Plasmalogens Week #3 – Aging

Continuing Plasmalogens Week with two 2025 papers, starting with a rodent study of plasmalogens’ effects on mitigating cognitive decline:

“We evaluated beneficial effects of plasmalogens (PLS), phosphatidylcholine (PC), and phosphatidylserine (PS) on age-associated cognitive decline. We established a mouse model of aging-associated cognitive impairment using the subcutaneous injection of d-galactose (D-gal) at a dosage of 400 mg/kg/day.

We randomly divided six-week-old female mice into nine groups: control, model, high-dose PLS (0.3 mg/kg/day), low-dose PLS (0.09 mg/kg/day), high-dose PC (200 mg/kg/day), low-dose PC (50 mg/kg/day), high-dose PS (200 mg/kg/day), low-dose PS (50 mg/kg/day), AMC-Plas (120 mg/kg/day; and functional component PLS (0.252 mg/kg/day).

We administered PLS, PC, and PS separately by oral gavage once daily. We extracted PLS from scallops according to the literature. AMC-Plas is a commercially available health supplement known for its neuroprotective properties and memory-enhancing effects. In this study, we included AMC-Plas as a positive control group to evaluate the effects of different phospholipids.

Synaptophysin (SYP), synapsin-1 (SYN-1), postsynaptic density protein 95 (PSD-95), and brain-derived neurotrophic factor (BDNF) play important roles in synapse formation and synaptic plasticity. Synaptic function alterations or losses are key pathological mechanisms that underlie development of cognitive impairment. Therapeutic strategies that attempt to restore synaptic function or promote synaptic remodeling are considered to be increasingly promising strategies to mitigate cognitive decline.

Results showed that:

  • PLS improved spatial memory performance by 44% and object recognition by 80% in D-galactose-induced cognitively impaired mice.
  • PLS significantly decreased glial fibrillary acidic protein (GFAP)-positive cells (an indicator of astrocyte activation) in the dentate gyrus (DG) of the hippocampus, an important result because the DG is a crucial neurogenesis region.
  • PLS alleviated neuronal damage and protected against synaptic injury, verified by a 228.01% increase in PSD-95 expression in the hippocampus.
  • PLS showed a more prominent role for the mitigation of age-related cognitive impairment compared with PC and PS.

In conclusion, the evaluation of PLS using both behavioral and neuropathological assessments in cognitively impaired mice highlighted its exceptional efficacy compared with other phospholipids. PLS at a remarkably low effective dose significantly ameliorated cognitive deficits in cognitively impaired mice. This result further emphasized its potential relevance in neurodegenerative disease research.

We found that PLS alleviated cognitive impairment potentially by improving synaptic function; however, the molecular mechanisms that underlie its effects on synaptic function warrant further investigation.”

https://www.sciencedirect.com/science/article/pii/S175646462500132X “Mitigating effects of plasmalogens on age-related cognitive impairment”

There was no disclosed chemical analysis of the PLS scallop extract’s plasmalogen types or other contents. Despite its name, I didn’t see that the AMC-Plas product contained plasmalogens or plasmalogen precursors.


A fruit fly study investigated plasmalogen effects on mitochondria during aging:

“We identify plasmalogens—endogenous ether-linked phospholipids—as key regulators of age-associated mitochondrial fission in Drosophila melanogaster. Loss of Kua (also known as plasmanylethanolamine desaturase (PEDS) / TMEM189 in mammals), the enzyme essential for plasmalogen biosynthesis, leads to inhibition of mitochondrial fission and impaired recruitment of the fission protein Drp1, similar to what is observed during aging.

Mitochondrial dynamics, comprising balanced cycles of fission and fusion, are essential for preserving organelle quality, metabolic flexibility, and cellular homeostasis throughout life. Aging disrupts this balance, with multiple studies reporting a decline in mitochondrial fission that contributes to the accumulation of enlarged and dysfunctional mitochondria.

These morphological changes are linked to impaired mitophagy, altered energy production, and tissue dysfunction. Midlife induction of Drp1—the dynamin-related GTPase that drives mitochondrial division—has been shown to reverse age-related mitochondrial defects and prolong lifespan in Drosophila.

To determine whether plasmalogen biosynthesis is essential for mitochondrial fission, we used KuaMI04999, a hypomorphic allele. Western blot analysis revealed significantly reduced Kua protein levels in KuaMI04999/+ heterozygotes compared to wild-type controls.

