FOXO4-DRI
FOXO4-DRI
FOXO4-DRI: Senolytic Peptide for Aging and Cellular Senescence Research
Disclaimer
Products described here are supplied for research use only and are not intended for diagnostic, therapeutic, or clinical application. All statements regarding biological activity reflect preclinical and in vitro findings exclusively and have not been evaluated by the U.S. Food and Drug Administration. These materials and related content are provided for educational and investigational purposes only and are intended solely for qualified researchers in laboratory and academic settings.
Introduction: Revolutionary Breakthrough in Senescent Cell Research
FOXO4-DRI represents a paradigm shift in aging research, emerging from groundbreaking investigations by Dr. Peter de Keizer and colleagues at Erasmus University Medical Center that fundamentally changed our understanding of cellular senescence and its research targeting. When their seminal research was published in *Cell* in March 2017, it introduced the first precision senolytic peptide capable of selectively eliminating senescent cells while sparing healthy tissue, opening unprecedented opportunities for studying aging mechanisms and developing research tools for age-related cellular dysfunction. This D-retro-inverso peptide specifically disrupts the FOXO4-p53 protein interaction that maintains senescent cell viability, providing researchers with a sophisticated tool for investigating cellular senescence biology and its role in aging processes.
Laboratory investigations have revealed FOXO4-DRI's exceptional selectivity for senescent cells, with research demonstrating that more than 80% of senescent cells undergo apoptosis while healthy cells remain unaffected. This unprecedented precision stems from the peptide's targeting of a unique vulnerability in senescent cell biology, where FOXO4 prevents p53 from executing its normal pro-apoptotic function. The compound's design as a competitive inhibitor fused with HIV-TAT for cellular uptake represents advanced peptide engineering that enables intracellular delivery and specific protein-protein interaction disruption, providing researchers with tools for studying senescence mechanisms that were previously inaccessible.
Contemporary research applications span diverse fields including aging biology, tissue regeneration, cancer research, and regenerative medicine, with laboratory models demonstrating remarkable restoration of tissue homeostasis in both accelerated and natural aging conditions. The compound's ability to reverse age-related deficits in fitness, fur density, renal function, and reproductive health in animal models provides researchers with unprecedented opportunities to study the causal relationships between cellular senescence and aging phenotypes. FOXO4-DRI's well-characterized mechanism and expanding research applications position it as an essential research tool for advancing our understanding of senescence biology and developing next-generation approaches to studying aging and age-related diseases in laboratory settings.
Molecular Design and Protein Interaction Targeting
Research investigations reveal FOXO4-DRI's sophisticated molecular architecture, designed through advanced peptide engineering principles to achieve selective disruption of senescent cell survival mechanisms. The compound functions as a synthetic D-retro-inverso peptide carrying a shortened version of the FOXO4 segment that binds to p53, acting as a competitive inhibitor of this critical protein-protein interaction. Laboratory studies demonstrate that the peptide was engineered as a fusion with HIV-TAT, a basic and hydrophilic sequence that enables energy-independent cellular uptake, ensuring effective intracellular delivery for precise molecular targeting in experimental settings.
Structural analysis demonstrates how FOXO4-DRI exploits the unique architecture of senescent cells, where FOXO4 latches onto p53 and prevents it from executing its normal pro-apoptotic function. Research shows that in senescent cells, p53 levels are maintained low through ubiquitination while its pro-apoptotic activity is repressed through nuclear segregation by FOXO4. The peptide's competitive inhibition restores p53 activity by promoting nuclear exclusion, allowing p53 to execute its pro-apoptotic program and eliminate senescent cells through targeted pathway activation.
Advanced molecular studies reveal that FOXO4-DRI promotes p53 nuclear exclusion and subsequent mitochondrial targeting, where p53 interacts with BCL-2 family proteins to promote mitochondrial outer membrane permeabilization and cytochrome c release. This mechanism initiates the intrinsic apoptosis pathway specifically in senescent cells, while healthy cells maintain different p53 regulation mechanisms and remain unaffected by the peptide administration. Recent solution NMR structural studies have provided detailed molecular models of the p53 transactivation domain in complex with both the FOXO4 forkhead domain and FOXO4-DRI, advancing understanding of the precise molecular interactions that enable selective senescent cell targeting in research applications.
