Why We Age: Stem Cell Exhaustion
Stem cells are essential for maintaining the body’s ability to repair and regenerate tissues. These unique cells can divide and produce specialized cell types, replacing damaged or dying cells and supporting tissue growth. Found in various parts of the body, including bone marrow, muscles, and skin, stem cells play a central role in preserving the health and function of organs over a lifetime [1].
One of stem cells’ key features is their ability to remain in a state of readiness, called quiescence until activated by signals that trigger their division and specialization. This balance between dormancy and activation ensures that stem cell reserves are preserved for future use while still responding to immediate repair needs. Stem cells help prevent organ deterioration and contribute to long-term health by sustaining tissue homeostasis, or equilibrium [1].
Stem cells are broadly categorized into embryonic stem cells, which have the potential to develop into any cell type in the body, and adult stem cells, which are more specialized and limited to regenerating specific tissues. Adult stem cells, such as hematopoietic stem cells in the bone marrow, are crucial for producing blood and immune cells. Similarly, mesenchymal stem cells contribute to the repair of bone, cartilage, and fat tissues, while neural stem cells support brain function and cognitive health [1].
The ability of stem cells to sustain tissue repair declines with age. Over time, stem cells experience stress, accumulate damage, and lose their capacity to regenerate effectively. This process, known as stem cell exhaustion, is now recognized as a hallmark of aging. It impairs tissue maintenance and increases disease susceptibility, including anemia, osteoporosis, and neurodegenerative conditions. Understanding the mechanisms behind this decline and developing therapies to counteract it have become critical goals in regenerative medicine [2].
Causes and mechanisms
Stem cell exhaustion is driven by multiple biological processes that gradually impair stem cells’ ability to function effectively. Over time, stem cells are exposed to internal and external stresses that compromise their genetic stability, metabolic function, and ability to repair damage [2]. A close look at these mechanisms provides insight into why stem cells decline with age and emphasizes opportunities for developing therapies to counteract this process.
DNA damage and accumulation of mutations
One of the primary causes of stem cell exhaustion is the accumulation of DNA damage. Throughout life, stem cells are exposed to environmental factors such as radiation, toxins, and metabolic byproducts that generate reactive oxygen species. These reactive molecules cause oxidative stress, damaging DNA, proteins, and cellular membranes. Although stem cells possess repair mechanisms to fix DNA damage, these systems become less efficient with age, allowing errors to accumulate over time [3].
The accumulation of DNA mutations not only reduces stem cells’ regenerative capacity but also increases the risk of abnormal cell growth and cancer. As their ability to maintain genetic stability declines, stem cells may enter a state of senescence, in which they stop dividing altogether. This process serves as a protective mechanism to prevent damaged cells from propagating but also contributes to the gradual depletion of functional stem cells [3].
Epigenetic alterations
In addition to DNA damage, stem cell exhaustion is influenced by epigenetic changes that alter gene expression without modifying the genetic code. Epigenetic mechanisms, such as DNA methylation and histone modifications, regulate which genes are active or inactive, controlling the identity and function of stem cells [4].
With age, these epigenetic patterns become disrupted, leading to inappropriate activation or silencing of genes. For example, DNA methylation may suppress genes involved in DNA repair or cell cycle regulation, impairing stem cells’ ability to maintain tissue health. Similarly, changes in histone modifications can alter chromatin structure, making it more difficult for transcription factors to access DNA and activate necessary genes [4].
These epigenetic alterations lock stem cells into dysfunctional states, reducing their flexibility and regenerative capacity. Furthermore, changes in chromatin organization may contribute to a loss of cell identity, causing stem cells to lose their ability to differentiate into specialized cell types. This process accelerates tissue deterioration and limits the effectiveness of repair mechanisms [4].
DNA damage and epigenetic changes create a feedback loop that reinforces stem cell exhaustion. As cells become less capable of repairing themselves and maintaining proper gene expression, their ability to sustain tissue health diminishes, highlighting the need for therapies that target these underlying mechanisms [4].
Metabolic dysfunction and mitochondrial decline
Metabolic dysfunction plays a central role in stem cell exhaustion as aging cells experience a decline in their ability to produce energy efficiently. Mitochondria, often called the cell’s powerhouses, generate adenosine triphosphate (ATP), which fuels cellular activities. Over time, mitochondrial function deteriorates, reducing energy output and increasing harmful byproducts such as reactive oxygen species. These byproducts cause oxidative damage to proteins, lipids, and DNA, further impairing cellular processes and accelerating stem cell aging [5].
Metabolic pathways, particularly AMP-activated protein kinase (AMPK) and sirtuins like SIRT1, are crucial regulators of energy balance and stress responses in stem cells. AMPK acts as a sensor for low energy levels, activating pathways that restore ATP production and repair cellular damage. SIRT1, on the other hand, influences gene expression, promotes mitochondrial health, and protects cells from oxidative stress. With age, the activity of these pathways declines, leaving stem cells vulnerable to energy depletion and metabolic stress. As a result, aging stem cells struggle to balance self-renewal and differentiation, contributing to exhaustion and reduced regenerative capacity [5, 6].
Inflammaging and chronic inflammation
Inflammaging, a term used to describe the chronic low-grade inflammation associated with aging, is another factor that accelerates stem cell exhaustion. Over time, the immune system produces elevated levels of inflammatory cytokines, including interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α). These molecules create a pro-inflammatory environment that disrupts normal stem cell signaling and function [7-9].
Inflammatory cytokines interfere with signaling pathways that regulate stem cell quiescence and activation. For example, TNF-α has been shown to impair hematopoietic stem cell function by promoting differentiation over self-renewal, depleting the stem cell pool. IL-6, another key cytokine, drives the expansion of myeloid cells at the expense of lymphoid cells, leading to immune system imbalances. Chronic exposure to these signals increases oxidative stress, accelerates DNA damage, and promotes senescence, further contributing to stem cell dysfunction [9].
