Plasmalogens Week #2 – Childhood Development

Continuing Plasmalogens Week with three 2025 papers, starting with a human study of plasmalogens’ effects of decreasing breastfed infants’ infections and inflammation:

“Mothers reported on breastfeeding and infant infections in questionnaires collected at 1 month, 3 months, 6 months, 12 months, and 18 months post-birth. Parent-reported infection burden was defined as the total number of infant respiratory tract infections, gastroenteritis, conjunctivitis, and acute otitis media episodes reported by mothers between birth and 6 months for 6-month analyses, and between birth and 12 months for 12-month analyses.

We constructed a causal mediation model to estimate the proportion of effects explained by a direct effect of breastfeeding on inflammation, measured via glycoprotein acetyls (GlycA)—the average direct effect (ADE)—and the proportion that was mediated by metabolomic biomarkers/lipid—the average causal mediation effect (ACME).

Breastfeeding is negatively associated with GlycA, positively associated with plasmalogens, and plasmalogens are negatively associated with GlycA. However, the positive association between breastfeeding and plasmalogens is stronger than the negative direct association between breastfeeding and inflammation, resulting in an ACME that exceeds the total effect. This pattern indicates that plasmalogens may play a dominant role in mediating the relationship between breastfeeding and systemic inflammation.

We have recently developed a plasmalogen score that is associated with a range of cardiometabolic outcomes, including type 2 diabetes and CVD.

  • At 6 months, the plasmalogen score was estimated to mediate 162% of the total effect (proportion mediated: 1.62, i.e. average causal mediation effect (ACME) to total effect ratio of 1.62, resulting in a percentage > 100%) of breastfeeding on GlycA.
  • At 12 months, the plasmalogen score mediated an estimated 75% of the total effect of breastfeeding on GlycA.

Any breastfeeding, regardless of supplementary feeding, was associated with lower inflammation, fewer infections, and significant, potentially beneficial changes in metabolomic and lipidomic markers, particularly plasmalogens. There was evidence of bidirectional mediation: metabolomic biomarkers and lipids mediated breastfeeding’s effects on inflammation, while inflammation partly mediated breastfeeding’s impact on certain metabolites and lipids.”

https://bmcmedicine.biomedcentral.com/articles/10.1186/s12916-025-04343-0 “The protective effect of breastfeeding on infant inflammation: a mediation analysis of the plasma lipidome and metabolome”

Reference 48 was the 2024 plasmalogen score study.


A second study by many of the first study’s researchers used the same cohort as the first study to investigate effects of maternal obesity on infant obesity:

“We aimed to investigate associations between maternal pre-pregnancy body mass index (pp-BMI), lipidomic profiles of mothers, human milk, and infants, and early life growth. We were particularly interested in ether lipids as they are higher in breastfed infants compared to formula-fed infants, are enriched in human milk compared to infant formula, and are involved in metabolic health and inflammation in adult populations.

Maternal plasmalogen score was negatively associated with pp-BMI and positively associated with plasmalogens in human milk and infant plasmalogen scores from birth to four years of age. We were unable to establish clear links between plasmalogen score and infant BMI within the first 4 years.

These findings position plasmalogens and ether lipids as potential biomarkers or intervention targets for reducing transmission of obesity from mother to infant. Optimising lipid profiles through reducing maternal pp-BMI and dietary or supplemental ether lipids may represent a novel strategy for mitigating early-life obesity risk.”

https://www.researchsquare.com/article/rs-7089146/v1 “Maternal BMI and infant obesity risk: a lipidomics perspective on the developmental origins of obesity”

There was a lot of hand waving and weasel-wording (i.e., could, may, potential, associated with) but little causal evidence in this preprint. Reference 42 was the preprint version of the first study.


A third paper investigated 9- to 12-year-olds’ plasmalogen levels and molecular types:

“The importance of plasmalogens (Pls) in several cellular processes is known, one of which is their protective effect against oxidative damage. The physiological role of Pls in human development has not been elucidated. This study is the first report on plasmalogen levels and molecular types in children’s plasma.

Ethanolamine plasmalogen (PlsEtn 16:0/20:5) and choline plasmalogen (PlsCho 16:0/20:5), both carrying eicosapentaenoic acid (EPA, ω-3), were significantly lower in girls than in boys. There was no significant difference observed among the 9, 10, 11, and 12-year-old groups between girls and boys in their levels of PlsEtn 16:0/20:5. However, a significant decrease in the levels of PlsCho 16:0/20:5 was observed for 9, 10 and 12-year-old groups of girls compared to boys.

