SS-31
SS-31
SS-31 (Elamipretide): Mitochondrial-Targeted Cardiolipin-Binding Peptide
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.
Scientific Discovery and Mitochondrial Targeting Innovation
SS-31, scientifically designated as Elamipretide (molecular formula C₃₂H₄₉N₉O₅, 639.8 g/mol), represents a groundbreaking advancement in mitochondrial-targeted therapeutics, engineered by Hazel Szeto and Peter Schiller as the lead compound in their revolutionary peptide series. This synthetic tetrapeptide with the sequence D-Arg-dimethylTyr-Lys-Phe-NH₂ demonstrates unprecedented selectivity for mitochondrial targeting through its unique cardiolipin-binding mechanism, establishing it as a cornerstone research tool for investigating cellular bioenergetics and mitochondrial dysfunction across diverse pathological states. The peptide's alternating aromatic-basic amino acid motif enables it to selectively penetrate cellular and mitochondrial membranes without requiring specific transporters, distinguishing it from conventional antioxidants that distribute non-specifically throughout cellular compartments.
The discovery of SS-31's cardiolipin-binding capacity fundamentally transformed understanding of mitochondrial membrane dynamics and opened new avenues for targeted mitochondrial intervention. Unlike traditional antioxidants that provide broad cellular protection, SS-31's precise targeting of the inner mitochondrial membrane allows researchers to dissect the specific contributions of mitochondrial dysfunction to various pathological processes. This targeting specificity stems from the peptide's cationic nature and its ability to recognize the unique lipid environment of the inner mitochondrial membrane, where cardiolipin constitutes approximately 20% of total phospholipids and serves as both a structural component and functional cofactor for respiratory chain complexes.
The peptide's breakthrough significance lies in its ability to address mitochondrial dysfunction at the source, offering researchers unprecedented precision in studying cellular energetics, oxidative stress responses, and mitochondrial membrane integrity. Research investigations have demonstrated SS-31's capacity to restore mitochondrial function across multiple experimental models, from cellular energy production to complex tissue-level pathological processes, establishing it as an essential research tool for laboratories investigating aging mechanisms, cardiovascular pathophysiology, neurodegeneration, and metabolic disorders. This remarkable selectivity and efficacy have positioned SS-31 as a leading research compound for advancing scientific understanding of mitochondrial biology and developing novel research strategies targeting cellular powerhouses.
Cardiolipin-Binding Mechanism and Membrane Stabilization
SS-31's primary mechanism centers on its high-affinity binding to cardiolipin, a unique four-acyl chain phospholipid exclusively localized to the inner mitochondrial membrane where it plays critical roles in maintaining respiratory chain supercomplex organization and membrane stability. Research demonstrates that SS-31 binding to cardiolipin occurs with remarkable specificity, shielding this essential phospholipid from oxidative degradation while preserving the structural integrity necessary for optimal electron transport chain function. The peptide's dimethyltyrosine residue acts as a localized antioxidant, forming unreactive tyrosine radicals that subsequently couple to create di-tyrosine linkages, effectively neutralizing harmful reactive oxygen species at their mitochondrial source without interfering with physiological redox signaling pathways essential for normal cellular function.
The cardiolipin-SS-31 interaction stabilizes respiratory chain supercomplexes, particularly the association of Complexes I, III, and IV into tightly organized assemblies that facilitate efficient electron transfer while minimizing electron leak and subsequent ROS generation. Laboratory studies reveal that SS-31 binding prevents cardiolipin peroxidation, a critical early event in mitochondrial dysfunction that leads to respiratory chain disassembly and compromised ATP synthesis. This protective mechanism maintains the optimal lipid environment necessary for proper protein-lipid interactions within the respiratory chain, preserving both structural organization and functional efficiency of oxidative phosphorylation. Research indicates that cardiolipin oxidation typically occurs early in pathological processes, making SS-31's protective mechanism particularly valuable for investigating mitochondrial dysfunction initiation and progression.
The peptide's targeting mechanism involves electrostatic interactions between its cationic residues and the anionic headgroups of cardiolipin, followed by insertion of aromatic residues into the lipid bilayer environment. This dual interaction model explains SS-31's remarkable selectivity for cardiolipin over other phospholipids and its ability to accumulate specifically in mitochondrial membranes at concentrations 1000-fold higher than in cytoplasm. Research investigations using fluorescently labeled SS-31 analogs demonstrate rapid mitochondrial uptake within minutes of exposure, with sustained residence times that correlate with protective effects lasting hours after peptide administration. The stability of the cardiolipin-SS-31 complex allows for sustained protection against oxidative damage while maintaining the membrane fluidity necessary for proper respiratory chain function, providing researchers with an excellent tool for studying long-term mitochondrial stability and function in various experimental paradigms.
Mitochondrial Bioenergetics and ATP Production Enhancement
Research demonstrates that SS-31 significantly enhances mitochondrial bioenergetic efficiency through multiple complementary mechanisms that optimize cellular energy production and metabolic flexibility. Laboratory investigations reveal that SS-31 administration improves ADP sensitivity in aged mitochondria by enhancing adenine nucleotide translocator (ANT) function, the rate-limiting step in ADP/ATP exchange across the inner mitochondrial membrane. Studies show that SS-31 directly binds to both ANT and ATP synthase, resulting in improved ADP uptake and enhanced ATP production capacity that can restore youthful bioenergetic profiles in aged cellular models. This mechanism proves particularly valuable for research investigations studying age-related decline in cellular energy metabolism and the role of mitochondrial dysfunction in metabolic disorders.
The peptide's effects on respiratory chain supercomplex organization contribute significantly to improved bioenergetic efficiency by facilitating optimal electron transfer and reducing energy dissipation as heat. Research findings indicate that SS-31 administration preserves the structural integrity of respiratory supercomplexes, maintaining the spatial organization necessary for efficient electron channeling between Complex I and Complex III, and subsequently to Complex IV. This optimization reduces electron leak and minimizes the formation of reactive oxygen species that can damage respiratory chain components and compromise energy production. Laboratory studies demonstrate that SS-31-treated mitochondria exhibit improved respiratory control ratios and enhanced P/O ratios, indicating more efficient coupling of oxygen consumption to ATP synthesis.
Comprehensive bioenergetic analyses reveal that SS-31 enhances both maximal respiratory capacity and respiratory reserve capacity, providing cells with greater metabolic flexibility to respond to changing energy demands. Research investigations show that SS-31 administration improves mitochondrial membrane potential stability while maintaining appropriate proton gradient for ATP synthesis, preventing the excessive hyperpolarization that can lead to increased ROS production. Studies in aged animal models demonstrate that SS-31 administration restores muscle endurance and exercise capacity by improving cellular energy production efficiency, with improvements correlating directly with enhanced mitochondrial bioenergetic parameters. These findings establish SS-31 as an exceptional research tool for investigating the relationship between mitochondrial function and cellular energy metabolism across various experimental conditions and disease models.
Cardiovascular Research Applications and Cardioprotection
Laboratory investigations consistently demonstrate SS-31's remarkable cardioprotective properties across diverse experimental models of cardiac injury and dysfunction, establishing it as a premier research tool for cardiovascular mitochondrial biology studies. Research in animal models of ischemia-reperfusion injury reveals that SS-31 administration significantly reduces infarct size by 25-40% when administered before, during, or immediately after ischemic events, indicating research windows relevant for translational research applications. The peptide's protective mechanism involves preservation of mitochondrial membrane integrity, prevention of mitochondrial permeability transition pore opening, and maintenance of calcium homeostasis during oxidative stress conditions that typically characterize cardiac injury models.
Comprehensive studies in canine models of heart failure demonstrate that chronic SS-31 administration produces sustained improvements in left ventricular systolic function, with ejection fraction improvements of 15-20% observed after 8-12 weeks of administration. Research investigations reveal that these functional improvements correlate with normalization of cardiac mitochondrial morphology, including restoration of cristae architecture and reduction in mitochondrial swelling that characterizes failing myocardium. Laboratory analyses show that SS-31 administration reverses the metabolic remodeling associated with heart failure, restoring fatty acid oxidation capacity and improving glucose utilization efficiency that becomes impaired in diseased cardiac tissue. These findings provide researchers with valuable insights into the role of mitochondrial dysfunction in cardiac pathophysiology and potential research targets for cardiovascular diseases.
Studies examining SS-31's effects on cardiac aging reveal significant prevention of age-related mitochondrial dysfunction and preservation of cardiac performance in aged animal models. Research demonstrates that SS-31 administration reduces age-associated increases in protein S-glutathionylation, a marker of oxidative protein modification that accumulates with aging and contributes to mitochondrial dysfunction. Laboratory investigations show that the peptide preserves cardiac mitochondrial calcium handling capacity and maintains appropriate stress-response mechanisms that become impaired with aging. Long-term studies indicate that SS-31 administration prevents the development of cardiac hypertrophy and fibrosis associated with aging, maintaining cardiac compliance and diastolic function that typically deteriorate with advanced age. These comprehensive cardiovascular research applications establish SS-31 as an invaluable tool for investigating cardiac mitochondrial biology and developing interventions targeting cardiovascular aging and disease progression.
Neuroprotective Mechanisms and Brain Research Applications
Research investigations reveal SS-31's potent neuroprotective properties across multiple experimental models of neurodegeneration, neuroinflammation, and cognitive dysfunction, establishing it as a critical research tool for neuroscience laboratories studying mitochondrial contributions to brain pathology. Studies in lipopolysaccharide-induced neuroinflammation models demonstrate that SS-31 administration significantly ameliorates learning and memory impairments while preserving hippocampal mitochondrial function, including maintenance of membrane potential and ATP production capacity that become compromised during inflammatory responses. Laboratory analyses show that SS-31 administration reduces microglial activation by 40-60% and decreases pro-inflammatory cytokine production, including TNF-α, IL-1β, and IL-6, while promoting anti-inflammatory mediator expression that supports neuronal survival and synaptic integrity.
Comprehensive research in Alzheimer's disease models reveals that SS-31 provides protection against amyloid-beta toxicity through multiple complementary mechanisms that preserve neuronal mitochondrial function and reduce oxidative stress-mediated damage. Studies demonstrate that SS-31 administration reduces amyloid-beta accumulation in neuronal cultures while protecting against the mitochondrial dysfunction typically induced by amyloid peptide exposure. Laboratory investigations show that the peptide preserves synaptic protein expression, including synaptophysin and PSD-95, and maintains dendritic spine density that becomes reduced in Alzheimer's disease models. Research findings indicate that SS-31 administration supports brain-derived neurotrophic factor (BDNF) signaling pathways essential for synaptic plasticity and cognitive function, providing researchers with valuable insights into mitochondrial contributions to neurodegenerative disease progression.
Studies in Parkinson's disease models demonstrate SS-31's ability to protect dopaminergic neurons against toxin-induced damage while preserving motor function and reducing neuroinflammatory responses characteristic of this neurodegenerative condition. Research investigations reveal that SS-31 administration maintains mitochondrial biogenesis capacity in aging brain tissue by supporting PGC-1α activation through improved cellular energy status and reduced oxidative suppression of mitochondrial DNA replication. Laboratory studies show that the peptide preserves cognitive performance in aged animal models, with improvements in spatial memory, working memory, and executive function tasks that correlate with maintained mitochondrial function in brain regions critical for cognitive processing. These extensive neuroprotective research applications establish SS-31 as an essential tool for investigating brain mitochondrial biology and developing potential interventions for neurodegenerative diseases and cognitive aging.
Aging Research and Cellular Longevity Applications
Laboratory investigations demonstrate SS-31's remarkable ability to address fundamental mechanisms of cellular aging by targeting mitochondrial dysfunction, a primary hallmark of the aging process that contributes to declined physiological function across multiple organ systems. Research studies in aged animal models reveal that 8-week SS-31 administration protocols almost completely reverse age-related increases in protein S-glutathionylation, a critical marker of oxidative protein modification that accumulates with aging and impairs cellular function. Studies show that SS-31 administration restores muscle endurance capacity in aged mice to levels approaching those of young animals, with improvements correlating directly with enhanced mitochondrial bioenergetic parameters and reduced oxidative damage markers throughout skeletal muscle tissue.
Comprehensive aging research reveals that SS-31 administration improves cellular conditions that favor PGC-1α activation, the master regulator of mitochondrial biogenesis that becomes suppressed with aging due to oxidative stress and metabolic dysfunction. Laboratory analyses demonstrate that SS-31 enhances AMPK activation pathways while improving NAD+/NADH ratios and reducing oxidative suppression of mitochondrial DNA replication, creating favorable conditions for mitochondrial renewal and expansion. Research investigations show that these improvements in mitochondrial biogenesis capacity translate to enhanced cellular energy production, improved stress resistance, and restored metabolic flexibility that characterizes youthful cellular phenotypes. Studies indicate that SS-31 administration preserves telomere length and reduces cellular senescence markers, providing researchers with valuable insights into mitochondrial contributions to cellular longevity and aging mechanisms.
Long-term research studies demonstrate that SS-31 administration prevents many age-associated pathological changes, including muscle atrophy, cognitive decline, and cardiovascular dysfunction that typically develop with advanced aging. Laboratory investigations reveal that the peptide preserves stem cell function and regenerative capacity by maintaining mitochondrial integrity in stem cell populations that become increasingly dysfunctional with aging. Research findings show that SS-31 administration maintains appropriate inflammatory responses while preventing the chronic low-grade inflammation (inflammaging) that contributes to age-related disease development and functional decline. Studies examining lifespan effects indicate that SS-31 administration extends healthspan and improves quality of life measures in aged animal models, providing researchers with powerful tools for investigating interventions targeting healthy aging and longevity enhancement. These comprehensive aging research applications establish SS-31 as an indispensable compound for laboratories studying fundamental mechanisms of aging and developing potential anti-aging interventions.
Renal Research Applications and Kidney Protection
Research investigations demonstrate SS-31's significant renoprotective properties across diverse models of kidney injury and disease, establishing it as a valuable research tool for studying mitochondrial contributions to renal pathophysiology and developing potential research strategies. Laboratory studies in acute kidney injury models reveal that SS-31 administration reduces tubular damage by 50-70% while preserving glomerular filtration rate and preventing the mitochondrial dysfunction that characterizes acute renal injury. Research demonstrates that the peptide protects against ischemia-reperfusion-induced kidney damage by maintaining mitochondrial membrane integrity, preserving ATP production capacity, and reducing oxidative stress-mediated cellular injury that typically leads to acute tubular necrosis and functional impairment.
Comprehensive studies in chronic kidney disease models show that SS-31 administration prevents progressive renal dysfunction by preserving mitochondrial structure and function in both tubular and glomerular cells. Laboratory investigations reveal that the peptide reduces proteinuria and albuminuria while maintaining appropriate podocyte function and glomerular barrier integrity that becomes compromised in progressive kidney disease. Research findings indicate that SS-31 administration prevents the metabolic remodeling associated with chronic kidney disease, maintaining appropriate fatty acid oxidation and glucose utilization that becomes impaired in diseased renal tissue. Studies demonstrate that the peptide reduces renal fibrosis development by preventing mitochondrial dysfunction-induced cellular senescence and inflammatory responses that contribute to progressive kidney damage.
Research in diabetic nephropathy models reveals that SS-31 provides protection against hyperglycemia-induced mitochondrial dysfunction while preserving renal metabolic flexibility and reducing oxidative stress-mediated damage characteristic of diabetic kidney disease. Laboratory analyses show that SS-31 administration maintains appropriate renal mitochondrial biogenesis capacity and prevents the accumulation of damaged mitochondria that contributes to diabetic nephropathy progression. Studies indicate that the peptide preserves renal vascular function and reduces inflammation while maintaining appropriate blood pressure regulation that becomes impaired in kidney disease models. These extensive renal research applications provide investigators with powerful tools for studying kidney mitochondrial biology and developing potential interventions targeting renal disease progression and preservation of kidney function.
Ophthalmologic Research and Retinal Protection Studies
Laboratory investigations reveal SS-31's remarkable protective properties in retinal research models, establishing it as an important tool for studying mitochondrial contributions to retinal degeneration and developing potential interventions for ophthalmologic conditions. Research studies in age-related macular degeneration models demonstrate that SS-31 administration protects retinal pigment epithelium (RPE) cells against oxidative damage while preserving phagocytic function essential for photoreceptor maintenance and visual function. Laboratory analyses show that the peptide maintains mitochondrial function in RPE cells under oxidative stress conditions, preventing the mitochondrial dysfunction that contributes to drusen formation and geographic atrophy characteristic of dry AMD progression.
Comprehensive research in diabetic retinopathy models reveals that SS-31 provides protection against hyperglycemia-induced retinal damage by preserving retinal vascular integrity and reducing inflammatory responses that contribute to diabetic eye disease progression. Studies demonstrate that SS-31 administration maintains appropriate retinal blood flow and reduces vascular permeability while preventing the mitochondrial dysfunction in retinal cells that leads to neurodegeneration and vision loss. Laboratory investigations show that the peptide preserves retinal ganglion cell function and reduces apoptotic cell death in models of retinal ischemia and oxidative stress, providing researchers with valuable tools for studying retinal neuroprotection mechanisms.
Research studies in glaucoma models demonstrate SS-31's ability to protect retinal ganglion cells against elevated intraocular pressure-induced damage while preserving optic nerve function and visual field integrity. Laboratory investigations reveal that the peptide maintains mitochondrial bioenergetics in retinal neurons under stress conditions and reduces the oxidative damage that contributes to glaucomatous neurodegeneration. Studies show that SS-31 administration preserves retinal thickness and reduces inflammation while maintaining appropriate retinal function across various ophthalmologic research models. These ophthalmologic research applications establish SS-31 as a valuable compound for investigating retinal mitochondrial biology and developing potential interventions for vision-threatening conditions characterized by mitochondrial dysfunction and oxidative stress.
Research Value and Scientific Investigation Potential
SS-31 represents an exceptional research tool that has fundamentally advanced scientific understanding of mitochondrial biology and provided researchers with unprecedented capabilities for investigating cellular bioenergetics, oxidative stress responses, and mitochondrial contributions to disease pathogenesis across diverse experimental systems. The peptide's unique targeting mechanism and established safety profile make it an ideal compound for laboratory investigations seeking to dissect the specific roles of mitochondrial dysfunction in various pathological processes while maintaining experimental precision and reproducibility. Research applications span multiple disciplines including cardiovascular biology, neuroscience, aging research, nephrology, and ophthalmology, providing investigators with versatile tools for advancing scientific knowledge in these critical areas of biomedical research.
The comprehensive research database surrounding SS-31, including over 35 peer-reviewed publications from leading institutions, establishes it as a well-characterized research compound with validated mechanisms and reproducible effects across multiple experimental paradigms. This extensive scientific foundation provides researchers with confidence in experimental design and interpretation while offering opportunities for novel research directions and mechanistic investigations. The peptide's development through rigorous preclinical studies and progression to clinical trials demonstrates its potential for translational research applications, allowing investigators to study mechanisms that may be relevant for human health and disease. SS-31's unique properties continue to reveal new insights into mitochondrial biology and provide researchers with powerful tools for advancing our understanding of cellular energetics and developing innovative research strategies targeting mitochondrial dysfunction across diverse pathological conditions.
Sources & Further Reading
- Birk, A.V., et al. (2020). Mitochondrial protein interaction landscape of SS-31. *Proceedings of the National Academy of Sciences* 117(25):14306-14313.
- Liu, S., et al. (2019). Elamipretide (SS-31) improves mitochondrial dysfunction, synaptic and memory impairment induced by lipopolysaccharide in mice. *Journal of Neuroinflammation* 16(1):230.
- Campbell, M.D., et al. (2023). The mitochondrially targeted peptide elamipretide (SS-31) improves ADP sensitivity in aged mitochondria by increasing uptake through the adenine nucleotide translocator (ANT). *GeroScience* 45(5):2813-2832.
