UA-45667900-1

Thursday 6 September 2018

Ketones and Autism Part 4 – Inflammation, Activated Microglia, CtBP, the NLRP3 Inflammasome and IL-1β




This series of posts on ketones and the ketogenic diet (KD) is nearly finished and I am glad that I made favourable comments about the KD earlier on in this blog, before I knew all the nitty-gritty of the science. (no re-editing required)




Inflammasome Inhibition: Putting Out the Fire                                                                                                                       


There is more than one anti-inflammatory mechanism involved in the ketogenic diet (KD); in Part 3 we covered Niacin Receptor HCA2, today in Part 4 we look at NLRP3 and CtBP.
The reason I am going into all this detail is because if you knew why someone responds to ketones in a favourable way, there might actually be an even more potent therapy using an entirely different substance.
CtBP represses the transcription of certain tumour supressing genes and some other genes involved in the development of cancer, i.e. they promote tumorigenesis.  CtBP is often overexpressed in certain cancers and indicates a worse prognosis. In these cancers you would want to inhibit CtBP.
Just to complicate matters, CtBP also supresses the activity of certain inflammatory genes. So, in certain diseases like diabetes you might benefit from keeping CtBP permanently in its active state. In particular, this would apply to when the microglia are activated, which is the case in much autism.
The coconut oil doctors have the idea that the key problem in autism is activated microglia in the brain.  Microglia mediate immune responses in the central nervous system, clearing cellular debris and dead neurons via a process called phagocytosis. These doctors propose coconut oil to calm the microglia.
Microglia can be in a resting or activated state, the research suggests that in much autism the microglia are permanently activated.
Some research suggests that microglia act like an “immunostat” reflecting not just what is going on in the brain, but elsewhere in the body.  I favour this view.
A small trial using a drug to calm the microglia did not impact autism.
Personally, I believe that microglia being activated is not a good thing, but that it is part of a much more complex picture than the coconut doctors suggest. 
As we learn later in this post, to get the CtBP benefit to microglia, it appears that you need the kind of ketosis you achieve only in the full ketogenic diet, not the transient mild ketosis that you achieve from two heaped tablespoons of coconut oil, or any of the keto supplements. 

NLRP3 inflammasome
The complicated-sounding NLRP3 inflammasome relates to diseases where the proinflammatory cytokine IL-1β is elevated; this includes Alzheimer’s, MS, Inflammatory Bowel Disease (IBD) and often autism.
For the details of how NLRP3 works see below; the important thing to note is that the result is elevated levels of IL-1β, which, at least in blood, is easy to measure.  It is an open question whether this represents the level inside the brain.  If your child has elevated IL-1β then it is worth studying NLRP3.




Schematic illustration of the NLRP3 inflammasome activation. Upon exposure to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs), Toll-like receptors (TLRs) are phosphorylated and subsequently activate NF-κB. In the nucleus, NF-κB promotes the transcription of NLRP3, proIL-1β, and proIL-18, which, after translation, remain in the cytoplasm in inactive forms. Thus, this signal (depicted in red as “Signal 1”) is a priming event. A subsequent stimulus (shown as “Signal 2” in black) activates the NLRP3 inflammasome by facilitating the oligomerization of inactive NLRP3, apoptosis-associated speck-like protein (ASC), and procaspase-1. This complex, in turn, catalyzes the conversion of procaspase-1 to caspase-1, which contributes to the production and secretion of the mature IL-1β and IL-18. Three models have been proposed to describe the second step of inflammasome activation: (1) Extracellular ATP can induce K+/potassium efflux through a purogenic P2X7-dependent pore, which, leads to the assembly and activation of the NLRP3 inflammasome. Calcium flux is also involved in this process. (2) PAMPs and DAMPs trigger the generation of ROS that promote the assembly and activation of the NLRP3 inflammasome. (3) Phagocytosed environmental irritants form intracellular crystalline or particulate structures leading to lysosomal rupture (magenta box) and release of lysosomal contents like cathepsin B. These induce NLRP3 inflammasome assembly and activation. In addition, other factors and mechanisms have been implicated in the assembly and activation of the NLRP3 inflammasome, including mitochondrial damage, autophagic dysfunction, and thioredoxin-interacting protein (TXNIP).



Proinflammatory cytokine IL-1β 
My public enemy number 1 cytokine is actually IL-6, today we primarily look at IL-1β, which for many people with a neurological disorder is a big part of their problem. IL-6 and IL-1β are actually interrelated, as we see later.
For a summary of the role of this cytokine in autism, I will leave it to Paul Ashwood:-  


Interleukin (IL)-1B

IL-1Β is an inflammatory cytokine expressed very early in immune responses. In tissue, IL-1Β propagates inflammation by activating local immune cells and the vascular endothelium. Systemically, IL-1Β stimulates IL-6 production and eventually an acute phase response in the liver. Systemic IL-1Β can cross the blood brain barrier and stimulate its own expression in the hypothalamus, which leads to neuroendocrine changes associated with fever and sickness behavior . IL-1Β receptors are structurally related to toll-like receptors (TLRs), and signaling is achieved through NF-κB and MAP kinase (MAPK) signaling cascades. IL-1Β belongs to an evolutionarily conserved family of proteins that function beyond immunity. It shares structural homology with fibroblast growth factors, which are critical in embryonic neurodevelopment, and are implicated in autism and schizophrenia.
Genes for IL-1Β, its receptor, and its receptor-associated proteins are associated with intellectual disability, schizophrenia, and autism. Children and adults with autism have increased plasma IL-1Β and skewed cellular IL-1Β responses following stimulation. Compared to controls, monocytes from children with ASD produce excessive IL-1Β following LPS exposure, and lower levels following exposure to TLR 9 agonists. The IL-1 antagonist, IL-1ra, is also increased among ASD subjects. IL-1ra reduces inflammation by competing for the IL-1Β receptor, and increased levels may represent an attempt to counteract inflammation in ASD. Postmortem brains from ASD subjects had normal IL-1Β levels, but given that peripheral IL-1Β can enter the brain, increased systemic levels could directly impact neurological processes
IL-1Β disruption can have a variety of neurological consequences relevant to autism. The cytokine and its receptors are found throughout the nervous system during critical developmental periods. IL-1Β induces neural progenitor cell proliferation in some CNS regions, while inhibiting it in others. This could contribute to the region-specific overgrowth and undergrowth observed in the ASD brain. Excitatory synapse formation is partially mediated by the IL-1 receptor and receptor-associated proteins.
Altering these proteins can tip the balance between excitatory and inhibitory signaling, which might underlie neurological features of autism. Increased IL-1ra in autism suggests an attempt to counterbalance IL-1Β and may or may not be beneficial. Following brain injury, IL-1ra upregulation serves a neuroprotective role by dampening excessive inflammation. However, if administered during critical windows of neurodevelopment, IL-1ra can negatively impact neurogenesis, brain morphology, memory consolidation, and behavior. This shows that some level of IL-1B signaling is essential during development. In adulthood, IL-1Β is implicated in CNS disorders like Alzheimer’s disease and the advancement of amyloid-containing plaques. While excessive IL-1B contributes to pathology in some cases, it may have a protective role in others. For example, IL-1Β limits neuronal damage following excitotoxic exposures, and mice lacking IL-1Β fail to undergo remyelination following experimental autoimmune encephalitis (EAE) induction. IL-1Β is involved in higher order brain processes and is induced in the hippocampus during learning processes, and is critical for maintenance of long-term potentiation (LTP) Both over expression and under expression of IL-1 beta are associated with impairments in memory and learning.