The pro-inflammatory environment also disrupts the stem cell niche, the supportive microenvironment required for stem cell maintenance. The niche becomes less effective in aged tissues at shielding stem cells from inflammatory signals, exacerbating their decline. This process creates a feedback loop in which inflammation promotes stem cell exhaustion and exhausted stem cells fail to repair tissues, perpetuating damage and dysfunction [8].
Telomere shortening and senescence
Telomeres, the protective caps at the ends of chromosomes, are critical in preserving genetic stability during cell division. Each time a cell divides, its telomeres shorten slightly. In stem cells, mechanisms like telomerase activity help replenish telomere length, but this capacity diminishes with age. Once telomeres become critically short, cells enter a state of senescence or undergo programmed cell death (apoptosis) [10].
Senescent stem cells lose their ability to divide and contribute to tissue repair, while also secreting inflammatory factors that further damage the surrounding microenvironment. This condition, the senescence-associated secretory phenotype (SASP), amplifies chronic inflammation and accelerates tissue aging. In addition to limiting regenerative potential, telomere erosion makes stem cells more susceptible to DNA damage, compounding the effects of oxidative stress and metabolic dysfunction [11, 12].
Telomere shortening acts as a biological clock, signaling when cells have reached the end of their replicative lifespan. While this mechanism helps prevent the propagation of damaged cells, it also leads to a gradual depletion of functional stem cells, particularly in tissues that rely heavily on constant regeneration, such as the skin, gut, and blood. Therapies that preserve telomere length or enhance telomerase activity are being investigated as potential strategies to delay stem cell exhaustion and extend tissue health [10].
By examining the roles of metabolic decline, chronic inflammation, and telomere shortening, researchers are uncovering new approaches to mitigate stem cell exhaustion and improve regenerative therapies for aging and disease.
Changes in the stem cell niche
The stem cell niche, a specialized microenvironment that supports stem cell function, plays a critical role in maintaining the balance between quiescence, activation, and differentiation [13]. However, aging contributes to this niche’s significant deterioration, contributing to stem cell exhaustion. The bone marrow microenvironment, which houses hematopoietic stem cells, undergoes structural and functional changes that reduce its ability to support stem cell maintenance [14].
One notable shift is the conversion of supportive stromal cells into inflammatory fat cells. As stromal cells lose their regenerative potential, they are increasingly replaced by adipocytes, or fat cells, which secrete inflammatory signals. This transformation alters the composition of the niche, creating a pro-inflammatory environment that disrupts stem cell signaling and impairs its ability to regenerate. The loss of structural support and growth factors from stromal cells further exacerbates stem cell decline [15, 16].
In addition to physical changes, aged niches accumulate oxidative stress and DNA damage, weakening the ability of niche cells to provide protective signals. These alterations compromise the ability of stem cells to remain quiescent, forcing them into cycles of activation and proliferation that accelerate exhaustion [12]. Looking into how these niche changes affect stem cell behavior has opened pathways for research into treatments that rejuvenate the microenvironment and restore its supportive function.
Specific tissue types
Hematopoietic stem cells
Hematopoietic stem cells (HSCs) produce all blood and immune cells throughout life. As these cells age, their ability to generate new blood cells declines, leading to reduced immunity and an increased risk of anemia. Older HSCs tend to favor myeloid cell production over lymphoid cells, resulting in imbalances that weaken the immune response. This myeloid skewing reduces the diversity of immune cells, leaving the body more vulnerable to infections and inflammatory conditions [17].
Aging HSCs also accumulate DNA damage and epigenetic changes, which impair their ability to self-renew and differentiate appropriately. These defects increase the risk of developing blood disorders, including myelodysplastic syndromes and leukemia. Furthermore, the bone marrow niche’s decline exacerbates HSC dysfunction, as it fails to provide the necessary signals to maintain healthy stem cell populations. Restoring HSC function is a focus of regenerative medicine, with therapies targeting inflammation, DNA repair, and metabolic pathways showing promise [17].
Neural stem cells
Neural stem cells reside in the brain and are essential for generating new neurons and supporting cognitive function. Aging significantly reduces neural stem cell activity, contributing to a decline in the production of new nerve cells (neurogenesis). This reduction is linked to cognitive decline, memory loss, and an increased risk of neurodegenerative diseases such as Alzheimer’s and Parkinson’s [18].
Factors such as inflammation, oxidative stress, and changes in metabolic signaling influence the decline of neural stem cells. Chronic inflammation in the brain, often associated with microglial activation, creates a hostile environment that impairs neural stem cell function. Additionally, mitochondrial dysfunction reduces the energy needed for neurogenesis, further limiting stem cell activity [19].
In Alzheimer’s disease, the loss of neural stem cells contributes to the buildup of amyloid plaques and tau tangles, which are hallmarks of the disease that interfere with neural communication [20, 21]. In Parkinson’s disease, the decline in dopaminergic neurons is partially linked to impaired neural stem cell replacement [22]. By addressing the changes in the stem cell niche and understanding the impact on specific tissue types, researchers aim to develop therapies that can reverse stem cell exhaustion and restore regenerative potential in aging tissues.
Mesenchymal stem cells
Mesenchymal stem cells (MSCs) are multipotent cells found in bone marrow, fat, and other tissues. They play a critical role in maintaining skeletal integrity and promoting wound healing. However, with age, MSCs experience a decline in number and function, reducing their ability to repair bone and cartilage. This decline contributes to age-related conditions such as osteoporosis, in which bones become brittle and prone to fractures. Impaired MSC function also leads to slower healing of wounds, as these cells are less effective in coordinating tissue regeneration [23].
Metabolic dysfunction, oxidative stress, and inflammatory signals within their microenvironment influence MSC deterioration. MSCs often exhibit reduced proliferation rates in aging tissues and impaired differentiation capacity, limiting their ability to form new bone or cartilage. Additionally, changes in the extracellular matrix surrounding MSCs further restrict their ability to migrate and interact with damaged tissues. Researchers are exploring therapies that restore MSC activity, including treatments that target inflammation, metabolic pathways, and extracellular signaling to rejuvenate these vital stem cells [24].
