025): 29.
  327. Buckley, Matthew T., Eric D. Sun, Benson M. George, Ling Liu, Nicholas Schaum, Lucy Xu, Jaime M. Reyes et al. "Cell-type-specific aging clocks to quantify aging and rejuvenation in neurogenic regions of the brain." Nature Aging 3, no. 1 (2023): 121-137.
  328. Trapp, Alexandre, Csaba Kerepesi, and Vadim N. Gladyshev. "Profiling epigenetic age in single cells." Nature Aging 1, no. 12 (2021): 1189-1201.
  329. Urban, Anatoly, Denis Sidorenko, Diana Zagirova, Ekaterina Kozlova, Aleksandr Kalashnikov, Stefan Pushkov, Vladimir Naumov et al. "Precious1GPT: multimodal transformer-based transfer learning for aging clock development and feature importance analysis for aging and age-related disease target discovery." Aging (Albany NY) 15, no. 11 (2023): 4649.
  330. Sidorenko, Denis, Stefan Pushkov, Akhmed Sakip, Geoffrey Ho Duen Leung, Sarah Wing Yan Lok, Anatoly Urban, Diana Zagirova et al. "Precious2GPT: the combination of multiomics pretrained transformer and conditional diffusion for artificial multi-omics multi-species multi-tissue sample generation." npj Aging 10, no. 1 (2024): 37. [CrossRef]
  331. Galkin, Fedor, Vladimir Naumov, Stefan Pushkov, Denis Sidorenko, Anatoly Urban, Diana Zagirova, Khadija M. Alawi et al. "Precious3GPT: Multimodal Multi-Species Multi-Omics Multi-Tissue Transformer for Aging Research and Drug Discovery." bioRxiv (2024): 2024-07.
  332. Ying, Kejun, Hanna Liu, Andrei E. Tarkhov, Marie C. Sadler, Ake T. Lu, Mahdi Moqri, Steve Horvath, Zoltán Kutalik, Xia Shen, and Vadim N. Gladyshev. "Causality-enriched epigenetic age uncouples damage and adaptation." Nature aging 4, no. 2 (2024): 231-246. [CrossRef]
  333. Kamya, Petrina, Ivan V. Ozerov, Frank W. Pun, Kyle Tretina, Tatyana Fokina, Shan Chen, Vladimir Naumov et al. "PandaOmics: an AI-driven platform for therapeutic target and biomarker discovery." Journal of chemical information and modeling 64, no. 10 (2024): 3961-3969. [CrossRef]
  334. Ivanenkov, Yan A., Daniil Polykovskiy, Dmitry Bezrukov, Bogdan Zagribelnyy, Vladimir Aladinskiy, Petrina Kamya, Alex Aliper, Feng Ren, and Alex Zhavoronkov. "Chemistry42: an AI-driven platform for molecular design and optimization." Journal of chemical information and modeling 63, no. 3 (2023): 695-701.
  335. Aliper, Alex, Roman Kudrin, Daniil Polykovskiy, Petrina Kamya, Elena Tutubalina, Shan Chen, Feng Ren, and Alex Zhavoronkov. "Prediction of clinical trials outcomes based on target choice and clinical trial design with multi-modal artificial intelligence." Clinical Pharmacology & Therapeutics 114, no. 5 (2023): 972-980.
  336. Wagle, Manoj M., Chunlei Liu, Zunpeng Liu, Yongheng Wang, Manolis Kellis, Ellis Patrick, and Pengyi Yang. "Interpretable deep generative ensemble learning for single-cell omics with Hydra." bioRxiv (2025): 2025-08.
  337. Xiong, Lei, Tianlong Chen, and Manolis Kellis. "scCLIP: Multi-modal single-cell contrastive learning integration pre-training." In NeurIPS 2023 AI for Science Workshop. 2023.
  338. Kong, Zhenglun, Mufan Qiu, John Boesen, Xiang Lin, Sukwon Yun, Tianlong Chen, Manolis Kellis, and Marinka Zitnik. "SPATIA: Multimodal Model for Prediction and Generation of Spatial Cell Phenotypes." ArXiv (2025): arXiv-2507.
  339. Queen, Owen, Yepeng Huang, Robert Calef, Valentina Giunchiglia, Tianlong Chen, George Dasoulas, LeAnn Tai et al. "Procyon: A multimodal foundation model for protein phenotypes." BioRxiv (2024): 2024-12.
  340. Quon, Jacob, Alex Reynolds, Nalu Tripician, Eric Rynes, Athanasios Teodosiadis, Manolis Kellis, and Wouter Meuleman. "Epilogos: information-theoretic navigation of multi-tissue functional genomic annotations." bioRxiv (2025).