Our findings reveal a previously unrecognized lipid-based mechanism that controls mitochondrial fission during aging and position plasmalogens as key effectors linking membrane composition to mitochondrial homeostasis. It is not merely expression or stability of Drp1 that is affected, but rather its recruitment to the mitochondrial surface, which is a critical activation step for fission.

While our study highlights the requirement of plasmalogen biosynthesis for Drp1 recruitment, further work is needed to understand how plasmalogens mechanistically facilitate this interaction.”

https://www.researchsquare.com/article/rs-7330024/v1 “Plasmalogen Biosynthesis Controls Mitochondrial Fission via Drp1 Recruitment during Aging”

This study didn’t analyze or characterize specific plasmalogens.


Ancient DNA fragments enable adult neurogenesis

A 2025 rodent study investigated mechanisms by which erythropoietin (EPO) enables adult neurogenesis and cognitive function:

“We mapped epigenomic and transcriptional landscapes of adult mouse hippocampus under recombinant human EPO (rhEPO) treatment. We discovered significant lineage-specific remodelling of chromatin accessibility predominantly in newly formed pyramidal neurons, highlighting a robust EPO-driven neurogenic response. Notably, transposable elements (TEs), particularly ancient LINEs and SINEs, emerged as critical cis-regulatory elements (cCREs).

EPO is known to be upregulated in the brain under hypoxic or injury conditions, and it has been considered a natural neuroprotective agent. We demonstrated that EPO, a traditionally hematopoietic hormone, can profoundly reprogram the adult neural epigenome to drive neurogenesis.

EPO may activate a specific subclass of dormant regulatory elements to drive nearby genes. Such a mechanism would represent a previously unappreciated mode of gene regulation: the de novo recruitment of ancient genomic elements to drive a contemporary cellular response.

Our data support the model that EPO drives differentiation of progenitors rather than inducing widespread cell division. The net effect is an enrichment of pyramidal neurons at the cost of interneurons. Pyramidal neurons integrate in the hippocampal circuitry, leading to potential implications for mood, memory, cognitive enhancement, and recovery from brain injury.

We propose a conserved evolutionary mechanism at play: ancient TEs embedded in the genome have been repurposed as cCREs in neural cells, and during an EPO-induced neurogenic stimulus, the brain taps into this reservoir of regulatory elements to rapidly reshape gene expression. In evolutionary terms, this represents an efficient strategy.”

https://www.biorxiv.org/content/10.1101/2025.10.13.682070v1.full “Transposable Element-Mediated Epigenomic Remodeling Drives Erythropoietin-Induced Neurogenesis in the Adult Hippocampus”


Sulforaphane as a senotherapy, Part 2

A 2025 rodent study by the same group as Part 1 investigated similar subjects from a different experimental angle of senotherapy effects on brain and behavior rather than cardioprotective effects of dasatinib / quercetin (a senolytic combination) and sulforaphane (senomorphic):

“This is the first study to analyze the effect of senotherapy in the brain of a model of chronic obesity in middle-aged female rats. D + Q reduced the pro-inflammatory cytokines evaluated in the obesity model. It did not improve memory and learning nor the expression of molecules associated with the maintenance of synapses.

In contrast, sulforaphane (SFN), which without eliminating senescent cells, decreased pro-inflammatory factors, increased IL-10, as well as brain-derived neurotrophic factor BDNF, synaptophysin (SYP), and postsynaptic density protein 95 (PSD-95), which, in turn, were associated with an improvement in behavioral tests in obese rats. This suggests that modulating the senescence-associated secretory phenotype (SASP), rather than eliminating senescent cells, might have better effects.”

https://www.sciencedirect.com/science/article/pii/S0014488625001955 “Senotherapy as a multitarget intervention in chronic obesity: Modulation of senescence, neuroinflammation, dysbiosis, and synaptic integrity in middle-aged female Wistar rats”


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.

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”


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”

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


Too dangerous to investigate?

This blog’s 1100th curation is a clinical trial of ergothioneine’s effects on cognitive decline:

“We recruited participants aged between 60–90 years of age, from three study cohorts diagnosed with mild cognitive impairment (MCI) and provided them with ergothioneine (ET)  (25 mg capsules administered orally three times a week) or placebo in a double-blinded and randomized manner. Blood samples were collected at baseline and quarterly (visits 1, 4, 7, 10, 14) for clinical safety assessment and biomarker analyses). Neuro-cognitive assessments were conducted biannually (visits 7 and 14).

