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

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.

Oat sprout stressors

Two 2023 Avena sativa oat sprout studies, starting with one that found different effects during germination from varying temperature and relative humidity:

“This study evaluated effects of temperature (20, 25, and 30°C) and relative humidity (RH, 55, 60, and 65%) as abiotic stressors during oat germination. We identified eighty polyphenols, nine avenanthramides, twelve lignans, and five phytosterols.

  • 100% germination was achieved at 25°C/60% RH from day 3, yielding the longest radicle size.
  • The highest content of most phenolic acids, avenanthramides, and lignans occurred at 30°C/65% RH, where 100% germination was attained by day 5, but with a shorter radicle size.
  • The best flavonoid and phytosterol profile was obtained at 20°C/55% RH, achieving only a 67% germination rate by day 5.

sprouted oat temp rh

By considering germination conditions, end-users can harness the versatility of oat sprouting to meet their specific needs and maximize potential benefits of this promising cereal crop. For instance, manufacturers of functional foods and beverages could consider using sprouts from conditions that yield high polyphenol content for products targeting antioxidant benefits, whereas nutraceutical manufacturers could focus on sprouting conditions that result in elevated levels of avenanthramides, well-known for their health-promoting properties.”

https://www.sciencedirect.com/science/article/abs/pii/S0308814623027917Impact of temperature and humidity conditions as abiotic stressors on the phytochemical fingerprint of oat (Avena sativa L.) sprouts” (not freely available) Thanks to Dr. Iza F. Pérez-Ramírez for providing a copy.


Another study compared and contrasted eight sprouted grains to their ungerminated grains and to each other. I’ll highlight oat sprout results:

“The method used was germination for up to 72 h at temperatures ranging from 19–23°C. Oat germination rate was 80%.

Linoleic acid (omega-6) was the predominant fatty acid in oat grain powder, followed by similar amounts of oleic and palmitic acids and smaller amounts of stearic and linolenic (omega-3) acids. Since omega-6 content remained unchanged and omega-3 quantity increased slightly in sprouted oats, the omega-6/omega-3 ratio decreased.”

https://www.mdpi.com/2304-8158/12/17/3306 “Effect of Germination on Fatty Acid Composition in Cereal Grains”


My kitchen cupboard’s oat sprouting conditions are closer to this second study’s temperature, where relative humidity wasn’t specified. I doubt that kitchen winter-time relative humidity ever rises to the 55% lower threshold of the first study for more than a few minutes.

At this time of year in Sprouting hulled oats, I got a 97% germination rate over three days with an estimated 21°C (70°F) and a relative humidity closer to 30% than 55%. Couldn’t tell you why the first study’s germination rate with 20°C/55% RH was only 67% at day 5, or why the second study’s germination rate was only 80% at day 3 with 19–23°C.


PXL_20231223_175744134

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

Plasmalogens, Part 1

The person who knows the most about this subject is Dayan Goodenowe, PhD. Some recent publications include:

https://www.frontiersin.org/articles/10.3389/fcell.2022.864842/full “Targeted Plasmalogen Supplementation: Effects on Blood Plasmalogens, Oxidative Stress Biomarkers, Cognition, and Mobility in Cognitively Impaired Persons”

https://www.frontiersin.org/articles/10.3389/fcell.2022.866156/full “Brain ethanolamine phospholipids, neuropathology and cognition: A comparative post-mortem analysis of structurally specific plasmalogen and phosphatidyl species”

plasmalogens and cognition


A sample of links freely available at https://drgoodenowe.com/.

1. Presentations to professional groups. Have your mouse ready to click the pause button.

https://drgoodenowe.com/dr-goodenowe-presents-at-the-iagg2023-in-yokohama-japan/ “A rare children’s disease that may be the key to reversing neurological decline in aging”

Includes videos of a treatment’s effects on a child.

https://neomarkgroup.wistia.com/medias/0qln0wy93t “The most influential biomarkers for aging and disease”

Despite the title, a considerable number of studies were presented on prenatal, infant, and early childhood development. He misspoke a few times, so read the slides.

Phenotype is reality. Genotype is possibility. Communications links between different fields are very poorly connected in science.

Peroxisomes are islands. They don’t have DNA like your mitochondria do. Peroxisomal transport issues are important things to understand.

All aging-related cross-sectional analyses are on the rate of decline. You’re declining from a previous well state. Age-matched controls are the most ridiculous thing to do.”