  • In both sexes, the plasmalogen levels for the 12-year-old children were lower than those for the 9-year-old children.
  • PlsCho (16:0/18:2) linoleic acid (ω-6)-derived was lower in the overweight children than in the normal-weight children for both sexes.
  • Arachidonic acid (ω-6)-containing PlsEtn (18:0/20:4) was the most abundant ethanolamine-type plasmalogen in both sexes.

This study has many limitations as follows:

  1. Non-fasting plasma samples were collected from the children’s plasma and used for analysis; since diet can influence Pls levels, the result may be affected by the sample collection method.
  2. Physical activity was also not monitored, which could have an influence on plasma levels, and
  3. A limited number of plasmalogen molecular species were quantified in this study.

A follow-up study may be essential to determine the plasma Pls in the same population when they are adolescents.”

https://www.mdpi.com/2075-4418/15/6/743 “Application of Liquid Chromatography/Tandem Mass Spectrometry for Quantitative Analysis of Plasmalogens in Preadolescent Children—The Hokkaido Study”


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”


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”

A sulforaphane review

Here’s a 2025 review where the lead author is a retired researcher whose words readers might interpret as Science. As a reminder, unlike study researchers, reviewers are free to:

  • Express their beliefs as facts;
  • Over/under emphasize study limitations; and
  • Disregard and misrepresent evidence as they see fit.

Reviewers also aren’t obligated to make post-publication corrections for their errors and distortions. For examples:

1. After the 7. Conclusions section, there’s an 8. Afterword: I3C and DIM section. The phrase “As detailed in our earliest work on broccoli sprouts..” indicated a belief carried over from last century of the low importance of those research subjects.

Then, contrary to uncited clinical trials such as Our model clinical trial for Changing to a youthful phenotype with broccoli sprouts and Eat broccoli sprouts for DIM, “Broccoli sprouts had next to no indole glucosinolates.” And in the middle of downplaying I3C and DIM research, they stated: “There are 149 clinical studies on DIM and 11 on I3C listed on clinicaltrials.gov, suggesting a good safety profile. Potential efficacy and mode of action in humans are a subject of intense current investigation, though definitive answers will not come for some time.” 🧐

2. In the 3. Sulforaphane section, they asserted: “Glucosinolates such as glucoraphanin are ‘activated’ or converted to isothiocyanates such as sulforaphane by an enzyme called myrosinase, which is present in that same plant tissue (e.g., seed, sprout, broccoli head, or microgreen) and/or in bacteria that all humans possess in their gastrointestinal tracts.” and cited a 2016 book they coauthored that I can’t access.

The first 2021 paper of Broccoli sprout compounds and gut microbiota didn’t assert that “all humans” had certain gut microbiota that converted glucosinolates to isothiocyanates. That paper instead stated: “Human feeding trials have shown inter-individual variations in gut microbiome composition coincides with variations in ITC absorption and excretion, and some bacteria produce ITCs from glucosinolates.”

3. Nearly half of their cited references were in vitro cancer papers. I rarely curate those types of studies because of their undisclosed human-irrelevant factors. For example, from the second paper of Polyphenol Nrf2 activators:

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.

applsci-15-00522-g001-550

https://www.mdpi.com/2076-3417/15/2/522 “The Impact of Sulforaphane on Sex-Specific Conditions and Hormone Balance: A Comprehensive Review”

Failed aging paradigms

A 2024 paper with 81 coauthors presented different views of aging:

“This article highlights the lack of consensus among aging researchers on fundamental questions such as the definition, causes, and onset of aging as well as the nature of rejuvenation. Our survey revealed broad disagreement and no majority opinion on these issues.

We obtained 103 responses (∼20% of which were submitted anonymously). The respondents included 29.8% professors, 25% postdoctoral fellows, 22.1% graduate students, 13.5% industry professionals, and 9.6% representing other categories (a total of eight additional groups).

When does aging begin? At 20 years (22%), gastrulation (18%), conception (16.5%), gametogenesis (13%), 25 years (11%), birth (8%), 13 years (5%), and 9 years (4%). Nobody chose the only remaining option (30 years).

m_pgae499f3

It is clear from responses that aging remains an unsolved problem in biology. While most scientists think they understand the nature of aging, apparently their understanding differs. Where some may stress the importance of targeting underlying mechanisms, others focus on ameliorating the phenotypes.”

https://academic.oup.com/pnasnexus/article/3/12/pgae499/7913315?login=false “Disagreement on foundational principles of biological aging”


I’ll assert that these researchers were unable to incorporate information outside of their chosen paradigm. This would explain why only 18% understood the embryonic stage of gastrulation as aging’s start, although the 2022 paper Epigenetic profiling and incidence of disrupted development point to gastrulation as aging ground zero in Xenopus laevis provided epigenetic clock evidence that:

“It is not birth, marriage, or death, but gastrulation which is truly the most important time in your life.”