Following ET intake, an increase in Z-scores was observed in the Rey Auditory Verbal Learning Test (RAVLT) (immediate and delayed recalls), which evaluates learning ability and memory.

ravlt

wbc

Participants in both ET and placebo groups recorded a lower total white blood cell count compared to baseline at visit 7, both of which recovered subsequently. The reasons for this anomaly are unclear but values were all still within the expected range for their age.”

https://journals.sagepub.com/doi/epub/10.1177/13872877241291253 “Investigating the efficacy of ergothioneine to delay cognitive decline in mild cognitively impaired subjects: A pilot study”


I rated this study a waste of time and money for the researchers’ incurious lack of following where their data led. Significant WBC signals of both treatment and placebo subjects’ immune system responses were shrugged off with an “expected range” non-explanation.

Polyphenol Nrf2 activators

Two 2024 reviews by the same group that published Sulforaphane in the Goldilocks zone investigated dietary polyphenols’ effects as “hormetic nutrients”:

“Polyphenols display biphasic dose–response effects by activating at a low dose the Nrf2 pathway resulting in the upregulation of antioxidant vitagenes [see diagram]. We aimed to discuss hormetic nutrients, including polyphenols and/or probiotics, targeting the Nrf2 pathway and vitagenes for the development of promising neuroprotective and therapeutic strategies to suppress oxidative stress, inflammation and microbiota deregulation, and consequently improve cognitive performance and brain health.

antioxidants-13-00484-g001

Hormetic nutrition through polyphenols and/or probiotics targeting the antioxidant Nrf2 pathway and stress resilient vitagenes to inhibit oxidative stress and inflammatory pathways, as well as ferroptosis, could represent an effective therapy to manipulate alterations in the gut microbiome leading to brain dysfunction in order to prevent or slow the onset of major cognitive disorders. Notably, hormetic nutrients can stimulate the vagus nerve as a means of directly modulating microbiota-brain interactions for therapeutic purposes to mitigate or reverse the pathophysiological process, restoring gut and brain homeostasis, as reported by extensive preclinical and clinical studies.”

https://www.mdpi.com/2076-3921/13/4/484 “Hormetic Nutrition and Redox Regulation in Gut–Brain Axis Disorders”


I’m not onboard with this study’s probiotic assertions because most of the cited studies contained unacknowledged measurement errors. Measuring gut microbiota, Part 2 found:

“The fecal microbiome does not represent the overall composition of the gut microbiome. Despite significant roles of gut microbiome in various phenotypes and diseases of its host, causative microbes for such characteristics identified by one research fail to be reproduced in others.

Since fecal microbiome is a result of the gut microbiome rather than the representative microbiome of the GI tract of the host, there is a limitation in identifying causative intestinal microbes related to these phenotypes and diseases by studying fecal microbiome.”

These researchers also erroneously equated isothiocyanate sulforaphane’s Nrf2-activating mechanisms with polyphenols activating Nrf2.


This research group did better in clarifying polyphenols’ mechanisms in a review of hormetic dose-response effects of the polyphenol rosmarinic acid:

“This article evaluates whether rosmarinic acid may act as a hormetic agent, mediating its chemoprotective effects as has been shown for similar agents, such as caffeic acid, a derivative of rosmarinic acid.

Rosmarinic acid enhanced memory in institute of cancer research male mice in the Morris water maze (escape latency).

untitled

Of importance in the evaluation of rosmarinic acid are its bioavailability, metabolism, and tissue distribution (including the capacity to affect and/or cross the BBB and its distribution and half-life within the brain). In the case of polyphenols, including rosmarinic acid, they are typically delivered at low doses in the diet and, in most instances, they do not escape first-pass metabolism, with the prominent chemical forms being conjugates of glucuronides and sulfates, with or without methylation.

These conjugated metabolites are chemically distinct from the parent compound, showing considerable differences in size, polarity, and ionic form. Their biological actions are quite different from the parent compound.