Senescent Cell Biology and Selective Targeting Mechanisms
Laboratory research demonstrates that FOXO4-DRI's selectivity stems from exploiting fundamental differences between senescent and healthy cell biology, particularly the paradoxical state where senescent cells are primed for apoptosis but protected by specific survival mechanisms. Research reveals that senescent cells show upregulation of pro-apoptotic initiators PUMA and BIM while anti-apoptotic guardian BCL-2 is reduced, suggesting these cells are ready to undergo apoptosis but the execution is restrained by the FOXO4-p53 interaction. This cellular priming provides the mechanistic foundation for FOXO4-DRI's selective targeting, enabling researchers to eliminate senescent cells without affecting healthy tissue function.
Experimental investigations show that senescent cells typically exhibit permanent cell cycle arrest mediated by p53/p21 signaling, creating a unique cellular state that depends on specific survival mechanisms for viability. FOXO4-DRI's disruption of FOXO4-p53 interactions specifically targets these survival mechanisms without affecting normal cell cycle regulation in healthy cells, providing researchers with precision tools for studying senescence biology. Laboratory protocols demonstrate that under optimal dosing conditions, the peptide eliminates senescent cells while destroying an undetectable number of healthy cells, representing exceptional selectivity compared to earlier senolytic approaches.
Research models reveal that senescent cells accumulate with aging and in response to various stressors, secreting inflammatory factors known as the senescence-associated secretory phenotype (SASP) that promotes chronic inflammation and can induce senescence in normal cells. FOXO4-DRI's ability to selectively eliminate these cells provides researchers with tools for studying how senescent cell burden contributes to aging phenotypes and age-related diseases. Laboratory studies demonstrate that elimination of senescent cells through FOXO4-DRI administration can break the cycle of senescence induction and chronic inflammation, offering insights into interventions for age-related cellular dysfunction and tissue deterioration.
Laboratory Model Applications and Research Protocols
Extensive laboratory research demonstrates FOXO4-DRI's effectiveness across diverse experimental systems, with radiation-induced senescent IMR90 mammalian fibroblasts serving as a foundational model for studying peptide selectivity and mechanism. Research protocols show that after single-dose administration, more than half of senescent cells disappear while healthy cells remain intact, providing researchers with quantitative measures of senolytic efficacy. Laboratory investigations using ionizing radiation-induced senescent cells demonstrate that FOXO4-DRI causes p53 nuclear exclusion and mitochondrial targeting, validating the proposed mechanism of selective senescent cell elimination through targeted protein-protein interaction disruption.
Animal model research reveals remarkable efficacy in both accelerated and natural aging conditions, with XpdTTD/TTD mice (accelerated aging model) showing restoration of multiple age-related deficits including fitness, fur density, and renal function following FOXO4-DRI administration. Laboratory protocols demonstrate that the peptide neutralizes doxorubicin-induced chemotoxicity and shows that research targeting of senescent cells is feasible even after health decline has occurred. Research in naturally aged mice demonstrates measurable improvements in physical performance, with treated animals showing increased running endurance and improved overall fitness metrics compared to untreated aged controls.
Advanced research applications include tissue-specific studies where FOXO4-DRI has demonstrated broad regenerative effects across multiple organ systems. Laboratory investigations show restored kidney function in both fast aging and naturally aged mouse models, suggesting senescent cell elimination can reverse organ-specific age-related decline. Research protocols demonstrate robust improvement of fur density through restored hair follicle function, with quantitative assessment using infrared-measured abdominal surface temperature showing that elevated temperatures in aging mice (due to fur loss) were reduced following administration. These diverse applications provide researchers with validated experimental frameworks for studying senescence contributions to age-related phenotypes across multiple tissue types and organ systems.