- Chatfield, K.C., et al. (2021). Elamipretide (SS-31) administration attenuates age-associated post-translational modifications of heart proteins. *Aging* 13(18):21900-21909.
- Karaa, A., et al. (2018). Randomized dose-escalation trial of elamipretide in adults with primary mitochondrial myopathy. *Neurology* 90(14):e1212-e1221.
- Karaa, A., et al. (2023). Efficacy and Safety of Elamipretide in Individuals With Primary Mitochondrial Myopathy: The MMPOWER-3 Randomized Clinical Trial. *Neurology* 101(7):e689-e701.
- Reynolds, B.A., et al. (2021). A phase 2/3 randomized clinical trial followed by an open-label extension to evaluate the effectiveness of elamipretide in Barth syndrome, a genetic disorder of mitochondrial cardiolipin metabolism. *Genetics in Medicine* 23(3):471-478.
- Szeto, H.H. and Schiller, P.W. (2011). Novel therapies targeting inner mitochondrial membrane - from discovery to clinical development. *Pharmaceutical Research* 28(11):2669-2679.
- Zhao, K., et al. (2020). The mitochondria-targeted peptide SS-31 binds lipid bilayers and modulates surface electrostatics as a key component of its mechanism of action. *Journal of Biological Chemistry* 295(21):7452-7462.
- Kloner, R.A., et al. (2012). Reduction of ischemia/reperfusion injury with bendavia, a mitochondria-targeting cytoprotective peptide. *Journal of the American Heart Association* 1(3):e001644.
- Daubert, M.A., et al. (2017). Novel mitochondria-targeting peptide in heart failure research: A randomized, placebo-controlled investigation of elamipretide. *Circulation: Heart Failure* 10(4):e003389.
- Siegel, M.P., et al. (2013). Mitochondrial-targeted peptide rapidly improves mitochondrial energetics and skeletal muscle performance in aged mice. *Aging Cell* 12(5):763-771.
- Manczak, M., et al. (2010). Mitochondria-targeted antioxidants protect against amyloid-beta toxicity in Alzheimer's disease neurons. *Journal of Alzheimer's Disease* 20(s2):S609-S631.
- Yang, L., et al. (2009). The mitochondria-targeted peptides protect neurons against amyloid-β peptide. *Antioxidants & Redox Signaling* 11(11):2717-2727.
- Eirin, A., et al. (2017). Mitochondrial protection restores renal function in swine atherosclerotic renovascular disease. *Cardiovascular Research* 103(4):461-472.
- Szeto, H.H., et al. (2011). Protection of mitochondria prevents high-fat diet-induced glomerulopathy and proximal tubular injury. *Kidney International* 80(1):23-33.
- Sabbah, H.N., et al. (2016). Chronic administration of elamipretide (MTP-131), a novel mitochondria-targeting peptide, improves left ventricular and mitochondrial function in dogs with advanced heart failure. *Circulation: Heart Failure* 9(2):e002206.
- Shi, J., et al. (2018). SS-31 ameliorates renal injury and oxidative stress in protein overload nephropathy. *American Journal of Nephrology* 47(4):245-254.
- Ali, S.S., et al. (2024). Application research of novel peptide mitochondrial-targeted antioxidant SS-31 in mitigating mitochondrial dysfunction. *Free Radical Biology and Medicine* 210:285-297.
- Escribano-Lopez, I., et al. (2025). Elamipretide: A Review of Its Structure, Mechanism of Action, and Therapeutic Potential. *International Journal of Molecular Sciences* 26(3):944.
Last reviewed:
| CAS Number | 736992-21-5 |
|---|---|
| Molecular Formula | C32H49N9O5 |
| Molecular Weight | 639.8 g/Mol |
| Purity | 99.7% |
| Lot Number | 25034 |
| Quantity | 11.30mg |
| Sequence | H-D-Arg-Tyr(2,6-diMe)-Lys-Phe-NH2 |
Cosmic Peptides
Certificate of Analysis
Unable to load certificate. Download COA