Bioavailability studies reveal that maximum concentrations in plasma typically do not exceed 1 µM following consumption of 10–100 mg of a single phenolic compound, with the maximum concentration occurring typically less than 2 h after ingestion, then dropping quickly thereafter. In the case of the in vitro studies assessed herein, and with few exceptions, most of the studies employed concentrations >10 µM with some studies involving concentrations in the several hundred µM range, with the duration of exposure typically in the range of 24–72 h, far longer duration than the very short time interval of a few minutes to several hours in human in vivo situations.

We strongly recommend that all experiments using in vitro models to study biological responses to dietary polyphenols use only physiologically relevant flavonoids and their conjugates at appropriate concentrations, provide evidence to support their use, and justify any conclusions generated. When authors fail to do this, referees and editors must act to ensure that data obtained in vitro are relevant to what might occur in vivo.”

https://www.degruyter.com/document/doi/10.1515/med-2024-1065/html “The chemoprotective hormetic effects of rosmarinic acid”

An elevator pitch for plasmalogen precursors

An excerpt from the latest video at Dr. Goodenowe’s Health Matters podcast, Episode 7 “The Truth about Parkinson’s”, starting at 50:30:

“What’s exciting about this community medicine focus that we’ve switched to which basically says: How do we develop technologies in a way that they can be incorporated into a community model versus a pharmaceutical drug model? People can actually do I would say self-experiment just the way you self-experiment with your own diet because these are fundamentally dietary nutrition molecules.

Could you give me an elevator pitch because there are probably people listening who are thinking what is this plasmalogen precursor and for sure how is it having this dramatic effect?

Plasmalogens are the most important nutrient that nobody knows about. Normally you don’t know about it because the body is usually pretty good at making them. What makes plasmalogens unique is that your body makes them kind of like cannon fodder, the first group of people that go into war. Your body throws them out for destruction. They absorb oxidative stress and get destroyed in the process.

They’re stored in your cell membranes. 50% of the membranes of your heart are these plasmalogen molecules. When your heart gets inflamed, what your heart does is it dumps these plasmalogens out of its membranes to douse the flame of inflammation. After inflammation is under control, your body naturally builds these things back up again.

But if you have an inability to make enough plasmalogens, these inflammation events knock you down and keep you down. So plasmalogen precursors are critical for maintaining high levels of plasmalogens across your body, not just in your brain (30% of the lipids in your brain) but in your heart, your lungs, your kidneys.”


PXL_20241117_185248742~2

Brain restoration with plasmalogens, Part 2

This September 2024 presentation adds data points and concepts to Part 1:

supplementation

  1. “Your brain is dynamically connected to and adaptively responsive to its environment.
  2. You are in control of this environment (nutrition, stimulation, adversity).
  3. Need to measure the environment (lab testing, physiology) and adaptive response to the environment (MRI) to optimize your environment (nutrition, lifestyle) to achieve optimal brain structure, function, health, and longevity.

neurovascular

From a global cortical volume and thickness perspective, 17 months of high dose plasmalogens reversed about 15 years of predicted brain deterioration. 31 months reversed almost 20 years. So you can get more out of life.”

https://drgoodenowe.com/immortal-neurology-building-maintaining-an-immortal-brain/


Dr. Goodenowe also added case studies of two patients:

1. A 50-year-old woman with MS who had been legally blind in one eye for 32 years who regained sight in that eye after eight months of supplementation.

“This is the adaptability of the human brain. Her eye is not actually impaired. What’s impaired is the ability, the adaptability of the brain to the signal of light, to actually start interpreting what that light signal is.”

2. A 61-year-old man with dementia from firefighting work for the U.S. Navy in a toxic environment with head injuries after nine months of supplementation.

“The brain can heal itself is the point of the story. His executive function skills in everyday life are getting better.”

Activate Nrf2 to reduce biological age

A 2024 primate study investigated effects of an off-patent drug on age-related changes:

“We evaluated geroprotective effects of metformin on adult male cynomolgus monkeys. The study encompassed a comprehensive suite of physiological, imaging, histological, and molecular evaluations, substantiating metformin’s influence on delaying age-related phenotypes at the organismal level.

monkey nrf2

Results highlighted a significant slowing of aging indicators, notably a roughly 6-year regression in brain aging. Metformin exerts a substantial neuroprotective effect, preserving brain structure and enhancing cognitive ability.

Geroprotective effects on primate neurons were partially mediated by activation of Nrf2, a transcription factor with anti-oxidative capabilities.”

https://www.cell.com/cell/abstract/S0092-8674(24)00914-0 “Metformin decelerates aging clock in male monkeys” (not freely available). Thanks to Dr. Pradeep Reddy for providing a copy.


