• Welcome to Phoenix Rising!

    Created in 2008, Phoenix Rising is the largest and oldest forum dedicated to furthering the understanding of and finding treatments for complex chronic illnesses such as chronic fatigue syndrome (ME/CFS), fibromyalgia (FM), long COVID, postural orthostatic tachycardia syndrome (POTS), mast cell activation syndrome (MCAS), and allied diseases.

    To become a member, simply click the Register button at the top right.

Ponderings and speculations about purinergic signaling, in pursuit of a unified ME/CFS theory

necessary8

Senior Member
Messages
134
Ponderings and speculations about purinergic signaling, in pursuit of a unified ME/CFS theory, Part 1

Introduction

ATP, while inside the cell serving for energy transport, when outside of the cells has signaling functions. Those functions differ between cell types, but a lot of it is pro-inflammatory, where extracellular ATP serves as a danger signal, so-called Damage Associated Molecular Pattern, DAMP. This is because ATP should normally be mostly contained inside the cells, and spills outside when a cell is ruptured, either by mechanical means or by an invading pathogen, therefore making a good danger signal to start the immune response we know as inflammation.

Dr. Naviaux proposed that overactive purinergic signaling sustains many chronic diseases, including ME/CFS and autism. In the case of autism he's also proven that this seems to be the case, by causing symptom remissions with IV administration the anti-purinergic drug suramin. He thinks the same might be happening in ME/CFS, and Alan Light's research which indicates that extracellular ATP is a key ingredient in facilitating the feeling of fatigue itself, supports this hypothesis.

For me, Naviaux's theories seemed fascinating, and made a lot of sense, but many questions arose, like how do we get different diseases from a unified danger response, how does it tie in with other observations in ME/CFS, how exactly does this cause our symptoms, etcetera. Naviaux never explained his theories in detail, only gave an incredibly simplified version at the Stanford symposium. Some details can be understood reading his papers about CDR and oxidative shielding, but those prompted even more questions in me. So I did the crazy thing and decided to dive into Google Scholar trying to understand this stuff myself by reading related papers. As it currently stands, I am very far from any form of full understanding of this subject, but nevertheless, I wanted to share my findings with you all, first to save some trouble other people interested in it, second because there are people here who understand biochemistry better than me (Im gonna tag a few - @nandixon, @Hip, @JaimeS), and might help me in refining this. And third, because our amazing Ron Davis apparently likes to get ideas from the community, which is why I'm tagging @Janet Dafoe (Rose49) to pass this post to him. I imagine he heard a lot of this from Naviaux and other scientists, but maybe a thing or two will spark a novel thought. I'm sorry it's so long, but I assure you I put a lot of thought in which of my findings include here and which not.

Biochemical background for the uninitiated

Purinergic receptors, which detect the presence of ATP and other purines outside the cell, consist of P1, P2X and P2Y families. P2X receptors are the ones most specific to ATP and seem to be most well studied.

ATP release from the cells can also occur without the cell rupturing, but rather to send the danger signal on purpose. ATP can be released vesicularly, but the more prominent and well studied method seem to be pannexin and connexin hemichannels, particularly Panx1. It's basically a protein on the surface of the cell, which when activated creates a large pore, permeable to molecules up to 900Da in size, including ATP. Those have been shown to open in various cell types in response to stressful stimuli - mechanical stretching[source], hypoxia[source]. This is in line with the model of eATP serving as a danger signal.

Interestingly, in some cells, Panx1 channels can be directly opened by activation of the P2X7 receptor.[source] They kinda bundle together and activation of the P2X7 activates Panx1 as well. By this mechanism, when a cell senses large amounts of extracellular ATP, it also releases ATP to propagate the danger signal to the next set of neighboring cells.

So what does eATP do to the cells, other than make them release more ATP? Well, depends on the cell. In this Part 1, I will mostly focus on the immune system.