I’ve cited Josh Mitteldorf’s work about aging a few times. His paradigm of aging is in his 2017 book Cracking the Aging Code: The New Science of Growing Old – And What It Means for Staying Young that:

“Aging has an evolutionary purpose: to stabilize populations and ecosystems.”

However, there isn’t evidence of such causal inheritance mechanisms that would begin an organism’s aging during embryogenesis, i.e., that an embryo’s development of aging elements at gastrulation is causally affected by population and ecosystem factors.


Dr. Goodenowe recently had a casual conversation Episode 8 – Perpetual Health, Exploring The Science Behind Immortality where he asserted items such as:

“What we’re all fighting is entropy. Entropy is the tendency of all things to reach a level of randomness. Aging is not a disease. It’s just apathy and entropy. The body just doesn’t care – people don’t pay attention.

This notion that we are programmed for death is wrong. We’re not programmed to die. We actually teach ourselves to die. The body learns how to die, so as your function decreases, it adjusts. It appears to be programmed because of the association with chronological age.”

I haven’t seen any of his papers that put these and his other assertions up for review. For example, I doubt the entropy-caused randomness assertion would survive peer review per Stochastic methylation clocks?:

“Entropic theories of aging have never been coherent, but they are nevertheless experiencing a resurgence in recent years, primarily because neo-Darwinist theories of aging are all failing. I find this ironic, because the neo-Darwinist theories arose precisely because scientists realized that the Second Law of Thermodynamics does not apply to living systems.”


The funny thing about failed aging paradigms is that quite a few of their treatments improve healthspan, but not lifespan. If they don’t “target aging underlying mechanisms” they “ameliorate aging phenotypes.” None so far have positively affected both human healthspan and lifespan.

PXL_20241129_174732711.MP~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.”

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 1

Three papers on myelin and oligodendrocytes, starting with a 2023 review:

“Myelin is the spiral ensheathment of axons by a lipid and cholesterol-rich glial cell membrane that reduces capacitance and increases resistance of the axonal membrane. Axonal myelination speeds up nerve conduction velocity as a function of axon diameter.

While myelination proceeds rapidly after birth in the peripheral nervous system, central myelination is a spatially and temporally more regulated process. Ongoing myelination of the human brain has been documented at up to 40 years of age. This late myelination in the adult cortex is followed by exhaustion of oligodendrocyte precursor cells (OPC) with senescence and a gradual loss of myelin integrity in the aging brain.

The brain is well known for its high energy demands, specifically in gray matter areas. In white matter tracts, energy consumption is lower. Myelination poses a unique challenge for axonal energy generation where myelin sheaths cover more than 95% of the axonal surface areas.

Oligodendrocytes help support axonal integrity. Oligodendrocytes survive well in the absence of mitochondrial oxidative phosphorylation, and without signs of myelin loss, cell death, neurodegeneration or secondary inflammation.

Glycolysis products of oligodendroglial origin are readily metabolized in axonal mitochondria. Oligodendroglial metabolic support is critical for larger and faster-spiking myelinated axons that also have a higher density of mitochondria. An essential requirement for the direct transfer of energy-rich metabolites from oligodendrocytes to the myelinated axonal compartment is ‘myelinic channels’ within the myelin sheath.

Interactions of oligodendrocytes and myelin with the underlying axon are complex and exceed the transfer of energy-rich metabolites. Continuous turnover of myelin membranes by lipid degradation and fatty acid beta-oxidation in mitochondria and peroxisomes leads to recycling of acetate residues by fatty acid synthesis and membrane biogenesis.

1-s2.0-S0959438823001071-gr2_lrg

In human multiple sclerosis (MS) and its animal model myelin oligodendrocyte glycoprotein-experimental autoimmune encephalomyelitis (MOG-EAE), acute inflammatory demyelination is followed by axonal degeneration in lesion sites that is mechanistically not fully understood. It is widely thought that demyelination and the lack of an axon-protective myelin sheath in the presence of numerous inflammatory mediators are the main causes of axon loss.

But unprotected axons improve rather than worsen the overall clinical phenotype of EAE mice which exhibited the same degree of autoimmunity. Thus, ‘bad myelin is worse than no myelin’ because MS-relevant myelin injuries perturb the integrity of myelinic channels and metabolic support.