2. I’ll highlight the longest of several interviews because there was plenty of room to expand on points. Maybe the best detailed explanations came as responses to that interviewer challenging with contrasting AD, traumatic brain injury, and cholesterol paradigms. Its transcript is more accurate than a usual YouTube interpretation, but there are still mistakes such as “fossil lipid” vs. phospholipid.

https://www.betterhealthguy.com/episode186 “Plasmalogens with Dr. Dayan Goodenowe, PhD”

“Science is how do you push things to its failure, until you can’t fail it again. We’ve lost that. It’s become more hypothesis proving.

Plasmalogens levels go up for a different reason than people think. The reason why it peaks in our 40s and 50s is because we’ve been myelinating. The white matter of our brain is still increasing. It’s not because we’re making more plasmalogens. It’s because the lake, the reservoir, gets full. What you’re measuring in blood is overflow from the lake. The lower plasmalogens start trickling down in your blood, the bigger drain that’s occurring on that system.

Low plasmalogens don’t just predict dementia in the elderly population. It predicts the rate of decline of that dementia. It predicts the rate of death.

The biggest drivers of plasmalogen manufacturing and the biggest reasons why they decrease with age, or in other circumstances is two things. One, the failure to maintain a fasting state of the human body. The second one is muscle atrophy.

Amyloid has absolutely nothing to do with Alzheimer’s, or dementia. It’s just a bystander on the road watching an accident happen.

Age-related cognitive decline is clearly where plasmalogens have the greatest impact. You’re always going to have mixed pathologies in the brain.

Nutritional availability of plasmalogens is virtually non-existent. As soon as they hit the hydrochloric acid of your stomach, they’re gone. They don’t make it past the stomach, or the upper intestine.”


I came across Dr. Goodenowe’s work last month from clicking a comment on this blog that linked back to her blog. Always be curious.

Continued in Part 2.

Canadian Thanksgiving

Canadians were in dire straights yesterday, needing something to not be depressed about. It isn’t that US citizens are less depressed, but we have our Bill of Rights 1st and 2nd amendments that we adhere to.

Several of the people on this three and a half hour conversation were Canadians. Will you listen to them?

I learned a lot, such as possible transgenerational inheritance of effects from worldwide coerced actions. Bhakdi shortly after the 1 hour 6 min point was the easiest to understand, and also the scariest.

So what were you thankful for yesterday? Exposing facts?


PXL_20230928_222908973

Neuritogenesis

Three 2023 papers on the initial stage of neuronal differentiation, starting with a rodent study of taurine’s effects:

“We aimed to assess the role of taurine (TAU) in axonal sprouting against cerebral ischemic injury, clarify the function of mitochondria in TAU-induced axonal sprouting, and further determine the underlying potential molecular mechanism.

experiment design

We determined that TAU improved motor function recovery and restored neurogenesis in ischemic stroke. This possibly occurred via improvements in mitochondrial function.

We investigated that the Sonic hedgehog (Shh) pathway exerted an important role in these effects. Our study findings highlighted the novel viewpoint that TAU promoted axonal sprouting by improving Shh-mediated mitochondrial function in cerebral ischemic stroke.”

https://www.scielo.br/j/acb/a/nxKvGXGk9g6gRkHxybMfbYJ/?lang=en “Taurine promotes axonal sprouting via Shh-mediated mitochondrial improvement in stroke”


A rodent study investigated effects of a soy isoflavone gut microbiota metabolite:

“Perinatally-infected adolescents living with HIV-1 (pALHIV) appear uniquely vulnerable to developing substance use disorders (SUD). Medium spiny neurons (MSNs) in the nucleus accumbens core (NAcc), an integrator of cortical and thalamic input, have been implicated as a key structural locus for the pathogenesis of SUD.

Treatment with estrogenic compounds (e.g., 17β-estradiol) induces prominent alterations to neuronal and dendritic spine structure in the NAcc supporting an innovative means to remodel neuronal circuitry. The carcinogenic nature of 17β-estradiol, however, limits its translational utility.

Plant-derived polycyclic phenols, or phytoestrogens, whose chemical structure resembles 17β-estradiol may afford an alternative strategy to target estrogen receptors. The phytoestrogen S-Equol (SE), permeates the blood-brain barrier, exhibits selective affinity for estrogen receptor β (ERβ), and serves as a neuroprotective and/or neurorestorative therapeutic for HIV-1-associated neurocognitive and affective alterations.