At the table in the kitchen, there were three bowls of porridge.  Goldilocks was hungry.  She tasted the porridge from the first bowl.
"This porridge is too hot!" she exclaimed.
So, she tasted the porridge from the second bowl.
"This porridge is too cold," she said
So, she tasted the last bowl of porridge.
"Ahhh, this porridge is just right," she said happily and she ate it all up. 

In summary, IL-1Β participates in neurological processes, and appears to have a role in both CNS pathology and healing. Normal, homeostatic levels of IL-1Β and its antagonist IL-1ra are necessary for proper brain development and function. This “Goldilocks” state is typical of many cytokines, where too much or too little is not desirable. Alterations in IL-1Β systems due to genetic mechanisms or environmental exposures may contribute to autism. 


CtBP (C-terminal-binding protein) 
In 2017 research led by Dr Raymond Swanson, a professor of neurology at the University of California, San Francisco, suggested CtBP as an additional possible mechanism by which the ketogenic diet can reduce brain inflammation.   CtBP activation turns off key inflammatory genes.
In the case of CtBP, I doubt that the very partial ketosis achieved with BHB and C8 supplements will be enough, I think you would need the full ketogenic diet. 
Restricting the glucose metabolism with the ketogenic diet lowers the NADH/NAD+ ratio which activates CtBP. There is no direct role played by ketones in this process, it is just the presence of large amounts of ketones reduces the role of glucose.



Factors that reduce glucose flux through glycolysis, such as reduced glucose availability or glycolytic inhibitors, reduce NADH levels and thereby reduce NADH:NAD+ ratio, whereas factors that inhibit oxidative metabolism, such as hypoxia and mitochondrial inhibitors, have the opposite effect. Glutamine provides ketone bodies (α-ketoglutarate) to fuel mitochondrial ATP production in the absence of glycolysis. Lactate dehydrogenase (LDH) maintains the lactate:pyruvate ratio in equilibrium with the cytosolic NADH:NAD+ ratio.


BHB is not directly a CtBP activator.
A drug that acts as an CtBP activator would be great for diabetes and anyone with brain inflammation.
Using BHB and C8 you would need to create enough ketones in your blood to reduce the glucose metabolism substantially, not by a trivial amount.
The easy to read version:- 

New research uncovers and replicates the mechanism by which a ketogenic diet curbs brain inflammation. The findings pave the way for a new drug target that could achieve the same benefits of a keto diet without having to actually follow one.

A keto state lowers brain inflammation
A keto diet changes the metabolism, or the way in which the body processes energy. In a keto diet, the body is deprived of glucose derived from carbs, so it starts using fat as an alternative source of energy.

In the new study, Dr Swanson and his colleagues recreated this effect by using a molecule called 2-deoxyglucose (2DG).
The 2DG molecule stopped glucose from metabolizing and created a ketogenic state in rodents with brain inflammation as well as in cell cultures. Levels of inflammation were drastically reduced - almost to healthy levels - as a result.
"We were surprised by the magnitude of our findings," said Dr Swanson. "Inflammation is controlled by many different factors, so we were surprised to see such a large effect by manipulating this one factor. It reinforces the powerful effect of diet on inflammation."
The restricted glucose metabolism lowered the so-called NADH/NAD+ ratio
"Cells convert NAD+ to NADH, as an intermediary step in generating energy from glucose, and thus increase the NADH/NAD+ ratio," he added.
When this ratio is lowered, the CtBP protein gets activated and attempts to turn off inflammatory genes. As Dr. Swanson told us, "CtBP is a protein that senses the NADH/NAD ratio and regulates gene expression depending on this ratio."
So, the scientists designed a molecule that stops CtBP from being inactive. This keeps the protein in a constant "watchful" state, blocking inflammatory genes in an imitation of the ketogenic state. Dr. Swanson said, "Our findings show that it is [...] possible to get the anti-inflammatory effect of a ketogenic diet without actually being ketogenic
The findings could apply to other conditions that are characterized by inflammation. In diabetes, for example, the excessive glucose produces an inflammatory response, and the new results could be used to control this dynamic.
"[The] ultimate therapeutic goal would be to generate a [drug] that can act on CtBP to mimic the anti-inflammatory effect of [the] ketogenic diet," Dr. Swanson concluded. 

Full Paper:- 


The innate inflammatory response contributes to secondary injury in brain trauma and other disorders. Metabolic factors such as caloric restriction, ketogenic diet, and hyperglycemia influence the inflammatory response, but how this occurs is unclear. Here, we show that glucose metabolism regulates pro-inflammatory NF-κB transcriptional activity through effects on the cytosolic NADH:NAD+ ratio and the NAD(H) sensitive transcriptional co-repressor CtBP. Reduced glucose availability reduces the NADH:NAD+ ratio, NF-κB transcriptional activity, and pro-inflammatory gene expression in macrophages and microglia. These effects are inhibited by forced elevation of NADH, reduced expression of CtBP, or transfection with an NAD(H) insensitive CtBP, and are replicated by a synthetic peptide that inhibits CtBP dimerization. Changes in the NADH:NAD+ ratio regulate CtBP binding to the acetyltransferase p300, and regulate binding of p300 and the transcription factor NF-κB to pro-inflammatory gene promoters. These findings identify a mechanism by which alterations in cellular glucose metabolism can influence cellular inflammatory responses.

The innate inflammatory response contributes to secondary injury in brain trauma and other disorders. Metabolic factors such as caloric restriction, ketogenic diet, and hyperglycemia influence the inflammatory response, but how this occurs is unclear. Here, we show that glucose metabolism regulates pro-inflammatory NF-κB transcriptional activity through effects on the cytosolic NADH:NAD+ ratio and the NAD(H) sensitive transcriptional co-repressor CtBP. Reduced glucose availability reduces the NADH:NAD+ ratio, NF-κB transcriptional activity, and pro-inflammatory gene expression in macrophages and microglia. These effects are inhibited by forced elevation of NADH, reduced expression of CtBP, or transfection with an NAD(H) insensitive CtBP, and are replicated by a synthetic peptide that inhibits CtBP dimerization. Changes in the NADH:NAD+ ratio regulate CtBP binding to the acetyltransferase p300, and regulate binding of p300 and the transcription factor NF-κB to pro-inflammatory gene promoters. These findings identify a mechanism by which alterations in cellular glucose metabolism can influence cellular inflammatory responses.

One way that CtBP regulates gene transcription is through interactions with the histone acetyltransferase HDAC1. 