Muscle stem cells (satellite cells)
Muscle stem cells, or satellite cells, are essential for muscle repair and regeneration. These cells reside in skeletal muscle and are activated in response to injury or stress, dividing and differentiating to form new muscle fibers. With age, satellite cells become less responsive to activation signals, leading to diminished muscle regeneration. This decline is a major contributor to sarcopenia, a condition characterized by the loss of muscle mass and strength, which increases the risk of frailty and physical disability [25].
Aging satellite cells become fewer in number and have impaired self-renewal capacity. They also experience disruptions in signaling pathways, including decreased activation of growth factors and increased inflammatory signals that interfere with regeneration. Mitochondrial dysfunction and oxidative stress further compromise their ability to maintain energy balance, making it harder for these cells to respond effectively to tissue damage [25].
Efforts to combat satellite cell exhaustion focus on enhancing their regenerative capacity through exercise, nutritional strategies, and pharmacological agents that improve mitochondrial function and reduce inflammation. Emerging therapies, including gene editing and stem cell-derived factors, aim to restore satellite cell function and promote muscle repair in aging people [26].
Therapeutic approaches
Lifestyle Interventions
Lifestyle modifications, such as exercise and dietary strategies, have shown promise in improving stem cell function and delaying exhaustion. Regular physical activity has been demonstrated to enhance stem cell proliferation, particularly in muscle and bone tissues. Exercise stimulates blood flow, increases oxygen delivery, and activates signaling pathways that promote cell growth and repair. Resistance training has been linked to improved muscle stem cell activation, supporting muscle regeneration and reducing the risk of sarcopenia [27-29].
Caloric restriction and intermittent fasting have also been studied for their effects on stem cell health. These dietary interventions reduce inflammation, lower oxidative stress, and activate metabolic pathways that improve cellular repair. Caloric restriction stimulates AMPK and SIRT1 activity, enhancing mitochondrial function and promoting DNA repair mechanisms. Fasting has been shown to increase the production of stem cells in the gut and immune system, improving tissue maintenance and immune resilience [30].
By incorporating exercise and dietary changes, people may be able to slow the progression of stem cell exhaustion and support regenerative health as they age. Researchers continue investigating how lifestyle interventions can be optimized to enhance stem cell function and delay the onset of age-related diseases.
Pharmacological therapies
Pharmacological approaches offer promising strategies to combat stem cell exhaustion by targeting cellular damage, inflammation, and metabolic dysfunction. Senolytics, a class of drugs designed to eliminate senescent cells, have gained attention for their ability to rejuvenate tissues. Senescent cells accumulate with age and release inflammatory molecules that damage surrounding tissues and impair stem cell function. By selectively clearing these cells, senolytics reduce inflammation and promote tissue regeneration, creating a more supportive environment for stem cell activity [31, 32].
Another pharmacological approach focuses on boosting mitochondrial function using NAD+ precursors. Nicotinamide adenine dinucleotide (NAD+) is a coenzyme essential for energy production and DNA repair. Levels of NAD+ decline with age, contributing to mitochondrial dysfunction and metabolic stress. Supplementation with NAD+ precursors, such as nicotinamide riboside (NR) or nicotinamide mononucleotide (NMN), has restored mitochondrial health and improved stem cell function in preclinical studies [33-35].
Drugs like metformin and resveratrol also show potential for improving metabolic health and enhancing stem cell longevity. Metformin, commonly used to treat diabetes, activates AMPK, a key regulator of energy balance, and reduces oxidative stress. Resveratrol, a compound found in grapes, activates SIRT1, which supports mitochondrial function and reduces inflammation. Both compounds have demonstrated anti-aging effects, making them promising candidates for preserving stem cell health [36, 37].
Gene editing and epigenetic modifications
Gene editing technologies have opened new possibilities for repairing genetic damage and restoring the function of aged stem cells. CRISPR-Cas9, a widely used gene-editing tool, allows scientists to precisely modify DNA sequences, correcting mutations that impair cell function [38]. This technology can potentially address inherited diseases and age-related genetic damage, restoring stem cells’ ability to regenerate tissues.
Epigenetic modifications, which regulate gene expression without altering the genetic code, are also being explored to reverse stem cell aging [4]. Techniques such as histone modification and DNA methylation adjustments can reset gene expression patterns, promoting a more youthful state in aged cells [39]. Reprogramming aged cells into induced pluripotent stem cells (iPSCs) is another approach that restores their ability to differentiate into various cell types. iPSCs may be a renewable source of healthy stem cells that can be used for transplantation and tissue repair [40].
Researchers are also investigating partial reprogramming, which rejuvenates aged cells without completely erasing their identities. This approach may allow for targeted restoration of function in specific tissues, minimizing risks associated with complete reprogramming, such as tumor formation [41, 42].
Stem cell-based therapies
Stem cell-based therapies remain a cornerstone of regenerative medicine, offering direct tissue repair and restoration solutions. Hematopoietic stem cell transplantation has been widely used to treat blood disorders, including leukemia and anemia. By replacing damaged or dysfunctional blood-forming cells with healthy ones, this therapy restores normal blood cell production and immune function [43].
MSC transplantation is another promising approach for repairing bone, cartilage, and connective tissues. MSCs secrete growth factors and anti-inflammatory molecules that promote tissue regeneration and modulate immune responses. Clinical trials continue to explore their effectiveness in treating osteoarthritis, cardiovascular disease, and autoimmune disorders [44, 45].
Induced pluripotent stem cells (iPSCs) represent an innovative advancement in stem cell therapies. These cells are reprogrammed from adult cells to regain pluripotency, allowing them to differentiate into any cell type. iPSCs provide a customizable and potentially limitless source of stem cells for regenerative medicine. They are being studied for treating neurodegenerative diseases, heart conditions, and metabolic disorders [40].
While stem cell transplantation holds significant potential, challenges like immune rejection, scalability, and safety concerns remain [46]. Advances in gene editing and biomaterials are being integrated into these therapies to enhance their effectiveness and reliability. Together, these approaches provide a multifaceted strategy for addressing stem cell exhaustion and improving regenerative capacity in aging tissues [47].
