Activate Nrf2 with far-infrared light

A 2025 rodent study investigated effects of far-infrared light on Alzheimer’s disease models. I’ll focus on its Nrf2 findings:

“Far-infrared radiation (FIR) is commonly utilized as a complementary treatment of a range of disease, for example, insomnia and rheumatoid arthritis. In this research, we explored how FIR light impacts cognitive functions of TgCRND8 AD mice and elucidated its underlying molecular mechanism.

Infrared radiation is a form of electromagnetic energy that has wavelengths between 750 nm and 1000 μm, which are longer than visible light. International Commission on Illumination categorizes infrared light as three sub-divisions according to the wavelength: (1) near-infrared radiation (0.7–1.4 μm), (2) middle infrared radiation (1.4–3.0 μm), and (3) far-infrared radiation (3.0–1000 μm).

Nrf-2/ HO-1 signaling, a key endogenous antioxidant system, helps mitigate oxidative stress and enhances expression of various endogenous genes. Activation of HO-1 during inflammatory conditions may serve as an adaptive response to reduce cytotoxicity through various mechanisms.

In this study, we applied EFFIT LITE® as the FIR spectrum transmitter which stably radiates an FIR spectrum with a wavelength of 4–20 μm, and the device was put within 1 cm directly above the head of the 3-month-old TgCRND8 mice for 30 min exposure once every day. FIR light notably enhanced cognitive function and spatial memory of TgCRND8 mice after 28-days consecutive treatment.

Underlying molecular mechanisms involve suppression of Aβ deposition, hyperphosphorylation of tau, and neuroinflammation through modulating Jak-2/Stat3 and Nrf-2/HO-1 pathways. Our current experimental findings amply indicate that FIR light is a potential non-pharmacological therapy for AD.”

https://link.springer.com/article/10.1007/s12017-025-08860-2“Far-Infrared Radiation Ameliorates the Cognitive Dysfunction in an Alzheimer’s Disease Transgenic Mouse via Modulating Jak-2/Stat3 and Nrf-2/HO-1 Pathways”


This study measured Nrf2 and its quickly-induced downstream enzyme HO-1 effects of daily far-infrared light exposure for 30 minutes. We’d have to see measurements of Nrf2’s more-slowly induced and longer-lasting downstream xenobiotic detoxifying enzyme NQO1 to compare far-infrared light Nrf2 activation effects with those of natural plant compounds.

Plasmapheresis doesn’t reduce biological age

A 2025 clinical trial investigated effects of plasmapheresis as measured with epigenetic clocks:

“This study aimed to assess whether plasmapheresis without volume replacement with young plasma or albumin affects epigenetic age and other biomarkers in healthy adults. No significant epigenetic rejuvenation was observed based on epigenetic clock measurements. Instead, plasmapheresis was associated with increases in DNAmGrimAge, the Hannum clock, and the Dunedin Pace of Aging.

  1. The relatively small sample size of 34 finishing participants comprising of first-time plasma donors limits the statistical power and generalizability of our findings.
  2. Our cohort was restricted to individuals aged 40 to 60 years in accordance with Czech regulatory guidelines, which, although intentional to focus on an older population where rejuvenating effects might be most apparent, constrains evaluation of age-related differences across a broader demographic.
  3. The 18-week duration of the study, while sufficient to detect rapid alterations in key biomarkers under an intensive plasmapheresis protocol, may not fully capture the long-term implications of these changes.
  4. Due to our trial taking place during spring and summer months, we cannot fully separate the effects of increased sunlight exposure, outdoor physical activity, and dietary changes from the observed rises in Vitamin D and concurrent shifts in DNAm-based aging metrics. We did not collect objective measures of activity or diet, so these factors remain potential confounders.

The protocol of donating plasma every two weeks, although deemed safe by many countries around the world, is not yet well researched and cannot therefore be marked as benefiting to the donor right now. Further refinement to balance clearance of pro-aging factors with maintenance of systemic homeostasis is needed.”

https://www.nature.com/articles/s41598-025-05396-0 “Human clinical trial of plasmapheresis effects on biomarkers of aging (efficacy and safety trial)”


Betaine as an exercise mimetic

A 2025 human study investigated effects of long-term exercise:

“Exercise has well-established health benefits, yet its molecular underpinnings remain incompletely understood. We conducted an integrated multi-omics analysis to compare effects of acute vs. long-term exercise in healthy males.

Acute exercise induced transient responses, whereas repeated exercise triggered adaptive changes, notably reducing cellular senescence and inflammation and enhancing betaine metabolism. Exercise-driven betaine enrichment, partly mediated by renal biosynthesis, exerts geroprotective effects and rescues age-related health decline in mice.

Betaine binds to and inhibits TANK-binding kinase 1 (TBK1), retarding the kinetics of aging.

Betaine effectively alleviated senescence phenotypes by reduced senescence-associated β-galactosidase (SA-β-Gal)-positive cells, decreased p21 expression, lowered DNA damage indicator γ-H2A.X, and elevated heterochromatin mark H3K9me3. Betaine treatment also enhanced cellular antioxidant capacity, as evidenced by increased NRF2 phosphorylation and reduced ROS accumulation.

These findings systematically elucidate the molecular benefits of exercise, and position betaine as an exercise mimetic for healthy aging.”

https://doi.org/10.1016/j.cell.2025.06.001 “Systematic profiling reveals betaine as an exercise mimetic for geroprotection” (not freely available) Thanks to Dr. Weimin Ci for providing a copy.


Eat broccoli sprouts for your HIIT, Part 2

A 2025 human study followed up Eat broccoli sprouts for your high intensity interval training using red kale sprouts:

“Glucosinolate-rich broccoli sprouts combined with intense exercise training for 7 days have been shown to reduce blood lactate concentrations during exercise, attenuate hypoglycemic events, improve physical performance, and reduce markers of oxidative stress. This study aimed to investigate the acute, dose-dependent effects of glucosinolate-rich red kale sprouts (GRS) on blood lactate and blood glucose following the ingestion of three different doses.