Taken together, our findings indicate that metabolic influences that alter the cytosolic NADH:NAD+ ratio regulate NF-κB transcriptional activity through an NADH-dependent effect on CtBP dimerization. Conditions that reduce glycolytic flux, such as ketogenic diet and caloric restriction, can thereby suppress NF-κB activity, while conditions that increase glycolytic flux may increase it. These interactions provide a mechanism for the suppressive effects of ketogenic diet and caloric restriction on brain inflammation after brain injury. By extension, these interactions may also contribute to the pro-inflammatory states associated with diabetes mellitus and metabolic syndrome. 



Inhibiting NLRP3 and/or activating CtBP

You do not need to be a genius to see that inhibiting NLRP3 and/or activating CtBP, using the ketogenic diet, is likely to benefit some people with autism.
On the flipside, someone with colon cancer, where CtBP is over-expressed to the point where the cancer depends on it for growth, certainly would not want the ketogenic diet.
This cancer flipside we have seen before, antioxidants like NAC and Sulforaphane (via activating the redox switch Nrf2) are chemoprotective for healthy people, but bad for you if you have developed cancer.  Oxidative stress is very damaging to cancer cells and so it becomes a good thing. Some people who develop cancer then choose to improve their diet to include new healthy foods, sadly for some people this may actually be counterproductive.
Estrogen is another case in point, it has many positive effects and has been suggested to be one reason why women like longer than men. If you develop estrogen positive breast cancer, more estrogen is the last thing you would want.  

Other NLRP3 inhibitors 

                          

Coll et al. (2015) discovered that MCC950, a diarylsulfonylurea-containing compound known to inhibit caspase-1-dependent processing of IL-1β, also inhibits both canonical and non-canonical activation of the NLRP3 inflammasome. MCC950 inhibits secretion of IL-1β and NLRP3-induced ASC oligomerization in mouse and human macrophages. It reduces secretion of IL-1β and IL-18, alleviating the severity of EAE and CAPS in mouse models. Coll et al. (2015) further showed that MCC950 acts specifically on the NLRP3 inflammasome

Note that MCC950 is the new name for a drug Pfizer originally called CP-456773 or CRID3, which was not successful as a treatment for arthritis, but now has a second chance

Youm et al. (2015) discovered that the ketone metabolite β-hydroxybutyrate (BHB), but not acetoacetate or the short-chain fatty acids butyrate and acetate, reduced IL-1β, and IL-18 production by the NLRP3 inflammasome in human monocytes. Like MCC950, BHB appears to block inflammasome activation by inhibiting NLRP3-induced ASC oligomerization. Their in vivo experiments showed that BHB or a ketogenic diet alleviate caspase-1 activation and caspase-1-mediated IL-1β production and secretion, without affecting the activation of NLRC4 or AIM2 inflammasomes. BHB inhibits NLRP3 inflammasome activation independently of AMP-activated protein kinase, ROS, autophagy, or glycolytic inhibition. These studies raise interesting questions about interactions among ketone bodies, metabolic products, and innate immunity. BHB levels increase in response to starvation, caloric restriction, high-intensity exercise, or a low-carbohydrate ketogenic diet. Vital organs such as the heart and brain can exploit BHB as an alternative energy source during exercise or caloric deficiency. Future studies should examine how innate immunity, particularly the inflammasome, is influenced by ketones and other alternative metabolic fuels during periods of energy deficiency 
Although both MCC950 and BHB inhibit NLRP3 inflammasome activation, their mechanisms differ in key respects. BHB inhibits K+ efflux from macrophages, while MCC950 does not. MCC950 inhibits both canonical and non-canonical inflammasome activation, while BHB affects only canonical activation. Nevertheless both inhibitors represent a significant advance toward developing therapies that target IL-1β and IL-18 production by the NLRP3 inflammasome in various diseases. 

Type I Interferon (IFN) and IFN-β

In contrast to these newly described, NLRP3-specific inflammasome inhibitors, type I interferons (IFNs), including IFN-α and IFN-β, have been used for some time to inhibit the NLRP3 and other inflammasomes in various auto-immune and auto-inflammatory diseases. These diseases include multiple sclerosis, systemic-onset juvenile idiopathic arthritis caused by gain-of-function NLRP3 mutations, rheumatic diseases and familial-type Mediterranean fever.

These studies highlight the efficacy of type I IFN therapy and the need for future studies to elucidate the mechanisms of NLRP3 inflammasome inhibition. This work may improve clinical approaches to treating multiple sclerosis and other auto-immune and auto-inflammatory diseases.

Other Kinds of NLRP3 Inflammasome Inhibitors
Several additional ways for inhibiting the NLRP3 inflammasome have opened up in recent years. Autophagy, a self-protective catabolic pathway involving lysosomes, has been shown to inhibit the NLRP3 inflammasome, leading researchers to explore the usefulness of autophagy-inducing treatments  

Cannabinoid receptor 2 (CB2R) is an already demonstrated therapeutic target in inflammation-related diseases (Smoum et al., 2015). Work from our own laboratory (Shao et al., 2014) has shown that autophagy induction may help explain why activation of the anti-inflammatory CB2R leads to inhibition of NLRP3 inflammasome priming
Thus CB2R agonists similar to the HU-308 used in our work may become an effective therapy for treating NLRP3 inflammasome-related diseases by inducing autophagy.
Several other microRNAs have been reported to be involved in the activation of the NLRP3 inflammasome, including microRNA-155, microRNA-377, and microRNA-133a-1. Reducing the levels of these factors may be useful for treating inflammasome-related disease 


Conclusion regarding NLRP3 inhibitors

At this point in time BHB is clearly the best choice; at some point it would be expected that Pfizer will commercialize MCC950. 

 Further relevant papers: 

Inflammasomes are newly recognized, vital players in innate immunity. The best characterized is the NLRP3 inflammasome, so-called because the NLRP3 protein in the complex belongs to the family of nucleotide-binding and oligomerization domain-like receptors (NLRs) and is also known as “pyrin domain-containing protein 3”. The NLRP3 inflammasome is associated with onset and progression of various diseases, including metabolic disorders, multiple sclerosis, inflammatory bowel disease, cryopyrin-associated periodic fever syndrome, as well as other auto-immune and auto-inflammatory diseases. Several NLRP3 inflammasome inhibitors have been described, some of which show promise in the clinic. The present review will describe the structure and mechanisms of activation of the NLRP3 inflammasome, its association with various auto-immune and auto-inflammatory diseases, and the state of research into NLRP3 inflammasome inhibitors. 

NLRP3-inflammasome activates caspase-1 and processes pro-IL-1β and pro-IL-18 into the active cytokines. Two recent studies describe specific inhibitors of NLRP3 inflammasome that inhibit IL-1β release and inflammation. The specificity and potency of these compounds gives hope that a targeted approach to inhibit NLRP3-driven inflammation may be just around the corner



Activation of the inflammasome is implicated in the pathogenesis of an increasing number of inflammatory diseases, including Alzheimer’s disease (AD). Research reporting inflammatory changes in post mortem brain tissue of individuals with AD and GWAS data have convincingly demonstrated that neuroinflammation is likely to be a key driver of the disease. This, together with the evidence that genetic variants in the NLRP3 gene impact on the risk of developing late-onset AD, indicates that targeting inflammation offers a therapeutic opportunity. Here, we examined the effect of the small molecule inhibitor of the NLRP3 inflammasome, MCC950, on microglia in vitro and in vivo. The findings indicate that MCC950 inhibited LPS + Aβ-induced caspase 1 activation in microglia and this was accompanied by IL-1β release, without inducing pyroptosis. We demonstrate that MCC950 also inhibited inflammasome activation and microglial activation in the APP/PS1 mouse model of AD. Furthermore, MCC950 stimulated Aβ phagocytosis in vitro, and it reduced Aβ accumulation in APP/PS1 mice, which was associated with improved cognitive function. These data suggest that activation of the inflammasome contributes to amyloid accumulation and to the deterioration of neuronal function in APP/PS1 mice and demonstrate that blocking assembly of the inflammasome may prove to be a valuable strategy for attenuating changes that negatively impact on neuronal function. 

Scientists say new treatments for inflammatory diseases could be on the way

New treatments for inflammatory diseases could be on the way thanks to a significant discovery made by an international group of scientists, including some at Trinity College Dublin. 
The treatments could be used for a whole range of inflammatory disease including arthritis, Alzheimer's, multiple sclerosis, Parkinson's, gout, asthma and Muckle-Wells syndrome.

The researchers have found that a molecule, previously developed and then abandoned by a multinational pharmaceutical company, can block one of the key drivers of a plethora of inflammatory conditions.
The molecule, MCC950, was produced by Pfizer two decades ago as a possible treatment for arthritis.
However, the company discontinued its efforts to bring the drug to market, and the intellectual property rights on it subsequently lapsed.
Around eight years ago, scientists at Trinity's Biomedical Sciences Institute led by Professor of Biochemistry Luke O'Neill came across the compound and began to explore its potential uses.
They subsequently discovered that it could effectively block the NLRP3 inflammasome.
Inflammasomes are a complex of molecules that trigger inflammation when exposed to infection or stress.
They have been identified as promising therapeutic targets for researchers in recent years.
The NLRP3 inflammasome has been found to be a common activator of a key process in certain inflammatory diseases.
The discovery by the research team, details of which are published in the journal Nature Medicine, confirms that all inflammatory diseases share a common process, although the part of the body which experiences the inflammation might differ.
The scientists subsequently carried out trials on mice and found that the molecule stopped the progression of multiple sclerosis and sepsis.
They also carried out testing on samples taken from humans with Muckle-Wells syndrome, a rare auto-inflammatory disorder, and discovered it was equally effective.
The scientists also say that it is likely the drug could produce fewer common side-effects, such as susceptibility to infection, than other anti-inflammatory drugs, and could prove cheaper and capable of being administered orally.
The next stage will involve testing the compound on humans and a wider group of diseases.
The researchers say for certain conditions, like Muckle-Wells syndrome and asthma, such trials could take place as early as two to three years from now, as the drug had already undergone some human testing by Pfizer.
However, even if the trials prove the drug is safe and effective, they stress that it could be ten-15 years before it could be fully approved for use in humans for the treatment of more complex diseases like multiple sclerosis or Alzheimer's.
They also stress that while the molecule could become an effective treatment, it will not be a cure, though it is possible it could be effective in undoing some of the damage done by well progressed cases of certain diseases.
Prof O'Neill and his team now plan to form a company to further develop and test the compound.
MCC950 is also currently being tested on mice in the US for anti-ageing properties, as there is a growing school of thought that inflammation is responsible for much of the ageing process - a theory which has come to be known as "inflammaging".
The study, part funded by Science Foundation Ireland and the European Research Council, was carried out by a collaboration of six institutions, including the Universities of Queensland, Michigan, Massachusetts and Bonn. 

Conclusion

I am amazed at all the potentially good things that ketones and KD can do for many people’s health and it is all based on science from very serious institutions. 






51 comments:

  1. Great post Peter. One thing I should point out that I know you already know, but may have overlooked is that Nicotanimide Riboside will directly raise NAD+ levels, thereby lowering the NADH/NAD+ ratio which is desirable for many reasons, especially with regards to the specific reasons you mention in this post. I give my son 250mg a day to address the issue of blocking L-Kynurenine into the brain (L-Kynurenine is necessary to produce NAD+ but the previous step in its synthesis involves Quinolinic Acid which can be exitotoxic in high levels if not converted into NAD+ by an enzyme and Vitamin B6) and sometimes wonder if more would do more. I know in an obesity study that I think is still ongoing, the participants were receiving 2000mg of NR per day. NR is getting cheaper due to competitive reasons in the marketplace but it is still not cheap. Of course, autism is very expensive as well. Also, high dose NR has been shown to be safe in adults, but obviously no studies on children, especially prepubescent children, have been done that I am aware of.

    ReplyDelete
    Replies
    1. Tyler, thanks for highlighting this. The question is how much Nicotanimide Riboside you would need to activate CtBP. Perhaps someone should suggest this to Dr Swanson at UC San Francisco?

      Delete
    2. I don't know the answer to that question. To get a guestimate of I suppose you would figure out how much the NADH/NAD+ ratio declines from NR which there is research on and then compare those numbers to this study. You also might get useful information searching around for NMN (Nicotanimide Mononucleide) which is what they inject directly into animals to raise NAD+ levels directly, and see if there are any studies looking at CtBt.

      Delete
  2. Here is a very large and very important new study concerning autism diagnosis via metabolite profiles which found a ratio between glutamine, glycine, and ornithine with BCAA's reliably predicted autism in 16.7% of the study participants.

    Press Release:

    https://www.sciencedaily.com/releases/2018/09/180906082016.htm

    Paper:

    https://www.biologicalpsychiatryjournal.com/article/S0006-3223(18)31793-1/fulltext

    The study in effect suggests (not directly) that BCAA supplementation may be useful in a subset of those with autism due to low levels of BCAA's in the blood, allowing other amino acids that compete with BCAA's (such as tyrosine, phenylalanine, and tryptophan) to more readily pass the blood-brain barrier and raise levels of these amino acids higher than they should be. Also, the levels of BCAA's themselves were not predictable of autism, rather the levels of glutamine, glycine, and ornithine, relative to BCAA's were inversely correlated with autism diagnosis. In other words, raising BCAA levels and lowering levels of glutamine, glycine, and ornithine "could" help a significant subset of people with autism. I stress "could" because correlation does not equal causation and a study employing translational research on this matter would need to be done first to draw any conclusions.

    Nevertheless, considering I give my son BCAA's for different reasons, this research is very interesting. I also give Hydrolyzed Collagen as well with his BCAA's which I may need to consider as Hydrolyzed Collagen is exceptionally high in Glycine which this study suggests might be wise to lower in the subtype of autism labeled in this study.

    ReplyDelete
  3. Hi Tyler, since I stopped Bcaas and started inulin my son developed a facial tic and difficulty in concentration, he started to regress after a month of making progress. I also give him magnesium glycinate. Do you think that going back to Bcaas and suspending inulin could help to return him to normal? Also magnesium glycinate can be worsening the problem?
    Valentina

    ReplyDelete
    Replies
    1. I have seen a LOT of regressions mentioned by parens connected to inulin. I think people who are doing okayish and need to take just a few more steps towards a place they are content with should not risk it. Especially since the regression is very hard to gain back, as far as people on various forums say. I personally have this attitude that if your kid is hitting walls and has a high atec, by all means drop therapy bombs, try anything under the sun. If you are at an atec of 20-40 and your child is soooo close to functioning normally, try only things without huge regression possibilities. Just not worth the risk at that point.

      Delete
    2. I give my child both inulin and BCAA's (not at the same time obviously as the powders tend to clump together) and have had none of these issues, but your child is older and has different symptoms and may still have dopamine receptor supersensitivity due to the D2 antagonist he was using for a long-time.

      Nonetheless, I don't think you will get facial tics from inulin though, at least not directly. Some people do have microbial overgrowth in their intestines and too many prebiotics can cause more harm than good, though with autism you usually have an issue of having a reduced diversity of bacteria and prebiotics can help maintain that diversity on up to perhaps adulthood where the gut microbiota stabilizes, though this is not an agreed upon fact as it had been thought for a long-time that the gut microbiota stabilizes by age 3, but a recent study on urban people (including children) living an indigenous lifestyle in Bolivia for a few months suggests that a hunter-gatherer diet and lifestyle can improve microbial diversity up to age 7 or 8. This is just one study of course.

      Also, is there a reason you believe there is a contraindication with regards to BCAA's and Inulin? BCAA's are rapidly absorbed while Inulin is not absorbed as it is fermented into short-chain fatty acids once it reaches the colon which can take 12 hours or so.

      Also, I have no idea why magnesium of any form would cause any issues other than the laxative issues some forms of magnesium can cause in large amounts.

      The facial tics again are likely a dopamine or a dopamine receptor issue in the striatum. If not that, then perhaps a GABAergic issue in the output structures of the basal ganglia (globus pallidus internus and substantia nigra pars reticulata). There is some evidence straight GABA (which is cheap) will help improve basal ganglia function so other than everything else you have tried so far, I would try that as well to alleviate the tics whether you are still using BCAA's or not.

      Delete
    3. Tatjana, your are absolutely right, there was no need to risk, my son is close to functioning normal, his issues are mainly sound hypersensitivity and also dopamine supersensitivity. I didn´t intend to follow NP, I got there by another way,looking for something to feed a bacteria with antiepileptic effect, and stambled upon it.
      Valentina

      Delete
    4. Tyler, thanks, my son´s profile was totally confirmed with this issue. I have GABA at home and will give it to him. I dont think that there is a contraindication with regards to BCAAs and inulin,good for your son, I shouldn´t have taken it out. I don´t know if start with BCAAs again at the same time as GABA or not. With regards to magnesium glycinate, if it had laxative effect would be great, my son has chronic constipation,I think it is used more for sleeping problems, that also has my son. I asked you because it was suggested to lower glycine in the study you mentioned.
      Valentina

      Delete
    5. Just to be clear, correlation does not equal causation, so the higher relative levels of irnithine, glycine, and glutamine could be the effect of a problem, rather than a cause. High levels of ornithine and glutamine could suggest a urea cycle disorder which for all I know could be the reason my son benefits from Aspartic Acid since it along with Ornithine helps converts ammonia to urea and glutamine. And again the absolute levels of these amino acids are not high in this study, but rather they are high relative to BCAAs.

      Delete
  4. Awesome post. Have anyone tried some of this possible treatments on his child? 2DG molecule, BHB or at least the kd diet. Apparently this should work better than bumetanide as goes directly to the brain.

    The only doubt is the long place effects on health when you give daily any drug.

    ReplyDelete
    Replies
    1. un tio, some children with epilepsy + autism are on the KD for many years. The KD is usually seen as medical diet, but the Modified Atkins Diet appears to achieve a similar result.

      Many people with autism in the US are taking MCT Oil or Coconut oil which will produce some BHB. There are lots of comments on the web.

      The more potent option to produce BHB is to use C8 oil (caprylic acid) and/or a BHB supplement.

      C8/MCT goes to your liver where BHB is produced and then it goes through your blood all over your body, just like bumetanide would. Only some of it will enter the brain.

      BHB/C8/KD is just another possible therapy. In some people it could be an add-on to bumetanide. It is not a case of either/or.

      Delete
    2. I agree about using both: bumetanide and BHB/C8/KD if a child is a responder and needs treatment.

      Long term safety is always of concern, but at least in my profoundly affected child, the risk of leaving things as they are is obviously higher.

      With regard to adverse effects, bumetanide is well studied and side effects are predictable/manageable. It has been successfully prescribed off label in France for about 10 years. KD has long been used in children, which I cannot say about BHB/C8. My son seems to tolerate BHB/C8 well, while I myself developed quite nasty side effects after switching to another C8 brand. You don't risk that with bumetanide, it's rx drug of pharmaceutical quality.

      Peter, are you aware about the European approval of bumetanide for autism? I did not follow that in details, but thought it's the matter of the near future. Actually, I recall 2017/2018 was once suggested. Now I've just learned Phase III trial by Servier will take another year:

      http://apps.who.int/trialsearch/Trial2.aspx?TrialID=EUCTR2017-004419-38-NL

      Do you know what is the real perspective?

      Delete
    3. Agnieszka, I have not recently asked about the bumetanide approval timetable.

      The trial dosage is too low in my opinion. They are using 0.5mg twice a day. That dose did not have a significant effect in my son. 1mg once a day was much more effective and for a long time I have been using 2mg once a day.

      It would be a shame if the trial fails.

      I am a fan of BHB/C8 as an add-on therapy. The BHB powder seems to have the same effect as Ketoforce liquid. I hope when my 10kg of C8 finally arrives it does not give side effects and just works like the first brand I tested.

      Delete
    4. Peter, I don't understand why they plan to use 0.5mg twice a day dose.

      The last year French trial conclusion was: "bumetanide improves the core symptoms of ASD and presents a favorable benefit/risk ratio particularly at 1.0 mg twice daily." Isn't it a step back to use less?

      I am glad to hear that you found BHB powder beneficial. Is it potassium BHB salt? Do you consider 6g dose optimal?

      Delete
    5. Agnieszka, I believe the problem is that children go hypokalemic and I expect Servier do not want the trial to fail because of this. I find this odd, since all you have to do in 80% of cases is to assume there will be a drop in K+ and prepare for it, by adding dietary potassium and a small supplement. I believe the French approach is to let the child go hypokalemic, measure their K+ and then give a K+ syrup. As a result instead of 20% having side effects, up to 100% will have them.

      So now the trial may fail because the dose is too weak to cross the BBB. Never mind.

      If I lived in the US I would use KetoBlitz (which includes some BHB as acid not a salt).

