SS-31: The Cardiolipin Guardian That's Revolutionizing Mitochondrial Medicine
Sometimes the most profound medical breakthroughs emerge not from grand theoretical leaps, but from the patient, methodical pursuit of understanding life's most fundamental processes.
Sometimes the most profound medical breakthroughs emerge not from grand theoretical leaps, but from the patient, methodical pursuit of understanding life’s most fundamental processes. In the case of SS-31—now known as elamipretide—we witness the rare convergence of elegant molecular design, rigorous scientific validation, and genuine therapeutic need that defines truly transformative medicine.
The story of SS-31 begins not with a eureka moment, but with a recognition that mitochondria, those cellular powerhouses we’ve long taken for granted, harbor secrets that could unlock treatments for some of humanity’s most challenging diseases. When researchers first synthesized this tetrapeptide with its alternating aromatic-cationic sequence, they weren’t just creating another experimental compound—they were engineering the first member of an entirely new class of therapeutics designed to work at the most fundamental level of cellular energy production.
What makes SS-31 extraordinary is not merely its ability to target mitochondria, but its exquisite specificity for cardiolipin, a unique phospholipid that serves as the architectural foundation for mitochondrial function. This isn’t the broad-spectrum approach of traditional antioxidants or the sledgehammer tactics of many pharmaceutical interventions. Instead, SS-31 represents precision medicine at the molecular level—a therapeutic that understands and works with the intricate machinery of cellular energy production rather than simply trying to override it.
The clinical journey of SS-31 has been marked by both the promise and the challenges inherent in developing treatments for ultra-rare diseases. From its early designation as an experimental peptide to its current status as elamipretide, a late-stage clinical candidate for Barth syndrome and other mitochondrial disorders, this compound has navigated the complex landscape of rare disease drug development with remarkable persistence. The 168-week TAZPOWER trial results, published in 2024, represent not just a clinical milestone but a validation of the entire approach of targeting mitochondrial dysfunction at its source [1].
SS-31, Sourced for Research — Peptide Partners. We supply research-grade SS-31 and complementary peptides. Each lot is verified by independent HPLC/MS, ships with a batch COA, and passes endotoxin screening per USP <85>. Browse inventory or request specifications.
Perhaps most remarkably, recent research has revealed that SS-31’s therapeutic effects extend far beyond its original intended applications. The 2024 discoveries linking SS-31 to ferroptosis protection in diabetic cardiomyopathy, its neuroprotective effects in epilepsy, and its ability to reverse aging-related muscle and heart dysfunction suggest that we may be witnessing the emergence of a truly foundational therapeutic approach—one that addresses the mitochondrial dysfunction underlying a vast spectrum of human disease [2].
The Molecular Precision of Cardiolipin Targeting
Understanding SS-31’s revolutionary approach requires appreciating the unique role of cardiolipin in mitochondrial function and the elegant precision with which this tetrapeptide has been designed to interact with this critical phospholipid. Cardiolipin is not just another membrane component—it is the architectural keystone that enables mitochondria to function as efficient energy-producing organelles, and SS-31’s ability to stabilize and protect cardiolipin represents a fundamentally new approach to treating mitochondrial dysfunction [3].
Cardiolipin’s structure is unlike any other phospholipid in human cells. With its two phosphate head groups and four acyl chains, cardiolipin creates the unique membrane environment necessary for optimal function of the respiratory complexes that drive ATP production. This unusual lipid is almost exclusively localized to the inner mitochondrial membrane, where it plays essential roles in maintaining cristae structure, facilitating respiratory complex assembly, and enabling the proton gradients that drive ATP synthesis. When cardiolipin function is compromised—whether through genetic mutations, oxidative damage, or age-related decline—the entire mitochondrial energy production system suffers [4].
SS-31’s molecular design represents a masterpiece of rational drug design, with each component of its tetrapeptide sequence serving a specific function in cardiolipin interaction. The alternating aromatic-cationic motif enables dual interactions with cardiolipin: hydrophobic interactions with the lipid’s acyl chains and electrostatic interactions with its anionic phosphate head groups. This dual binding mechanism allows SS-31 to stabilize cardiolipin in its optimal conformation while protecting it from oxidative damage that would otherwise compromise mitochondrial function [5].
The 2020 PNAS study that mapped SS-31’s protein interaction landscape provided unprecedented insight into how this cardiolipin-targeting approach translates into functional benefits. Using chemical cross-linking mass spectrometry, researchers identified twelve specific mitochondrial proteins that interact directly with SS-31, all of which are known cardiolipin binders involved in either ATP production through oxidative phosphorylation or 2-oxoglutarate metabolic processes. This discovery revealed that SS-31’s effects are not limited to cardiolipin stabilization but extend to direct modulation of the protein machinery responsible for mitochondrial energy production [6].
The spatial organization of these interactions is particularly revealing. The regions where SS-31 binds to mitochondrial proteins are consistently proximal to cardiolipin-protein interaction sites, suggesting that SS-31 functions as a molecular bridge that enhances the stability and function of cardiolipin-protein complexes. This mechanism explains how a single therapeutic intervention can simultaneously improve multiple aspects of mitochondrial function, from respiratory complex assembly to ATP synthesis efficiency to protection against oxidative damage [7].
Recent research has further illuminated the sophisticated ways in which SS-31 modulates mitochondrial membrane organization. The peptide’s ability to induce tighter curvatures in cristae membranes optimizes the spatial organization of respiratory supercomplexes, enhancing electron transfer efficiency and reducing the production of reactive oxygen species. This structural optimization represents a level of therapeutic precision that goes beyond simple biochemical intervention to encompass the physical architecture of cellular energy production [8].
The cardiolipin-targeting approach has proven particularly powerful in addressing the root causes of mitochondrial dysfunction in genetic diseases like Barth syndrome, where mutations in the TAFAZZIN gene disrupt cardiolipin remodeling. In these conditions, SS-31’s ability to stabilize existing cardiolipin and compensate for defective remodeling processes provides therapeutic benefits that address the fundamental pathophysiology rather than merely treating symptoms. The 2024 cardiac mitochondrial morphology studies demonstrated that SS-31 treatment can restore normal mitochondrial structure and function even in the presence of TAFAZZIN mutations [9].
The Clinical Validation Journey
The path from laboratory discovery to clinical validation for SS-31 represents one of the most comprehensive and rigorous development programs in mitochondrial medicine, with the TAZPOWER trial serving as a landmark study that has fundamentally changed how we think about treating mitochondrial diseases. The journey from initial preclinical studies to the 168-week long-term extension results published in 2024 demonstrates both the promise and the challenges of developing therapeutics for ultra-rare diseases [10].
The TAZPOWER trial was designed as a randomized, double-blind, placebo-controlled study specifically focused on Barth syndrome, an ultra-rare genetic disorder affecting fewer than 300 known patients worldwide. This patient population presented unique challenges for clinical trial design, requiring innovative approaches to demonstrate efficacy in a condition where traditional large-scale studies are impossible. The trial’s primary endpoint—improvement in the 6-minute walk test—was chosen as a functional measure that could capture the real-world impact of treatment on patients’ daily lives [11].
SS-31, Sourced for Research — Peptide Partners. We supply research-grade SS-31 and complementary peptides. Each lot is verified by independent HPLC/MS, ships with a batch COA, and passes endotoxin screening per USP <85>. Browse inventory or request specifications.
The initial 12-week randomized controlled phase of TAZPOWER provided the first rigorous clinical evidence of SS-31’s efficacy in humans. While the primary endpoint did not reach statistical significance in this short timeframe, the trial revealed important signals of benefit that became more pronounced in the long-term extension phase. This pattern—where benefits become more apparent with extended treatment—reflects the fundamental nature of mitochondrial dysfunction and the time required for cellular repair and adaptation processes to manifest as functional improvements [12].
The open-label extension phase of TAZPOWER, which ultimately extended to 168 weeks, provided unprecedented insight into the long-term effects of SS-31 treatment in Barth syndrome patients. The sustained improvements in functional assessments, cardiac function, and quality of life measures observed over this extended period represent some of the most compelling evidence for mitochondrial-targeted therapy in human disease. Particularly striking were the improvements in exercise tolerance and cardiac function, which continued to accrue over time rather than plateauing after initial treatment [13].
The natural history comparison studies conducted alongside TAZPOWER provided additional validation of SS-31’s therapeutic effects by comparing treated patients to the expected disease progression based on historical data. These analyses revealed that patients receiving SS-31 experienced significantly slower rates of functional decline compared to what would be expected based on the natural history of Barth syndrome. This approach to demonstrating efficacy—comparing treatment effects to natural disease progression—has become a model for rare disease drug development [14].
Safety data from the TAZPOWER program has been consistently reassuring, with SS-31 demonstrating an excellent tolerability profile even with extended treatment. The most common adverse events were mild injection site reactions, and no serious safety signals emerged even after 168 weeks of treatment. This safety profile is particularly important for a chronic treatment intended for use in pediatric and young adult populations, where long-term safety considerations are paramount [15].
The 2024 publication of the complete TAZPOWER results marked a watershed moment in mitochondrial medicine, providing the first definitive evidence that targeting mitochondrial dysfunction can produce meaningful clinical benefits in human disease. The sustained improvements in cardiac function, exercise capacity, and quality of life measures observed in this study have established SS-31 as the leading therapeutic candidate for Barth syndrome and have validated the broader approach of mitochondrial-targeted therapy [16].
Beyond Barth syndrome, SS-31’s clinical development has expanded to include other mitochondrial diseases, with ongoing trials in primary mitochondrial myopathy and Leber’s hereditary optic neuropathy. These studies are exploring whether the cardiolipin-targeting approach can provide benefits across the spectrum of mitochondrial disorders, potentially establishing SS-31 as a platform therapy for mitochondrial dysfunction regardless of its underlying cause [17].
The Ferroptosis Revolution and Expanded Applications
The discovery that SS-31 provides protection against ferroptosis—a recently identified form of programmed cell death driven by iron-dependent lipid peroxidation—has opened entirely new avenues for therapeutic application and fundamentally expanded our understanding of how mitochondrial-targeted therapy can address diverse disease processes. The 2024 research demonstrating SS-31’s ability to prevent ferroptosis in diabetic cardiomyopathy represents a paradigm shift in how we conceptualize mitochondrial dysfunction and its role in common diseases [18].
Ferroptosis differs fundamentally from other forms of cell death in its dependence on iron accumulation and lipid peroxidation, processes that are intimately connected to mitochondrial function and cardiolipin metabolism. The discovery that SS-31 can interrupt ferroptotic pathways through activation of mitochondrial glutathione peroxidase 4 (mitoGPX4) and enhancement of the mitochondrial glutathione system provides a mechanistic explanation for the peptide’s protective effects that extends far beyond its original cardiolipin-stabilizing properties [19].
The diabetic cardiomyopathy research has been particularly revelatory, demonstrating that SS-31’s ferroptosis protection can address one of the most common and devastating complications of diabetes. Diabetic heart disease affects millions of patients worldwide and represents a major cause of morbidity and mortality in diabetic populations. The finding that SS-31 can prevent the mitochondria-dependent ferroptosis that drives diabetic cardiac dysfunction suggests potential applications in a patient population orders of magnitude larger than the rare diseases for which SS-31 was originally developed [20].
The neurological applications of SS-31’s ferroptosis protection have proven equally compelling, with 2024 research demonstrating that the peptide can prevent seizures in epilepsy models by inhibiting ferroptosis in hippocampal neurons. This neuroprotective effect appears to be mediated through inhibition of p38 MAPK phosphorylation, revealing yet another mechanism by which SS-31 can provide cellular protection. The implications for treating epilepsy and other neurological disorders characterized by ferroptotic cell death are profound [21].
The aging research published in 2025 has added another dimension to SS-31’s therapeutic potential, with studies demonstrating that the peptide can reverse age-related muscle and heart dysfunction in animal models. Remarkably, these functional improvements occur without changes in epigenetic or transcriptomic markers of aging, suggesting that SS-31’s effects are mediated through direct restoration of mitochondrial function rather than through broader anti-aging mechanisms. This finding supports the hypothesis that mitochondrial dysfunction is a primary driver of age-related functional decline [22].
The ferroptosis protection mechanism has also provided new insights into SS-31’s effects in the original Barth syndrome indication. Recent research suggests that the cardiolipin deficiency characteristic of Barth syndrome may predispose cells to ferroptotic death, and that SS-31’s protective effects may involve both cardiolipin stabilization and ferroptosis prevention. This dual mechanism of action helps explain the sustained and progressive improvements observed in the TAZPOWER trial [23].
The breadth of diseases potentially addressable through ferroptosis protection is staggering, encompassing conditions as diverse as acute kidney injury, stroke, traumatic brain injury, and various forms of heart disease. The 2025 systematic review of SS-31’s clinical applications identified ferroptosis as a common pathway underlying many of the conditions where SS-31 has shown therapeutic promise, suggesting that ferroptosis protection may be a unifying mechanism that explains the peptide’s broad therapeutic potential [24].
The Regulatory Pathway and Commercial Reality
The regulatory journey of SS-31 through the FDA approval process represents a fascinating case study in rare disease drug development, highlighting both the opportunities and challenges inherent in bringing innovative therapies to patients with ultra-rare conditions. The February 2025 announcement that the FDA has identified a “path forward” for elamipretide in Barth syndrome, despite extending the review timeline, reflects the complex regulatory considerations involved in evaluating therapies for conditions affecting fewer than 300 patients worldwide [25].
The orphan drug designation granted to SS-31 for multiple indications has provided crucial regulatory advantages, including extended market exclusivity, reduced regulatory fees, and enhanced FDA interaction opportunities. These incentives, designed to encourage development of therapies for rare diseases, have been essential in making the substantial investment required for SS-31’s development economically viable. The multiple orphan designations—for Barth syndrome, primary mitochondrial myopathy, and Leber’s hereditary optic neuropathy—reflect the broad therapeutic potential of mitochondrial-targeted therapy [26].
The regulatory challenges faced by SS-31 highlight the unique difficulties of demonstrating efficacy in ultra-rare diseases. Traditional clinical trial designs, which rely on large patient populations and standardized endpoints, are often impractical or impossible in conditions affecting only hundreds of patients globally. The TAZPOWER trial’s innovative design, incorporating natural history comparisons and extended open-label phases, has helped establish new paradigms for rare disease clinical development [27].
SS-31, Sourced for Research — Peptide Partners. We supply research-grade SS-31 and complementary peptides. Each lot is verified by independent HPLC/MS, ships with a batch COA, and passes endotoxin screening per USP <85>. Browse inventory or request specifications.
The FDA’s extended review period for SS-31 reflects the agency’s careful consideration of the unique evidence package required for rare disease approvals. The combination of randomized controlled trial data, natural history comparisons, and extensive long-term safety information represents a comprehensive approach to demonstrating benefit-risk balance in a population where traditional large-scale studies are not feasible. The agency’s identification of a path forward suggests that this evidence package may ultimately prove sufficient for approval [28].
Stealth BioTherapeutics’ commercial strategy for SS-31 reflects the realities of rare disease drug development, with a focus on building specialized capabilities in ultra-rare disease commercialization. The company’s pipeline approach, developing SS-31 for multiple mitochondrial diseases simultaneously, maximizes the potential return on the substantial development investment while serving multiple patient populations with significant unmet medical need [29].
The pricing considerations for SS-31 reflect the economics of rare disease drug development, where high development costs must be recovered from small patient populations. The orphan drug pricing model, which typically involves premium pricing justified by the substantial unmet medical need and limited patient population, will likely apply to SS-31’s commercial launch. The demonstrated clinical benefits and excellent safety profile support the value proposition for payers and patients [30].
The global regulatory strategy for SS-31 involves coordinated submissions to regulatory agencies worldwide, leveraging the FDA’s review and decision as a foundation for approvals in other markets. The European Medicines Agency and other international regulators have shown increasing alignment with FDA approaches to rare disease drug evaluation, potentially facilitating global access for SS-31 once initial approval is achieved [31].
The Future of Mitochondrial Medicine
SS-31’s journey from experimental peptide to late-stage clinical candidate represents more than just the development of a single therapeutic—it embodies the emergence of mitochondrial medicine as a legitimate and powerful approach to treating human disease. The success of the cardiolipin-targeting strategy has validated fundamental assumptions about the role of mitochondrial dysfunction in disease and has opened pathways for an entire generation of mitochondrial-targeted therapeutics [32].
The mechanistic insights gained from SS-31 research are already informing the development of next-generation mitochondrial therapeutics. The understanding of cardiolipin’s central role in mitochondrial function, the importance of ferroptosis protection, and the potential for direct protein interactions are all contributing to more sophisticated approaches to mitochondrial targeting. Future therapeutics may combine multiple mechanisms—cardiolipin stabilization, ferroptosis protection, and direct protein modulation—to achieve even greater therapeutic effects [33].
The expansion of SS-31’s applications beyond rare diseases to common conditions like diabetic cardiomyopathy and age-related dysfunction suggests that mitochondrial-targeted therapy may have broad relevance across the spectrum of human disease. The recognition that mitochondrial dysfunction underlies many common diseases of aging—cardiovascular disease, neurodegeneration, metabolic disorders—positions mitochondrial medicine as a potentially transformative approach to addressing the health challenges of an aging global population [34].
The combination therapy opportunities for SS-31 are particularly exciting, with potential synergies with other mitochondrial interventions, gene therapies, and even lifestyle modifications like exercise and dietary interventions. The understanding that SS-31 works by optimizing mitochondrial function rather than simply replacing it suggests that combination approaches could achieve additive or even synergistic benefits [35].
The personalized medicine applications of SS-31 are beginning to emerge as researchers identify genetic and biomarker factors that predict treatment response. The development of companion diagnostics that can identify patients most likely to benefit from mitochondrial-targeted therapy could optimize treatment outcomes while minimizing unnecessary exposure in patients unlikely to respond [36].
Perhaps most importantly, SS-31’s success has demonstrated that it is possible to develop effective therapies for mitochondrial dysfunction, providing hope for the millions of patients worldwide affected by mitochondrial diseases. The validation of the mitochondrial targeting approach has energized the field and attracted increased investment and research attention to mitochondrial medicine, accelerating the development of additional therapeutic options [37].
References
[1] Long-term efficacy and safety of elamipretide in patients with Barth syndrome: 168-week open-label extension results of TAZPOWER. Genetics in Medicine. https://www.sciencedirect.com/science/article/pii/S1098360024000716
[2] New insight for SS-31 in treating diabetic cardiomyopathy. International Journal of Molecular Medicine. https://www.spandidos-publications.com/10.3892/ijmm.2024.5436
[3] First-in-class cardiolipin-protective compound as a therapeutic agent to restore mitochondrial bioenergetics. British Journal of Pharmacology. https://bpspubs.onlinelibrary.wiley.com/doi/abs/10.1111/bph.12461
[4] The mitochondria-targeted peptide SS-31 binds lipid bilayers and modulates surface electrostatics. Journal of Biological Chemistry. https://www.jbc.org/article/S0021-9258(17)50276-8/fulltext
[5] Elamipretide: a review of its structure, mechanism of action, and therapeutic potential. International Journal of Molecular Sciences. https://www.mdpi.com/1422-0067/26/3/944
[6] Mitochondrial protein interaction landscape of SS-31. PNAS. https://www.pnas.org/doi/10.1073/pnas.2002250117
[7] Contemporary insights into elamipretide’s mitochondrial mechanism of action and therapeutic effects. Biomedicine & Pharmacotherapy. https://www.sciencedirect.com/science/article/pii/S0753332225002501
[8] SS-31 treatment ameliorates cardiac mitochondrial morphology and function. Nature Scientific Reports. https://www.nature.com/articles/s41598-024-64368-y
[9] The mitochondrially targeted peptide elamipretide (SS-31) improves ADP sensitivity in aged mitochondria. GeroScience. https://link.springer.com/article/10.1007/s11357-023-00861-y
[10] Natural history comparison study to assess the efficacy of elamipretide in patients with Barth syndrome. Orphanet Journal of Rare Diseases. https://link.springer.com/article/10.1186/s13023-022-02469-5
[11] Temporal evolution of the heart failure phenotype in Barth syndrome and treatment with elamipretide. Future Cardiology. https://www.tandfonline.com/doi/abs/10.2217/fca-2023-0008
[12] Elamipretide Prevents the Natural History Progression of Cardiomyopathy in Barth Syndrome. Journal of Cardiac Failure. https://onlinejcf.com/article/S1071-9164(20)31144-1/fulltext
[13] Long-term efficacy and safety of elamipretide in patients with Barth syndrome. PubMed. https://pubmed.ncbi.nlm.nih.gov/38602181/
[14] Natural history comparison study to assess the efficacy of elamipretide. PMC. https://pmc.ncbi.nlm.nih.gov/articles/PMC9438322/
[15] Application research of novel peptide mitochondrial-targeted therapy. Science Direct. https://www.sciencedirect.com/science/article/abs/pii/S1567724924000047
[16] Elamipretide Clinical Studies and Trials. Stealth BioTherapeutics. https://stealthbt.com/programs-pipeline/
[17] Elamipretide - Friedreich’s Ataxia Research Alliance. FARA. https://www.curefa.org/drug-development/elamipretide/
[18] New insight for SS-31 in treating diabetic cardiomyopathy. PubMed. https://pubmed.ncbi.nlm.nih.gov/39364755
[19] Ferroptosis in diabetic cardiomyopathy: from its mechanisms to therapeutic targets. Frontiers in Endocrinology. https://www.frontiersin.org/journals/endocrinology/articles/10.3389/fendo.2024.1421838/pdf
[20] Mitochondria-targeting peptide SS-31 attenuates ferroptosis via inhibition of p38 MAPK. Brain Research. https://www.sciencedirect.com/science/article/abs/pii/S0006899324001367
[21] Mitochondria-targeting peptide SS-31 attenuates ferroptosis in epileptic rats. Brain Research. https://www.sciencedirect.com/science/article/pii/S0006899324001367
[22] Diabetes Drug Elamipretide Reverses Muscle and Heart Aging. NAD.com. https://www.nad.com/news/mitochondrial-drug-elamipretide-reverses-muscle-and-heart-aging-new-study-reveals
[23] Aging, mitochondrial dysfunction, and cerebral microhemorrhages. GeroScience. https://link.springer.com/article/10.1007/s11357-025-01634-5
[24] Application research of novel peptide mitochondrial-targeted therapy. Science Direct. https://www.sciencedirect.com/science/article/abs/pii/S1567724924000047
[25] FDA Extends Review of Elamipretide for Barth Syndrome. Rheumatology Advisor. https://www.rheumatologyadvisor.com/news/fda-extends-review-of-elamipretide-for-barth-syndrome/
[26] Stealth BioTherapeutics Announces “Path Forward” Despite Disappointing Delay. Stealth BioTherapeutics. https://stealthbt.com/stealth-biotherapeutics-announces-path-forward-despite-disappointing-delay-for-ultra-rare-barth-syndrome/
[27] tazpower - Barth Syndrome Foundation. BSF. https://www.barthsyndrome.org/research/clinicaltrials/tazpower.html
[28] The effects of a mitochondrial targeted peptide on BAX recruitment. BMC Research Notes. https://link.springer.com/article/10.1186/s13104-021-05613-9
[29] Elamipretide in the Management of Barth Syndrome. PubMed. https://pubmed.ncbi.nlm.nih.gov/40816230/
[30] Late-life restoration of mitochondrial function reverses cardiac dysfunction. eLife. https://elifesciences.org/articles/55513
[31] Mitochondrial Cardiolipin-Targeted Tetrapeptide, SS-31, Exerts Neuroprotective Effects. MDPI. https://www.mdpi.com/1422-0067/26/7/3327
[32] Elamipretide (SS-31) improves mitochondrial dysfunction, synaptic and memory impairment. Journal of Neuroinflammation. https://jneuroinflammation.biomedcentral.com/articles/10.1186/s12974-019-1627-9
[33] The mitochondrially targeted peptide elamipretide improves physiological function. GeroScience. https://pubmed.ncbi.nlm.nih.gov/37462785/
[34] Randomized clinical trial followed by an open-label extension to evaluate elamipretide. Genetics in Medicine. https://www.sciencedirect.com/science/article/pii/S1098360021049467
[35] Contemporary insights into elamipretide’s mitochondrial mechanism. Biomedicine & Pharmacotherapy. https://www.sciencedirect.com/science/article/pii/S0753332225002501
[36] Aging, mitochondrial dysfunction, and cerebral microhemorrhages evaluation. GeroScience. https://link.springer.com/article/10.1007/s11357-025-01634-5
[37] Elamipretide: a review of its structure, mechanism of action, and therapeutic potential. International Journal of Molecular Sciences. https://www.mdpi.com/1422-0067/26/3/944





Great article. But the peptides, the supplements, the NIR light, the strategies: of what consequence when we know that nanoplastics are lodged in mitochondrial cardiolipin layer, in the oxidative phosphorylation process, these plastics are absorbed by endocytosis into mitochondria. They do not conduct electricity. It seems to me that we have to work on detoxing the body from plastics before the full effect of the healing of dysfunctional mitochondria, which I believe is far more common than we know, this for me is an important step and why I continue the research
Phenomenal breakdown of the cardiolipin-protein complex stabilization. The 2020 PNAS cross-linking data showing SS-31's direct protein interactions is underappreciated, most ppl still think its just an antioxidant when really its acting as a molecular bridge at cardiolipin binding sites. I remember analyzing respiratory complex assembly data back in 2021 and the improvemnts in supercomplex formation were wild, way beyond what you'd expect from simple ROS reduction. One aspect that facinates me is the dose-response in Barth syndrome patients, why do benefits keep accruing past 100 weeks when cardiolipin stability should plateau earlier?