Fifteen healthy participants [11 females, 4 males] consumed 37.5 g or 75 g of GRS or an isocaloric placebo blended into a beverage on three separate occasions. The participants cycled on an ergometer at three submaximal work rates before and three hours after ingestion.

Intake of glucosinolate-rich sprouts acutely decreased blood lactate levels during submaximal cycling and increased blood glucose levels at rest. The largest reduction in blood lactate was observed at the 37.5 g dose compared to placebo.

To identify the dose of GRS that results in the lowest blood lactate concentration during submaximal exercise, we applied a quadratic modeling approach. The optimal dose for minimizing lactate accumulation was calculated as 44 g of GRS.

In our previous study, we found a tendency towards a lower respiratory exchange ratio after one week of supplementation. Moreover, studies have demonstrated that mitochondrial oxidation of long-chain and short-chain fatty acids is depressed in the absence of Nrf2, and accelerated when Nrf2 is constitutively active.

We observed a reduction in myeloperoxidase levels approximately three hours after GRS intake, suggesting a decrease in oxidative stress. This finding indicates that the adaptive compensatory system may act rapidly, likely within just a few hours of GRS consumption.

A limitation is that we did not assess whether the lower lactate levels translated into improved performance. Theoretically, if the reduction in lactate results from the activation of pyruvate dehydrogenase, it could enhance performance by channeling more pyruvate into mitochondria for efficient oxidation, reducing reliance on glycolysis, and thereby sparing muscle glycogen. Alternatively, if the lower lactate levels are due to increased activity of the hepatic Cori cycle, lactate could be more rapidly converted to glucose, possibly supporting glycogen resynthesis or maintaining blood glucose levels during exercise. Both mechanisms could potentially contribute to improved performance.

Acute intake of small doses of GRS followed by submaximal ergometer cycling results in changes in lactate and glucose metabolism that could be beneficial for exercise performance.”

https://www.biorxiv.org/content/10.1101/2025.04.15.648889v1.full “A glucosinolate-rich beverage lowers blood lactate concentrations during submaximal exercise”


These researchers chose red kale sprouts of undisclosed age over the predecessor study’s broccoli raab five-day old sprouts, and two other undisclosed cruciferous vegetable sprouts.

This study is in its preprint phase. Items that could be clarified before publishing in final form include:

  • In the Abstract section, reference findings to red kale sprouts rather than broccoli sprouts;
  • Characterize the lactate U-shaped dose-response curve as hormesis; and
  • Reference other hormesis findings for context.

Sulforaphane as a senotherapy

A 2025 rodent study investigated cardioprotective effects of dasatinib / quercetin (a senolytic combination) and sulforaphane (senomorphic):

“Senolytics are molecules that selectively eliminate senescent cells (SCs). Senomorphics are drugs that suppress or mitigate the pro-inflammatory effects of the senescence-associated secretory phenotype (SASP) without killing SCs. Senomorphics decrease the number of SCs by diminishing the paracrine effect of senescence induction in neighboring cells, and by enhancing elimination of SCs by the immune system, which reduces their harmful effects.

We used middle-aged female rats fed a hypercaloric diet (HD) from 21 days to 15 months of age. Under our experimental conditions, rats exhibited cardiac hypertrophy and fibrosis, accumulation of senescent cells, changes in mitochondrial morphology, and oxidative stress. Rats were treated for 2 months with senolytic (dasatinib + quercetin, DQ) or senomorphic (sulforaphane, SFN) agents.

A novel aspect of recent research has been the crosstalk between organelles, particularly between the mitochondria and endoplasmic reticulum (ER), through specialized contact sites (MERCSs). However, there is still no consensus on the optimal distance between MERCSs and their positive or negative effects on disease progression.

HD rats showed cardiac improvement after both treatments. Although both strategies improved cardiomyocyte size and cardiac fibrosis, only DQ decreased LDH levels, whereas SFN positively affected cardiac damage proteins.

In general, no changes in structure or damage-associated enzymes were observed in control rats treated with DQ or SFN, indicating that senotherapies do not promote adverse effects on the heart, reinforcing the concept that they are safe for application in the clinical field. Data suggest a possible link mechanism between Nrf2 activation and MERCSs preservation, activated by SFN rather than by the DQ combination, which allowed cardiac structure maintenance in HD rats decreasing harmful effects of senescent cells.”

https://www.sciencedirect.com/science/article/abs/pii/S0955286325000865 “Cardioprotective effect of senotherapy in chronically obese middle-aged female rats may be mediated by a MERCSs/Nrf2 interaction” (not freely available) Thanks to Dr. Alejandro Silva for providing a copy.


Eat broccoli sprouts for your eyes, Part 3

Two 2025 papers cited Precondition your defenses with broccoli sprouts, starting with a review of age-related macular degeneration:

“AMD progression from intermediate to late AMD leads to a point of irreversible retinal pigmented epithelium (RPE) degeneration where treatment becomes worthless. Treating patients at the early/intermediate stages presents a better therapeutic window opportunity for AMD as the disease could potentially be prevented or slowed down.

Strong evidence points to RPE dysfunction at these stages, mainly through redox imbalance and lysosomal dysfunction in RPE oxidative injury. Restoring oxidative balance and lysosomal function may act as preventive and therapeutic measures against RPE dysfunction and degeneration.

Due to interaction with KEAP1, NRF2 is a ubiquitously expressed protein with a high turnover and half-life of about 20 minutes. Because the turnover of NRF2 is faster than KEAP1, newly synthesized NRF2 does not have free KEAP1 to bind and is translocated into the nucleus. Once in the nucleus, NRF2 dimerizes with sMAF and the complex binds to antioxidant response element (ARE) sequences, promoting the expression of ARE genes.

There is NRF2 involvement in most of the hallmarks of aging. Key transcriptional regulatory factors of related pathways, such as transcription factor EB (TFEB) and NRF2, may be targeted to restore homeostasis and/or prevent further RPE degeneration.”

https://www.mdpi.com/2076-3921/14/5/596 “Targeting Lysosomal Dysfunction and Oxidative Stress in Age-Related Macular Degeneration”

There were other informative tidbits throughout this review, such as:

  • “Anti-inflammatory effects of most electrophilic NRF2 activators are thought to be at least partly NRF2-independent, suggesting that these compounds lacking specificity may be advantageous for multitargeted pathologies.
  • TFEB can activate NRF2 under conditions devoid of oxidative stress.”

This paper also cited Bridging Nrf2 and autophagy when discussing the above graphic.


In this human cell and rodent study, several coauthors of the original 2020 study tested sulforaphane and TFEB interactions for ameliorating effects of a rare disease:

“Mutations in genes encoding lysosomal proteins could result in more than approximately 70 different lysosomal storage disorders. Niemann–Pick disease type C (NPC) is a rare lysosomal storage disorder caused by mutation in either NPC1 or NPC2 gene. Deficiency in NPC1 or NPC2 protein results in late endosomal/lysosomal accumulation of unesterified cholesterol.

Clinical symptoms of NPC include hepatosplenomegaly, progressive neurodegeneration, and central nervous system dysfunction, that is, seizure, motor impairment, and decline of intellectual function. So far there is no FDA-approved specific therapy for NPC.

Under stress conditions, that is, starvation or oxidative stress, TFEB is dephosphorylated and actively translocates into the nucleus, promoting expression of genes associated with lysosome and autophagy. TFEB overexpression or activation results in increased number of lysosomes, autophagy flux, and exocytosis.

Pharmacological activation of TFEB by sulforaphane (SFN), a previously identified TFEB agonist, significantly promoted cholesterol clearance in human and mouse NPC cells, while genetic inhibition (KO) of TFEB blocked SFN-induced cholesterol clearance. This clearance effect exerted by SFN was associated with upregulated lysosomal exocytosis and biogenesis. SFN treatment has no effect on the liver and spleen enlargement of Npc1 mice.

SFN is reportedly BBB-permeable, assuring a good candidate for efficient delivery to the brain, which is essential for targeting neurodegenerative phenotypes in neurological diseases including NPC. This is the first time that SFN was shown to directly activate TFEB in the brain.

Collectively, our results demonstrated that pharmacological activation of TFEB by a small-molecule agonist can mitigate NPC neuropathological symptoms in vivo. TFEB may be a putative target for NPC treatment, and manipulating lysosomal function via small-molecule TFEB agonists may have broad therapeutic potential for NPC.”

https://elifesciences.org/articles/103137 “Small-molecule activation of TFEB alleviates Niemann–Pick disease type C via promoting lysosomal exocytosis and biogenesis”


Eat broccoli sprouts to alleviate diabetic heart disease

A 2025 rodent study investigated sulforaphane’s effects on diabetic cardiomyopathy:

“The protective effect of cruciferae-derived sulforaphane (SFN) on diabetic cardiomyopathy (DCM) has garnered increasing attention. However, no studies have specifically explored its mechanistic involvement in cardiac substrate metabolism and mitochondrial function.

To address this gap, Type 2 diabetes mellitus (T2DM) db/db mice were orally gavaged with vehicle or 10 mg/kg body weight SFN every other day for 16 weeks, with vehicle-treated wild-type mice as controls. SFN intervention (SFN-I) alleviated hyperglycemia, dyslipidemia, HOMA-IR, serum MDA levels, and liver inflammation. SFN-I improved the lipotoxicity-related phenotype of T2DM cardiomyopathy, manifested as attenuation of diastolic dysfunction, cardiac injury, fibrosis, lipid accumulation and peroxidation, ROS generation, and decreased mitochondrial complex I and II activities and ATP content.

Although not fully understood, multiple systemic and cardiac-local mechanisms contribute to DCM, encompassing hyperglycemia, dyslipidemia, insulin resistance (IR), disturbances in cardiac substrate metabolism, lipotoxicity, glucotoxicity, mitochondrial dysfunction, oxidative stress, inflammation, and endoplasmic reticulum (ER) stress. Nrf2 and its downstream metallothionein also mediated the preventive effect of SFN on DCM, and may underlie the synergistic effect of SFN and zinc in DCM.

These results suggest that chronic oral SFN-I protects against DCM by mitigating overall metabolic dysregulation and inhibiting cardiolipotoxicity. The latter might involve controlling cardiac fatty acid metabolism and improving mitochondrial function, rather than promoting glucose metabolism.”

https://www.mdpi.com/2076-3921/14/5/603 “Oral Sulforaphane Intervention Protects Against Diabetic Cardiomyopathy in db/db Mice: Focus on Cardiac Lipotoxicity and Substrate Metabolism”


This study had numerous charts like the above showing it was better to not have a deviation from health (Ctrl) rather than incur injury (DCM) then try to fix it with sulforaphane (DCM + SFN). But the control group was wild-type mice, not mice genetically inclined to diabetes like the treatment groups.

The subjects’ starting points were at nine-weeks-old (equivalent to 18-25 year-old humans), and duration was 16 weeks. Grok 3 said: “A 25-week-old db/db mouse is roughly equivalent to a human aged 30–35 years chronologically, though its metabolic condition may mimic older human physiological states in diabetes and obesity research.”

A human equivalent of a 10 mg/kg sulforaphane dose is (.081 x 10 mg) = 56 mg orally administered every other day. That’s about how much total sulforaphane I estimated I took every day (52 mg) from Week 6 through Week 56 by eating microwaved broccoli sprouts twice daily.

No rationale was provided for the sulforaphane dose or the every-other-day dosing regimen. Since I took ~52 mg every day for almost a year, I’ll guess that this study may have had more definitive results with daily dosing. Or maybe add zinc per Zinc and broccoli sprouts – a winning combination.

Nrf2 activators and transcriptomic clocks

Two preprint studies looked at making transcriptional aging clocks using Nrf2 activators. Let’s start with a 2025 nematode study that used constant exposure to sulforaphane at different concentrations:

“To explore the potential of sulforaphane as a candidate natural compound for promoting longevity more generally, we tested the dose and age-specific effects of sulforaphane on C. elegans longevity, finding that it can extend lifespan by more than 50% at the most efficacious doses, but that treatment must be initiated early in life to be effective. We then created a novel, gene-specific, transcriptional aging clock, which demonstrated that sulforaphane-treated individuals exhibited a “transcriptional age” that was approximately four days younger than age-matched controls, representing a nearly 20% reduction in biological age.

The clearest transcriptional responses were detoxification pathways, which, together with the shape of the dose-response curve, indicates a likely hormetic response to sulforaphane. The hormetic, stress-pathway inducing properties of sulforaphane may indicate that many beneficial dietary supplements work in a fairly generic fashion as mild toxins rather than being driven by the biochemical properties of the compounds themselves (e.g., as antioxidants).

These results support the idea that robust longevity-extending interventions can act via global effects across the organism, as revealed by systems level changes in gene expression.”

https://www.biorxiv.org/content/10.1101/2025.05.11.653363v1 “The broccoli derivative sulforaphane extends lifespan by slowing the transcriptional aging clock”

There are difficulties in researchers translating nematode studies to mammals and humans. Nematodes lack a homolog to the Keap1 protein, which is sulforaphane’s main mammalian target to activate Nrf2.


A 2024 study developed various mammalian epigenetic clocks:

“A unified transcriptomic model of mortality that encompasses both aging and various models of lifespan-shortening and longevity interventions (i.e., mortality clocks) has been lacking. We conducted an RNA-seq analysis of mice subjected to 20 compound treatments in the Interventions Testing Program (ITP).

We sequenced the transcriptomes of a large cohort of ITP mice subjected to various neutral and longevity interventions, expanded the dataset with publicly available gene expression data representing organs of mice and rats across various strains and lifespan-regulating interventions, connected these models with survival data, and performed a meta-analysis of aggregated 4,539 rodent samples, which allowed us to identify multi-tissue transcriptomic signatures of aging, mortality rate, and maximum lifespan.

Aging and mortality were characterized by upregulation of genes involved in inflammation, complement cascade, apoptosis, and p53 pathway, while oxidative phosphorylation, fatty acid metabolism, and mitochondrial translation were negatively associated with mortality, both before and after adjustment for age.

Utilizing the aggregated dataset, we developed rodent multi-tissue transcriptomic clocks of chronological age, lifespan-adjusted age, and mortality. While the chronological clock could distinguish the effect of detrimental genetic and dietary models, it did not show a decrease in biological age in response to longevity interventions. In contrast, clocks of lifespan-adjusted age and mortality both captured aging-associated dynamics and correctly predicted the effect of lifespan-shortening and extending interventions.

Transcriptomic biomarkers developed in this study provide an opportunity to identify interventions promoting or counteracting molecular mechanisms of mortality, and characterize specific targets associated with their effects at the level of cell types, intracellular functional components, and individual genes. Our study underscores the complexity of aging and mortality mechanisms, the interplay between various processes involved, and the clear potential for developing therapies to extend healthspan and lifespan.”

https://www.biorxiv.org/content/10.1101/2024.07.04.601982v1.full “Transcriptomic Hallmarks of Mortality Reveal Universal and Specific Mechanisms of Aging, Chronic Disease, and Rejuvenation”


This second study’s references included an ITP study curated in Astaxanthin and aging, which stated:

“Despite the fact that the average diet contained 1840 ppm astaxanthin (only 46% of the target), median lifespans of male UM-HET3 mice were significantly improved. Amounts of dimethyl fumarate (DMF) in the diet averaged 35% of the target dose, which may explain the absence of lifespan effects.”

So screw-ups in making both astaxanthin and DMF mouse chows ended up with study data that didn’t measure the full lifespan impacts of activating transcription factor Nrf2. I’ll assert that such faulty data may have deviated this second study by downplaying Nrf2 activation’s impact on aging, chronic disease, and rejuvenation.

Sponsors may be less likely to be presented sulforaphane and other Nrf2 activator candidates for future aging and chronic disease studies as this first study suggests, thinking that these have already been studied in mammals. Well, maybe these compounds haven’t been accurately studied. There’s no effective way to fix a rodent study’s missing DMF Nrf2 data and faulty astaxanthin Nrf2 data to train an epigenetic clock in this second study.

I could be wrong about this second study using faulty astaxanthin Nrf2 data. It was cited as Reference 27 in the Introduction as an ITP study, but not specifically cited in the Method section. I don’t know how findings such as one of Nrf2’s target genes (“Remarkably, one of the top genes positively associated with maximum lifespan and negatively associated with chronological age and expected mortality was Gpx1, encoding the selenoprotein glutathione peroxidase 1″) and a Nrf2 specific pathway (Phase II) (“Pathways positively associated with lifespan and negatively with mortality, both before and after adjustment for age, included..xenobiotic metabolism..”) were made without Reference 27. Neither of the above studies has been peer reviewed yet.


The third phase of reversing aging and immunosenescent trends

Here’s a 2025 interview with Dr. Greg Fahy:

“We found that we could statistically demonstrate thymic regeneration morphologically on single individuals at single time points. MRI changes really are detecting shifts from the fatty tissue infiltration state of the involuted thymus to the regenerated thymus with functional thymic epithelial cells.

When you go through puberty your thymus involutes so you don’t have much left even when you’re 40. Essentially the process consists of loss of functional thymic mass and replacement of that functional thymic mass with adipose tissue, that’s what thymic involution is. It continues throughout life, but you retain a small amount of functional thymic mass all the way out to the age of 107.

The function of the thymus is to essentially manufacture half of your immune system. You have precursor cells arise from the bone marrow. They either go into the meiotic lineage and turn into the innate immune system, or you have the lymphocytic cells for what turns into T cells that enter the thymus and are educated in the thymus to grow up into newborn T cells and they’re released into the bloodstream.

The thymus has two jobs. It manufactures these lovely T cells without which you die but it also has a secondary finishing school. In the thymus cortex you manufacture all these lovely T cells but in the thymus medulla the T cells go to the medulla and if they don’t pass the second examination that they have to pass before they release into the body they’re all killed off. That second examination is: Do you reject self? As we get older, the thymus weakens in both the functions of making the T cells and screening out the ones that attack self. It stands to reason as we get older and the thymus’ influence wanes, we’re going to get more autoimmune disorders.

It took people a while to catch on to the fact that this involution problem is really a significant issue because the T cells that you made when you were 12, and even 20 and 40, they’re probably lasting until you’re 60. But at some point they don’t get replaced as fast as they’re going out of existence, and then your immune system goes off the cliff. Between the ages of 62 and 78 you lose 98% of your ability to recognize foreign antigens, and you still have a lot of capacity left.

We had nine guys in the first trial. Second trial we had 18 men 6 women and 2 controls that happen to be contemporaneous with that group. We have some more controls now that are either finished or or nearing completion. The second population was older than the first population by about nine years, but based on the epigenetic clocks that we looked at, they were starting off biologically younger.

On this last data analysis for Triim XA we looked at 21 different aging clocks. One aspect of the noise that we’re talking about is that biological aging as measured by some of these clocks is circadian. If you measure your age at 4:00 a.m. versus 11:00 a.m. you’re going to get a different result. It’s dynamic and there’s a trend and over time you change in a certain direction, but over any short period of time you can bounce around a little bit. The clocks predict your probability of cognitive dysfunction, they predict your probability of having impairments in your daily life, and they also predict your mortality.

We’re pretty much wrapping up that second clinical trial and going into the third. As we look at more data we understand more and more things and we see more and more things that we previously were not aware of. We began to look at a phenomena that may be responsible for limiting the magnitude of responses that we’re seeing limiting the aging reversal.

Triim-XD which is the next flavor of Triim-X is going to be looking at shifting biochemical pathways in such a way that it optimizes effects of these three medications that we’re giving people [human growth hormone, DHEA, and metformin] and prevents contradictions between them and prevents side effects of each one of these things. That’s about all I can tell you right now.”


Charts regarding the discussed item of how long effects may last are covered in The next phase of reversing aging and immunosenescent trends which was the last time I curated this research effort.


Practice what you preach, or shut up

A 2025 review subject was sulforaphane and brain health. This paper was the latest in a sequence where the retired lead author self-aggrandized his career by citing previous research.

He apparently doesn’t personally do what these research findings suggest people do. The lead author is a few weeks older than I am, and has completely white hair per an interview (Week 34 comments). I’ve had dark hair growing in (last week a barber said my dark hair was 90%) since Week 8 of eating broccoli sprouts every day, which is a side effect of ameliorating system-wide inflammation and oxidative stress.

If the lead author followed up with what his research investigated, he’d have dark hair, too. Unpigmented white hair and colored hair are both results of epigenetics.

Contrast this lack of personal follow-through of research findings with Dr. Goodenowe’s protocol where he compared extremely detailed personal brain measurements at 17 months and again at 31 months. He believes enough in his research findings to personally act on them, and demonstrate to others how personal agency can enhance a person’s life.

It’s every human’s choice whether or not we take responsibility for our own one precious life. I’ve read and curated on this blog many of this paper’s references. Five years ago for example:

So do more with their information than just read.

https://www.mdpi.com/2072-6643/17/8/1353 “Sulforaphane and Brain Health: From Pathways of Action to Effects on Specific Disorders”

Year Five of Changing to a youthful phenotype with sprouts

1. I’ve continued daily practices from Year Four to experience another year without being sick! I’ll get a set of Labcorp tests in a week to see if anything is sneaking up on me.

Really think that Brassica clinical trials should last years, not weeks. Once people get over the fact that broccoli, red cabbage, and mustard sprouts will never taste good because their compounds are plants’ defenses against predators, they’ll overlook that in favor of health benefits. Avena sativa oat sprouts don’t have a palatability problem.

2. Daily supplements have changed a little:

  • Started taking a quercetin supplement suggested in a comment to Year One as helpful for seasonal allergies (it doesn’t do that for me). Repeatedly rinsing and soaking the salt out of capers for quercetin content became too much of a nuisance, and the results didn’t always taste right;
  • Stopped taking Prodrome supplements because of unsustainable high costs;
  • Started taking Ovega 3 algae oil DHA 420 mg/EPA 140 mg twice a day in their place;
  • Substituted flax oil 1400 mg once a day for Balance oil;
  • Started taking 2 g magnesium L-threonate;
  • Upped taurine intake from 5 to 6 grams;
  • Upped D3 by 25 mcg to a daily 4400 IU;
  • Reduced chondroitin sulfate by 1.8 g since my joints are doing fine;
  • Stopped soy lecithin in favor of eating three raw eggs.

3. I injured my left shoulder in May 2024 by overdoing upper body exercises, and stopped seven months to recover. Gained thirty pounds during that layoff, and have worked off ten pounds with new routines since then.

I’m no longer dogmatic about aerobic exercise / beach walks. I’ll go over to the beach before sunrise when it isn’t raining or windy, or wait until the afternoon for weather to improve, rather than walk 30 minutes a day irregardless.

2025 α-ketoglutarate research

I haven’t mentioned α-ketoglutarate for a while, although I’ve taken it twice a day for several years. Here are four 2025 papers on α-ketoglutarate, starting with a review of its role in bone health:

“α-Ketoglutarate (α-KG) serves as a pivotal intermediate in various metabolic pathways in mammals, significantly contributing to cellular energy metabolism, amino acid metabolism, and other physiological processes. α-KG may be a therapeutic target for a variety of bone-related diseases, such as osteoporosis, osteoarthritis, and rheumatoid arthritis, because of its role in maintaining metabolic balance of bone.

α-KG, as a rate-determining mitochondrial intermediate, is crucial in cell energy metabolism because it connects intracellular carbon and nitrogen metabolism between isocitrate and succinyl coenzyme A. Additionally, α-KG is closely involved in the amino acid cycle. As a precursor of amino acids such as glutamine and glutamic acid, α-KG plays a direct role in energy production and a wide range of cellular chemical reactions. α-KG provides an energy source, stimulating protein synthesis, inhibiting protein degradation in muscle, and serving as a significant metabolic fuel for gastrointestinal cells.

α-KG promotes osteogenic differentiation of stem cells, increases activity of osteoblasts to promote osteogenesis, and inhibits bone resorption activity of osteoclasts. α-KG in articular cartilage promotes differentiation and maturation of chondrocytes and formation of a cartilage matrix. The protective effect of α-KG on bone has practical value in treatment of abnormal bone loss symptoms in various bone tissue diseases.”

https://www.sciengine.com/ABBS/doi/10.3724/abbs.2025020 “Essential role of the metabolite α-ketoglutarate in bone tissue and bone-related diseases”


A rodent study explored adding α-KG to osteoarthritis treatment:

“Mesenchymal stem cell (MSC) therapy represents a promising treatment strategy for osteoarthritis (OA). Nevertheless, the therapeutic efficacy of MSCs may be attenuated under conditions of cellular senescence or when the available clinical quantity is insufficient. α-Ketoglutarate (AKG) exerts beneficial effects on skeletal tissues and activity of stem cells. The present study was designed to explore the potential of AKG in augmenting viability of MSCs and the potential of their combined utilization in treatment of OA.

AKG plays a crucial role in multiple biological processes. It is involved in regulating stem cell differentiation, exerts anti-apoptotic effects, modulates the body’s immune and inflammatory responses, contributes to muscle and bone development, and is essential for maintaining stability of the cartilage matrix.

Platelet-rich plasma (PRP) has been demonstrated to have protective effects on chondrocytes and can effectively repair damaged cartilage in OA. However, PRP has intractable problems in terms of product quality control and allogeneic application, and its long-term therapeutic effect gradually weakens.

Combining AKG’s regulation of cellular metabolism with the multi-directional differentiation and immunomodulatory functions of MSCs is likely to generate a synergistic effect. This combined treatment modality targets the complex pathological processes of OA, including cartilage damage, inflammatory responses, and extracellular matrix imbalance, in a more comprehensive manner than a single therapy.”

https://www.sciencedirect.com/science/article/pii/S2707368825000032 “The repair effect of α-ketoglutarate combined with mesenchymal stem cells on osteoarthritis via the hedgehog protein pathway”


A rodent study investigated whether α-KG has a role in determining frailty:

“Frailty is an age-related geriatric syndrome, for which the mechanisms remain largely unknown. We performed a longitudinal study of aging female (n = 40) and male (n = 47) C57BL/6NIA mice, measured frailty index, and derived metabolomics data from plasma samples.

We find that frailty related metabolites are enriched for amino acid metabolism and metabolism of cofactors and vitamins, include ergothioneine, tryptophan, and alpha-ketoglutarate, and present sex dimorphism. We identify B vitamin metabolism related flavin adenine dinucleotide and pyridoxate as female-specific frailty biomarkers, and lipid metabolism related sphingomyelins, glycerophosphoethanolamine and glycerophosphocholine as male-specific frailty biomarkers.

We were interested to observe whether metabolite abundance at any specific timepoint was associated with frailty at a future timepoint. Unfortunately, we didn’t observe any metabolites that showed an overall significant association with future FI (FIf) or future devFI (devFIf). When focusing only on the abundance of metabolites at the baseline time point (∼400 days), we found a single metabolite, alpha-ketoglutarate, was negatively associated with both FIf and devFIf.”

https://www.biorxiv.org/content/10.1101/2025.01.22.634160v1.full “Metabolomics biomarkers of frailty: a longitudinal study of aging female and male mice”


Wrapping up with a rodent study adding α-KG to exercise for its effects on depression and learning:

“aKG acts as a prophylactic and antidepressant to effectively counteract social avoidance behaviors by modulating BDNF levels in the hippocampus and nucleus accumbens. Exercise increases aKG levels in the circulation.

In mice, aKG supplementation prolongs lifespan and reduces aging-associated frailty. aKG supplementation also reverses aging in humans as measured by DNA methylation patterns.

aKG functions as a co-factor for epigenetic enzymes. Changes in the intracellular αKG/succinate ratio regulates chromatin modifications, including H3K27me3 and ten-eleven translocation (Tet)-dependent DNA demethylation. The ability of aKG to influence epigenetic status of cells may explain both its prophylactic and anti-depressant effects since transcriptional dysregulation and aberrant epigenetic regulation are unifying themes in psychiatric disorders. This may also explain its ability to differentially regulate BDNF expression in the hippocampus and NAc.

If exercise mediates its effects through aKG, aKG may be a pivotal component of an exercise pill along with lactate and BHB that can serve as both a prophylactic and antidepressant treatment for depression.”

https://www.sciencedirect.com/science/article/pii/S266717432500031X “α-ketoglutarate (aKG) is a circulatory exercise factor that promotes learning and memory recall and has antidepressant properties


Vitamin K2 and your brain

A 2025 review linked Vitamin K2‘s effects on vascular health with cognitive function:

“Cardiovascular disease (CVD) is negatively correlated with cognitive health. Arterial stiffness, in particular, appears to be a critical factor in the functional and structural brain changes associated with aging. We review the association between vitamin K and cerebral function, discussing novel developments regarding its therapeutic role in arterial stiffness and cognitive health.

Among the non-invasive measures of vascular stiffness, pulse wave velocity (PWV) is considered the gold standard. PWV measures arterial stiffness along the entire aortic pathway, providing a reliable, feasible, and accurate assessment of vascular health. Arterial stiffness, as measured by PWV, is negatively associated with total brain volume, brain atrophy, and cognitive function. Pathogenic mechanisms responsible for vascular stiffness recently shifted from collagen and elastin to the differentiation of vascular smooth muscle cells to osteoblastic phenotype, which is triggered by oxidative stress and inflammation, membrane mechanotransduction, lipid metabolism, genetic factors, and epigenetics.

Vitamin K-dependent proteins (VKDPs) rely on vitamin K to undergo γ-glutamylcarboxylation, a modification essential for their biological activity. This family of proteins includes hepatic VKDPs such as prothrombin, FVII, FIX, and FX, protein S and protein C as well as extrahepatic VKDPs such as matrix Gla-protein (MGP), which is involved in inhibiting vascular calcification, and osteocalcin, which plays a role in bone mineralization.

Structural differences between K1 and K2 influence their bioavailability, absorption, bioactivity, and distribution within tissues. Compared to vitamin K1, the K2 subtype menaquinone-7 (MK-7) has a significantly longer half-life, accumulates more effectively in blood, and exhibits greater biological activity, particularly in facilitating the carboxylation of extrahepatic VKDPs. Circulating dephosphorylated, uncarboxylated Matrix Gla protein (dp-ucMGP), a marker of extrahepatic vitamin K deficiency, could represent a novel therapeutic target for mitigating both arterial stiffness and cognitive decline.

Vascular calcification and arterial stiffness may represent pathophysiological mechanisms underlying the onset and progression of cognitive decline. Vitamin K deficiency is a key determinant of arterial health and, by extension, may influence cognitive function in the elderly.

To elucidate potential therapeutic benefits of MK-7 supplementation on cognitive function, future randomized controlled trials (RCTs) are needed. These trials should focus on using optimal dosages (>500 μg/day), ensuring long follow-up periods, and utilizing the most bioactive form of vitamin K (MK-7).”

https://www.frontiersin.org/journals/aging-neuroscience/articles/10.3389/fnagi.2024.1527535/full “The role of vitamin K2 in cognitive impairment: linking vascular health to brain health”


A coauthor Dr. Katarzyna Maresz took time on her weekend to answer a few questions:

1. Regarding the second paper of Part 2 of Vitamin K2 – What can it do?:

Hello Dr. Maresz. Did this trial ever happen? “Effects of Combined Vitamin K2 and Vitamin D3 Supplementation on Na[18F]F PET/MRI in Patients with Carotid Artery Disease: The INTRICATE Rationale and Trial Design” I haven’t seen a followup mention of it since 2021.

“Hello. The study never started. The capsules were produced for the study, but the research center experienced delays. Unfortunately, I’m afraid it won’t proceed. Regarding studies on aortic stenosis and vitamin K2, BASIC II has been completed, and the data from this pilot study are currently under analysis. (https://pubmed.ncbi.nlm.nih.gov/29561783/). There is also published study with K1: https://www.ahajournals.org/doi/10.1161/CIRCULATIONAHA.116.027011

2. Thank you! In your recent review of cognitive function and K2 (above), what influenced the heuristic that a >500 mcg K2 dose should be pursued in future RCTs?

“The optimal vitamin K dosage depends on the target population. Research in kidney patients has shown that 460 mcg daily was insufficient, that is why have hypothesis that at least 500 mcg should be used. The ongoing VIKIPEDIA study is using 1,000 mcg daily in peritoneal dialysis patients. In healthy young individuals, 180-360 mcg was effective in improving vitamin K status (British Journal of Nutrition (2012), 108, 1652–1657) . However, a one-year clinical study found that 180 mcg daily was sufficient for women but not for men. Additionally, older adults and individuals with metabolic disorders may require higher doses for optimal benefits. So it is pretty complicated situation. We do not have good marker of extrahepatic K status. dp-ucMGP seems to be valuable from CV perspective.”

3. Regarding Fat-soluble vitamin competition:

Thank you again Dr. Maresz! Would any consideration be given to dosing K2 separately from dosing another fat-soluble vitamin? A 2015 in vitro study found that vitamins D, A, and E outcompeted K1 intake when simultaneously dosed. I inferred from the one capsule of D3-K2 produced for the canceled trial that isn’t that much of a problem with K2?

“You are right, the key findings suggest that vitamin D, E, and K share common absorption pathways, leading to competitive interactions during uptake. However, I’m afraid we do not have human data. The majority of studies have focused on vitamin K2 alone. Recent research combining K2 and D3 showed an improvement in vitamin K status. Example: https://pubmed.ncbi.nlm.nih.gov/35465686/ or increase in D level: https://pubmed.ncbi.nlm.nih.gov/39861434/. We do not know if VKDP activation or absorption of D would be more effective if K2 were not supplemented with D3 at the same time. Unfortunately, I doubt anyone will fund such a study, as clinical trials are very expensive. In vitro data will always raise questions regarding their relevance to human physiology. In my opinion, for patients to fully benefit from optimal vitamin K status, vitamin D levels should also be optimized, as both have synergistic effects.”