Stem cell-conditioned media (secretome therapy)
Stem cell-conditioned media, often called secretome therapy, harnesses the regenerative properties of stem cells without requiring direct transplantation. Instead, this approach relies on the bioactive molecules secreted by stem cells, including growth factors, cytokines, and extracellular vesicles, to promote healing and reduce inflammation. These secreted components influence surrounding cells, enhancing tissue repair and modulating immune responses [48, 49].
One of the primary advantages of conditioned media is its reduced risk of immune rejection and tumor formation compared to live-cell therapies. Because it does not involve the introduction of living cells, conditioned media circumvents many of the challenges associated with stem cell transplantation, such as immune compatibility and safety concerns. This makes it a more accessible and scalable option for therapeutic use.
Conditioned media has shown promise in various applications, including cardiovascular repair [50], neuroprotection, and wound healing [48]. In cardiovascular research, it has been demonstrated to improve blood vessel formation and reduce tissue damage following heart attacks. In neurological studies, it has shown the potential to protect neurons and promote recovery in conditions such as stroke and neurodegenerative diseases [51]. For wound healing, the growth factors and cytokines within the media accelerate cell migration, collagen production, and tissue remodeling, making it a valuable tool for skin regeneration [48].
Stress erythropoiesis as a model
Stress erythropoiesis provides a useful model for understanding how stem cells respond to injury and disease. It refers to the process by which the body rapidly increases red blood cell production in response to anemia or other forms of stress. This process relies on activating progenitor cells primed to produce erythrocytes, demonstrating the potential for targeted therapies that stimulate specific stem cell populations [52].
Enhancing erythropoiesis has been explored as a therapeutic strategy for addressing anemia and blood disorders. Treatments that mimic the signals involved in stress erythropoiesis, such as erythropoietin and stem cell-derived growth factors, can promote red blood cell production and improve recovery following blood loss or bone marrow failure. By leveraging the principles of stress erythropoiesis, researchers aim to develop therapies that enhance the regenerative capacity of hematopoietic stem cells and address age-related declines in blood cell production [52].
Challenges in therapeutic development
Despite advances in stem cell research, translating therapies into clinical practice remains challenging. One major hurdle is the variability in stem cell behavior, which arises from differences in donor sources, culture conditions, and genetic backgrounds. This variability makes it difficult to standardize treatments and ensure consistent outcomes [53].
Standardization in therapy development is essential to address these issues. Researchers are working to create reproducible protocols for isolating, expanding, and modifying stem cells to maintain their therapeutic potential. Advances in quality control and manufacturing techniques are also being implemented to ensure that therapies meet the safety and efficacy standards required for clinical use [53].
Complexity of pathways and regulation
Stem cell function is governed by intricate signaling networks coordinating quiescence, activation, and differentiation. These pathways ensure that stem cells remain dormant when not needed, preserving their long-term viability, but can rapidly activate when repair or regeneration is required. Maintaining this balance is crucial, as excessive activation may lead to stem cell depletion, while prolonged quiescence can impair tissue repair and regeneration [54].
Key regulatory pathways include Wnt, Notch, and TGF-β signaling, which guide stem cell fate decisions. Crosstalk between these pathways adds complexity, making it challenging to design therapies that target a single mechanism without unintended consequences. For example, stimulating one pathway to promote regeneration may unintentionally suppress another, reducing stem cell survival. Research continues to explore methods to fine-tune these pathways, enabling precise control over stem cell activity without triggering exhaustion or dysfunction [55, 56].
Ethical and safety concerns
Ethical considerations are central to developing stem cell therapies, particularly those involving embryonic stem cells or genetic modifications. Using embryonic stem cells raises debates about the moral implications of sourcing cells from embryos, leading researchers to focus more on induced pluripotent stem cells (iPSCs) as an alternative. However, iPSCs carry their safety risks, including the potential for tumor formation due to their highly proliferative nature [57].
Gene editing technologies, such as CRISPR, also present ethical challenges. While they offer powerful tools for correcting genetic defects, concerns remain about unintended off-target effects and heritable genetic changes. These modifications may have unpredictable consequences, raising questions about long-term safety and the ethics of altering the human genome [58].
To address these concerns, regulatory frameworks emphasize rigorous preclinical testing, transparency in research, and public dialogue to build trust. Ensuring that therapies adhere to ethical guidelines while advancing scientific innovation remains a delicate balance.
Regulatory hurdles
The development and approval of stem cell therapies face significant regulatory challenges. These treatments require extensive preclinical testing to demonstrate safety and efficacy before progressing to clinical trials. Regulatory agencies, such as the FDA, require robust data on dosing, manufacturing consistency, and long-term effects, which can delay approvals and increase costs [59, 60].
Standardization is another obstacle, as variations in cell culture conditions, donor sources, and processing techniques can affect therapeutic outcomes. Establishing uniform protocols and quality control measures is essential for ensuring reproducibility and scalability.
Clinical trials for stem cell therapies often face additional scrutiny due to their potential for immune rejection, tumor formation, and unpredictable responses [59]. As a result, researchers must demonstrate therapeutic effectiveness and long-term safety in diverse patient populations [60]. Collaboration between scientists, regulatory bodies, and biotechnology companies is critical to overcoming these hurdles and bringing stem cell therapies to broader clinical use.
Future directions and emerging strategies
Advances in gene editing technologies, particularly CRISPR, are reshaping the future of stem cell research and therapies. CRISPR enables precise modifications to DNA, offering a means to correct genetic mutations that accumulate with age or cause inherited disorders. This approach allows researchers to restore normal function in exhausted stem cells by repairing damaged genes or removing harmful mutations [61]. The ability to tailor gene-editing treatments to individual patients opens the door to personalized medicine, in which therapies are customized to meet specific genetic profiles.