Beginning at approximately postnatal day (PD) 28, HIV-1 transgenic (Tg) animals were treated with a daily oral dose of 0.2 mg of SE. The SE dose of 0.2 mg was selected for two primary reasons, including:

  1. A dose-response experimental paradigm established 0.2 mg of SE as the most effective dose for mitigating neurocognitive deficits in sustained attention in the HIV-1 Tg rat; and
  2. The dose, which yielded a daily amount of 0.25–1.0 mg/kg/SE (i.e., approximately 2.5–10 mg in a 60 kg human), is translationally relevant (i.e., well below the daily isoflavone intake of most elderly Japanese.

Daily oral treatment continued through PD 90.

j_nipt-2023-0008_fig_002

HIV-1 Tg animals exhibited an initial increase in dendrite length (A) and the number of dendritic spines (B) early in development; parameters which subsequently decreased across time. In sharp contrast, dendrite length and the number of dendritic spines were stable across development in control animals.

Targeting these alterations with the selective ERβ agonist SE during the formative period induces long-term modifications to synaptodendritic structure, whereby MSNs in the NAcc in HIV-1 Tg animals treated with SE resemble control animals at PD 180.”

https://www.degruyter.com/document/doi/10.1515/nipt-2023-0008/html “Constitutive expression of HIV-1 viral proteins induces progressive synaptodendritic alterations in medium spiny neurons: implications for substance use disorders”


A rodent brain cell study investigated soy isoflavones’ effects on a different estrogen receptor:

“We evaluated effects of isoflavones using mouse primary cerebellar culture, astrocyte-enriched culture, Neuro-2A clonal cells, and co-culture with neurons and astrocytes. Soybean isoflavone-augmented estradiol mediated dendrite arborization in Purkinje cells.

These results indicate that ERα plays an essential role in isoflavone-induced neuritogenesis. However, G-protein-coupled ER (GPER1) signaling is also necessary for astrocyte proliferation and astrocyte–neuron communication, which may lead to isoflavone-induced neuritogenesis.

We highlight the novel possibility that isoflavones enhance dendritogenesis and neuritogenesis, indicating that they can be a useful supplementary compound during brain development or in the injured brain.”

https://www.mdpi.com/1422-0067/24/10/9011 “Isoflavones Mediate Dendritogenesis Mainly through Estrogen Receptor α”

Adverse Childhood Experiences, Part 2

A request was made to present studies that investigated epigenetic impacts of corporal punishments or physical trauma to children or adolescents. Here’s a follow-on of the 2015 Grokking an Adverse Childhood Experiences (ACE) score, since physical abuse is one factor of an ACE score.

1. The largest problem is that a person filling out an ACE questionnaire or Childhood Trauma Questionnaire can’t provide first-hand answers of their own experiences during womb life, infancy, and early childhood. These critical development periods are more impacted by adversity than are later life windows.

Human brains aren’t developed enough before age 3 to provide retrospective answers using cerebral memories. A self-reported ACE score can’t possibly address what happened during the times when we were most vulnerable to disrupted neurodevelopment. And good luck with parents providing factual histories of whether they physically or emotionally neglected, physically or emotionally abused, or otherwise adversely treated their fetus, infant, and young child.

2. Another problem is researchers can pretty much choose whatever questions they want as input criteria. I’ve seen pliable ACE scores developed from 5- to 25-item questionnaires.

Do these questionnaires cover all relevant adverse childhood experiences? For example, are researchers permitted to use as inputs societal-created adversities a child may have lived through such as the Khmer Rouge or Cultural Revolution? Studies are just starting to investigate adverse childhood experiences created by worldwide abuses of authority since 2020.

3. Other problems were discussed in a 2023 paper https://www.sciencedirect.com/science/article/abs/pii/S0145213423003162 “Adverse childhood experiences and adult outcomes using a causal framework perspective: Challenges and opportunities” (not freely available), two of which were:

  • Adding up ACE factors to a cumulative score ignores the impact of synergistic sets. For example, although both cumulative ACE scores are 2, a child who was physically and sexually abused would probably be more adversely affected than a child whose parents divorced or separated, and also had a family member incarcerated.
  • At any given time point, and especially with older people, there’s a potential selection bias against those most affected by adverse childhood experiences, such as those who died.