From this study’s Nrf2 activation findings:

“Metformin treatment resulted in increased nuclear phosphorylated Nrf2, accompanied by up-regulation of Nrf2 target genes like HO-1, NQO-1, SOD3, GPX2, and GPX1, which were generally suppressed and typically down-regulated during human neuron senescence.

Genes pivotal for neuronal function, such as dendrite morphogenesis/extension and synapse assembly (e.g., GSK3B, GRID2, and NRG3), were down-regulated during aging in excitatory neurons (ExN), inhibitory neurons (InN), oligodendrocytes (OL), oligodendrocyte progenitor cells (OPC), microglia, and astrocyte but were restored by metformin treatment. By contrast, pathways that were up-regulated during aging, including activation of the immune response, complement activation, and regulation of the TGF-b receptor signaling pathway, were reset to lower levels by metformin treatment.

metformin neuronal gene pathways

We verified that markers associated with brain aging and progression of neurodegenerative diseases were restored by metformin treatment to levels similar to those observed in young monkeys. Additionally, we observed that reduced myelin sheath thickness, a characteristic of aged monkeys, was rebuilt to a younger state following metformin treatment.

These findings align with the levels of nuclear-localized phosphorylated Nrf2, suggesting that Nrf2 pathway activation is a key mechanism in metformin’s role in delaying human neuronal aging and, by extension, brain aging. Consistent with our in vitro findings, Nrf2 pathway activation was also detected across multiple tissues in metformin-treated monkeys, including frontal lobe neurons.


At last count, I’ve curated 250+ papers this decade on cruciferous vegetables, and many of these explored relationships with Nrf2 activation. Basically, eating a clinically-relevant daily dose of 3-day-old cruciferous sprouts and taking off-patent metformin both induce Nrf2 activation effects.

Don’t expect to see many researchers highlighting this equivalency. They’d rather wait another decade to nitpick other studies with not-enough-subjects / not-exactly replicated / other nitpicks before expressing opinions urging caution from their nursing home beds.

But even then, they won’t get their facts straight. For example, a contemporaneous opinion article https://www.nature.com/articles/d41586-024-02938-w “The brain aged more slowly in monkeys given a cheap diabetes drug” attempted to summarize this study, and flubbed two points:

1. The study said: “We conducted a proof-of-concept study involving male cynomolgus monkeys (Macaca fascicularis) aged between 13 and 16 years, roughly equivalent to approximately 40–50 years in humans. Monkeys adhered to this regimen for a period of 1,200 days, approximately 3.3 years, which corresponds to about 10 years in humans.”

The opinion claimed: “Animals took the drug for 40 months, which is equivalent to about 13 years for humans.”

2. The opinion quoted a New York City researcher involved in a separate metformin study and employed at a medical school for:

“Research into metformin and other anti-ageing candidates could one day mean that doctors will be able to focus more on keeping people healthy for as long as possible rather than on treating diseases.”

This statement is a big break from the realities of medical personnel daily actions at least so far this decade, which is when I started to pay close attention:

  • Doctors have very little diet and exercise training in medical school. There’s no way they can give health advice. There’s no way that a “keeping people healthy” paradigm will emerge from the current medical system.
  • Fixing a disease doesn’t restore a patient’s health. Dr. (PhD) Goodenowe cites several examples in his talks, such as a study that compared colorectal cancer therapy with post-operation patient health.
  • If you listen to yesterday’s two-hour-long podcast, the currently injured person in the first hour gave plenty of contrary evidence of doctors’ focuses: behaviors of trying to blame and gaslight the patient, thinly-disguised punitive actions, CYA etc., all of which they will be sued for one day. The doctor in the second hour provided an example of the quoted researcher in her explanation of how doctors higher in the hierarchy either can’t see or can’t admit realities of doctor/patient interactions, and what therapies have actually benefited or harmed a patient.

Consequences of perinatal stress

A 2024 rodent study followed up earlier studies of perinatal stress:

“Stress is a multisystemic and multiscale reaction experienced by living beings in response to a wide range of stimuli, encompassing a highly complex order of biological and behavioral responses in mammals, including humans. In the present study, we evaluated changes in mRNA levels in 88 regions of interest (ROIs) in male rats both exposed to perinatal stress and not exposed.

Depending on critical life stage (e.g., perinatal life, infancy, childhood, adolescence, aging), duration, and type of stressor, different effects can be detected by examining behavioral and physiological functions. Stress is related to several cognitive processes, including spatial and declarative memory (involving the hippocampus), fear and memories of emotionally charged events (involving the amygdala), and executive functions and fear extinction (involving the prefrontal cortex).

This PRS paradigm is a well-characterized animal model in which offspring is exposed to stress during pregnancy and after birth because of receiving defective maternal care. Offspring exhibit behavioral hyperreactivity, as well as increased susceptibility to drug addiction and decreased risk-taking behavior.

Starting from day 11 of gestation until delivery, pregnant females were subjected to restraint in a transparent plastic cylinder and exposed to bright light during three daily sessions of 45 min. Since gestational stress induces a <40% reduction of maternal behavior in stressed mothers, we refer to the whole procedure as Perinatal Stress.

Intercorrelation between the orbitofrontal cortex (OFC) and various brain regions such as the thalamus and amygdala were found disrupted in the PRS group. These functional correlations appear to be associated with regulation of executive functions, goal-directed behavior, and directed attention. Also, discrete functional links between the OFC and limbic regions and striatum were lost in the PRS group.

Decreased expression of the Homer1a gene across multiple brain regions after perinatal stress exposure may derange normal architecture of glutamatergic synapses during neurodevelopment and after birth. Changes at the glutamatergic synapse have been considered pivotal in adaptive stress behaviors.

Our results show that PRS preferentially reinforces the centrality of subcortical nodes, resulting in increased centrality of structures such as amygdala, caudate-putamen, and nucleus accumbens, suggestive of reduced cortical control over these regions. In conclusion, when analyzing Homer gene expression after stress exposure not only in terms of quantitative changes compared to the control group, but also as a basis for conducting brain connectivity graph analysis, we observed that perinatal stress could significantly affect the functional connectivity of brain regions implicated in modeling pathophysiology of severe psychiatric disorders.”

https://www.sciencedirect.com/science/article/pii/S0278584624001003 “Perinatal stress modulates glutamatergic functional connectivity: A post-synaptic density immediate early gene-based network analysis”


PXL_20240528_094419674

Maintaining your myelin, Part 2

Continuing Part 1 with three 2024 preprint studies, starting with an investigation of neuroinflammation in high school athletes:

“Axons are long fibers conducting nerve impulses from nerve cells to synaptic ends. Like electric wires, axons are insulated by the myelin sheath produced by oligodendrocytes (ODC) in the brain or Schwann cells in the periphery. The myelin sheath is vulnerable to mechanical stresses after head injuries, as well as targets for autoimmune attack in multiple sclerosis and degeneration in various white matter diseases.

145850ce6289d06e5318d35f

It is challenging to definitively validate axonal neuroinflammation, because axonal neuroinflammation is only diagnosed at post-mortem autopsy, or wait for more than a decade to potentially witness progression to chronic traumatic encephalopathy, or white matter dementia. Advanced imaging analysis of computed tomography and magnetic resonance imaging are not sensitive enough to identify such microscopic abnormalities.

We developed a sandwich immunoassay detecting dual signals of myelin oligodendrocyte glycoprotein (MOG) and interleukin 1B (IL1B) in human plasma, [IL1B on MOG]. MOG is a transmembrane protein specifically expressed in ODC and Schwann cells membranes, and doesn’t freely exist in plasma. We found that serum from capillary blood is acceptable, and we tested control and athlete samples using only 5 mL samples. When we tested 63 control plasma samples, values were widely distributed over 2 logs, so we focused on longitudinal studies.

Damaged neurons are not easily detectable using conventional physical examinations, because the brain’s inherent adaptability allows it to compensate for localized damage by finding alternate routes. While this adaptability is advantageous, it also means that these concealed lesions can go unnoticed, potentially leading to future complications.

Elevation of [IL1B on MOG] was seen in some athletes who did not show concussion or traumatic brain injury (TBI). While the occurrence of concussion is relatively limited, potential prevalence of subconcussion or subconcussive condition is expected to be substantially higher.