Despite its promise, gene editing faces challenges, including concerns about off-target effects that may alter healthy DNA sequences. Researchers continue to develop techniques to improve precision and reduce risks, ensuring that gene-editing therapies can be safely applied to human cells. Ongoing efforts aim to combine CRISPR with other technologies, such as epigenetic modifications, to fine-tune gene expression and promote cellular rejuvenation without permanently altering the genome [62].
Biomaterials and tissue engineering offer innovative tools to enhance stem cell-based therapies. Biocompatible scaffolds made from synthetic or natural materials provide structural support, helping stem cells integrate into damaged tissues and improving their survival. These scaffolds mimic the extracellular matrix, promoting cell attachment, growth, and differentiation. In addition, controlled-release systems embedded in biomaterials deliver growth factors and cytokines to stimulate repair and reduce inflammation [63].
Tissue engineering strategies are being tested for bone, cartilage, and skin repair applications, with promising results. Researchers can create functional tissues that restore structural integrity and biological function by combining stem cells with biomaterials. These approaches benefit treating injuries and degenerative diseases where traditional therapies may fall short [47].
Artificial extracellular vesicles represent a cutting-edge approach to delivering therapeutic molecules without using live cells. These vesicles mimic the natural ability of extracellular vesicles to transfer proteins, RNA, and other bioactive compounds between cells. By engineering vesicles to carry specific therapeutic cargo, researchers can precisely target damaged tissues and signaling pathways [64].
The versatility of artificial vesicles allows them to be customized for different conditions, such as reducing inflammation, promoting regeneration, or modulating immune responses. Because they do not contain living cells, they present fewer safety risks, including immune rejection and tumor formation. As research advances, artificial vesicles may provide scalable and cost-effective solutions for regenerative medicine [64].
A multimodal approach that combines pharmacology, gene editing, and biomaterials is emerging as a powerful strategy to address the complexity of stem cell exhaustion. These therapies target multiple mechanisms simultaneously, offering synergistic effects that enhance regenerative potential. For example, combining NAD+ precursors to boost mitochondrial function with CRISPR-based gene editing to repair DNA damage can provide comprehensive support for aging cells [65].
Integrating biomaterials with pharmacological treatments also allows for sustained delivery of therapeutic agents, improving outcomes while minimizing side effects. This combination approach leverages the strengths of each therapy, creating flexible solutions tailored to specific conditions [63].
Inflammaging, the chronic low-grade inflammation associated with aging, is a key factor contributing to stem cell exhaustion. Future research aims to deepen our understanding of the molecular mechanisms driving inflammaging and identify biomarkers that predict susceptibility to inflammation-related decline [66].
Therapies targeting inflammaging focus on reducing pro-inflammatory cytokines, clearing senescent cells, and modulating immune responses. Preclinical studies have shown promise in anti-inflammatory drugs, senolytics, and dietary interventions such as fasting and caloric restriction. By addressing inflammation at its source, researchers hope to preserve stem cell function and delay the onset of age-related diseases [67].
Emerging strategies also involve identifying genetic and epigenetic regulators of inflammaging, providing new targets for drug development. Advances in this field may lead to earlier interventions and more personalized therapies, improving health outcomes as people age.
Future research will continue to build upon these strategies, offering innovative approaches to protecting and restoring stem cell health. By combining cutting-edge technologies with a deeper understanding of the biology of aging, scientists aim to expand the potential of regenerative medicine and extend healthy lifespans.
Conclusion
Stem cell exhaustion is a complex process influenced by genetic, metabolic, and environmental factors that limit the ability of stem cells to repair and regenerate tissues. Mechanisms such as DNA damage, mitochondrial dysfunction, chronic inflammation, and telomere shortening contribute to this decline. Advances in therapies, including pharmacological agents, gene editing, and stem cell-conditioned media, offer promising approaches to counteract these effects. Emerging strategies, such as artificial extracellular vesicles and tissue engineering, provide alternative treatments that address the challenges of stem cell exhaustion while minimizing risks associated with live cell transplantation.
The field of regenerative medicine continues to advance, offering new hope for reversing age-related declines in stem cell function. The development of personalized treatments, combined with breakthroughs in gene editing and biomaterials, can potentially improve healthspan and quality of life. Researchers are also exploring ways to target inflammation and improve the microenvironments that support stem cells, further enhancing their regenerative potential.
Stem cell exhaustion is a major aspect of aging, but advancements in science and technology provide a pathway toward addressing this challenge. By leveraging innovative therapies and continuing to explore the biology of aging, researchers are laying the foundation for extending healthspan and promoting healthier, more resilient tissues. The future of regenerative medicine holds great promise for improving outcomes and transforming how aging and age-related diseases are treated.
Literature
[1] Bueno, V. Stem Cell Exhaustion. Cellular and Molecular Aspects of Ageing 2024, 77–86.
[2] Chaudhary, J.K.; Rath, P.C. Stem Cells and Aging. Models, Molecules and Mechanisms in Biogerontology: Cellular Processes, Metabolism and Diseases 2020, 213–234.
[3] Vitale, I.; Manic, G.; De Maria, R.; Kroemer, G.; Galluzzi, L. DNA Damage in Stem Cells. Mol Cell 2017, 66, 306–319.
[4] Ren, R.; Ocampo, A.; Liu, G.H.; Izpisua Belmonte, J.C. Regulation of Stem Cell Aging by Metabolism and Epigenetics. Cell Metab 2017, 26, 460–474.
[5] Mansilla, E.; Díaz Aquino, V.; Zambn, D.; Marin, G.H.; Mártire, K.; Roque, G.; Ichim, T.; Riordan, N.H.; Patel, A.; Sturla, F.; et al. Could Metabolic Syndrome, Lipodystrophy, and Aging Be Mesenchymal Stem Cell Exhaustion Syndromes? Stem Cells Int 2011, doi:10.4061/2011/943216.
[6] Rajagopalan, K.S.; Kazeminia, S.; Glasstetter, L.M.; Farahani, R.A.; Zhu, X.Y.; Tang, H.; Jordan, K.L.; Chade, A.R.; Lerman, A.; Lerman, L.O.; et al. Metabolic Syndrome Induces Epigenetic Alterations in Mitochondria-Related Genes in Swine Mesenchymal Stem Cells. Cells 2023, 12, 1274.