      I am using Primaforce, which was the product without too much Ca2+, that is sold by iHerb. A Chinese producer of the bulk chemicals says that the calcium salt of BHB is not well absorbed and we know that Ca2+ is a core problem in autism. While calcium in food is not an issue, calcium supplements may have strange effects.

      I would ideally give more than 6g. I am happy to have 400mg of K+. I do consider buying 100% KBHB and adding a small amount (containing 200mg of K+) to my Primaforce product.

      Ideally I will use a larger dose of C8 as a means to producing BHB, without the problem of K,Na,Mg,Ca. This is the next phase of my investigation.

      Delete
  5. Many many thanks Peter and Agniezska! And what quantity daily of coconut oil or MCT or C8 should be taken??

    ReplyDelete
    Replies
    1. un tio, there is no easy answer to your question, because it depends on what the problem is. Some people use a tiny dose of MCT (eg 2.5ml) and say it is beneficial all day long. There is no way that is giving the brain a boost from ketones as a fuel, the dose is far too low. In Alzheimer's they suggest 40ml of C8 will give a tangible "fuel boost".

      I found 6g of BHB with 20ml of C8 gives a tangible, but not profound benefit. With a 50% lower dose the effect was minimal.

      Delete
  6. I am posting this here just because its the newest post, but the topic is not strictly connected - I see that a company called Rosemont Pharma the Uk has a syrup of Clonazepam which would make life for everyone trying to replicate Peters therapy oh so much easier. Anybody has any idea how to get it? I know how to get stuff which is available in my country, but I dont know the UK rules and who might be able to bend them and how.

    ReplyDelete
    Replies
    1. If you can get a prescription from a helpful local doctor, you can access drugs in most other European countries. Only in some southern countries in Europe are pharmacists willing to bend the rules. The UK is super strict, for example doctors will not/cannot prescribe off-label drugs to their own child with autism and write to me to ask what to do.

      Delete
    2. we actually know a pharmacist practising in a country nearby and I was thinking of asking him to perhaps make the liquid for us. i try to eliminate all complications before i start a therapy because I believe that sticking with a regimen of supplements, next to speech therapy, sports lessons, hair washing, plain parenting etc, can be very tiring and this is not a sprint, its a marathon.

      Delete
  7. Robert Naviaux who has been discuseed in length on this blog has a new paper out suggesting his CDR theory for autism can be more broadly applied to a whole range of chronic diseases:

    Press Release:

    https://www.sciencedaily.com/releases/2018/09/180909103211.htm

    Paper:

    https://www.sciencedirect.com/science/article/pii/S1567724918301053?via%3Dihub

    The paper is very long and very dense and worth a read even though much of what he presents is very controversial to some people in his field as are any hypotheses which go along the lines of "all disease is caused by this one reason". Nevertheless, the paper is open access so anyone can read it if they choose to without having to go to unorthodox or very expensive means of obtaining scientific publications.

    The press release also mentions he is planning a larger clinical trial for autism in 2019 of what I assume will be low-dose suramin to subjects with autism. He has been trying to get funding for quite a while now so it will be interesting to see how that goes.

    ReplyDelete
    Replies
    1. Tyler, very interesting paper indeed and I suggest everyone takes a look, at least at the press release version.

      Delete
    2. Thank you Tyler.
      Zoe

      Delete
  8. Tyler,
    I’m sure you’ve seen this:
    https://www.erchonia.com/erchonia-submits-data-to-us-fda-to-support-low-level-laser-510k-market-clearance-for-autism/
    Are you still using LLLT? What could be so special about this 640nm Erchonia Spectrum Laser as opposed to another device that emits 640nm LED? Bc my pulsaderm that I use for *my* skin emits 630-660nm. My son likes it but I haven’t tried any regimen for him with it.
    Thanks for any insights

    ReplyDelete
    Replies
    1. I don't user a laser, but rather an array of infrared LEDs that are typically used for security purposes in illuminating an area with infrared light for infrared security cameras to capture. The LEDS are 850nm. A longer wavelength will have more of the photons get through the skull and hit the brain. 660nm is likely to only penetrate the skin and therfore only useful for topical purposes.

      Delete
    2. I was at a therapy place called Terrasuit that uses the Masgutova reflex integration method on various people. I was mightily impressed by the family of a car crash victim who was left paralyzed down from his neck and was able to gain back the use of his hands by this method. But to come back to lasers - they also use for the same purpose something called a QRI laser. My at the time 3 year old extremely hyperactive and sound sensitive daughter would sit for a whole hour with bliss on her face while they did the reflex points laser therapy - while the laser emits a beeping high pitched sound. I will actually acquire one next month - it costs a lot but since her atec is now 32 and we are looking for small stepsntaking us down 3-4 points at a time withou much risk, this is it. I can say that from my experience and what other people also tell me, it works wonders.

      Delete
    3. Tatjana, there actually is a very recent study using cold/low-level laser therapy in autism.

      Effects of Low-Level Laser Therapy in Autism Spectrum Disorder.
      https://www.ncbi.nlm.nih.gov/pubmed/29956199

      Delete
    4. I have seen it, it has been widely shared on the QRI facebook group. A lot of parents have the problem that their spouse wants a scientific study to back up every single treatment they include so this study is important for many of them. The QRI costs 3200 usd so its a serious purchase but on the other hand the effects can be gauged from the fact that you would find maybe 2-5 of them being resold used per year, mostly for reasons of someone being financially in hardship.

      Delete
  9. Here is some very interesting new research directly pertinent to this blog post which shows beta-hydroxybutyrate prevents vascular senescence, thereby indirectly providing an anti-aging effect as vascular issues are the harbinger of other diseases to come, just as high blood pressure is the number one indicator of impending mortality:

    Press Release:

    https://www.sciencedaily.com/releases/2018/09/180910160626.htm

    Paper:

    https://www.sciencedirect.com/science/article/pii/S1097276518306051?via%3Dihub

    Vascular issues are obviously a very important topic with regards to autism, as is oxidative stress on endothelial cells which can cause reduced blood flow to the brain and impart poor cognitive function in someone with autism who has oxidative stress issues. It looks like beta-hydroxybutyrate seems to be protective in vascular function when challenged by stress, such as the stress incurred naturally from aging, or the hypothesized vascular stress from some autism phenotypes.

    The obvious question is would just supplementing ketone BHB supplements directly improve vascular function in the manner described in this paper, or do you need to constantly be in a state of ketosis which has mixed health effects, or do you actively need to fast every day for 24 hours.

    It is an open question, but one that would be very helpful if answered with regards to autism interventions such as fasting, the ketogenic diet, and BHB/Caprylic Acid supplementation.

    ReplyDelete
  10. The obvious question is would just supplementing ketone BHB supplements directly improve vascular function in the manner described in this paper, or do you need to constantly be in a state of ketosis which has mixed health effects, or do you actively need to fast every day for 24 hours.
    approximately inflammation

    ReplyDelete
  11. It’s intriguing that so many roads to autism (and speech dysfunction) lead to the microglial NLRP3 inflammasome and its vicinity. I know you have written extensively about this before Peter but maybe the topic deserves a revisit.

    “A conservative estimate based on the research suggests that at least 69% of individuals with an ASD diagnosis have microglial activation or neuroinflammation”
    (Relevance of Neuroinflammation and Encephalitis in Autism, https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4717322/)

    From the blogpost above and later comments from the community we learned that both keto/BHB therapy and clemastine can raise the amount of produced communication - even in cases with very different etiology. BHB is an antagonist of the NLRP3, while clemastine regulates the purinergic P2X7 receptor that can activate the NLRP3 (“Signal 2”).

    Another, perhaps unexpected, intervention here is mefenamic acid, which acts as a selective antagonist of the NLRP3 inflammasome (https://www.nature.com/articles/ncomms12504). We know it is now developed to treat “minimally verbal pediatric autism”.

    It is almost impossible to not mention Suramin here too, which isn’t a very useful intervention but still is very effective at making nonverbal autistic kids talk after several years of mutism. Suramin antagonizes purinergic P2X and P2Y receptors that sense certain DAMPs/danger-associated molecular patterns (more specifically ATP) and then activate NLRP3.

    A couple of the community’s favorite interventions also act at the NLRP3 and its neighborhood.

    Statins, proposed by Peter, attenuate TLR4 and NF-κB expression (simvastatin) and/or inhibit the release of the DAMP HMGB1 (both simvastatin and atorvastatin).

    Cannabinoids have a role in attenuating the activation of microglia / release of inflammatory cytokines (please anyone fill me in why/how)

    Verapamil has neuroprotective effects via binding to/inhibiting microglial NADPH oxidase (and I think memantine does this too).

    SIRT1 inducers inhibit a coactivator of NF-κB.

    Resveratrol, quercetin, curcumin, propolis, ginseng, sulforaphane; all have different effects (See https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5031844/)

    Pushing it a bit, even the inhibition of aquaporins (Bumetanide inhibits AQP-1 and AQP-4) can limit the release of IL­1β from NLRP3­activated macrophages.

    I also found this alternative figure of the NLRP3 inflammasome: https://www.frontiersin.org/files/Articles/487654/fimmu-10-02538-HTML/image_m/fimmu-10-02538-g001.jpg
    It suggests that a substance called parthenolide, found in the supplement Fewerfew also could act beneficially at several targets. The bioavailability is said to be low, but according to anecdotal reviews Fewerfew has an effect on migraine (microglia are involved in pain?).

    ---------------------------
    I think more than one intervention is needed to silence NLRP3 enough so it stops killing brain cells. Several pathways (autoimmunity, high glucose, bacterial infections, epilepsy, sterile inflammation) lead down to it from different angles. Most likely one needs to shut several doors to get optimal results, but of course without hazarding a required baseline inflammatory response.
    In one sentence, I think the NLRP3 inflammasome deserves more attention as it to me seems very central.

    /Ling

    ReplyDelete
    Replies
    1. I should have added the link to the paper with the NLRP3 inflammasome figure too, as it is relevant:

      Pharmacological Inhibitors of the NLRP3 Inflammasome (2019)
      https://www.frontiersin.org/articles/10.3389/fimmu.2019.02538/full

      /Ling

      Delete
    2. The role of Cl− and K+ efflux in NLRP3 inflammasome and innate immune response activation | American Journal of Physiology-Cell Physiology
      https://journals.physiology.org/doi/full/10.1152/ajpcell.00421.2021

      Delete
    3. Nitazoxanide also decreases nlrp3 inflammasome, il-1b, and il-6.

      https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.15949

      -Stephen

      Delete
    4. Wouldn’t then mefenamic acid actual activate NLRP3, since it activates K+ efflux?

      Delete
    5. Thats a great question, I don't have the answer but Peter might. I just recently met with Dr. Jyonuchi and she has termed a interesting form of ASD.
      "Autoinflammatory syndrome affecting inflammasomes."

      "Activation of inflammasome
      is regulated by multiple
      proteins like NLRP3.
      Inflammasome is activated
      by non-specific mediators
      derived from microbes or
      tissue injuries. Some
      vaccines also can activate
      inflammasomes causing
      sterile inflammation - fever,
      headache, joint ache, etc.
      This is attributed in part to
      the release of inflammatory
      cytokines (IL-1ß, IL-18)from
      inflammasomes."

      Treatment-colchicine/anakinra

      Lab-
      Monocytes to diagnose

      -Stephen

      Delete
    6. I guess Emodin also works on nlrp3 inflammasome.

      https://pubmed.ncbi.nlm.nih.gov/25867480/

      Delete
  12. Ling, thanks for posting - it is very interesting.

    There is also glyburide/glibenclamide mentioned in the picture. While it is suggested that too high dose would be needed to target inflammasome pathway, low dose which does not induce hypoglycemia was found neuroprotective here:

    https://pubmed.ncbi.nlm.nih.gov/19023097-protective-effect-of-delayed-treatment-with-low-dose-glibenclamide-in-three-models-of-ischemic-stroke/

    Well, not in the humans with autism and maybe not in this mechanism.

    Feverfew looks very interesting to me considering benefits reported in migraine and cluster headache.

    Regarding several doors to be shut, I wonder what can be done on trigger avoidance level?

    ReplyDelete
    Replies
    1. Ah, I didn't even bother to check those other substances in the figure, but you are right and now I see that glyburide is a drug for treating diabetes. Looks like the NLRP3 inflammasome is highly involved in Type 2 diabetes. I also missed to check dimethylfumarate, but it also has a suppressive effect on NLRP3.

      I'm not sure what you mean with 'trigger avoidance level'? I'm thinking there are too many triggers to control (allergy, gut issues, epilepsy, sterile inflammation, HSPs, unfolded proteins, ATP...)

      /Ling

      Delete
    2. I'm not going to pretend I understand much of this article, but I wish I did. The reason I found it was because MFA inhibits the NLRP3 inflammasome via blocking the "volume regulated anion channel" (VRAC) and I had not heard about that channel ever before.

      Volume-regulated anion channel – a frenemy within the brain
      https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4752865/

      The reason I bring it up even though I can't decipher it myself is that it touches on several things mentioned above. It mentions this VRAC in association with microglia, bumetanide, NKCC1/KCC2, migraines and even purinergic signaling (and accidentally even hyponatremia).
      If anyone wants to feel smarter than me, please go ahead and explain some of it. :)

      /Ling

      Delete
    3. Glibenclamide, mentioned in Agnieszka's comment above, resurfaces as a treatment that possibly could alleviate opioid-induced pain. The reason I find this article interesting is because it mentions many things we've learned about on this blog: ER stress, all three arms of the unfolded protein response (PERK/eIF2α, ATF6, IRE1α/XBP1), heat shock protein 70 (HSP70), NMDAr, PKA and an ATP-gated potassium channel.

      To be honest, it is a bit over my head how this could be useful in autism, but I'll put the link here for anyone to find as it hits too many paths to be ignored.