  341. Johnson, Kevin B., Wei-Qi Wei, Dilhan Weeraratne, Mark E. Frisse, Karl Misulis, Kyu Rhee, Juan Zhao, and Jane L. Snowdon. "Precision medicine, AI, and the future of personalized health care." Clinical and translational science 14, no. 1 (2021): 86-93. [CrossRef]
  342. Linzer, Mark, Asaf Bitton, Shin-Ping Tu, Margaret Plews-Ogan, Karen R. Horowitz, Mark D. Schwartz, and Association of Chiefs and Leaders in General Internal Medicine (ACLGIM) Writing Group*. "The end of the 15–20 minute primary care visit." Journal of general internal medicine 30, no. 11 (2015): 1584-1586. [CrossRef]
  343. Bohata, Kirsti, Alexandra Jones, Mike Mantin, and Steven Thompson. “Medicalising Miners? Medicine, Care and Rehabilitation.” In Disability in Industrial Britain: A Cultural and Literary History of Impairment in the Coal Industry, 1880–1948 [Internet]. Manchester University Press, 2020. https://www.ncbi.nlm.nih.gov/books/NBK553581/.
  344. Wojtara, Magda, Emaan Rana, Taibia Rahman, Palak Khanna, and Heshwin Singh. "Artificial intelligence in rare disease diagnosis and treatment." Clinical and Translational science 16, no. 11 (2023): 2106-2111.
  345. He, Ting, Kory Kreimeyer, Mimi Najjar, Jonathan Spiker, Maria Fatteh, Valsamo Anagnostou, and Taxiarchis Botsis. "Artificial Intelligence-assisted Biomedical Literature Knowledge Synthesis to Support Decision-making in Precision Oncology." In AMIA Annual Symposium Proceedings, vol. 2024, p. 513. 2025.
  346. Esteva, Andre, Brett Kuprel, Roberto A. Novoa, Justin Ko, Susan M. Swetter, Helen M. Blau, and Sebastian Thrun. "Dermatologist-level classification of skin cancer with deep neural networks." nature 542, no. 7639 (2017): 115-118.
  347. Liopyris, Konstantinos, Stamatios Gregoriou, Julia Dias, and Alexandros J. Stratigos. "Artificial intelligence in dermatology: challenges and perspectives." Dermatology and Therapy 12, no. 12 (2022): 2637-2651.
  348. “АЙРА Лабс.” Accessed October 28, 2025. https://ira-labs.ru/.
  349. Neuron23 Inc. “A Phase 2, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety and Efficacy of NEU-411 in Companion Diagnostic-Positive Participants With Early Parkinson's Disease.” Clinical trial registration. clinicaltrials.gov, October 14, 2025. https://clinicaltrials.gov/study/NCT06680830.
  350. Neuron23. “Neuron23.” Accessed October 28, 2025. https://neuron23.com/neuron23-announces-96-5-million-series-d-financing-and-first-patient-dosed-in-global-phase-2-neulark-clinical-trial-of-neu-411-for-early-parkinsons-disease/.
  351. Wasserkort, Reinhold, Alexandra Kalmar, Gabor Valcz, Sandor Spisak, Manuel Krispin, Kinga Toth, Zsolt Tulassay, Andrew Z. Sledziewski, and Bela Molnar. "Aberrant septin 9 DNA methylation in colorectal cancer is restricted to a single CpG island." BMC cancer 13, no. 1 (2013): 398. [CrossRef]
  352. Hariharan, Rohit, and Mark Jenkins. "Utility of the methylated SEPT9 test for the early detection of colorectal cancer: a systematic review and meta-analysis of diagnostic test accuracy." BMJ open gastroenterology 7, no. 1 (2020): e000355. [CrossRef]
  353. ToxGenSolutions. “ToxGenSolution BV.” Accessed October 28, 2025. https://toxgensolutions.eu/.
  354. DiamiR. “microRNA Testing.” Accessed October 28, 2025. https://www.diamirbio.com/micrornatesting/.
  355. “Neuro-miR – Centre for Systems Medicine.” Accessed October 28, 2025. https://www.systemsmedicineireland.ie/research/neuroscience/neuro-mir/.
  356. Azam, Hafiz Muhammad Husnain, Rosa Ilse Rößling, Christiane Geithe, Muhammad Moman Khan, Franziska Dinter, Katja Hanack, Harald Prüß et al. "MicroRNA biomarkers as next-generation diagnostic tools for neurodegenerative diseases: a comprehensive review." Frontiers in molecular neuroscience 17 (2024): 1386735.
  357. Commissioner, Office of the. “FDA Clears First Blood Test Used in Diagnosing Alzheimer’s Disease.” FDA, May 28, 2025. https://www.fda.gov/news-events/press-announcements/fda-clears-first-blood-test-used-diagnosing-alzheimers-disease.
  358. “The Anatomy of a Personal Health Agent.” Accessed October 28, 2025. https://research.google/blog/the-anatomy-of-a-personal-health-agent/.
  359. Tempus. “Tempus | AI-Enabled Precision Medicine.” Accessed October 28, 2025. https://www.tempus.com/.
  360. “Luria Health | Be the Driver of Your Health.” Accessed October 28, 2025. https://luria.health/.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated