LongeCityNews
Last Updated:
21 November 2025 - 08:38 AM
Injected Oxytocin Slows Cognitive Decline in Aged Mice 20 November 2025 - 07:22 PM
Circulating oxytocin levels are known to decline with age, and a number of research groups have focused on upregulation of oxytocin as an approach to treating aging. A couple of papers published a few months ago are indicative of the animal studies presently taking place, the first focused on increased longevity in mice achieved via the combined reduction of TGF-β and increase in oxytocin, and the second evaluating intranasal delivery of oxytoxin as a route to improve function in the aging brain.
Today's paper reports on another example of oxytocin delivery in aged mice. These researchers are also focused on the brain, but in this case the oxytocin is delivered via intraperitoneal injection. As with most peptide or protein therapies, the effects are limited in scope as the delivered molecules have a short half-life. Repeated treatments are required, often daily, as is the case here. Given further progress towards the clinic, however, we might expect that the community of developers presently assessing gene therapies to safely transform a small number of cells into long-lasting factories that produce a desired circulating molecule (such as klotho or follistatin) will add oxytocin to their list.
Brain aging is characterized by progressive structural and functional deterioration, leading to cognitive decline and impaired social functioning. A key factor in this process is the age-related decline in adult neurogenesis, particularly in the hippocampal dentate gyrus, which is linked to deficits in learning, memory, and increased social anxiety. Oxytocin, a neuropeptide synthesized in the hypothalamus, regulates social behavior, cognition, and emotion by acting on brain regions including the hippocampus. Importantly, oxytocin levels decrease with age, potentially contributing to cognitive impairment.
Here, we examined whether chronic intraperitoneal oxytocin administration could attenuate cognitive decline in aged mice. Twelve-month-old mice received oxytocin injections (0.5 mg/kg) five times weekly for 13 weeks. Behavioral testing at 12 weeks of treatment using the object-place recognition task showed enhanced spatial learning and recognition memory in oxytocin-treated mice compared with saline controls. Immunohistochemistry revealed significantly increased doublecortin (DCX)-positive cells in the hippocampus, indicating enhanced neurogenesis. Furthermore, oxytocin treatment upregulated the expression of glutamate receptor 1 (GluR1) and N-methyl-D-aspartate receptor subunit 2B (NMDAR2B), which are markers of synaptic plasticity.
These findings suggest that chronic oxytocin treatment is associated with enhanced neurogenesis and synaptic plasticity, which may contribute to improved cognition in aged mice. Our results support oxytocin as a potential therapeutic agent for age-related cognitive decline.
View the full article at FightAging
How Senescent Cells Encourage Melanoma Growth 20 November 2025 - 06:04 PM
Researchers publishing in Aging Cell have documented a key reason why older people are much more likely to get melanoma.
Why older people have significantly worse melanoma cases
While melanoma is much more treatable now than in the past, it still remains a serious danger. Melanoma can develop resistance to otherwise effective techniques [1], meaning that they only delay instead of permanently stop the disease.
Fortunately, the origins of melanoma are largely well-understood. The first thing that drives most melanoma cases is a point mutation of the BRAF gene [2]. This does not trigger melanoma by itself, but further mutations of other genes lead to malignancy [3].
The severity of melanoma is measured by its thickness, as it has been known for over half a century that thicker melanomas are much more dangerous [4]. The cancer’s transition from horizontal to vertical growth is associated with increasing mutation frequency [5], and, unsurprisingly, older people are typically diagnosed with considerably thicker melanomas than younger people are [6].
The researchers of this study have pinpointed cellular senescence as the most likely driver of this increase in severity, and significant previous work has been done to establish the connection between these cells’ potentially dangerous SASP secretions and melanoma [7]. However, that previous work did not completely elucidate the biological relationship between the two, which is what this study was created to find.
Melanoma is attracted to senescent cells
In their first experiment, the researchers confirmed a direct relationship between the prevalence of senescent skin fibroblasts and the incidence of melanoma. While p16 is a tumor suppressor and appears in both the benign and malignant portions of melanoma, it is bypassed by other cancerous mutations [8]. Injecting melanoma cells together with senescent or non-senescent fibroblasts into the skin of mice confirmed this relationship: the mice given senescent fibroblasts had tumors that were ten times as thick.
The researchers then looked into why this is the case. Cultivating melanoma in conditioned media that contains secretions from senescent fibroblasts, but not non-senescent fibroblasts, greatly increases the cancer’s growth. The researchers found two compounds of particular interest: GCL-2 and ENA-78, which melanoma cells were discovered to actively grow towards in a chemotaxic response, resulting in independent, unanchored growth. Neutralizing these compounds in conditioned media through antibodies greatly reduces the growth of melanoma, and enhancing GCL-2 production in non-senescent cells causes their related conditioned media to encourage the growth of this cancer.
These two compounds, which are considerably more abundant in the skin of older adults compared to young adults, are generated by senescent fibroblasts and not significantly by other cell types.
Further work found that GCL-2 is considerably more important than ECL-78 in encouraging the growth of cancer cells, as silencing GCL-2 had significant effects on the growth of melanoma in mice but silencing ECL-78 did not. Furthermore, senescent fibroblasts were confirmed to be the source of the harmful GCL-2, as silencing this compound in the melanoma itself had no significant effect.
The protein that drives melanoma’s growth
A more in-depth examination found this to be due to the phosphorylation of the cAMP-responsive element binding protein (CREB). CREB activation leads to tumor progression in melanoma, activating several related cancer genes, and GCL-2 was found to significantly drive this effect, with ECL-78’s effect being much weaker. Removing GCL-2 from the environment of melanoma was found to almost completely stop CREB activation, both in conditioned media and in mice.
The researchers found that significant CREB activation occurs only in the malignant part of melanoma, not the benign part. It drives glycolysis, a form of energy use that encourages cancer progression. Directly inhibiting this process, either by suppressing CREB or suppressing glycolysis in these cells, prevents the related acceleration of cancer, thus providing strong evidence that this is indeed the biological cause. An examination of naturally occurring melanomas confirmed their glycolytic nature.
As the researchers note, their data “allows several options for novel strategies for therapeutic intervention.” While fighting fibroblast senescence itself would be an ideal solution, targeting GCL-2 or its receptors offers a few potential avenues, and directly targeting CREB offers another. As targeting GCL-2 receptors is already being investigated in the context of other cancers [9], it may be that translating these drugs to melanoma is on the short-term horizon.
Literature
[1] Kim, K. B., Kefford, R., Pavlick, A. C., Infante, J. R., Ribas, A., Sosman, J. A., … & Lewis, K. D. (2013). Phase II study of the MEK1/MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. Journal of Clinical Oncology, 31(4), 482-489.
[2] Gray-Schopfer, V. C., Dias, S. D. R., & Marais, R. (2005). The role of B-RAF in melanoma. Cancer and Metastasis Reviews, 24(1), 165-183.
[3] Dankort, D., Curley, D. P., Cartlidge, R. A., Nelson, B., Karnezis, A. N., Damsky Jr, W. E., … & Bosenberg, M. (2009). Braf V600E cooperates with Pten loss to induce metastatic melanoma. Nature genetics, 41(5), 544-552.
[4] Breslow, A. (1970). Thickness, cross-sectional areas and depth of invasion in the prognosis of cutaneous melanoma. Annals of surgery, 172(5), 902.
[5] Greene, V. R., Johnson, M. M., Grimm, E. A., & Ellerhorst, J. A. (2009). Frequencies of NRAS and BRAF mutations increase from the radial to the vertical growth phase in cutaneous melanoma. Journal of investigative dermatology, 129(6), 1483-1488.
[6] Kruijff, S., Bastiaannet, E., Francken, A. B., Schaapveld, M., Van Der Aa, M., & Hoekstra, H. J. (2012). Breslow thickness in the Netherlands: a population-based study of 40 880 patients comparing young and elderly patients. British journal of cancer, 107(3), 570-574.
[7] Liu, J., Zheng, R., Zhang, Y., Jia, S., He, Y., & Liu, J. (2023). The cross talk between cellular senescence and melanoma: From molecular pathogenesis to target therapies. Cancers, 15(9), 2640.
[8] Mooi, W. J., & Peeper, D. S. (2006). Oncogene-induced cell senescence—halting on the road to cancer. New England Journal of Medicine, 355(10), 1037-1046.
[9] Campbell, L. M., Maxwell, P. J., & Waugh, D. J. (2013). Rationale and means to target pro-inflammatory interleukin-8 (CXCL8) signaling in cancer. Pharmaceuticals, 6(8), 929-959.
View the article at lifespan.io
Mixed Results in a Meta-Analysis of Epigenetic Clocks and Frailty 20 November 2025 - 11:22 AM
Epigenetic clocks have existed for long enough for numerous large study databases to include data on their use. Thus meta-analysis papers are emerging to assess this body of data as a whole. This is a necessary part of the process of gaining confidence in the ability of epigenetic clocks, and indeed aging clocks in general, to rapidly assess the potential of any novel form of intervention intended to slow or reverse aspects of aging. This is a much needed capability. In many ways, efforts to treat aging as a medical condition proceed blindly, given just how much time and funding is required in order to understand whether one approach is better or worse than another. If there was a way to quickly assess the quality of an anti-aging therapy immediately after its application, then the field could adjust quickly to pursue the best paths forward. The hope is that aging clocks can be that tool - but we are clearly not there yet.
Frailty is an age-related condition characterised by multisystem physiological decline, which increases vulnerability to adverse outcomes. Biomarkers of ageing might identify individuals at risk and enable early interventions. This systematic review and meta-analysis aimed to examine cross-sectional and longitudinal associations between DNA methylation-based biological age metrics (eg, DNA methylation age, epigenetic-age acceleration [EAA], and age deviation) and frailty.
24 studies met the inclusion criteria (17 cross-sectional studies, one longitudinal study, and six studies that were both cross-sectional and longitudinal), encompassing 28,325 participants (14,757 female; median of mean age 65.2 years). DNA methylation age and age deviation showed no association with frailty. In cross-sectional meta-analyses, higher Hannum EAA (nine studies; n=11,162; standardised β coefficient 0.06), PhenoAge EAA (eight studies; n=10,371; standardised β coefficient 0.07), GrimAge EAA (eight studies; n=10,371; standardised β coefficient 0.11), and pace of ageing (five studies; n=7,895; standardised β coefficient 0.10) were significantly associated with higher frailty. In longitudinal meta-analyses, higher GrimAge EAA (five studies; n=6,143; standardised β coefficient 0.02) was significantly associated with increases in frailty, whereas PhenoAge EAA and pace of ageing were not significantly associated with frailty.
In conclusion, higher GrimAge EAA is consistently associated with higher frailty. Future research should focus on developing and validating DNA methylation clocks that integrate molecular surrogates of health risk and are specifically trained to predict frailty in large, harmonised, longitudinal cohorts, enabling their translation into clinical practice.
Link: https://doi.org/10.1016/j.lanhl.2025.100773
View the full article at FightAging
MicroRNA-126 Expression as a Way to Prevent TDP-43 Aggregation in Amyotrophic Lateral Sclerosis 20 November 2025 - 11:11 AM
TDP-43 is one of a small number of proteins in the brain that can misfold or otherwise become altered in ways that allow toxic aggregates to form, or even encourage other molecules of the same protein to become dysfunctional in the same way. TDP-43 aggregation in later life is a relatively recent discovery, and has a neurodegenerative condition newly named for it, limbic-predominant age-related TDP-43 encephalopathy (LATE). It has also been found that TDP-43 is likely important in amyotrophic lateral sclerosis (ALS), and thus progress on that front seems likely to help with other forms of TDP-43 pathology. Here, researchers report a promising discovery in the biochemistry of TDP-43 aggregation in the context of ALS.
Amyotrophic lateral sclerosis (ALS) is a lethal adult-onset motor neuron disease, characterized by disruption of neuromuscular junctions (NMJs), axonal degeneration and neuronal death. Most ALS cases are linked to TDP-43 pathology, characterized by its mislocalization from the nucleus to the cytoplasm and the formation of phosphorylated aggregates. TDP-43 is a multifunctional DNA-binding/RNA-binding protein with roles in transcriptional and splicing regulation, RNA processing and RNA transport/subcellular localization.
Recently, we showed that TDP-43 co-localizes with the core stress granule component G3BP1 in axonal condensates of patients with ALS and mice. These TDP-43-G3BP1 condensates sequester RNA and inhibit local protein synthesis, resulting in mitochondrial malfunction and NMJ disruption with subsequent axonal degeneration. Furthermore, recent studies revealed aggregation of TDP-43 in peripheral motor axons of patients with ALS during initial diagnosis. Thus, axonal TDP-43 condensates exert pathological regulation over essential local synthesis events.
Here, we studied the localized accumulation of TDP-43 in axons and NMJs. Our findings highlight the presence of distal TDP-43 pathology in patients with SOD1 ALS and mouse models. We found that TDP-43 accumulates at NMJs due to aberrant local synthesis triggered by a reduction in miR-126a-5p within muscle extracellular vesicles. This chain of events ultimately initiates neurodegeneration. Notably, delivery of miR-126 is neuroprotective in neuromuscular co-cultures, delays TDP-43 accumulation at NMJs, and postpones the onset of motor symptoms in the SOD1G93A mouse model of ALS.
Link: https://doi.org/10.1038/s41593-025-02062-6
View the full article at FightAging
How Much of the Aging of the Gut Microbiome is Induced by Pharmaceutical Use? 19 November 2025 - 07:06 PM
The human gut microbiome is made up of a few thousand distinct microbial species. The relative sizes of these populations shift in response to day to day circumstances, such as variations in diet, but one would expect consistency from one year to the next. Over longer spans of time, the gut microbiome ages. Populations producing beneficial metabolites are reduced in number, while populations that provoke the immune system into chronic inflammatory behavior grow in number. This data is derived from both animal and human studies. One can control what happens to a mouse over the course of its life, but for human data the detailed history of any particular individual is largely a mystery.