Epigenetic clock analysis of a clinical trial

A 2025 paper performed post-hoc epigenetic clock analyses of a supplement and exercise clinical trial completed earlier this decade:

“We report results of a post hoc analysis among 777 participants of the DO-HEALTH trial on the effect of vitamin D (2,000 IU per day) and/or omega-3 (1 g (330 mg EPA plus 660 mg DHA from marine algae) per day) and/or a home exercise program (a strength-training exercise program performed for 30 min three times per week) on four next-generation DNA methylation (DNAm) measures of biological aging (PhenoAge, GrimAge, GrimAge2 and DunedinPACE) over 3 years. Omega-3 alone slowed the DNAm clocks PhenoAge, GrimAge2 and DunedinPACE, and all three treatments had additive benefits on PhenoAge.

Inclusion criteria were age 70 years and older, living at home, having no major health events (no cancer or myocardial infarction) in the 5 years before enrollment, having sufficient mobility to visit the study centers without help and having good cognitive function with a Mini-Mental State Examination score of at least 24. 777 provided consent for these analyses and had samples available after the application of the exclusion criteria. This group of individuals formed our analysis sample, which had the following characteristics: 59% were women; the mean age at baseline was 75 years; 30% had 25-hydroxyvitamin D (25(OH)D) levels of <20 ng ml−1; 53% were healthy agers as defined in the Nurses’ Health Study (free of major chronic diseases, disabilities, cognitive impairments and mental health limitations); and 88% were physically active (29% were active one to three times per week, and 59% were active more than three times per week). The Swiss participant subgroup represents a healthier and more active subgroup within the total DO-HEALTH population.

Overall, from baseline to year 3, standardized effects ranged from 0.16 to 0.32 units (2.9–3.8 months). In summary, our trial indicates a small protective effect of omega-3 treatment on slowing biological aging over 3 years across several clocks, with an additive protective effect of omega-3, vitamin D, and exercise based on PhenoAge.”

https://www.nature.com/articles/s43587-024-00793-y “Individual and additive effects of vitamin D, omega-3 and exercise on DNA methylation clocks of biological aging in older adults from the DO-HEALTH trial”

These epigenetic clock measurements of a subset of trial subjects was interesting, although I didn’t find it particularly relevant to what I do. I take twice as much Vitamin D and omega-3s everyday, do resistance exercises once or twice a week whenever I’ve recovered from the previous session, walk a few miles on the beach if the weather is nice, and other things.

I don’t bother with epigenetic clock measurements anymore because the free one (PhenoAge) is too variable to be personally accurate. For other clocks, it would be meaningless if all I got was a 2-3 month improvement over a three year period like this trial. Studies usually find that the most deficient subjects at the beginning are the ones that show the greatest improvements with effective treatments. Unhealthiness on any epigenetic clock parameter probably wouldn’t be my starting point, so I may not show even a one-month improvement over three years.


Dr. Goodenowe offered his opinion on the paper:

“DHA is a polyunsaturated fatty acid that is essential for maintaining youthful fluidity of the body’s membranes. While our bodies can make DHA from the essential omega-3 dietary fatty acid, as we get older, our ability to make DHA decreases and oxidative stress on our bodies increases. These two factors contribute to our membranes becoming stiffer and less pliable as we age, in other words, ‘older.’

Because getting older and losing function appear to go hand in hand, we equate aging with a loss of function. As such, we think that aging causes this loss of function, like a disease. Instead, the opposite is true, and it’s the loss of function that causes aging. To slow aging you need to focus on maintaining function.”

https://www.prevention.com/health/a63850396/vitamin-exercise-boost-longeivty-study/ “Scientists Find Taking This Vitamin Boosts Longevity, Add Years to Your Life”

Prevention magazine’s editors need to better proof their writers’ work before it gets published. Unlike the headline, the trial had nothing to do with adding years to human lifespan.

Broccoli antihypertensive peptides

This 2025 rodent cell study investigated effects of broccoli peptides:

“ACE is a pivotal enzyme that has a regulatory effect on blood pressure in human renin-angiotensin system (RAS). Inhibiting ACE activity can reduce production of angiotensin II (Ang II), which binds to receptors on the vascular wall, causing vasoconstriction.

Development of natural ACE inhibitors with low side effects is an urgent need for cardiovascular therapy. Many natural angiotensin-converting enzyme inhibitory (ACEI) peptides have been widely studied. However, their stability in vivo is poor in most cases.

In this study, peptides were initially digested from broccoli in vitro, and absorption was simulated by Caco2 cells transport and then analyzed by peptideomics and molecular docking. ACEI activity of broccoli crude peptide increased after digestion.

Subsequently, mechanisms were verified using a high glucose-induced vascular smooth muscle cells (VSMCs) dysfunction model. Five peptides not only inhibited proliferation, migration, and apoptosis of VSMCs by inhibiting ERK and p38 MAPK phosphorylation, but also restrained the activities of ACE and AT1R, prominently reducing Ang II levels within VSMCs under high glucose.

This research provides valuable insights into the production of novel ACEI peptides derived from broccoli protein, and offers directions for utilization of these antihypertensive peptides in health applications.”

https://www.frontiersin.org/journals/nutrition/articles/10.3389/fnut.2025.1528184/fullIn vitro gastrointestinal digestion simulation screening of novel ACEI peptides from broccoli: mechanism in high glucose-induced VSMCs dysfunction”