[7] López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of Aging: An Expanding Universe. Cell 2023, 186, 243–278.
[8] Bogeska, R.; Mikecin, A.M.; Kaschutnig, P.; Fawaz, M.; Büchler-Schäff, M.; Le, D.; Ganuza, M.; Vollmer, A.; Paffenholz, S. V.; Asada, N.; et al. Inflammatory Exposure Drives Long-Lived Impairment of Hematopoietic Stem Cell Self-Renewal Activity and Accelerated Aging. Cell Stem Cell 2022, 29, 1273-1284.e8.
[9] Clapes, T.; Lefkopoulos, S.; Trompouki, E. Stress and Non-Stress Roles of Inflammatory Signals during HSC Emergence and Maintenance. Front Immunol 2016, 7.
[10] Fathi, E.; Charoudeh, H.N.; Sanaat, Z.; Farahzadi, R. Telomere Shortening as a Hallmark of Stem Cell Senescence. Stem Cell Investig 2019, 6, 7.
[11] Allen, N.D.; Baird, D.M. Telomere Length Maintenance in Stem Cell Populations. Biochim Biophys Acta Mol Basis Dis 2009, 1792, 324–328.
[12] Zhang, J.; Ju, Z. Telomere, DNA Damage, and Oxidative Stress in Stem Cell Aging. Birth Defects Res C Embryo Today 2010, 90, 297–307.
[13] Mannino, G.; Russo, C.; Maugeri, G.; Musumeci, G.; Vicario, N.; Tibullo, D.; Giuffrida, R.; Parenti, R.; Lo Furno, D. Adult Stem Cell Niches for Tissue Homeostasis. J Cell Physiol 2022, 237, 239–257.
[14] Cain, T.L.; Derecka, M.; McKinney-Freeman, S. The Role of the Haematopoietic Stem Cell Niche in Development and Ageing. Nature Reviews Molecular Cell Biology 2024 26:1 2024, 26, 32–50.
[15] Farkhondeh, T.; Llorens, S.; Pourbagher-Shahri, A.M.; Ashrafizadeh, M.; Talebi, M.; Shakibaei, M.; Samarghandian, S. An Overview of the Role of Adipokines in Cardiometabolic Diseases. Molecules 2020, 25.
[16] Li, Y.; Jin, D.; Xie, W.; Wen, L.; Chen, W.; Xu, J.; Ding, J.; Ren, D. PPAR-γ and Wnt Regulate the Differentiation of MSCs into Adipocytes and Osteoblasts Respectively. Curr Stem Cell Res Ther 2017, 13, 185–192..
[17] de Haan, G.; Lazare, S.S. Aging of Hematopoietic Stem Cells. Blood 2018, 131, 479–487.
[18] Nicaise, A.M.; Willis, C.M.; Crocker, S.J.; Pluchino, S. Stem Cells of the Aging Brain. Front Aging Neurosci 2020, 12, 563285.
[19] Silva-Vargas, V.; Crouch, E.E.; Doetsch, F. Adult Neural Stem Cells and Their Niche: A Dynamic Duo during Homeostasis, Regeneration, and Aging. Curr Opin Neurobiol 2013, 23, 935–942.
[20] Coronel, R.; Palmer, C.; Bernabeu-Zornoza, A.; Monteagudo, M.; Rosca, A.; Zambrano, A.; Liste, I. Physiological Effects of Amyloid Precursor Protein and Its Derivatives on Neural Stem Cell Biology and Signaling Pathways Involved. Neural Regen Res 2019, 14, 1661–1671.
[21] Bernabeu-Zornoza, A.; Coronel, R.; Palmer, C.; Monteagudo, M.; Zambrano, A.; Liste, I. Physiological and Pathological Effects of Amyloid-β Species in Neural Stem Cell Biology. Neural Regen Res 2019, 14, 2035–2042.
[22] Liu, G.H.; Qu, J.; Suzuki, K.; Nivet, E.; Li, M.; Montserrat, N.; Yi, F.; Xu, X.; Ruiz, S.; Zhang, W.; et al. Progressive Degeneration of Human Neural Stem Cells Caused by Pathogenic LRRK2. Nature 2012 491:7425 2012, 491, 603–607.
[23] Fehrer, C.; Lepperdinger, G. Mesenchymal Stem Cell Aging. Exp Gerontol 2005.
[24] Singer, N.G.; Caplan, A.I. Mesenchymal Stem Cells: Mechanisms of Inflammation. Annual Review of Pathology: Mechanisms of Disease 2011, 6, 457–478.
[25] Jiang, H.; Liu, B.; Lin, J.; Xue, T.; Han, Y.; Lu, C.; Zhou, S.; Gu, Y.; Xu, F.; Shen, Y.; et al. MuSCs and IPCs: Roles in Skeletal Muscle Homeostasis, Aging and Injury. Cellular and Molecular Life Sciences 2024 81:1 2024, 81, 1–18.
[26] Yingao, X.; Jianda, K.; Yun, C.; Zhilin, L.; Peng, X. Biological Mechanism of Satellite Cell Aging in Skeletal Muscles and Potential Coping Strategies. Chinese Journal of Tissue Engineering Research 2024, 28, 4094.
[27] Guo, Q.; Luo, Q.; Song, G. Control of Muscle Satellite Cell Function by Specific Exercise-Induced Cytokines and Their Applications in Muscle Maintenance. J Cachexia Sarcopenia Muscle 2024, 15, 466–476.
[28] Rayagiri, S.S.; Ranaldi, D.; Raven, A.; Mohamad Azhar, N.I.F.; Lefebvre, O.; Zammit, P.S.; Borycki, A.G. Basal Lamina Remodeling at the Skeletal Muscle Stem Cell Niche Mediates Stem Cell Self-Renewal. Nat Commun 2018, 9.