Antibiotics and a range of other pharmaceuticals produce dramatic short-term effects on the composition of the gut microbiome. To a large degree, the gut microbiome restores itself once the pharmaceutical is no longer present. That said, we might well ask how much of the observed human data on gut microbiome aging is produced by, say, antibiotic use. Researchers have observed gut microbiome changes in population studies taking place as early as the mid-30s, and it is hard to envisage any form of meaningful degeneration taking place at that age. But exposure to antibiotics and other pharmaceuticals? That is very prevalent. In later life, given the presence of chronic diseases of aging, there is a great deal more chronic pharmaceutical use, as well as the employment of pharmaceuticals with meaningful side effects. We might again ask how much of the observed aging of the human gut microbiome at the population level results from the use of these treatments versus mechanisms of aging.
Drug-mediated disruption of the aging gut microbiota and mucosal immune system
The dynamic relationship of gut microbiota, mucosal immunity, aging, and pharmaceutics interventions has a significant impact on overall physiological functions and disease susceptibility. Aging is associated with changes in the gut microbiome including decreased microbial diversity, reduced short-chain fatty acid (SCFA) production, and elevated pathobiont proportions. These changes are associated with impaired mucosal immunity, increased intestinal permeability, and heightened systemic inflammation in the host, which can exacerbate age-related disorders.
Medications such as proton pump inhibitors (PPIs), metformin, nonsteroidal anti-inflammatory drugs (NSAIDs), corticosteroids, and antibacterials also influence gut microbiota function. PPIs may alter microbial colonies and induce overgrowth of pathogenic bacteria, which compromises the mucosal defenses and in severe cases, the resulting infections may cause ulcers. Metformin, through its metabolic benefits, causes Akkermansia muciniphila to increase in relative abundance, which is associated with improved gut barrier composition. NSAIDs, because of their strong anti-inflammatory properties, disturb gut homeostasis by increasing intestinal permeability, reducing prostaglandin synthesis, and inducing dysbiosis in the host. Corticosteroids, through their immunosuppressive mechanisms, reduce microbial diversity and secretory immunoglobulin A levels, impairing mucosal immunity and enhancing the host's susceptibility to infections. Antibacterials are a major disruptor of the gut microbiota, causing a decline in beneficial bacteria and an increased risk for opportunistic infections such as Clostridium difficile.
To address drug-induced dysbiosis, probiotics and prebiotics products may be helpful to restore microbial balance, enhance SCFA production, and reinforce mucosal defenses. Individualized gut microbiota profiling may enable safer medication usage by identifying patients that are at an increased risk for dysbiosis-related complications. Additionally, development of microbiota-sparing medications and targeted therapies may help to enhance gut health outcomes in aging populations. Future research should address the long-term effects of pharmacological agents on gut microbiota and mucosal immunity in aging populations, as well as identification of connections between microbiota, immune function, and the effects of medications. Integrating microbiome-conscious approaches into clinical practice could allow healthcare providers to optimize patient care
View the full article at FightAging
The Impact of Plant Polyphenols on Ovarian Aging 19 November 2025 - 05:01 PM
A recent review in the Journal of Ovarian Research summarizes current knowledge of the impact of various polyphenols on different aspects of ovarian aging. The researchers discuss that polyphenol supplementation could be used as an intervention to delay ovarian aging [1].
Every woman’s problem
Ovarian aging and cessation of proper ovarian functioning precede aging in other organs. Ovarian aging is related to a reduction of oocyte quality and quantity, which is the main reason for age-related infertility.
Beyond the fertility problems, ovarian aging impacts lifespan and is also linked to many age-related conditions, such as osteoporosis, cardiovascular disease, and neurodegenerative disorders. Therefore, finding ways to delay it is in the interest of every female, regardless of her childbearing goals.
At this moment, hormone replacement therapy and assisted reproductive technologies are used to address ovarian aging-related problems; however, they are unable to reverse female reproductive aging and the declining ovarian reserve. Longevity researchers are currently seeking ways to extend the female reproductive span, but before effective therapies are on the market, lifestyle factors can be utilized to address ovarian aging.
The authors of this review highlight polyphenols, naturally occurring metabolites found in fruits, vegetables, nuts, seeds, herbs, spices, and medicinal plants, as one possible intervention. Polyphenols exhibit strong biological activities, including antioxidant, anti-inflammatory, antibacterial, and antiviral properties, and demonstrate numerous beneficial effects. Studies also suggest that they can reduce the risk of cardiovascular disease and neurodegenerative disorders [2].

Delaying ovarian aging
The first part of this review discusses resveratrol, a plant-derived polyphenol that has antioxidant and anti-apoptotic properties. Multiple studies have shown that resveratrol’s dietary supplementation or oral administration ”alleviated ovarian oxidative stress damage, restored hormone levels, reduced ovarian cell apoptosis, and improved reproductive performance in animals” [3-7]. It also positively affected epigenetic changes and gene expression in the aging ovary.
Polyphenols extracted from tea leaves were shown to reduce inflammatory responses and oxidative stress and to improve ovarian reserve and ovarian function in animal models of induced ovarian damage [8, 9]. Human, mice, and other mammalian research also suggested their benefits in “alleviating ovarian aging and improving reproductive performance by enhancing oocyte quality and reducing oxidative stress” [10-12].
Quercetin was described as having strong antioxidant properties. It can promote in vitro maturation of oocytes from aged mice and humans [13] and slow down the aging of human ovarian cells [14]. Experiments in middle-aged mice also showed that oral administration improved estrous cycles, pregnancy rate, and ovarian reserve [14]. In polycystic ovary syndrome (PCOS) models, quercetin reversed many detrimental changes, such as irregular ovulation and hormone secretion, or increased ovarian cell apoptosis and inflammation [15, 16]. Similar beneficial effects were also seen in experiments in livestock and poultry animals.
Proanthocyanidins are among the most potent natural antioxidants and possess several other beneficial characteristics. They have been linked to ovarian health in multiple studies. Research in rodents and human cells reported that proanthocyanidins reduced oxidative stress, inhibited ovarian cells apoptosis, improved oocyte viability and quality, modulated hormone levels, and alleviated PCOS [17-20].
Less commonly studied polyphenols also mitigate ovarian aging. Curcumin “has been found to delay the ovarian aging process by increasing follicular number, modulating hormone secretion, reducing oxidative stress, enhancing oocyte maturation and embryo development in an aged mouse model” [21]. Other polyphenols, such as chlorogenic acid, ferulic acid, and pterostilbene, have been shown to positively impact ovarian reserve, ovarian function, and oocyte quality by mitigating oxidative stress, reducing ovarian cell apoptosis, and reducing DNA damage [22-25].
It is worth noting that several studies have shown that a moderate dose of polyphenols can make a profound difference, and high doses of polyphenols can be toxic and cause ovarian damage and impair oocyte maturation. [17, 26-29].
Many mechanisms, one goal
The beneficial effects of polyphenols can be achieved through modulations of many molecular pathways. One of them involves oxidative stress and the excessive production of reactive oxygen species (ROS), which can contribute to an increase in inflammation and disrupt the redox balance, leading to damage to mitochondrial function, telomere shortening, apoptosis, and inflammation, all of which compromise the proper functioning of ovaries and contribute to ovarian aging.
Since polyphenols have antioxidant properties, the researchers investigated their role in delaying ovarian aging by examining the effects of resveratrol, quercetin, and epigallocatechin gallate (EGCG). Those polyphenols have been shown to modulate multiple molecular signaling pathways related to oxidative stress, improve ovarian antioxidant capacity, and reduce ovarian cell apoptosis [30-32]. A human randomized controlled trial also showed a positive role of plant polyphenols (curcumin or resveratrol supplementation) in alleviating ovarian aging by reducing oxidative stress [33, 34].
Polyphenols’ anti-inflammatory properties can also help alleviate ovarian aging by reducing inflammation, which negatively impacts ovarian health. These benefits were demonstrated in animal and human models of ovarian damage [8,35]. Reduction of inflammation and improved oocyte and embryo quality resulted from polyphenol supplementation in women with PCOS who received quercetin [36]. However, there is still a need for research on the impact of polyphenols on inflammation markers in naturally aging ovaries.
Aging-related hormonal dysregulation significantly impacts ovarian aging. The hypothalamic-pituitary-ovary (HPO) axis controls hormones related to the reproductive system. Aging-related dysregulation of the HPO axis contributes to the depletion of the ovarian reserve and decreased quality and quantity of ovarian cells [37]. A growing body of research suggests that plant polyphenols regulate the sensitivity and secretion of reproductive hormones, potentially mitigating ovarian aging. The impact of polyphenols on hormonal balance and ovarian health was tested in preclinical and clinical studies of women with PCOS, showing their benefits for ovarian health through the regulation of hormone secretion [38-40].
The microenvironment surrounding the ovaries is also essential. This environment normally supports ovary and oocyte maturation, but aging leads to the accumulation of metabolites that might disrupt the homeostasis. The gut microbiota and its metabolites influence the ovarian microenvironment via the gut-ovary axis. Through this axis, the ovary communicates with the gut microbiota via hormone secretion. On the other hand, gut microbes produce metabolic signalling molecules that can impact ovarian function. Studies that used fecal microbiota transplants suggest that maintaining “a youthful gut microbiota may help preserve ovarian function and reproductive health” [41]. Similarly, experiments with laying hens as a model suggested that polyphenol supplementation could improve ovarian function by modulating the gut microbiota [27, 42].

There appear to be many mechanisms and pathways through which polyphenols impact ovarian aging. This is unsurprising since polyphenols are a broad group of molecules with diverse chemical structures, resulting in distinct bioactivity profiles.
This knowledge can be used to design safe plant polyphenol-based interventions for female reproductive longevity that can be used alone or in combination with other treatments.
Literature
[1] Gong, H., Zhang, H., Liu, Y., Mao, X., & Wang, J. (2025). Role and mechanisms of plant polyphenols in ovarian aging. Journal of ovarian research, 18(1), 239.
[2] Potì, F., Santi, D., Spaggiari, G., Zimetti, F., & Zanotti, I. (2019). Polyphenol Health Effects on Cardiovascular and Neurodegenerative Disorders: A Review and Meta-Analysis. International journal of molecular sciences, 20(2), 351.
[3] Yong, W., Jiao, J., Kou, Z., Wang, C., & Pang, W. (2021). Resveratrol ameliorates malathion-induced estrus cycle disorder through attenuating the ovarian tissue oxidative stress, autophagy and apoptosis. Reproductive toxicology (Elmsford, N.Y.), 104, 8–15.
[4] Okamoto, N., Sato, Y., Kawagoe, Y., Shimizu, T., & Kawamura, K. (2022). Short-term resveratrol treatment restored the quality of oocytes in aging mice. Aging, 14(14), 5628–5640.
[5] Wu, H., Xue, J., Di, H., Lv, C., Hao, Y., & Nie, Z. (2022). Resveratrol improves ovarian function in aged rat by inhibiting oxidative stress and activating the Sirt1. General physiology and biophysics, 41(1), 53–61.
[6] Herrero, Y., Velázquez, C., Pascuali, N., May, M., Abramovich, D., Scotti, L., & Parborell, F. (2023). Resveratrol alleviates doxorubicin-induced damage in mice ovary. Chemico-biological interactions, 376, 110431.
[7] Zhu, H., Li, X., Qiao, M., Sun, X., & Li, G. (2023). Resveratrol Alleviates Inflammation and ER Stress Through SIRT1/NRF2 to Delay Ovarian Aging in a Short-Lived Fish. The journals of gerontology. Series A, Biological sciences and medical sciences, 78(4), 596–602.
[8] Fabbri, R., Macciocca, M., Vicenti, R., Caprara, G., Piccinni, M. P., Paradisi, R., Terzano, P., Papi, A., & Seracchioli, R. (2019). Epigallocatechin-3-gallate inhibits doxorubicin-induced inflammation on human ovarian tissue. Bioscience reports, 39(5), BSR20181424.
[9] Chen, Q., Xu, Z., Li, X., Du, D., Wu, T., Zhou, S., Yan, W., Wu, M., Jin, Y., Zhang, J., & Wang, S. (2021). Epigallocatechin gallate and theaflavins independently alleviate cyclophosphamide-induced ovarian damage by inhibiting the overactivation of primordial follicles and follicular atresia. Phytomedicine : international journal of phytotherapy and phytopharmacology, 92, 153752.
[10] Zhang, H., Su, W., Zhao, R., Li, M., Zhao, S., Chen, Z. J., & Zhao, H. (2024). Epigallocatechin-3-gallate improves the quality of maternally aged oocytes. Cell proliferation, 57(4), e13575.
[11] Fan, Z., Xiao, Y., Chen, Y., Wu, X., Zhang, G., Wang, Q., & Xie, C. (2015). Effects of catechins on litter size, reproductive performance and antioxidative status in gestating sows. Animal nutrition (Zhongguo xu mu shou yi xue hui), 1(4), 271–275.
[12] Zhou, C., Zhang, X., ShiYang, X., Wang, H., & Xiong, B. (2019). Tea polyphenol protects against cisplatin-induced meiotic defects in porcine oocytes. Aging, 11(13), 4706–4719.
[13] Cao, Y., Zhao, H., Wang, Z., Zhang, C., Bian, Y., Liu, X., Zhang, C., Zhang, X., & Zhao, Y. (2020). Quercetin promotes in vitro maturation of oocytes from humans and aged mice. Cell death & disease, 11(11), 965.
[14] Wu, M., Tang, W., Chen, Y., Xue, L., Dai, J., Li, Y., Zhu, X., Wu, C., Xiong, J., Zhang, J., Wu, T., Zhou, S., Chen, D., Sun, C., Yu, J., Li, H., Guo, Y., Huang, Y., Zhu, Q., Wei, S., … Wang, S. (2024). Spatiotemporal transcriptomic changes of human ovarian aging and the regulatory role of FOXP1. Nature aging, 4(4), 527–545.
[15] Jiao, Y., Wang, Y., Jiang, T., Wen, K., Cong, P., Chen, Y., & He, Z. (2022). Quercetin protects porcine oocytes from in vitro aging by reducing oxidative stress and maintaining the mitochondrial functions. Frontiers in cell and developmental biology, 10, 915898.
[16] Shah, M. Z. U. H., Shrivastva, V. K., Mir, M. A., Sheikh, W. M., Ganie, M. A., Rather, G. A., Shafi, M., Bashir, S. M., Ansari, M. A., Al-Jafary, M. A., Al-Qhtani, M. H., Homeida, A. M., & Al-Suhaimi, E. A. (2023). Effect of quercetin on steroidogenesis and folliculogenesis in ovary of mice with experimentally-induced polycystic ovarian syndrome. Frontiers in endocrinology, 14, 1153289.
[17] Barbe, A., Ramé, C., Mellouk, N., Estienne, A., Bongrani, A., Brossaud, A., Riva, A., Guérif, F., Froment, P., & Dupont, J. (2019). Effects of Grape Seed Extract and Proanthocyanidin B2 on In Vitro Proliferation, Viability, Steroidogenesis, Oxidative Stress, and Cell Signaling in Human Granulosa Cells. International journal of molecular sciences, 20(17), 4215.
[18] Luo, Y., Zhuan, Q., Li, J., Du, X., Huang, Z., Hou, Y., & Fu, X. (2020). Procyanidin B2 Improves Oocyte Maturation and Subsequent Development in Type 1 Diabetic Mice by Promoting Mitochondrial Function. Reproductive sciences (Thousand Oaks, Calif.), 27(12), 2211–2222.
[19] Zhou, Y., Lan, H., Dong, Z., Cao, W., Zeng, Z., & Song, J. L. (2021). Dietary proanthocyanidins alleviated ovarian fibrosis in letrozole-induced polycystic ovary syndrome in rats. Journal of food biochemistry, 45(5), e13723.
[20] Zhou, S., Zhao, A., Wu, Y., Mi, Y., & Zhang, C. (2022). Protective Effect of Grape Seed Proanthocyanidins on Oxidative Damage of Chicken Follicular Granulosa Cells by Inhibiting FoxO1-Mediated Autophagy. Frontiers in cell and developmental biology, 10, 762228.
[21] Azami, S. H., Nazarian, H., Abdollahifar, M. A., Eini, F., Farsani, M. A., & Novin, M. G. (2020). The antioxidant curcumin postpones ovarian aging in young and middle-aged mice. Reproduction, fertility, and development, 32(3), 292–303.
[22] Yin, Y. J., Zhang, Y. H., Wang, Y., Jiang, H., Zhang, J. B., Liang, S., & Yuan, B. (2023). Ferulic acid ameliorates the quality of in vitro-aged bovine oocytes by suppressing oxidative stress and apoptosis. Aging, 15(21), 12497–12512.
[23] Qian, F., Zhu, Z., Luo, C., Qi, R., Wei, L., Bo, L., Jiang, W., & Mao, C. (2025). Chlorogenic Acid Ameliorates Chronic Unpredictable Stress-Induced Diminished Ovarian Reserve Through Ovarian Renin-Angiotensin System. Molecular nutrition & food research, 69(5), e202400814.
[24] Chu, Y., Zhao, J., Zhao, Y., Li, Z., Yang, S., Chen, N., Liu, Y., Zhang, J., Zhou, L., & Chen, X. (2025). Multi-Omics Reveal the Effects and Regulatory Mechanism of Dietary Magnolol Supplementation on Production Performance of Post-Peak Laying Hens. Journal of agricultural and food chemistry, 73(7), 4027–4041.
[25] Chen, F., Zhang, H., Du, E., Jin, F., Zheng, C., Fan, Q., Zhao, N., Guo, W., Zhang, W., Huang, S., & Wei, J. (2021). Effects of magnolol on egg production, egg quality, antioxidant capacity, and intestinal health of laying hens in the late phase of the laying cycle. Poultry science, 100(2), 835–843.
[26] Gao, W., Jin, Y., Hao, J., Huang, S., Wang, D., Quan, F., Ren, W., Zhang, J., Zhang, M., & Yu, X. (2021). Procyanidin B1 promotes in vitro maturation of pig oocytes by reducing oxidative stress. Molecular reproduction and development, 88(1), 55–66.
[27] Moreira-Pinto, B., Costa, L., Felgueira, E., Fonseca, B. M., & Rebelo, I. (2021). Low Doses of Resveratrol Protect Human Granulosa Cells from Induced-Oxidative Stress. Antioxidants (Basel, Switzerland), 10(4), 561.
[28] Liang, Y., Xu, M. L., Gao, X., Wang, Y., Zhang, L. N., Li, Y. C., & Guo, Q. (2023). Resveratrol improves ovarian state by inhibiting apoptosis of granulosa cells. Gynecological endocrinology : the official journal of the International Society of Gynecological Endocrinology, 39(1), 2181652.