Using flawed, squishy, cumulative ACE scores as inputs, here are two 2023 studies that found epigenetic associations:

“We tested the following pre-registered hypotheses: Mothers’ adverse childhood experiences are correlated with DNA methylation (DNAm) in peripheral blood during pregnancy (hypothesis 1) and in cord blood samples from newborn infants (hypothesis 2), and women’s depression and anxiety symptoms during pregnancy mediate the association between mothers’ ACE exposure and prenatal/neonatal DNA methylation (hypothesis 3).

  1. Hypothesis 1: In 896 mother−infant pairs with available methylation and ACE exposure data, there were no significant associations between mothers’ ACE score and DNAm from antenatal peripheral blood, after controlling for covariates.
  2. Hypothesis 2: In infant cord blood, there were 5 CpG sites significantly differentially methylated in relation to mothers’ ACEs (false discovery rate < .05), but only in male offspring. Effect sizes were medium. CpG sites were in genes related to mitochondrial function and neuronal development in the cerebellum.
  3. Hypothesis 3: There was no mediation by maternal anxiety/depression symptoms found between mothers’ ACEs score and DNAm in the significant CpG sites in male cord blood.”

https://www.jaacap.org/article/S0890-8567(23)00313-1/fulltext “Epigenetic Intergenerational Transmission: Mothers’ Adverse Childhood Experiences and DNA Methylation”


“In this study, the effect of cumulative ACEs experienced on human maternal DNAm was estimated while accounting for interaction with domains of ACEs in prenatal peripheral blood mononuclear cell samples. Intergenerational transmission of ACE-associated DNAm was explored used paired maternal and neonatal cord blood samples. Replication in buccal samples was also explored.

We used a four-level categorical indicator variable for ACEs exposure: none (0 ACEs), low (1–3 ACEs), moderate (4–6 ACEs), and high (> 6 ACEs). 🙄

125a4c3cacfe4b922e5b864c

https://www.researchsquare.com/article/rs-2977515/v1 “Effect of Parental Adverse Childhood Experiences on Intergenerational DNA Methylation Signatures”

Paradigms determine findings

This 2023 rodent study from Dr. Michael Skinner’s labs at Washington State University investigated epigenetic transgenerationally inherited differential DNA methylation regions (DMRs). I’ll focus on a paradigm shift that enabled some of this study’s findings:

“The current study was designed to assess if morula embryos escape the erasure of DDT-induced transgenerational sperm DMR methylation. Observations demonstrate:

  • 98% of transgenerational sperm DMR sites retain DNA methylation and are not erased, appearing similar to imprinted-like sites.
  • Maintenance of DNA methylation on a variety of imprinted sites in a comparison of sperm versus morula methylation levels using methylated DNA immunoprecipitation (MeDIP) followed by next-generation sequencing (MeDIP-Seq).
  • The majority of low-density CpG genomic sites had a significant increase in DNA methylation in the morula embryo compared to sperm.

The general erasure of DNA methylation during embryogenesis appears applicable to high-density DNA methylation sites (e.g. CpG islands) but neither to transgenerational DMR methylation sites nor to low-density CpG deserts, which constitute the vast majority of the genome’s DNA methylation sites.

dvad003f1

Bisulfite procedures have been extensively used followed by next-generation sequencing (BS-Seq) to assess genome-wide DNA methylation in early embryonic development. This has led to the concept that DNA methylation erasure occurs during early embryo development and primordial germ cell development.

A limitation with BS-Seq is that it is often biased toward detecting changes in higher-density CpG sites with >5 CpG/100 bp. A critical technical limitation to BS-Seq is that bioinformatics protocols used remove low-density (<3 CpG/100 bp) regions from the genome prior to analysis. In contrast, MeDIP-Seq analysis is biased to low-density CpG sites with <5 CpG/100 bp that constitute >90% of the genome.

Alteration of morula stem cell epigenetics will impact epigenomes and transcriptomes of all subsequently derived somatic cells. This is the molecular basis for epigenetic transgenerational inheritance phenotypes and pathologies.