If [IL1B on MOG] levels remain unchanged during this early post-concussion period (2-4 weeks), it may suggest that neuroinflammation has not been induced, potentially providing reassurance for the athletes to return to play. Conversely, if [IL1B on MOG] levels increase within this timeframe, it may indicate the need for intervention or closer monitoring. Thus, there is compelling potential for incorporating this test into concussion guidelines.”

https://www.researchsquare.com/article/rs-3997676/v1 “An approach for the analysis of axonal neuroinflammation by measuring dual biomarkers of oligodendrocytes and inflammatory cytokine in human plasma”


A rodent study investigated the immune system’s influence on oligodendrocyte lineage cells after TBI:

“White matter injury is thought to be a major contributor to long-term cognitive dysfunctions after TBI. This damage occurs partly due to apoptotic death of oligodendrocyte lineage cells (OLCs) after injury, triggered directly by the trauma or in response to degenerating axons.

Our data indicates that depletion of the gut microbiota after TBI impaired remyelination, reduced OLCs proliferation, and required the presence of T cells. This suggests that T cells are an important mechanistic link by which the gut microbiota modulate oligodendrocyte response and white matter recovery after TBI.

Our findings suggest that oligodendrocytes are not passive in the neuroinflammatory and degenerative environment caused by brain trauma, but instead could exert an active role in modulation of immune response.”

https://www.researchsquare.com/article/rs-4289147/v1 “Gut Microbiota Shape Oligodendrocyte Response after Traumatic Brain Injury”


A rodent study investigated whether oligodendrocyte precursor cells had myelination-independent roles in brain aging:

“OPCs, the source cells of myelin-forming cells in the central nervous system, have been linked to brain aging by their compromised differentiation and regeneration capability. Our results demonstrate that macroautophagy influx declines in aged OPCs, which results in the accumulation of senescent OPCs in aged brains. Senescent OPCs impair neuronal plasticity and exacerbate neurodegeneration, eventually leading to cognitive decline.

Inactivation of autophagy in OPCs exhibits a limited effect on myelin thickness but a loss of myelin in middle-aged mice. The loss of myelin observed is an adaptational change to suppressed neuronal plasticity. However, neither the number of OLs nor oligodendrogenesis is altered by inactivation of autophagy in adult OPCs.

The present study indicates that the intervention of senescent OPCs is an additional promising therapeutic strategy for aging and aging-related cognitive deficits. Autophagy regulates senescence by impairing protein turnover, mitochondrial homeostasis, oxidative stress, and maintaining senescence-associated secretory phenotype. Further investigation remains on whether autophagy in OPCs shares the exact mechanism to promote senescence as that in other types of cells.

Considering autophagy declines with aging, our study brings a novel mechanism in brain aging. Declined autophagy causes senescence of OPCs, which impairs neuronal plasticity and exacerbates neurodegeneration via CCL3/5-CCR5 signaling.”

https://www.researchsquare.com/article/rs-3926942/v1 “Impaired Macroautophagy in Oligodendrocyte Precursor Cells Exacerbates Aging-related Cognitive Deficits via a Senescence Associated Signaling”


PXL_20240418_104114528.MP

Ergothioneine dosing, Part 2

Continuing Part 1 with a 2024 rodent healthspan and lifespan study:

“We investigated the effects of daily oral supplementation of ergothioneine (ERGO) dissolved in drinking water on lifespan, frailty, and cognitive impairment in male mice from 7 weeks of age to the end of their lives. Ingestion of 4 ~ 5 mg/kg/day of ERGO remarkably extended the lifespan of male mice.

11357_2024_1111_Fig1_HTML

The ERGO group showed significantly lower age-related declines in weight, fat mass, and average and maximum movement velocities at 88 weeks of age. This was compatible with dramatic suppression by ERGO of age-related increments in plasma biomarkers. ERGO also rescued age-related impairments in learning and memory ability.

Ingestion of ERGO may promote longevity and healthy aging in male mice, possibly through multiple biological mechanisms.”

https://link.springer.com/article/10.1007/s11357-024-01111-5 “Ergothioneine promotes longevity and healthy aging in male mice”

Subjects’ plasma ergothioneine levels of an estimated 4 ~ 5 mg/kg daily dose were:

11357_2024_1111_Fig3_HTML

A human equivalent daily dose is an estimated 22 mg to 28 mg (4 or 5 mg x .081 x 70 kg).