Dysfunctional or injured oligodendrocytes that do not allow for compensation by any other cell types turn the affected myelin ensheathment into a burden of the underlying axonal energy metabolism, which causes irreversible axon loss. Any loss of myelin integrity, as seen acutely in demyelinating disorders or more gradually in the aging brain, becomes a risk factor for irreversible neurodegeneration.”

https://www.sciencedirect.com/science/article/pii/S0959438823001071 “Expanding the function of oligodendrocytes to brain energy metabolism”


A 2024 review focused on myelin and oligodendrocyte plasticity:

“This review summarizes our current understanding of how myelin is generated, how its function is dynamically regulated, and how oligodendrocytes support the long-term integrity of myelinated axons.

Apart from its unique ultrastructure, there are several other exceptional features of myelin. One is certainly its molecular composition. Another is its extraordinary stability. This was compellingly illustrated when 5000-year-old myelin with almost intact ultrastructure was dissected from a Tyrolean Ice Man.

Myelin is a stable system in contrast to most membranes. However, myelin is compartmentalized into structurally and biochemically distinct domains. Noncompacted regions are much more dynamic and metabolically active than tightly compacted regions that lack direct access to the membrane trafficking machinery of oligodendrocytes.

The underlying molecular basis for stability of myelin is likely its lipid composition with high levels of saturated, long chain fatty acids, together with an enrichment of glycosphingolipids (∼20% molar percentage of total lipids) and cholesterol (∼40% of molar percentage of total lipids). In addition, myelin comprises a high proportion of plasmalogens (ether lipids) with saturated long-chain fatty acids. In fact, ∼20% of the fatty acids in myelin have hydrocarbon chains longer than 18 carbon atoms (∼1% in the gray matter) and only ∼6% of the fatty acids are polyunsaturated (∼20% in gray matter).

With maturation of oligodendrocytes, the plasma membrane undergoes major transformations of its structure. Whereas OPCs are covered by a dense layer of large and negatively charged self-repulsive oligosaccharides, compacted myelin of fully matured oligodendrocytes lacks most of these glycoprotein and complex glycolipids.

Schematic depiction of an oligodendrocyte that takes up blood-derived glucose and delivers glycolysis products (pyruvate/lactate) via monocarboxylate transporters (MCT1 and MCT2) to myelinated axons. Oligodendrocytes and myelin membranes are also coupled by gap junctions to astrocytes, and thus indirectly to the blood–brain barrier.

oligodendrocyte

Adaptive myelination refers to dynamic events in oligodendroglia driven by extrinsic factors such as experience or neuronal activity, which subsequently induces changes in circuit structure and function. Understanding how these adaptive changes in neuron-oligodendroglia interactions impact brain function remains a pressing question for the field.

Transient social isolation during adulthood results in chromatin and myelin changes, but does not induce consequent behavioral alterations. When mice undergo a social isolation paradigm during early life development, they similarly exhibit deficits in prefrontal cortex function and myelination, but these deficiencies do not recover with social reintroduction. This implicates a critical period for social deprivation effects on myelin dynamics. Experience-dependent changes in myelin dynamics may depend on not only the age, brain region, and cell type studied, but also the specific myelin structural change assessed.

Local synaptic neurotransmitter release along an axon not only affects the number of OPCs and oligodendrocytes associated with that axon and local synthesis of myelin proteins, but also drives preferential selection of active axons for myelination over the ensheathment of electrically silenced neighboring axons. Neuronal activity–induced plasticity may preferentially impact brain regions that remain incompletely myelinated compared to more fully myelinated tracts.

Whereas the myelin sheath has been regarded for a long time as an inert insulating structure, it has now become clear that myelin is metabolically active with cytoplasmic-rich pathways, myelinic channels, for movement of macromolecules into the periaxonal space. The myelin sheath and its subjacent axon need to be regarded as one functional unit, which are not only morphological but also metabolically coupled.”

https://cshperspectives.cshlp.org/content/early/2024/04/15/cshperspect.a041359 “Oligodendrocytes: Myelination, Plasticity, and Axonal Support” (not freely available) Thanks to Dr. Klaus-Armin Nave for providing a copy.


A 2024 rodent study investigated oligodendrocyte precursor cell transcriptional and epigenetic changes:

“We used single-cell RNA sequencing (scRNA-seq), single-cell ATAC sequencing (scATAC-seq), and single-cell spatial transcriptomics to characterize murine cortical OPCs throughout postnatal life. One group (active, or actOPCs) is metabolically active and enriched in white matter. The second (homeostatic, or hOPCs) is less active, enriched in gray matter, and predicted to derive from actOPCs. Relative to developing OPCs, both actOPCs and hOPCs are less active metabolically and have less open chromatin.