Tissue Regeneration and Organ Function Research
Research investigations demonstrate FOXO4-DRI's capacity to restore tissue homeostasis across multiple organ systems, providing researchers with tools for studying the causal relationships between senescent cell burden and age-related functional decline. Laboratory studies in renal function demonstrate that FOXO4-DRI administration restores kidney function in both accelerated aging models and naturally aged mice, indicating that senescent cell elimination can reverse organ-specific age-related deterioration. These findings provide researchers with evidence that senescence contributes directly to organ dysfunction and that targeted interventions can restore functional capacity even after decline has occurred.
Reproductive health research reveals significant applications in studying age-related endocrine dysfunction, with laboratory studies showing that FOXO4-DRI improves the testicular microenvironment and alleviates age-related testosterone secretion insufficiency by targeting senescent Leydig cells in aged male mice. Research protocols demonstrate quantitative improvements in testosterone levels and testicular function, providing insights into mechanisms of reproductive aging and potential interventions for age-related hormonal decline. These studies offer researchers validated models for studying endocrine aging and senescence contributions to reproductive dysfunction.
Cartilage and joint research applications demonstrate FOXO4-DRI's utility in tissue engineering and regenerative medicine studies. Laboratory investigations using mammalian chondrocytes expanded in vitro show that the senolytic peptide selectively removes senescent cells while significantly reducing senescence-associated β-galactosidase staining. Research confirms no noticeable cell loss in cultures of non-senescent chondrocytes, validating selective targeting capabilities. This selectivity makes FOXO4-DRI valuable for tissue engineering applications where elimination of senescent cells from expanded cell populations could improve research outcomes and provide researchers with enhanced cell preparation methods for regenerative medicine investigations.
Cancer Research and p53 Pathway Studies
Laboratory research demonstrates FOXO4-DRI's significant utility in cancer research applications, particularly for studying p53 biology and developing combination investigational approaches. Research investigations have shown that targeted peptides can specifically eliminate FOXO4+ senescent cancer cells, with implications for eradicating residual disease and serving as combination therapy for frontline cancer administration in experimental models. Laboratory studies demonstrate that senescent cancer cells maintain similar FOXO4-p53 interactions that protect them from apoptosis, making them susceptible to FOXO4-DRI-mediated elimination while providing researchers with tools for studying cancer cell senescence and survival mechanisms.
Combination therapy research reveals promising applications where FOXO4-DRI enhances the efficacy of conventional cancer administrations. Laboratory studies demonstrate that combinations of improved senolytic peptides (such as ES2, which shows 3-7 times greater efficacy than FOXO4-DRI) with chemotherapeutic agents like Dabrafenib result in greater elimination of both cancer and senescent cells. Research protocols show significant improvement in survival compared to chemotherapy alone in preclinical melanoma models, providing researchers with frameworks for studying "one-two punch" strategies that combine senolytic and targeted cancer therapies.
Research applications include investigation of chemotherapy-induced senescence and its role in administration resistance and tumor recurrence. Laboratory studies demonstrate that FOXO4-DRI can eliminate chemotherapy-induced senescent cells that may contribute to administration resistance, while the timing of senolytic administration relative to chemotherapy requires careful optimization in experimental designs. Research reveals that some chemotherapy-induced senescent cells may initially contribute to administration efficacy through SASP-mediated immune activation, but their persistence can promote tumor recurrence and administration resistance. These findings provide researchers with insights into optimizing combination approaches and understanding the complex roles of senescence in cancer administration outcomes.
Advanced Research Applications and Emerging Directions
Current research developments focus on expanding FOXO4-DRI applications beyond traditional aging studies to include investigation of fibrosis, neurodegeneration, and immune system aging. Laboratory research demonstrates efficacy in bleomycin-induced pulmonary fibrosis mouse models, with treated groups showing milder pathologic changes and less collagen deposition compared to controls. Research investigations reveal that senescent cells contribute to fibrosis through SASP-mediated inflammation, and their elimination can reduce fibrotic progression, providing researchers with tools for studying senescence contributions to fibrotic diseases and developing interventions for fibrosis research models.