When a pathogen enters the body, macrophages and dendritic cells (which are in the tissues) detect them through Toll-Like Receptors, becoming activated. They then secrete Il-1β and TNF-α, major cytokines that start the inflammatory response. This secretion process is hugely dependent on co-stimulation by eATP mainly through the P2X7 receptor. eATP also serves as a signal for those cells to migrate to the infection site in the first place.[source] It also is a necessary factor for neutrophils to get there, as it potentiates the Il-8 mediated chemotaxis through the P2Y2 receptors.[source] The secretion of Il-8 from activated monocytes also requires P2Y2 and P2Y6 stimulation in them.[source]
Macrophages and dendritic cells engulf the pathogens, break them down, and present their peptides on MHC proteins to T cells, activating them if their receptors correspond to the presented peptide. T cells express P2X1, P2X4, P2X7 receptors, and Panx1. All of those, with the exception of P2X7, translocate to the immune synapse (the connection between the T cell and the antigen presenting cell), and ATP is released from the T cell, amplifying the activation signal by acting on its own P2 receptors.[source] The activated T cell produces IL-2, which acts on the T cell itself, triggering it's clonal expansion. Presence of eATP outside of the immune synapse, acting on P2X7, can also increase IL-2 secretion in activated T cells.[source] Th cells then activate B cells which produce antibodies, and cytotoxic T cells go killing infected cells in the body. B cells also express purinoreceptors, but the effects of their activation have not been studied.

Basically, extracellular ATP is an important co-factor in almost every step of immune activation, having pro-inflammatory effects. Except for NK cells. For NK cells, the presence of extracellular ATP actually reduces their cytotoxic activity.[source] (Sound familiar?)


The good stuff

ATP is not the only pro-inflammatory molecule, the immune system has a large number of cytokines to regulate itself as well as other Damage Associated Molecular Patterns, and Pathogen Associated Molecular Patterns. In a lot of studies assessing purinergic signaling in immune cells, they found that many immune cell types have two or more pathways for activation of their primary functions - one through purinergic receptor stimulation by ATP, and other through direct contact with a pathogen, or through cytokines. Those pathways are often needed to act in synergy to achieve a full-blown immune activation.[source] Given all this, it is possible, that a "kind of" inflammatory response is perpetuated in ME/CFS by purinergic signaling. There was an infection (or other triggers), which initiated classical immune response, which resolved the infection, and initiated anti-inflammatory immune resolution, but that resolution succeeded only partially. We might not see very elevated cytokines in ME/CFS because that kind of signaling has already been mostly resolved, while eATP pro-inflammatory signaling still persists. Now, obviously those are not fully separate, but it's very possible they're not always completely in sync either. It could be that they're only kinda-connected. This could explain the finding of Montoya's and Mark Davis's cytokine profiling, that more severe patients have higher pro-inflammatory cytokines, but are still not high enough to distinguish from controls.[source] Those severe patients would have stronger eATP-mediated inflammation, which elicits higher cytokine levels, but because that connection is weak, the cytokine levels don't quite raise above the norm.

But why doesn't this happen in healthy people? What's the difference between them and ME/CFS patients? To answer this, there is a simple approach and a complex approach.

The simple approach is to go "Wait, eATP makes cells release more ATP? This is a positive feedback loop. There must be some inhibitory mechanism of this, otherwise the whole immune system would go into runaway endless inflammation". And there is. ATP can be hydrolyzed to AMP and then to adenosine, by enzymes called nucleotidases. Nucleotidases present outside the cell, or ecto-nucleotidases, are indeed expressed by many cells which also have purinergic receptors. The main such ecto-nucleotidase, which seem to be responsible for most of eATP breakdown is NTPDase1, also called CD39.[source1][source2] It was shown in many studies to downregulate purinergic signaling, first by reducing eATP concentrations, and second, along with 5'-NT (CD73), it produces adenosine, activating some of the P1 receptors, which have anti-inflammatory functions in many immune cells.[source]

The obvious hypothesis from this is that CD39 function is impaired in ME/CFS. Now, I'm unsure of this because I don't know the specifics of the test, but I'm under the impression that this, to some extent, can be tested in Ron's impedance meter. First, you can introduce recombinant CD39 into the plasma and see if it reduces the signal in ME/CFS cells. Second, you can take healthy cells (in healthy plasma) and treat them with the CD39 inhibitor, ARL 67156, and see if the impedance response starts resembling that of ME/CFS. It might be necessary to add extracellular ATP also, to mimic nearby cells secreting it, as it (presumably) happens in ME/CFS. You'd do two variants - one with just eATP, and one with eATP and ARL 67156. Another way might be to add a non-hydrolyzable ATP derivative, which would activate P2 receptors, but won't be hydrolyzed by CD39. The obvious limitation of this is that it only tests white blood cells, and what's happening in the tissues might be a different story.