Future studies need to re-evaluate the current dogma of a genome-wide erasure of DNA methylation, and consider a more dynamic regulation of early embryonic stem cell epigenetic development.”

https://academic.oup.com/eep/article/9/1/dvad003/7190131 “Transgenerational sperm DMRs escape DNA methylation erasure during embryonic development and epigenetic inheritance”


PXL_20230614_014127763

Transgenerational transmission of stress

This 2023 rodent study found that effects of stress during mid-late gestation were epigenetically transmitted to the first, second, and third female generations:

“We investigated effects of gestational chronic variable stress (CVS) in rats using restraint and social isolation stress in the parental F0 generation. Only the F0 pregnant dams were subjected to stress.

When a pregnant female experiences adversity, impacts of that stress affect exposed somatic tissues (F0 generation), the fetuses (F1 generation), and the fetuses’ germline (F2 generation). A true transgenerational inheritance arises when germline epimutations are transmitted to unexposed F3 offspring.

A subset of F1 rats was housed in an enriched environment (EE) to mitigate adverse effects of CVS. F2 offspring reared in EE had increased birth weights, but their uterine gene expression patterns remained comparable to those of stressed animals.

ijms-24-03734-g001

We provide evidence that psychological and psychosocial CVS alters inflammatory status and endocrine markers in uteri of adult dams through transgenerational programming of the female germline. EE therapy in prenatally stressed F1 offspring had no beneficial effects on uterine expression of inflammatory and endocrine markers for them or their future offspring.”

https://www.mdpi.com/1422-0067/24/4/3734 “Environmental Enrichment Promotes Transgenerational Programming of Uterine Inflammatory and Stress Markers Comparable to Gestational Chronic Variable Stress”


PXL_20230611_100728709

Taurine’s effects on healthspan and lifespan

A 2023 human / primate / rodent / worm study with 56 coauthors exhaustively investigated taurine effects:

“We measured the blood concentration of taurine during aging and investigated the effect of taurine supplementation on healthspan and lifespan in several species.

  • In C57Bl/6J wild-type (WT) mice, serum taurine concentrations declined from 132.3 ± 14.2 ng/ml at 4 weeks to 40.2 ± 7.1 ng/ml at 56 weeks.
  • In 15-year-old monkeys, serum taurine concentrations were 85% lower than in 5-year-old monkeys.
  • Taurine concentrations in elderly humans were decreased by more than 80% compared with concentration in serum of younger individuals.

Regardless of their sex, taurine-fed mice survived longer than control mice. The median lifespan increase was 10 to 12%, and life expectancy at 28 months increased by 18 to 25%.

Improved survival of taurine-fed mice was not a consequence of low survival of control animals or differences in diet. Taurine deficiency is a driver of aging in mice because its reversal increases lifespan.

lifespan extension starting taurine in middle age

We investigated the health of taurine-fed middle-aged mice and found an improved functioning of bone, muscle, pancreas, brain, fat, gut, and immune system, indicating an overall increase in healthspan. Taurine reduced cellular senescence, protected against telomerase deficiency, suppressed mitochondrial dysfunction, decreased DNA damage, and attenuated inflammation.

An association analysis of metabolite clinical risk factors in humans showed that lower taurine, hypotaurine, and N-acetyltaurine concentrations were associated with adverse health, such as increased abdominal obesity, hypertension, inflammation, and prevalence of type 2 diabetes. We found that a bout of exercise increased concentrations of taurine metabolites in blood, which might partially underlie antiaging effects of exercise.

Taurine abundance decreases during aging. A reversal of this decline through taurine supplementation increases healthspan and lifespan in mice and worms, and healthspan in monkeys.”

https://www.science.org/doi/10.1126/science.abn9257 “Taurine deficiency as a driver of aging”


One area curiously not investigated in this study was that taurine supplementation freed up cysteine to do things other than synthesize taurine, like synthesize glutathione, an idea in Treating psychopathological symptoms will somehow resolve causes? An introductory article brought up this point:

“One of the most studied mechanisms of action for taurine is an increase in antioxidant capacity. Although oxidative damage is not clearly linked to mammalian lifespan, it plays a role in many age-associated pathologies.

Taurine is a poor scavenger of reactive oxygen species, with the exception of hypochlorite, which it detoxifies to N-chlorotaurine. N-Chlorotaurine is anti-inflammatory and induces expression of antioxidant enzymes in mice and humans.

Taurine supplementation might also cause an increase in levels of its precursors, including the antioxidants hypotaurine and cysteine. An interesting corollary is that up-regulating endogenous taurine synthesis would have the opposite result—consuming hypotaurine and cysteine.”

https://www.science.org/doi/10.1126/science.adi3025 “Taurine linked with healthy aging”


A human equivalent taurine dose is (1 g x .081) x 70 kg = 5.67 grams. Dose tests from supplementary data were:

“Dose and frequency of taurine administration was selected based on a pilot study, which showed that when given once daily to middle-aged WT mice, this regimen increased peak blood taurine concentrations to baseline concentrations in young (4-week-old) mice.”

taurine dose

I’ve taken 2 grams every day for the past three years, and will now bump that up to 5 grams. My diet doesn’t regularly include any foods high in taurine.

I recommend reading the study rather than commentaries. Its publisher did a very good job of linking figures so that images can be viewed, then the reader returned to the right context.

Gatekeepers are out in full force on this study, and their viewpoints are probably what you’ll see first, to include unevidenced statements like “the study’s main authors cautioned the public not to self-dose with the supplement” and the above introductory article’s unreferenced “equivalent doses used in the study by Singh et al. would be very high in humans.” Pretty pathetic that such ‘authorities’ are even publicized after recent years of deliberately misleading the world about science and medicine.

This study and all commentaries called for clinical trials that are NOT going to happen:

  • Drug companies can’t make money from a research area that’s cheap, not patentable, and readily accessible.
  • Government sponsors are likewise not incentivized to act in the public’s interest per their recent behavior.

Take responsibility for your own one precious life. See Part 2 for a sample of citing papers.

PXL_20230601_181526429

Amphibian epigenetic clocks

This 2023 study of two frog species expanded one of the cited studies in Epigenetic clocks so far in 2022 to include post-embryonic epigenetic clock measurements:

“We generated DNA methylation data from African clawed frogs (Xenopus laevis) and Western clawed frogs (Xenopus tropicalis) and built multiple epigenetic clocks. Dual species clocks were developed that apply to both humans and frogs (human-clawed frog clocks), supporting that epigenetic aging processes are evolutionary conserved outside mammals.

The two species underlying our Xenopus clocks have markedly different maximum lifespans (30.3 for X. laevis and 16 for X. tropicalis), and average ages of sexual maturity (1 year for laevis and 0.375 for tropicalis). When building our Xenopus clocks, we addressed this fact in two ways:

  • In our pan-clock, we used a log-linear transformation of age that effectively normalizes ages with respect to age at sexual maturity.
  • In our relative pan-clock, we instead estimate relative age (chronological age divided by maximum lifespan), which normalizes ages with respect to maximum lifespan.

We also created dual-species clocks, referred to as human-clawed frog clocks, for estimates of chronological age and relative age. Relative age is the ratio of chronological age to maximum lifespan, and takes on values between 0 and 1. Maximum lifespan observed for humans was 122.5 years.

The relative age clock allows for alignment and biologically meaningful comparison between species with different lifespans.

relative age

Previous studies in humans showed that a hallmark of age-related CpGs is their association with target sites of Polycomb repressive complex 2 (PRC2), which gain methylation with age. This feature is fully recapitulated in Xenopus, and physiological significance of this association is an important open question.

PRC2 plays a prominent role during embryonic development and consequently, many aging-clock-associated genes relate to developmental processes. Given its evolutionary conservation from frogs to humans, methylation status of PRC2 targets supports some critical causal relationship to systemic aging.

Since the association with PRC2 with aging stems from analyses of adult postmitotic cells, and of different tissue origin rather than from embryonic cells, it is tempting to speculate that adult methylation status will get important input during embryonic development, the very phase when PRC2 target gene expression is prominent.

Genes associated with both positive and negative age-related CpGs relate to neural processes, although in somewhat opposite direction. While DNAm increase is linked to neural developmental genes, DNAm decrease links to synaptic transmission, roughly corresponding to processes of immature vs. mature neuronal cells, respectively. This leads to the counter-intuitive suggestion that studying Xenopus neural development may yield new insights into biological aging.”

https://link.springer.com/article/10.1007/s11357-023-00840-3 “DNA methylation clocks for clawed frogs reveal evolutionary conservation of epigenetic aging”


I’ve seen dual-species epigenetic clocks – introduced in A rejuvenation therapy and sulforaphane – referenced elsewhere, most recently in Selective Breeding for High Intrinsic Exercise Capacity Slows Pan-Tissue Epigenetic Aging in Rats. These clocks still aren’t in wide use by researchers, though. Don’t know what it will take to persuade researchers to use dual-species relative age clocks in their model organism studies so that they can justifiably invoke human applicability.

PXL_20230601_182718629

Nrf2 Week #3: Epigenetics

To follow the Nrf2 Week #2 finding that chromatin accessibility parallels Nrf2 expression, this 2023 cell study explored how Nrf2 influences other epigenetic processes:

“We identified antioxidant response element sequences in promoter regions of genes encoding several epigenetic regulatory factors, such as histone deacetylases (HDACs), DNA methyltransferases (DNMTs), and proteins involved in microRNA biogenesis.

  • We treated cells with dimethyl fumarate (DMF), an activator of the NRF2 pathway through both the KEAP1 and GSK-3 pathways. NRF2 is able to modulate expression of HDAC1, HDAC2, HDAC3, and SIRT1 in different cell types.
  • DMF treatment induced DNMT1 and DNMT3b at both mRNA and protein levels. For DNTM3a, there was a slight induction of mRNA levels but not at the protein level.

antioxidants-12-00641-g007

  • Our data indicate that of all miRNAs analyzed, only miR-27a-3p, miR-27b-3p, miR-128-3p, and miR-155-5p associate with Nfe2l2 mRNA. NRF2 causes degradation of miR-155-5p, which is implicated in neuroinflammation and other pathologies, and is the main miRNA induced by LPS treatment in microglia. miR-155 alters expression of genes that regulate axon growth, supporting the bioinformatic prediction that miR-155 can regulate expression of genes involved in central nervous system development and neurogenesis.

Todate we only understand how epigenetic modifications affect expression and function of the NRF2 pathway. The fact that NRF2 can promote expression of type I HDACs, DNMTs, and proteins involved in miRNA biogenesis opens new perspectives on the spectrum of actions of NRF2 and its epigenetic influences.”

https://www.mdpi.com/2076-3921/12/3/641 “The Transcription Factor NRF2 Has Epigenetic Regulatory Functions Modulating HDACs, DNMTs, and miRNA Biogenesis”


PXL_20230515_185958612

Nrf2 Week #2: Neurons

To follow the Nrf2 Week #1 suggestion that Nrf2 target neurological disorders, this 2023 cell study investigated Nrf2 expression in neurons:

“Oxidative metabolism is inextricably linked to production of reactive oxygen species (ROS), which have the potential to damage all classes of macromolecules. Yet ROS are not invariably detrimental. Several properties make ROS useful signaling molecules, including their potential for rapid modification of proteins and close ties to cellular metabolism.

We used multiple single cell genomic datasets to explore Nrf2 expression and regulation in hundreds of neuronal and non-neuronal cell types in mouse and human. With few exceptions, Nrf2 is expressed at far lower levels in neurons than in non-neuronal support cells in both species.

This pattern is maintained in multiple disease states, and the chromatin accessibility landscape at the Nrf2 locus parallels these expression differences. These results imply that Nrf2 activity is limited in almost all neurons of the mouse and human central nervous system (CNS).

nrf2 expression

We separated cell types into neuron or non-neuronal ‘support’ cell categories. The general ‘support’ term is not meant to minimize the functional relevance of non-neuronal cells in the CNS, but is an umbrella term meant to cover everything from glial cell types (astrocytes, microglia, oligodendrocytes) to endothelial cells.

It is not clear why an important, near ubiquitous cytoprotective transcription factor like Nrf2 remains off in mature neurons, especially considering oxidative stress is a driver of many diseases. The simplest explanation is that Nrf2 activity also disrupts normal function of mature neurons.

ROS play a key role in controlling synaptic plasticity in mature neurons. These activity-dependent changes in synaptic transmission, which are important for learning and memory, are disrupted by antioxidants.

A subset of important Nrf2-targeted antioxidant genes (e.g., Slc3a2, Slc7a11, Nqo1, Prdx1) are also low in neurons. So it is likely that these and/or other Nrf2 targets must remain low or non-ROS-responsive in mature neurons. Future work exploring why this expression pattern persists in mature neurons will inform our models on roles of antioxidant genes in normal neuronal physiology and in neurological disorders.

https://www.biorxiv.org/content/10.1101/2023.05.09.540014v1.full “Limited Expression of Nrf2 in Neurons Across the Central Nervous System”


PXL_20230520_182827767