Neurodegeneration research applications explore whether selective elimination of senescent glial cells could restore immune function and reduce neuroinflammation in aging brain models. Laboratory studies investigate how senescent microglia and astrocytes contribute to neuroinflammation and disease progression in neurodegenerative disease models, while FOXO4-DRI provides researchers with tools for testing whether senescent cell elimination can restore brain function and reduce neuroinflammatory markers. These emerging applications offer researchers opportunities to study senescence contributions to neurodegeneration and develop novel approaches for studying brain aging and neurodegenerative disease mechanisms.
Immunosenescence research investigates whether selective elimination of senescent immune cells could restore immune function in aging models, potentially improving vaccine responses and immune surveillance capabilities. Laboratory protocols explore how senescent T cells, B cells, and other immune cells contribute to age-related immune dysfunction, while FOXO4-DRI administration provides researchers with methods for testing immune system restoration following senescent cell elimination. Research applications include studying how senescent immune cells affect vaccine efficacy, immune surveillance, and infection resistance in aging models, providing insights into interventions for age-related immune decline and immunosenescence mechanisms.
Next-Generation Senolytic Development and Optimization
Research advancements have led to development of improved senolytic peptides with enhanced efficacy compared to first-generation FOXO4-DRI. Laboratory studies demonstrate that ES2 peptides show 3-7 times more potent senolytic activity than FOXO4-DRI in both in vitro and in vivo experimental systems, while CPP-CAND demonstrates approximately 1.2-fold higher selectivity despite shorter peptide length. These improvements provide researchers with enhanced tools for studying senescent cell elimination and enable investigation of dose-response relationships and selectivity mechanisms in various experimental models.
Structural optimization research focuses on understanding molecular determinants of peptide efficacy and selectivity through detailed protein-protein interaction studies. Recent solution NMR structural investigations provide molecular models of p53 transactivation domain interactions with both FOXO4 and senolytic peptides, enabling rational design approaches for next-generation compounds. Laboratory research utilizes these structural insights to develop peptides with improved cellular uptake, enhanced selectivity, and reduced off-target effects, providing researchers with increasingly sophisticated tools for senescence research applications.
Research pipeline development through companies like Cleara Biotech provides researchers with insights into translational senolytic development while maintaining focus on research applications. Laboratory investigations continue to optimize peptide formulations, delivery methods, and dosing protocols to enhance research utility and enable investigation of chronic senescent cell elimination effects. Research protocols explore combination approaches with other senolytic compounds such as dasatinib and quercetin, providing frameworks for studying multi-target senolytic strategies and understanding synergistic effects in various experimental models and disease research applications.
Research Protocol Considerations and Experimental Design
Laboratory protocol development for FOXO4-DRI research requires careful consideration of dosing strategies, administration timing, and experimental endpoints to maximize scientific insights while ensuring reproducible results. Research protocols typically employ intermittent dosing strategies (once weekly or 3-day cycles) designed to prevent excessive apoptosis while enabling effective senescent cell elimination, with doses ranging from 5-10 mg/kg body weight in animal studies. Cell culture applications utilize concentrations of 10-50 μM for effective senescent cell elimination, with administration durations typically 24-72 hours depending on experimental objectives and cellular models.
Experimental design considerations include the importance of timing senolytic administration relative to senescence induction or disease progression, as the biological context affects both efficacy and interpretation of results. Laboratory studies demonstrate that FOXO4-DRI effectiveness depends on senescent cell burden, tissue type, and the specific mechanisms maintaining senescent cell viability in different experimental conditions. Research protocols must account for potential beneficial roles of some senescent cells in wound healing and immune responses, requiring careful consideration of administration timing and duration in experimental designs.
Safety and monitoring considerations for research applications include assessment of potential effects on tissue regeneration, immune function, and wound healing capacity, as senescent cells may play beneficial roles in some biological processes. Laboratory protocols should include appropriate controls and monitoring strategies to distinguish between beneficial elimination of harmful senescent cells and potential adverse effects on beneficial senescent cell populations. Research applications require consideration of the peptide's selectivity mechanisms and potential off-target effects, while the well-characterized mechanism provides foundation for power calculations and experimental planning in senescence research applications across diverse biological systems and disease models.
Future Research Directions and Scientific Applications
Current research trajectories focus on understanding fundamental questions about senescence biology, aging mechanisms, and the development of precision interventions for age-related diseases using FOXO4-DRI as a research tool. Laboratory investigations explore how different types of senescent cells contribute to various aging phenotypes, while the peptide's selectivity enables researchers to test causal relationships between senescent cell burden and age-related functional decline. Research protocols investigate how senescent cell elimination affects stem cell function, tissue regeneration capacity, and organ homeostasis, providing insights into mechanisms of aging and potential interventions for age-related diseases.
Advanced research applications investigate the temporal dynamics of senescent cell accumulation and elimination in various disease models, with FOXO4-DRI providing tools for studying how senescent cell burden changes over time and in response to different stressors. Laboratory studies explore optimal administration regimens for different research objectives, including acute versus chronic senescent cell elimination and the effects of repeated administrations on tissue function and aging markers. Research investigations focus on understanding individual variation in senescent cell accumulation and responses to senolytic administration, providing foundation for developing personalized approaches to senescence research.
Emerging research directions include investigation of senescence heterogeneity and tissue-specific senescent cell populations, with FOXO4-DRI serving as a tool for studying how different senescent cell types contribute to various pathological processes. Laboratory applications explore the development of combination approaches that target multiple senescence mechanisms simultaneously, while research into biomarkers of senescent cell burden and senolytic administration efficacy enables better experimental monitoring and outcome assessment. Future research priorities include understanding the long-term effects of senescent cell elimination on tissue homeostasis, regenerative capacity, and overall healthspan, providing foundation for developing sophisticated approaches to aging research and intervention strategies.
Conclusion: Research Implications and Scientific Utility
FOXO4-DRI represents a transformative breakthrough in senescence research that has fundamentally changed our understanding of cellular aging and opened unprecedented opportunities for studying age-related diseases and interventions. From its origins in Dr. Peter de Keizer's pioneering research at Erasmus University Medical Center to its current status as a leading senolytic research tool, FOXO4-DRI demonstrates how precise molecular targeting can enable selective elimination of harmful cellular populations while preserving healthy tissue function. The peptide's exceptional selectivity for senescent cells, combined with its well-characterized mechanism of FOXO4-p53 interaction disruption, provides researchers with powerful capabilities for investigating senescence biology that were previously impossible to study.
Laboratory research has established FOXO4-DRI as an invaluable tool for investigating fundamental questions in aging biology, regenerative medicine, cancer research, and tissue engineering, with its proven efficacy across diverse experimental models enabling researchers to test causal relationships between senescence and age-related phenotypes. The compound's ability to restore tissue homeostasis and reverse age-related functional decline in animal models provides direct evidence for the role of senescent cells in aging processes, while its applications in cancer research offer insights into senescence mechanisms in disease progression and administration resistance. The ongoing development of next-generation senolytic peptides builds upon FOXO4-DRI's foundation while providing researchers with increasingly sophisticated tools for senescence research.
Future research applications will likely expand to include investigation of senescence heterogeneity, tissue-specific senescent cell populations, and the development of personalized approaches to senolytic intervention based on individual senescent cell burden and characteristics. The compound's role in advancing our understanding of aging mechanisms, developing intervention strategies, and exploring the research potential of senescent cell elimination positions it at the forefront of aging research priorities. For researchers investigating cellular senescence, aging biology, regenerative medicine, and age-related diseases, FOXO4-DRI offers a sophisticated tool that combines mechanistic precision with practical utility, enabling advancement in our understanding of senescence biology and the development of innovative approaches to studying aging and developing research tools for age-related cellular dysfunction.