So, if CD39 activity really was inhibited in ME/CFS, what could be the reason? I see three possibilities, and to talk about them, I first need to talk about Treg cells. T regulatory cells, formerly known as T suppressive cells, are responsible for control and suppression of the immune response. They are also the most confusing little shits ever.[source] To a lot of questions about them, the answer is "data is not conclusive". But we do know that they are the main source of CD39 and CD73 in the immune system, expressing them in higher amounts than other cells.In fact, breaking down ATP to adenosine with those two enzymes is thought to be one of the main ways in which Tregs suppress the immune system.[source1][source2][source3]

So the three possibilities are:

1. Not enough Tregs positive in CD39 and CD73 are being produced.
Not all Tregs express CD39, only a specific CD45RO+CCR6+ effector/memory-like subset of them, also called TREM. Interesting observations have been made in multiple sclerosis, where the overall number of Tregs was not much different from controls, but the amount of Tregs positive in CD39 was much lower in MS patients.[source] It is possible that a similar thing is happening in ME/CFS, perhaps by a different mechanism.

2. Not enough CD39+ Tregs are being activated. Tregs, like any other T cells, have a T Cell Receptor (TCR) which upon stimulation activates the cell to its primary function. It has been observed in mice, that Tregs exhibit CD39 activity only when activated.[source] It probably works similarly in humans. As for when Tregs become activated, that is where the data starts being non-conclusive[source], so I'll leave it at that.
This could also lead to a lowered number of them, as Tregs highly express the P2X7 receptor, and are very sensitive to high concentrations of eATP. Without the protection of CD39-mediated ATP hydrolysis, they easily die, or their suppresive capabilites are inhibited.[source]

3. Direct obstruction of CD39 activity.

This third option is the most interesting to me, as it ties some stuff together, but I also don't know if this is possible the way I think it is.
Let's go back for a moment to Ron's impedance assay. The plasma switch results indicated that it is something in the blood that is making the cells behave differently than healthy ones. After reading all this you might think that this something is eATP, but it is most probably not. First, I have seen no publications indicating possible endocrine ATP signaling, only short-range paracrine (to neighboring cells) and autocrine (to self). Second, the filtration results showed that this factor in the plasma is larger than 10kDa, which ATP is not.
But then you also have the result that adding suramin "greatly reduced" the sickly response. Suramin is a non-selective P2 antagonist, blocking many of the P2X and P2Y receptors, to varying degrees. So how can a similar response be achieved by blocking ATP signaling as by filtering out large stuff from the plasma?

The answer can be an anti-CD39 antibody. This antibody would be present in ME/CFS blood and would inhibit ATP breakdown, increasing the activation of P2 receptors, opening of Panx1 channels, resulting in loss of ATP from the cell, constant sort-of-inflammation and many of our symptoms. When it is filtered out, CD39 can work and hydrolyze ATP, returning the cells to normal. When P2 receptors are blocked directly, the cells also return to normal.
This would explain why B cell depletion with Rituximab helps (activated B cells produce antibodies) and why TGF-β was one of the only two elevated cytokines in Montoya's and Mark Davis's findings[source]; TGF- β is one of the other ways in which Tregs suppresses the immune system. It might be so, that the Tregs are working hard to resolve the inflammation, but cannot do it fully because CD39 is blocked by an antibody.
This is an idea that the immune system is basically attacking its own stop button. Or rather a part of it.

There are two problems with this hypothesis, and I have no idea if they can be resolved. First, ME/CFS plasma makes healthy cells behave like ME/CFS cells. In this hypothesis the factor this plasma introduces is the anti-CD39 antibody. But for this antibody to do anything at all to the cell, eATP must be already present. And the cells are healthy, from a human without an active infection. Do certain levels of ATP release occur at all times enough for the blockage of CD39 to make a difference? Or was some eATP floating around in the ME/CFS plasma? I don't know, but it seems like a stretch.

The second problem is the humoral immunity profiling ME/CFS study done by Lombardi et al.[source] They used random peptide microarrays to check all the antibodies in the blood and what peptides they bind to. The aminoacid pattern they identified as a common thing was GVALSG. I checked, it doesn't match the sequence of CD39 at all. Now, I don't know the technical limitations of that study and the possibility that it would miss a key antibody.

Recombinant (synthetic) anti-CD39 antibodies are a thing though, so maybe this can be tested in the impedance assay.

There is another similar possibility that doesn't involve CD39 at all, and that is that the P2 receptors are being activated by some other agonist, not ATP, and that agonist also is larger than 10kDa and is our mysterious plasma factor. Maybe it could be an antibody. Just in case, I compared the sequences of all seven P2X receptors to the GVALSG pattern, and the best I got was 3 aminoacids in a pattern. I don't know much about antibody binding, but I find it hard to imagine a sequence of 3 would make for any meaningful binding. As for a non-antibody protein capable of P2X agonism, the only two I've found so far (mostly by accident, as I almost haven't touched this angle yet) are biglycan[source] (43kDa) and Serum Amyloid A[source] (11-14kDa, depending on subtype). (This is a super-long stretch, and probably not related, but I thought I would also mention it - particles of the NLRP3 inflammasome, which is activated in macrophages by eATP to release IL-1β, can also be released extracellularly, and probably are also a DAMP.[source])

Another possibility that I haven't explored in detail, is that the mysterious blood factor upregulates P2 receptor expression in cells, making them more vulnerable to eATP signaling. Alan Light did find upregulated P2X4 and P2X5 expression in ME/CFS patients post-exercise.[source1][source2] This would have the same problem as the anti-CD39 hypothesis in the sense that it doesn't seem to explain healthy cells becoming sickly in ME/CFS plasma.

So what about other inhibitory mechanisms of purinergic signaling, other than ecto-nucleotidases? The only other thing that I found, were some observations that P2X7 activation can inhibit Panx1-mediated ATP release.[source] Yes, this is the reverse of what I said at the beginning. There were two studies that reported this, one on murine cells, other on human HEK293 cells. This is a cell line cultured from embryonic kidney cells, often used in research because it's easy to modify them to do what you want, so they have different properties based on how you "bake them". In this case, Panx1 was artificially introduced to those cells through transfection.

Those observations raise two possibilities. First, that depending on the cell type, P2X7 activation might open or close Panx1 channels. The cell types in which P2X7 activation has been observed as leading to Panx1 channel opening are macrophages[source], astrocytes[source], and enteric neurons[source]. (And maybe more, I just haven't found the papers about it.) The P2X7-mediated Panx1 activation seems to be widely accepted in literature, but citations about it are often not present. It is very possible that in some cells P2X7 activation opens Panx1 and in other types closes them.
The second, more interesting possibility, is that there is some other factor, which decides if P2X7 activation in a cell opens or closes Panx1 channels. A distinction on P2X7 splice variants might be it.[source] Or it might be some yet undiscovered thing, possibly our mysterious blood factor, or something tied to it biochemically. We don't really know.

On this anticlimactic note, I want to conclude Part 1.

Possible future ponderings

What about the complex approach that I mentioned earlier? The complex approach is to understand exactly how does resolution of inflammation work. How does antibody synthesis stop. When are Tregs activated and when are they suppressed. How do Panx1 channels close. How does this all tie together in a system that "knows" when pathogen extermination is over and when it's time to suppress the inflammatory response. Then we can try to find out what in that sequence might have gone wrong in ME/CFS. My thoughts on this will come in Part 2, if I don't get overwhelmed by it. Probably will take me quite a long while.

Now, in the title, I put "in pursuit of a unified ME/CFS theory". That's the goal, but I'm nowhere near it. I feel like before we can talk about even a speculatory unified theory of ME/CFS, four other areas need to be addressed in detail.

First is the connection to the gut. In the gut, weird things happen with the immune system. For example, intestinal CD8+ T cells have higher P2X7 receptor expression than CD8+ (cytotoxic) T cells found in other places of the body. This causes them to be very sensitive to eATP, in a manner similar to Tregs. I wonder what's their CD39 expression like. The digestive system also has its own variation of Tregs to not allow rampant immune reactions to the food passing through it, and to the natural intestinal bacteria. Could it be possible that reduced CD39 activity causes more pathological immune response in the gut, resulting in our digestive symptoms and destroying good bacteria? Maybe. Could it also tie back into why the CD39 activity is lower, in a feedback loop? I don't know. I need to read more.

Second is the connection to the brain. In the nervous system eATP is a neurotransmitter. It also activates microglia, which are basically the nervous system's macrophages. Jarred Younger's research points to microglia activation in ME/CFS and it could also explain some, or all, of our neurological symptoms, as well as the common dysautonomia. There could also be a feedback loop in here.

Third is the connection to exercise and PEM. The exacerbation of symptoms after exercise is a very distinct, hallmark symptom of ME/CFS and is the main thing that makes ME/CFS such a disabling condition. I have yet to see a plausible biochemical model of PEM, and I believe this is a compulsory requirement for any general theory of ME/CFS. The delayed nature of PEM and the fact that mental exercise can also cause it, can be important clues to what it is biochemically.

Fourth is the intracellular stuff. This is the hard part, because of how complex it is, as there are hundreds/thousands of proteins involved in the functioning of any cell. But if we are to tie this to PDH inhibition, glycolysis inhibition, and to the metabolomics findings, we need to understand what does purinergic signaling do inside the cell, how it impacts the cellular metabolism. The impedance assay results I think show that the signal difference is due to insufficient ATP inside the cell to pump out the salt. This is the only thing injecting ATP or pyruvate into the cell, and adding suramin to the plasma have in common - they increase the ATP in the cell. The question is - is the insuffucient ATP due to the loss through pannexin channels, or due to impaired ATP production, or both? If both, are they connected? In which direction? I have some ideas about how to answer those questions with the impedance assay, but I would have to know more about the specifics of it first, because I might have some incorrect assumptions here (Ron, if you're reading this, can I ask you a few questions about the assay?)
Theorizing about intracellular stuff is even more difficult because it will differ depending on cell type, but a common core is very possible. Dr Naviaux certainly seems to think this is the case, because his Cell Danger Response theory is exactly this. I still don't understand many things about it, like why are the metabolomics results in ME/CFS the reverse of an acute CDR. But it certainly makes sense in a lot of ways.

So, as for future continuations of this. The gut and brain connections I will probably explore in detail when I have the time and energy for it, and write part 3 and part 4 about it. The PEM aspect is more difficult to search for existing publications. The best way would be for a specialist in exercise physiology to look over all of the indicated pathways, and tell if something rings a bell as something they know as changing with physical exercise. (If any of you know good papers about cell-to-cell communication during exercise, send them my way) And the intracellular thing... .___. I don't wanna do it. That stuff is hard. But if someone else wants to try, I want to present to them the most fucking confusing paper I have yet found. It states that CD39 (which is outside the cells), makes the cells have less ATP *inside*. Yes, this is exactly opposite to everything I said here. Further, it states that Tregs have lower intracellular ATP that Th cells, and those low levels make cyclophosphamide able to selectively kill Tregs, sparing other T cells. Yes, cyclophosphamide is the drug currently being trialed in Norway (along with Rituximab) as a treatment for ME/CFS based on some case studies showing that it helps. Now, this specific confusion I can resolve, it's just dose-dependent. Tregs are more sensitive to cyclophosphamide, so it only kills them, boosting the immune response (used in cancer), but high-dose cyclo kills all T cells, suppressing the immune response (used in autoimmune disease). As for why the rest makes no sense - I have no idea. It can be an error in the methodology of the study, or the results might be correct and tie into our illness in a clever way that I'm too stupid to understand. Or it might be totally unrelated.