In adulthood, these two groups are transcriptionally but not epigenetically distinct, indicating that they may represent different states of the same OPC population. If that is the case, then one model is that the parenchymal environment maintains adult OPCs within an hOPC state, whereas those OPCs recruited into white matter or exposed to demyelinated axons may transition toward an actOPC state in preparation for making new oligodendrocytes. We do not yet know the functional ramifications of these differences, but this finding has clear implications for the development of therapeutic strategies for adult remyelination.

opcs

Another finding is that developing but not adult actOPC chromatin is preferentially open for binding motifs associated with neural stem cells, transit-amplifying precursors, and neurogenesis. Although this may simply reflect their origin as the immediate progeny of neonatal neural precursor cells, it may also explain why developing but not adult OPCs have the capacity to make neurons in culture.

If we could, at least in part, reverse the global chromatin shutdown that occurs between development and adulthood, then perhaps adult OPCs may reacquire the ability to make neurons or become better able to generate new oligodendrocytes for remyelination.”

https://www.cell.com/stem-cell-reports/fulltext/S2213-6711(24)00077-8 “Single-cell approaches define two groups of mammalian oligodendrocyte precursor cells and their evolution over developmental time”

Continued in Part 2.


PXL_20240414_103442372

What can be done today to fulfill early unmet needs?

Got agitated earlier this week watching Tucker Carlson’s freely-available interview with a maniac who thinks he’s graduated into a higher state by worshiping the Great AI (Artificial Intelligence, aka Automated Internet, inhabited solely by robots) which will dictate every aspect of what to do with his life. Nevermind that behind the Great AI curtain are the same people who have lied to billions of us, especially during every day of this decade.

Are his current set of beliefs better than previous ones he had of putting a chip into everybody’s brain? What’s wrong with getting to live your own life?

5000

What I saw expressed in the interview was an exhausting pursuit of substitutes for feeling loved. I doubt that many others saw the same, because feeling unloved is so devastating we’ll do anything to avoid it.

But re-experiencing early memories and feelings of unmet needs in a therapeutic setting is the way to keep them from subsequently running our lives. Otherwise, we’ll develop unfulfilling substitutes for what we missed, with misdirected ideas and beliefs accompanied by their unconscious act-outs.

While speaking with a mother who is doing a terrific job of meeting her six-month-old’s needs, I attempted to contrast this interview with the experiences she and her husband are giving their child. Maybe if they read this post, my poor explanation will become clearer.


Wild persimmon trees’ eclipse shadows

PXL_20240408_192336638

Sulforaphane’s effects on autism and liver disease

Here are two more papers that cited Precondition your defenses with broccoli sprouts, starting with a 2024 human / rodent study investigating gut microbiota / sulforaphane’s effects on autism:

“Sulforaphane (SFN) has been found to alleviate complications linked with several diseases by regulating gut microbiota (GM), while the effect of GM on SFN for autism spectrum disorders (ASD) has not been studied. We evaluated therapeutic effects of SFN on maternal immune activation (MIA)-induced ASD-like rat model and pediatric autism patients aged 4–7 years.

OSU-SO for social interactive OSU behavioral subscores, OSU-CO for non-verbal communicative OSU behavioral [significant] subscores, and OSU-ST for repetitive or ritualistic OSU behavioral subscores:

fnut-10-1294057-g0005

Although gut microbiota composition was significantly altered in SFN-treated ASD-like rats, alteration of GM was not evident in ASD patients after 12 weeks of SFN treatment. Limitations in this study:

  1. Studies were conducted in male rats and boys only;
  2. The sample size of our clinical study is relatively small [6 SFN-treated boys] and needs to be further expanded in the future; and
  3. This study only uncovered a potential link between gut flora and the therapeutic effects of SFN on ASD.

SFN treatment alleviates social deficits in MIA-induced ASD-like rats and ASD patients, and improvements might be associated with gut microbiota.”

https://www.frontiersin.org/articles/10.3389/fnut.2023.1294057/full “Therapeutic efficacy of sulforaphane in autism spectrum disorders and its association with gut microbiota: animal model and human longitudinal studies”

The 2022 Efficacy of Sulforaphane in Treatment of Children with Autism Spectrum Disorder: A Randomized Double-Blind Placebo-Controlled Multi-center Trial (not freely available) was referenced for sulforaphane (actually, glucoraphanin with myrosinase enzyme) doses:

“Dosing was weight-based:

  • Two tablets/day for 10–29 lb;
  • Three tablets/day for 30–49 lb;
  • Four tablets/day for 50–69 lb.

An estimated delivery of approximately 24, 36, and 48 μmol of sulforaphane daily was expected in the respective SF dosage groups.”

Weights of the above μmol estimated dose amounts per https://pubchem.ncbi.nlm.nih.gov/compound/sulforaphane are 4.3, 6.4, and 8.5 mg, respectively. An average weight of a 4-year-old boy is 36 lbs / 16.3 kg, and a 7-year-old boy is 51.1 lbs / 23.2 kg.