      /Ling

      Delete
    4. If I forgot the link in my previous comment on Glibenclamide, it's here:
      https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7243285/

      /Ling

      Delete
  13. One of the DAMPs mentioned above that can trigger the NLRP3 is HMGB1. It figures as a common cause/pathway affected in cognitive decline in a range of conditions:

    HMGB1: A Common Biomarker and Potential Target for TBI, Neuroinflammation, Epilepsy, and Cognitive Dysfunction
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6142787/

    "Inhibiting the HMGB1/RAGE/TLR4 signaling axis could be a novel therapeutic strategy against several HMGB1 mediated conditions like TBI, neuroinflammation, epilepsy and cognitive dysfunction"

    Prolonged elevation of plasma HMGB1 is associated with cognitive impairment in intensive care unit survivors

    "Cognitive impairment is an important part of post-intensive care syndrome [..]. The underlying pathophysiology of cognitive impairment in ICU survivors is not well understood, but experimental evidence indicates that prolonged inflammation may play an important role
    [..]
    High Mobility Group Box 1 (HMGB1), an alarmin released in tissue damage and inflammation, remains elevated long after a severe systemic inflammatory insult and causes hippocampal inflammation and cognitive impairment in experimental animals. Interestingly, anti-HMGB1 therapy administered several days after critical illness can attenuate cognitive decline in mice"

    Another pathway that triggers the NLRP3 goes via TXNIP.
    Picture-googling is best to get a quick overview, like here: https://www.researchgate.net/figure/A-potential-role-for-TXNIP-in-hyperglycemia-induced-oxidative-stress-ER-stress_fig1_332384020

    This pathway can be activated by for example diabetes/hyperglycemia, aberrant UPR (PERK and IRE-1 branches) and oxidative stress. There are several inhibitors here, like metformin, quercetin, icariin, lipoic acid (downstreams), fatty acids, verapamil, PPARa agonists and caloric restriction... probably even more.

    I find it useful to look at the NLRP3 from different gateways, because if you can match what interventions have had best impact so far you might be able to follow that gateway to the source of inflammation in your case.

    /Ling

    ReplyDelete
  14. Here's an article on the relation between NRF2 and NLRP3 - it is not yet clear.
    https://onlinelibrary.wiley.com/doi/full/10.1002/eji.201646665

    "Here, we present evidence that Nrf2 ablation as well as Nrf2 activating compounds block activation of the NLRP3 inflammasome. Whereas Nrf2 ablation only dampens inflammasome activation, Nrf2 activators are able–‐dependent on the dose–‐to completely block caspase‐1 activation. This suggests that two different molecular mechanisms are underlying NLRP3 inflammasome inhibition by Nrf2 ablation and Nrf2 activating compounds.
    (..)
    NLRP3 inflammasome activation induces rapid Nrf2 degradation (..)
    Cell death induced by inflammasome activation can be essential for immunity due to killing of intracellular pathogens. In addition, this cell death contributes to the limitation of the inflammasome‐dependent inflammatory response. Nrf2 activation would be detrimental in this context, because it supports survival of cells.
    (..)
    The mechanisms of action of DMF in patients suffering from psoriasis or MS are only poorly characterized. Our results suggest that DMF exerts its anti‐inflammatory activity—at least in part—via inflammasome inhibition. This raises the possibility that DMF may also be useful for the treatment of other inflammatory diseases with an involvement of inflammasomes. In addition, SFN might be even more efficient in these patients, as our results demonstrate inflammasome inhibition at lower SFN concentrations in vitro and in vivo compared to DMF."

    /Ling

    ReplyDelete
  15. Social stress, propranolol and NLRP3? Maybe not all three at the same time. But an intriguing link to the spleen…

    I saw here that propranolol is useful for anxiety and language/speech issues in autism. As these all are high-priority topics for me I couldn't resist to check for connections to the NLRP3.

    According to the below thesis and cited papers, propranolol blocks neuroinflammatory responses in the brain but the NLRP3 does not mediate the anxious behavior caused by previous social stress.

    “Specifically, increased production of monocytes in the bone marrow, storage in the spleen, and increased trafficking to the brain is associated with the neuroinflammatory stress response. This has been confirmed using propranolol to block monocyte circulation and accumulation, resulting in decreased pro-inflammatory cytokine production. [..]
    Here we show that the peripheral immune response to stress is preserved when NLRP3 is genetically knocked out. This is confirmed by the increase of myeloid cell production in the bone marrow and increased myeloid cell populations in the spleen and in circulation. We also show that the NLRP3 inflammasome is not necessary for development of anxiety-like behaviour in response to stress.”

    https://kb.osu.edu/bitstream/handle/1811/81629/1/Sobol_Thesis.pdf
    ----------------

    Interestingly, I found another paper that partly mirrors the above one.
    Here they replaced microglia after stress induction in mice to see what would happen. The priming of microglia was responsible for initiating and preserving the inflammatory ‘memory’ of the previous stress experience. However, microglia repopulation after stress did not affect the behavioral hypersensitivity to stress.

    The authors mention previous work where removal of the spleen blocked recurrence of anxiety and suggest that myeloid cells of the spleen act as a sensitized reservoir of monocytes that can be trafficked to the brain upon stress (mediated by the sympathetic nervous system).

    https://www.researchgate.net/publication/328524514_The_Influence_of_Microglial_Elimination_and_Repopulation_on_Stress-Sensitization_Induced_by_Repeated_Social_Defeat

    Removing the spleen sounds drastical when propranolol is around.

    /Ling

    ReplyDelete
  16. Another NLRP3 inhibitor at the horizon, Dapansutrile, this time for acute gout and possibly for heart failure too:

    The first phase 2A proof-of-concept study of a selective NLRP3 inflammasome inhibitor, dapansutrile™ (OLT1177™), in acute gout

    https://ard.bmj.com/content/78/Suppl_1/A70

    /Ling

    ReplyDelete
  17. And let's add a link from the NLRP3 inflammasome to those Tregs that act as the thermostat of our immune system:

    "In this study, we described that NLRP3-deficient mice generate increased Treg populations. Moreover, NLRP3 knockdown in CD4+ T cells exhibits increased Treg differentiation
    ()
    These results demonstrated that pro-inflammatory NLRP3 can repress the differentiation of Tregs by diminishing Foxp3 expression and suggest a role of NLRP3 as a new negative regulator of Treg differentiation."
    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6879348/

    It's not a surprise to find another one of the FOXP genes involved here too, though I think this one is not as involved with language as the others.

    /Ling

    ReplyDelete
  18. You can use colchicine the gout med for nlrp3

    https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10013297/

    ReplyDelete
  19. You can also use sodium benzoate to decreases nlrp3 as well as increasing BBB integrity.

    https://www.mdpi.com/2077-0383/11/13/3716

    -Stephen

    ReplyDelete
    Replies
    1. Yes sodium benzoate is very interesting, it is cheap and classed as a food additive. Note that people who lack DAO and so are histamine intolerant have to avoid it.

      Definitely should be trialed to see if there is a benefit.

      Delete

Post a comment