References
- Baar, M.P. et al. (2017). Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 169(1):132-147. https://doi.org/10.1016/j.cell.2017.02.031
- Gorgoulis, V. et al. (2019). Cellular senescence: defining a path forward. Cell 179(4):813-827. https://doi.org/10.1016/j.cell.2019.10.005
- van Deursen, J.M. (2014). The role of senescent cells in ageing. Nature 509(7501):439-446. https://doi.org/10.1038/nature13193
- López-Otín, C. et al. (2013). The hallmarks of aging. Cell 153(6):1194-1217. https://doi.org/10.1016/j.cell.2013.05.039
- Kirkland, J.L. & Tchkonia, T. (2017). Cellular senescence: a translational perspective. EBioMedicine 21:21-28. https://doi.org/10.1016/j.ebiom.2017.04.013
- Tchkonia, T. et al. (2013). Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. Journal of Clinical Investigation 123(3):966-972. https://doi.org/10.1172/JCI64098
- Baker, D.J. et al. (2011). Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479(7372):232-236. https://doi.org/10.1038/nature10600
- Xu, M. et al. (2018). Senolytics improve physical function and increase lifespan in old age. Nature Medicine 24(8):1246-1256. https://doi.org/10.1038/s41591-018-0092-9
- Zhu, Y. et al. (2015). The Achilles' heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell 14(4):644-658. https://doi.org/10.1111/acel.12344
- Chang, J. et al. (2016). Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nature Medicine 22(1):78-83. https://doi.org/10.1038/nm.4010
- Yosef, R. et al. (2016). Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nature Communications 7:11190. https://doi.org/10.1038/ncomms11190
- Fuhrmann-Stroissnigg, H. et al. (2017). Identification of HSP90 inhibitors as a novel class of senolytics. Nature Communications 8:422. https://doi.org/10.1038/s41467-017-00314-z
- Zhu, Y. et al. (2017). Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 16(4):815-817. https://doi.org/10.1111/acel.12618
- Hickson, L.J. et al. (2019). Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 47:446-456. https://doi.org/10.1016/j.ebiom.2019.08.069
- Justice, J.N. et al. (2019). Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-mammalian, open-label, pilot study. EBioMedicine 40:554-563. https://doi.org/10.1016/j.ebiom.2018.12.052
- Johmura, Y. et al. (2021). Senolysis by glutaminolysis inhibition ameliorates various age-associated disorders. Science 371(6526):265-270. https://doi.org/10.1126/science.abb5916
- González-Gualda, E. et al. (2021). Galactose-modified duocarmycin prodrugs as senolytics. Aging Cell 20(4):e13338. https://doi.org/10.1111/acel.13338
- Triana-Martínez, F. et al. (2019). Identification and characterization of Cardiac Glycosides as senolytic compounds. Nature Communications 10:4731. https://doi.org/10.1038/s41467-019-12888-x
- Guerrero, A. et al. (2019). Cardiac glycosides are broad-spectrum senolytics. Nature Metabolism 1(11):1074-1088. https://doi.org/10.1038/s42255-019-0122-z
- Samaraweera, L. et al. (2017). A high content drug screen identifies idelalisib as a selective inducer of apoptosis in senescent cells. Aging Cell 16(4):813-814. https://doi.org/10.1111/acel.12617
Last reviewed: October 2025
| CAS Number | 2460055-10-9 |
|---|---|
| Molecular Formula | C228H388N86O64 |
| Molecular Weight | 5358.06 g/Mol |
| Sequence | D-(Leu-Thr-Leu-Arg-Lys-Glu-Pro-Ala-Ser-Glu-Ile-Ala-Gln-Ser-Ile-Leu-Glu-Ala-Tyr-Ser-Gln-Asn-Gly-Trp-Ala-Asn-Arg-Arg-Ser-Gly-Gly-Lys-Arg-Pro-Pro-Pro-Arg-Arg-Arg-Gln-Arg-Arg-Lys-Lys-Arg-Gly) |
Cosmic Peptides
Certificate of Analysis
Unable to load certificate. Download COA