This study’s maternal immune activation was done by injecting lipopolysaccharide into pregnant rats. Would injecting pregnant women with immune-activating substances have similar harmful effects on the fetus? We don’t have evidence because unbiased and unconflicted studies looking for such effects weren’t sponsored and/or published before immune-activating substances’ deployments.


A 2024 rodent study investigated sulforaphane’s effects on diabetic liver damage:

“We investigated whether sulforaphane, an Nrf2 activator and antioxidant, prevents diabetes-induced hepatic ferroptosis, and the mechanisms involved. Results showed that diabetes-induced inactivation of Nrf2 and decreased expression of its downstream antiferroptotic molecules critical for:

  • Antioxidative defense (catalase, superoxide dismutases, thioredoxin reductase);
  • Iron metabolism (ferritin heavy chain (FTH1), ferroportin 1);
  • Glutathione (GSH) synthesis (cystine-glutamate antiporter system, cystathionase, glutamate-cysteine ligase catalitic subunit, glutamate-cysteine ligase modifier subunit, glutathione synthetase); and
  • GSH recycling – glutathione reductase (GR)

were reversed/increased by sulforaphane treatment.

Diabetes-induced increases in serum glucose and triglyceride levels were also significantly reduced by sulforaphane. Taken together, our results demonstrate a potent effect of SFN in inhibiting ferroptotic death of hepatocytes under diabetic conditions in vivo, thereby alleviating liver injury.

This is the first study to demonstrate the protective role of SFN against ferroptosis in the liver of diabetic mice. This nominates sulforaphane as a promising phytopharmaceutical for the prevention/alleviation of ferroptosis in diabetes-related pathologies.”

https://iubmb.onlinelibrary.wiley.com/doi/10.1002/biof.2042 “Sulforaphane prevents diabetes-induced hepatic ferroptosis by activating Nrf2 signaling axis”

Improving peroxisomal function

A 2024 review provided details about “mysteries” in peroxisome research:

“Peroxisomes are key metabolic organelles with essential functions in cellular lipid metabolism (e.g., β-oxidation of fatty acids and synthesis of ether phospholipids, which contribute to myelin sheath formation), and metabolism of reactive oxygen species (ROS), particularly hydrogen peroxide. Loss of peroxisomal function causes severe metabolic disorders in humans.

Additional non-metabolic roles of peroxisomes have been revealed in cellular stress responses, regulation of cellular redox balance and healthy ageing, pathogen and antiviral defence, and as cellular signalling platforms. New findings also point to a role in regulation of immune responses.

In our previous reviews, we addressed the role of peroxisomes in the brain, in neurological disorders, in development of cancer, and in antiviral defence. To avoid repetition, we refer to those articles where appropriate, and to more specialised recent reviews on peroxisome biology.

418_2023_2259_Fig5

Proper functioning of peroxisomes in metabolism requires the concerted interaction with other subcellular organelles, including the endoplasmic reticulum (ER), mitochondria, lipid droplets, lysosomes, and the cytosol. A striking example of peroxisome-ER metabolic cooperation is de novo biosynthesis of ether phospholipids.

Metabolic activities of peroxisomes, such as ɑ- and β-oxidation of fatty acids, plasmalogen synthesis, and ROS/reactive nitrogen species metabolism, have been linked to numerous immune-related pathways. Roles for peroxisomes in immune and defence mechanisms have opened a new field of peroxisome research, and highlight once more how important peroxisomes are for human health and disease.

It is still not fully understood how peroxisomal functions and abundance are regulated, what kinases/phosphatases are involved, or how peroxisomes are linked to cellular signalling pathways and how they act as signalling platforms.”

https://link.springer.com/article/10.1007/s00418-023-02259-5 “The peroxisome: an update on mysteries 3.0”


Last Friday was Day 90 of a 90-day trial of plasmalogens coincident with improving peroxisomal function via resistance exercise and time-restricted eating. A sticking point has been leg resistance exercises. Ankle issues are interfering with progress, although beach walks aren’t similarly affected. I’m almost back to an upper body exercise routine of five years ago, and I’ve added a half-dozen abs exercises.

I’ll continue taking the two Prodrome plasmalogen precursor supplements (ProdromeGlia and ProdromeNeuro) and with efforts to improve peroxisomal function. Since achieving effective resistance exercise levels is taking longer than expected, and my crystal ball is out-of-commission, I don’t have a realistic end time estimate for stopping the supplements.