The third paper in Part 1 cited a 2017 clinical trial that provided 5 mg and 25 mg ergothioneine doses for 7 days, resulting in these plasma ergothioneine levels:

figure 3

The first paper of Part 1 referenced a 2020 human study where the dose was 5 mg/day for 12 weeks, but I don’t have access to it. It’s unclear whether humans could continually raise ergothioneine levels by daily consumption throughout our lives as did this rodent study.


A 2024 paper reviewed the importance of ergothioneine to humans:

“We propose that the diet-derived compound ergothioneine (ET) is an important nutrient in the human body, especially for maintenance of normal brain function, and that low body ET levels predispose humans to significantly increased risks of neurodegenerative and possibly other age-related diseases.

Work by multiple groups has established that low ET levels in humans are associated not only with cognitive impairment/AD but also with other age-related conditions, including frailty, Parkinson’s disease, vascular dementia, chronic renal disease, cardiovascular disease, and macular degeneration. Low ET levels also correlate with increased risk of developing preeclampsia in pregnant women [53].

Plasma ET levels from healthy (age-matched) vs unhealthy individuals in Singapore – Mild cognitive impairment (MCI); Alzheimer’s disease (AD); vascular dementia (VaD); Parkinson’s disease (PD); age-related macular degeneration (AMD):

1-s2.0-S0891584924001357-gr2_lrg

  • Does low ET cause or contribute to age-related neurodegeneration, or
  • Does disease cause low ET, or
  • Low ET and increased disease risk are both caused by something else, as yet unidentified?

Prevention of neurodegeneration is especially important, since by the time dementia is usually diagnosed damage to the brain is extensive and likely irreversible.”

https://www.sciencedirect.com/science/article/pii/S0891584924001357 “Are age-related neurodegenerative diseases caused by a lack of the diet-derived compound ergothioneine?”

Whether or not the healthy individuals ate mushrooms daily in the above graphic was lost while conglomerating multiple studies.

Note that scales of the above two human graphics are a thousand times smaller than the above rodent graphic. I thought that maybe the rodent study made a plasma ergothioneine calculation error, but didn’t see one in the provided Supplementary data.


Reference 53 of the second paper is a 2023 human study:

“We analysed early pregnancy samples from a cohort of 432 first time mothers. Of these 432 women, 97 went on to develop pre-term or term pre-eclampsia (PE).

If a threshold was set at the 90th percentile of the reference range in the control population (≥462 ng/ml), only one of these 97 women (1%) developed PE, versus 96/397 (24.2%) whose ergothioneine level was below this threshold. One possible interpretation of these findings, consistent with previous experiments in a reduced uterine perfusion model in rats, is that ergothioneine may indeed prove protective against PE in humans.”

https://portlandpress.com/bioscirep/article/43/7/BSR20230160/233119/Relationship-between-the-concentration-of “Relationship between the concentration of ergothioneine in plasma and the likelihood of developing pre-eclampsia”

Eyeballing the Healthy individuals in the above graphic, none of those 544 people were below this study’s 462 ng threshold.


A 2023 companion article analyzed the third paper’s unusual findings:

“These results suggest that there might be a dichotomized association between ergothioneine concentrations and preeclampsia; and only a high ergothioneine level over 90th percentile of the control population could be protective against preeclampsia.

Univariable results showed that ergothioneine had a significant non-linear association with preeclampsia and it would start to offer protective effect from 300 ng/ml onward. Analysis also confirmed that body mass index was significantly associated with an increased risk of preeclampsia.

A large observational study could strengthen the causal association between ergothioneine and preeclampsia. If confirmed, a randomized controlled trial (RCT) assessing whether ergothioneine supplementation can reduce risk of preeclampsia will be imminently feasible. Ideally, such RCT should compare placebo with a range of different doses of ergothioneine to identify the best or minimal effective dose, given its good safety records, including in pregnancy, with a no-observed-adverse-effect level (NOAEL) of 800 mg/kg body weight per day.”

https://portlandpress.com/bioscirep/article/43/8/BSR20231076/233395/Dose-related-relationship-between-ergothioneine “Dose-related relationship between ergothioneine concentrations and risk of preeclampsia”

My daily mushroom ergothioneine dose is around 7 mg, and I weigh about 70 kg. I don’t think a daily 800 mg/kg ergothioneine dose would be desirable for anybody, regardless of what experts say.

How many times have public health employees been wrong this decade? Would you bet your or your child’s health on their advice?


PXL_20240316_182330822