[29] Chen, J.; Zhou, R.; Feng, Y.; Cheng, L. Molecular Mechanisms of Exercise Contributing to Tissue Regeneration. Signal Transduct Target Ther 2022, 7.
[30] Puca, F.; Fedele, M.; Rasio, D.; Battista, S. Role of Diet in Stem and Cancer Stem Cells. International Journal of Molecular Sciences 2022, Vol. 23, Page 8108 2022, 23, 8108.
[31] Rose, S.C.; Larsen, M.; Xie, Y.; Sharfstein, S.T. Salivary Gland Bioengineering. Bioengineering 2024, Vol. 11, Page 28 2023, 11, 28.
[32] Grezella, C.; Fernandez-Rebollo, E.; Franzen, J.; Ventura Ferreira, M.S.; Beier, F.; Wagner, W. Effects of Senolytic Drugs on Human Mesenchymal Stromal Cells. Stem Cell Res Ther 2018, 9, 1–6.
[33] Zhang, J.; Shang, J.; Ding, H.; Li, W.; Li, Z.; Yuan, Z.; Zheng, H.; Lou, Y.F.; Wei, Z.; Zhou, H.; et al. Nicotinamide Riboside Promotes the Proliferation of Endogenous Neural Stem Cells to Repair Spinal Cord Injury. Stem Cell Rev Rep 2024, 20, 1854–1868.
[34] Wang, H.; Sun, Y.; Pi, C.; Yu, X.; Gao, X.; Zhang, C.; Sun, H.; Zhang, H.; Shi, Y.; He, X. Nicotinamide Mononucleotide Supplementation Improves Mitochondrial Dysfunction and Rescues Cellular Senescence by NAD+/Sirt3 Pathway in Mesenchymal Stem Cells. Int J Mol Sci 2022, 23, 14739.
[35] Igarashi, M.; Miura, M.; Williams, E.; Jaksch, F.; Kadowaki, T.; Yamauchi, T.; Guarente, L. NAD+ Supplementation Rejuvenates Aged Gut Adult Stem Cells. Aging Cell 2019, 18, e12935.
[36] Liu, X.; Li, Z.; Liu, L.; Zhang, P.; Wang, Y.; Ding, G. Metformin-Mediated Effects on Mesenchymal Stem Cells and Mechanisms: Proliferation, Differentiation and Aging. Front Pharmacol 2024, 15, 1465697.
[37] Jiang, L.L.; Liu, L. Effect of Metformin on Stem Cells: Molecular Mechanism and Clinical Prospect. World J Stem Cells 2020, 12, 1455.
[38] Pulecio, J.; Verma, N.; Mejía-Ramírez, E.; Huangfu, D.; Raya, A. CRISPR/Cas9-Based Engineering of the Epigenome. Cell Stem Cell 2017, 21, 431–447.
[39] Sun, Y.; Zhang, H.; Qiu, T.; Liao, L.; Su, X. Epigenetic Regulation of Mesenchymal Stem Cell Aging through Histone Modifications. Genes Dis 2023, 10, 2443–2456.
[40] Esteves, F.; Brito, D.; Rajado, A.T.; Silva, N.; Apolónio, J.; Roberto, V.P.; Araújo, I.; Nóbrega, C.; Castelo-Branco, P.; Bragança, J.; et al. Reprogramming IPSCs to Study Age-Related Diseases: Models, Therapeutics, and Clinical Trials. Mech Ageing Dev 2023, 214, 111854.
[41] Wang, C.; Rabadan Ros, R.; Martinez-Redondo, P.; Ma, Z.; Shi, L.; Xue, Y.; Guillen-Guillen, I.; Huang, L.; Hishida, T.; Liao, H.K.; et al. In Vivo Partial Reprogramming of Myofibers Promotes Muscle Regeneration by Remodeling the Stem Cell Niche. Nature Communications 2021 12:1 2021, 12, 1–15.
[42] Sahu, S.K.; Reddy, P.; Lu, J.; Shao, Y.; Wang, C.; Tsuji, M.; Delicado, E.N.; Esteban, C.R.; Belmonte, J.C.I. Targeted Partial Reprogramming of Age-Associated Cell States Improves Markers of Health in Mouse Models of Aging. Sci Transl Med 2024, 16.
[43] Swartzrock, L.; Dib, C.; Denis, M.; Willner, H.; Ho, K.; Haslett, E.; Han, J.; Pan, W.; Byrne-Steele, M.; Brown, B.; et al. In Utero Hematopoietic Stem Cell Transplantation for Fanconi Anemia. Blood Adv 2024, 8, 4554–4558.
[44] Kumar, R.; Mishra, N.; Tran, T.; Kumar, M.; Vijayaraghavalu, S.; Gurusamy, N. Emerging Strategies in Mesenchymal Stem Cell-Based Cardiovascular Therapeutics. Cells 2024, Vol. 13, Page 855 2024, 13, 855.
[45] Ma, W.Q.; Zhuo, A.P.; Xiao, Y.L.; Gao, M.; Yang, Y.T.; Tang, L.C.; Wu, Y.H.; Tian, D.; Fu, X.F. Human Bone Marrow Derived-Mesenchymal Stem Cells Treatment for Autoimmune Premature Ovarian Insufficiency. Stem Cell Rev Rep 2024, 20, 538–553.
[46] Lindsay, J.O.; Hind, D.; Swaby, L.; Berntsson, H.; Bradburn, M.; Bannur C, U.; Byrne, J.; Clarke, C.; Desoysa, L.; Dickins, B.; et al. Safety and Efficacy of Autologous Haematopoietic Stem-Cell Transplantation with Low-Dose Cyclophosphamide Mobilisation and Reduced Intensity Conditioning versus Standard of Care in Refractory Crohn’s Disease (ASTIClite): An Open-Label, Multicentre, Randomised Controlled Trial. Lancet Gastroenterol Hepatol 2024, 9, 333–345.
[47] Rose, S. A 3D Co-Culture Model of Senescent Cell Burden and Salivary Gland Fibrosis. Electronic Theses & Dissertations (2024 – present) 2024.