Building your plasmalogen savings account

A webinar from earlier this week with Dr. Goodenowe, a clinical trial facilitator, and a physician:

From the Q&A segment:

“Is there a particular age where it’s recommended to test for plasmalogen levels? And what levels would be considered normal?

That’s a good question. That actually raises this whole concept of optimal health and this concept of aging.

The best way to think about it – we talked about this paycheck-to-paycheck situation, where as long as our bills are paid every day, technically we think we’re normal. But we still feel this sense of health anxiety – if you will – like we just don’t know if my car breaks down, or my water heater breaks down, do I have enough money to pay these events in my life?

That’s what health feels like to a lot of people, because they’re just kind of getting by. From a health perspective, they’re considered normal, but they have no reserve capacity, and they have no vitality in terms of health.

Plasmalogens are a type of molecule that you build a savings account of, over years, over decades. Your heart builds them up, your brain builds them up, and you slowly accumulate them. Then when you get an oxidative stress like what’s happening now in today’s world with all the covid and myocarditis and brain fog – a lot of these things are being caused because that reserve of plasmalogens has been depleted.

We want plasmalogens for a longevity perspective. There are other situations that can have low plasmalogens, other things can really knock your plasmalogens down.

So you want to start early, you want to build a savings account, and you want to maintain it. Maintain health and function, and create a sustained surplus for optimal health, for optimal neuromuscular performance.”


PXL_20231207_185012059

Brain restoration with plasmalogens

In this 2023 presentation for a professional audience, Dr. Dayan Goodenowe showed an example of what could be done (in the form of what he personally did at ages 53-54) to restore and augment brain structure and function over a 17-month period by taking plasmalogens and supporting supplements:

https://drgoodenowe.com/recording-of-dr-goodenowes-presentation-from-the-peptide-world-congress-2023-is-now-available/

Follow the video along with its interactive transcript. Restorative / augmentative supplements included:

1. Nutritional Supplementation Strategy

Forms of MRI used to document brain structure and function changes were:

2. Advanced MRI Technologies

Brain volume decreases are the rule for humans beginning at age 40. Dr. Goodenowe documented brain volume increases, which aren’t supposed to happen, but did per the below slide of overall results:

3. Reversing Brain Shrinkage

“From a global cortical volume and thickness perspective, 17 months of high-dose plasmalogens reversed ~15 years of predicted brain deterioration.”


Specific increased adaptations in brain measurements over 17 months included:

  1. Cortical thickness .07/2.51 = +3%.
  2. White matter microstructure fractional anisotropy +8%.
  3. Nucleus accumbens volume +30%.
  4. Dopaminergic striatal terminal fields’ volume +18%.
  5. Cholinergic cortical terminal fields’ volume +10%.
  6. Occipital cortex volume +10%.
  7. Optic chiasm volume +225%.
  8. Nucleus basalis connectivity.
  9. Neurovascular coupling signal controlled by noradrenaline integrity.
  10. Amygdala volume +4% and its connectivity to the insula, indicating ongoing anxiety and emotional stress response.
  11. Parahippocampus volume +7%.
  12. Hippocampus fractional anisotropy +5%.

No changes:

  1. Amygdala connectivity to the ventral lateral prefrontal cortex, the same part of the brain that relates to placebo effect.
  2. Hippocampus connectivity.

Decreased adaptations in brain measurements included:

  1. White matter microstructure radial diffusivity -10%.
  2. Amygdala connectivity to the anterior cingulate cortex to suppress / ignore / deny anxiety response.
  3. Amygdala connectivity to the dorsal lateral prefrontal cortex.
  4. Entorhinal cortex volume -14%.
  5. Hippocampus volume -6%.
  6. Hippocampus mean diffusivity (white matter improved, with more and tighter myelin) -4%.

The other half of this video was a lively and wide-ranging Q&A session.


The referenced 2023 study of 653 adults followed over ten years showed what brain deterioration could be expected with no interventions. Consider these annual volume decrease rates to be a sample of a control group:

etable 3

https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2806488 “Characterization of Brain Volume Changes in Aging Individuals With Normal Cognition Using Serial Magnetic Resonance Imaging”

Also see a different population’s brain shrinkage data in Prevent your brain from shrinking.


The daily plasmalogen precursor doses Dr. Goodenowe took were equivalent to 100 mg softgel/kg, double the maximum dose of 50 mg softgel/kg provided during the 2022 clinical trial of cognitively impaired old people referenced in Plasmalogens Parts 1, 2, and 3.

He mentions taking 5 ml in the morning and 5 ml at night because he used the Prodrome oil products. 1 ml of a Prodrome oil plasmalogen precursor product equals 900 mg of their softgel product.


“My brain is trying to minimize long-term effects of pain/stress by suppressing my memory of it. But this can only go on for so long before it becomes an entrenched state.