[29] Huang, Z., Pang, Y., Hao, H., Du, W., Zhao, X., & Zhu, H. (2018). Effects of epigallocatechin-3-gallate on bovine oocytes matured in vitro. Asian-Australasian journal of animal sciences, 31(9), 1420–1430.
[30] Li, N., & Liu, L. (2018). Mechanism of resveratrol in improving ovarian function in a rat model of premature ovarian insufficiency. The journal of obstetrics and gynaecology research, 44(8), 1431–1438.
[31] Yan, Z., Dai, Y., Fu, H., Zheng, Y., Bao, D., Yin, Y., Chen, Q., Nie, X., Hao, Q., Hou, D., & Cui, Y. (2018). Curcumin exerts a protective effect against premature ovarian failure in mice. Journal of molecular endocrinology, 60(3), 261–271.
[32] Barberino, R. S., Santos, J. M. S., Lins, T. L. B. G., Menezes, V. G., Monte, A. P. O., Gouveia, B. B., Palheta, R. C., Jr, & Matos, M. H. T. (2020). Epigallocatechin-3-gallate (EGCG) reduces apoptosis of preantral follicles through the phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) signaling pathway after in vitro culture of sheep ovarian tissue. Theriogenology, 155, 25–32.
[33] Heshmati, J., Moini, A., Sepidarkish, M., Morvaridzadeh, M., Salehi, M., Palmowski, A., Mojtahedi, M. F., & Shidfar, F. (2021). Effects of curcumin supplementation on blood glucose, insulin resistance and androgens in patients with polycystic ovary syndrome: A randomized double-blind placebo-controlled clinical trial. Phytomedicine : international journal of phytotherapy and phytopharmacology, 80, 153395.
[34] Ardehjani, N. A., Agha-Hosseini, M., Nashtaei, M. S., Khodarahmian, M., Shabani, M., Jabarpour, M., Fereidouni, F., Rastegar, T., & Amidi, F. (2024). Resveratrol ameliorates mitochondrial biogenesis and reproductive outcomes in women with polycystic ovary syndrome undergoing assisted reproduction: a randomized, triple-blind, placebo-controlled clinical trial. Journal of ovarian research, 17(1), 143.
[35] Ardehjani, N. A., Agha-Hosseini, M., Nashtaei, M. S., Khodarahmian, M., Shabani, M., Jabarpour, M., Fereidouni, F., Rastegar, T., & Amidi, F. (2024). Resveratrol ameliorates mitochondrial biogenesis and reproductive outcomes in women with polycystic ovary syndrome undergoing assisted reproduction: a randomized, triple-blind, placebo-controlled clinical trial. Journal of ovarian research, 17(1), 143.
[36] Vaez, S., Parivr, K., Amidi, F., Rudbari, N. H., Moini, A., & Amini, N. (2023). Quercetin and polycystic ovary syndrome; inflammation, hormonal parameters and pregnancy outcome: A randomized clinical trial. American journal of reproductive immunology (New York, N.Y. : 1989), 89(3), e13644.
[37] Wu, C., Chen, D., Stout, M. B., Wu, M., & Wang, S. (2025). Hallmarks of ovarian aging. Trends in endocrinology and metabolism: TEM, 36(5), 418–439.
[38] Hu, H., Zhang, J., Xin, X., Jin, Y., Zhu, Y., Zhang, H., Fan, R., Ye, Y., & Li, D. (2024). Efficacy of natural products on premature ovarian failure: a systematic review and meta-analysis of preclinical studies. Journal of ovarian research, 17(1), 46.
[39] Ali Fadlalmola, H., Elhusein, A. M., Al-Sayaghi, K. M., Albadrani, M. S., Swamy, D. V., Mamanao, D. M., El-Amin, E. I., Ibrahim, S. E., & Abbas, S. M. (2023). Efficacy of resveratrol in women with polycystic ovary syndrome: a systematic review and meta-analysis of randomized clinical trials. The Pan African medical journal, 44, 134.
[40] Malik, S., Saeed, S., Saleem, A., Khan, M. I., Khan, A., & Akhtar, M. F. (2024). Alternative treatment of polycystic ovary syndrome: pre-clinical and clinical basis for using plant-based drugs. Frontiers in endocrinology, 14, 1294406.
[41] Xu, L., Zhang, Q., Dou, X., Wang, Y., Wang, J., Zhou, Y., Liu, X., & Li, J. (2022). Fecal microbiota transplantation from young donor mice improves ovarian function in aged mice. Journal of genetics and genomics = Yi chuan xue bao, 49(11), 1042–1052.
[42] Zhang, T., Bai, S., Ding, X., Zeng, Q., Xuan, Y., Xu, S., Mao, X., Peng, H., Zhang, K., & Wang, J. (2024). Pu-erh tea theabrownin improves the ovarian function and gut microbiota in laying hens. Poultry science, 103(7), 103795.
View the article at lifespan.io