[48] Riazifar, M.; Pone, E.J.; Lotval, J.; Zhao, W. Stem Cell Extracellular Vesicles: Extended Messages of Regeneration. Annu Rev Pharmacol Toxicol 2017, 57, 125–154, doi:10.1146/ANNUREV-PHARMTOX-061616-030146/CITE/REFWORKS.
[49] Jafarzadeh, A.; PourMohammad, A.; Goodarzi, A. A Systematic Review of the Efficacy, Safety and Satisfaction of Regenerative Medicine Treatments, Including Platelet-Rich Plasma, Stromal Vascular Fraction and Stem Cell-Conditioned Medium for Hypertrophic Scars and Keloids. Int Wound J 2024, 21, e14557, doi:10.1111/IWJ.14557.
[50] Li, J.; Sun, S.; Zhu, D.; Mei, X.; Lyu, Y.; Huang, K.; Li, Y.; Liu, S.; Wang, Z.; Hu, S.; et al. Inhalable Stem Cell Exosomes Promote Heart Repair after Myocardial Infarction. Circulation 2024, 150, 710–723.
[51] Subbarayan, R.; Murugan Girija, D.; Raja, S.T.K.; Krishnamoorthy, A.; Srinivasan, D.; Shrestha, R.; Srivastava, N.; Ranga Rao, S. Conditioned Medium-Enriched Umbilical Cord Mesenchymal Stem Cells: A Potential Therapeutic Strategy for Spinal Cord Injury, Unveiling Transcriptomic and Secretomic Insights. Mol Biol Rep 2024, 51, 1–14.
[52] Țichil, I.; Mitre, I.; Zdrenghea, M.T.; Bojan, A.S.; Tomuleasa, C.I.; Cenariu, D. A Review of Key Regulators of Steady-State and Ineffective Erythropoiesis. Journal of Clinical Medicine 2024, Vol. 13, Page 2585 2024, 13, 2585.
[53] Maličev, E.; Jazbec, K. An Overview of Mesenchymal Stem Cell Heterogeneity and Concentration. Pharmaceuticals 2024, Vol. 17, Page 350 2024, 17, 350, doi:10.3390/PH17030350.
[54] Cain, T.L.; Derecka, M.; McKinney-Freeman, S. The Role of the Haematopoietic Stem Cell Niche in Development and Ageing. Nature Reviews Molecular Cell Biology 2024 26:1 2024, 26, 32–50.
[55] Gumede, D.B.; Abrahamse, H.; Houreld, N.N. Targeting Wnt/β-Catenin Signaling and Its Interplay with TGF-β and Notch Signaling Pathways for the Treatment of Chronic Wounds. Cell Communication and Signaling 2024 22:1 2024, 22, 1–17.
[56] Aval, S.F.; Lotfi, H.; Sheervalilou, R.; Zarghami, N. Tuning of Major Signaling Networks (TGF-β, Wnt, Notch and Hedgehog) by MiRNAs in Human Stem Cells Commitment to Different Lineages: Possible Clinical Application. Biomedicine & Pharmacotherapy 2017, 91, 849–860.
[57] King, N.M.P.; Perrin, J. Ethical Issues in Stem Cell Research and Therapy. Stem Cell Res Ther 2014, 5, 1–6.
[59] Malhotra, P.; Saraswat, S.K.; Khatri, N.; Verma, R.; Datt, R.; Pandey, S. Stem Cell-Based Products and the Regulatory Hurdle Race. Stem Cells: an Alternative Therapy for COVID-19 and Cytokine Storm 2024, 371–38,.
[60] Knoepfler, P.S. From Bench to FDA to Bedside: US Regulatory Trends for New Stem Cell Therapies. Adv Drug Deliv Rev 2015, 82–83, 192–196.
[61] Yoon, D.E.; Lee, H.; Kim, K. Recent Research Trends in Stem Cells Using CRISPR/Cas-Based Genome Editing Methods. Int J Stem Cells 2023, 17, 1–14.
[62] Abusalah, M.A.H.; Priyanka; Abd Rahman, E.N.S.E.; Choudhary, O.P. Evolving Trends in Stem Cell Therapy: An Emerging and Promising Approach against Various Diseases. International Journal of Surgery 2024.
[63] Ramesh, P. Biomimetic Scaffolds Targeting Remediation of Fibrosis and Regeneration of the Salivary Gland. 2022.
[64] Zmievskaya, E.A.; Mukhametshin, S.A.; Ganeeva, I.A.; Gilyazova, E.M.; Siraeva, E.T.; Kutyreva, M.P.; Khannanov, A.A.; Yuan, Y.; Bulatov, E.R. Artificial Extracellular Vesicles Generated from T Cells Using Different Induction Techniques. Biomedicines 2024, 12, 919.
[65] Salazar-Noratto, G.E.; Luo, G.; Denoeud, C.; Padrona, M.; Moya, A.; Bensidhoum, M.; Bizios, R.; Potier, E.; Logeart-Avramoglou, D.; Petite, H. Understanding and Leveraging Cell Metabolism to Enhance Mesenchymal Stem Cell Transplantation Survival in Tissue Engineering and Regenerative Medicine Applications. Stem Cells 2020, 38, 22–33.
[66] Tatullo, M.; Rengo, S.; Sammartino, G.; Simeone, M.; Valletta, A.; Gasparro, R. Inflammaging, Epigenetic Damage, Mitochondrial Dysfunctions, and Macrophage Alterations Are the Main Missing Pieces in the Complex Mosaic of Stem Cell Physiology. Discov Med 2024, 36, 1772.
[67] Andonian, B.J.; Hippensteel, J.A.; Abuabara, K.; Boyle, E.M.; Colbert, J.F.; Devinney, M.J.; Faye, A.S.; Kochar, B.; Lee, J.; Litke, R.; et al. Inflammation and Aging-Related Disease: A Transdisciplinary Inflammaging Framework. GeroScience 2024 2024, 1–28.