I have solved the sustenance side of the equation. I need to work harder to solve the environmental side.”

While I agree that we each have a responsibility to ourselves to create an environment that’s conducive to our health, the above phenomenon isn’t necessarily resolvable by changing an individual’s current environment. My understanding is that long-term effects of pain, stress, and related human experiences are usually symptoms of causes that started much earlier in our lives.

Adjusting one’s present environment may have immediate results, but probably won’t have much therapeutic impact on long-term issues. Early life memories and experiences are where we have to gradually go in order to stop being driven by what happened back then.

See Dr. Arthur Janov’s Primal Therapy for its principles and explanations. I started Primal Therapy at a similar age, 53, and continued for three years.


Continued with Part 2.

Plasmalogens, Part 3

The 2022 plasmalogen clinical trial mentioned in Parts 1 and 2 bypassed peroxisome metabolism of cognitively impaired people per discussion of the below diagram:

fcell-10-864842-g003

Increasing the body’s fasting state with time-restricted eating, and preventing muscle atrophy with resistance exercise, were offered as the two most important ways to improve peroxisomal function.

I didn’t find any relevant 2023 human studies (where I could access the full study) on different non-drug treatments that I was willing to do. A 2023 review outlined aspects of peroxisomes, to include a few older human studies:

“Peroxisomes are small, single-membrane-bound organelles, which are dynamic and ubiquitous. Peroxisomes directly interact with other organelles, such as endoplasmic reticulum, mitochondria, or lysosomes. Peroxisomes exert different functions in various cells through both catabolic and anabolic pathways.

The main functions of peroxisomes can be categorized as reactive oxygen species (ROS) metabolism, lipid metabolism, and ether-phospholipid biosynthesis. Peroxisomes also play important roles in inflammatory signaling and the innate immune response.”

1-s2.0-S2667325823001425-gr3_lrg

https://www.sciencedirect.com/science/article/pii/S2667325823001425 “Peroxisome and pexophagy in neurological diseases”


1. Since I haven’t recently tried the two main ways to improve peroxisomal function, I’ll give them a go over the next three months:

  • Expect to get my feeding timeframe to within eight hours. Don’t know about making it short like 6 hours, because my first meal of the day is 35 calories of microwaved cruciferous sprouts, then I wait an hour before eating anything else.
  • Resistance exercise progress should be measurable, as I recorded exercises during the first ten weeks of eating broccoli sprouts every day 3.5+ years ago.

2. Don’t know that I’ll recognize any cognitive improvements to the extent I did during Week 9.

  • I don’t have a young brain anymore, and I’m sure some decline could be measured in memory tests. But I’m not going to become a lab rat.
  • There’s an occasional annoyance that’s been going on for some time, especially when I’m distracted. It happens when I think of something to do, and it somehow becomes a short-term memory that I did it, instead of going into a Things To Do queue. It’s largely self-correcting. For example, regardless of what I paid, I’ll drive back to the grocery store self-checkout to retrieve a third bag that didn’t make it home. A pink-haired employee said young people leave their paid-for groceries behind all the time. It’s usually more of a reality disconnect for me than forgetfulness, because I have a memory that I performed the action. Definitely room for improvement.

3. Don’t know that I’d see biochemical changes such as some described in Part 1. Maybe I’ll move up an annual physical to compare it with the last one in May?

  • I already have very little oxidative stress, very little inflammation, low triglycerides, high HDL, and no major improvements are indicated on CBC / CMP / lipid panels.
  • Take supplements to ensure other things like acetylcholine neurotransmitter availability, one-carbon / methylation metabolism, vitamin / mineral adequacy.

4. I started the two Prodrome plasmalogen precursor supplements (ProdromeGlia and ProdromeNeuro) a week ago, and take their standard doses. My thought is that resultant plasmalogens won’t degrade very much if their primary use isn’t to immediately address oxidative stress and inflammation. That could give these extra plasmalogens a chance to make larger homeostatic contributions in myelin and membrane areas.

I don’t expect any particular effects to manifest. But I’m interested to see if these two areas would be affected:

  • My left ulnar nerve has been giving me problems for over five years, and several resistance exercises aggravate it. I’ve had two nerve continuity tests during that time to confirm. Numbness and pain are intermittent, though.
  • I still take acetaminophen several times a day for other pain.

None of the above treatments are specifically indicated. But if time-restricted feeding and/or extra plasmalogens have an effect on left ulnar or other pain, maybe I’ll be able to make better progress on resistance exercise.

Update #1 11/13/2023

Update #2 11/22/2023

Update #3 12/13/2023 comments

Update #4 1/30/2024

Update #5 3/31/2024