Implications for treatment

To close things of, I wanted to touch a bit on treatment, albeit very carefully. Please don't take my words as medical advice.

If rampant eATP signaling is at the core of ME/CFS, then suramin by blocking it could indeed by very beneficial. However. If the reason for this is impaired ATP hydrolysis through ecto-nucleodidases, there is another thing to consider. Suramin, other than its P2 antagonism, was also shown to inhibit ecto-nucleotidases.[source] The question is if this effect is less or more potent than the P2 antagonism. I would imagine less, based on how the autism trial went. But there is also the issue of adenosine not being produced and not activating the P1 anti-inflammatory receptors. Now, I'm not saying that suramin is a bad idea and we shouldn't try it in ME/CFS. My very uninformed guess is that overall the effect would still be positive. But it might not be. So I just wanted to point this out.

There's also couple other interesting things I found in regard to treatment.
Firstly, a couple of years ago, the AstraZeneca pharmaceutical company filed a patent for AZD9056, a potent, orally bioavailable, selective P2X7 inhibitor. It was to be used in rheumatoid arthritis. The drug was tolerated well, but failed to bring any symptomatic change in a phase IIb clinical trial, so it never reached the market. I have no idea if they would be interested in doing this, but what if AstraZeneca could pay for an ME/CFS clinical trial of AZD9056?
Secondly, it seems that the food dye FD&C Blue No. 1, also known as Brilliant Blue FCF, or E133, is a Panx1 inhibitor with neutral effect on P2X7.[source] I don't really know what dose would be needed for it to have any meaningful effect, and if that dose is still known to be safe, but it might be possible to calculate this if we can somehow find out its bioavailability. Just please don't go eating some dangerous amount of this food dye, okay? I'm kinda wary of sharing this last thing in an open forum, but I'm gonna trust you guys to be responsible.
Thirdly, there are some herbs known to be P2 inhibitors. @Jesse2233 beat me to the punch in making a thread about it, as he usually does. There are more than just the two he mentioned though. I might write a post about them after I look more into it.

Thanks for reading.
 
Last edited:

A.B.

Senior Member
Messages
3,780
The answer can be an anti-CD39 antibody. This antibody would be present in ME/CFS blood and would inhibit ATP breakdown, increasing the activation of P2 receptors, opening of Panx1 channels, resulting in loss of ATP from the cell, constant sort-of-inflammation and many of our symptoms. When it is filtered out, CD39 can work and hydrolyze ATP, returning the cells to normal. When P2 receptors are blocked directly, the cells also return to normal.
This would explain why B cell depletion with Rituximab helps (activated B cells produce antibodies) and why TGF-β was one of the only two elevated cytokines in Montoya's and Mark Davis's findings; TGF- β is one of the other ways in which Tregs suppresses the immune system. It might be so, that the Tregs are working hard to resolve the inflammation, but cannot do it fully because CD39 is blocked by an antibody.
This is an idea that the immune system is basically attacking its own stop button. Or rather a part of it.

Interesting idea. @Jonathan Edwards' model of B cell autoimmunity is conceptually similar I believe (immune signals that harm the body while simultaneously driving further immune response and thereby creating a vicious cycle of autoimmunity).

A while ago I had a similar thought about an autoimmune loop that passes through B cells and P2X receptors, although I speculated about an anti-P2X receptor antibody (I'm not an expert, just someone who can't help but to think about these things). If Naviaux and Fluge & Mella are both correct then simplest explanation seems to be indeed that there is an antibody that activates these pathways while simultaneously promoting production of more this antibody.
 

Jesse2233

Senior Member
Messages
1,942
Location
Southern California
Absolutely fascinating @necessary8 thank you for synthesizing this into a coherent framework. It must've taken hours

I will need to read this a few times to thoroughly digest it, but a few questions jumped out initiallly.

1. Is there an existing readily available assay that can measure serum eATP (or a closely related biomaker?)

2. If hypoxia is one cause of eATP release might amerliorating that through hyperbaric oxygen therapy be one of the mechanisms through which that treatment improves some ME patients?

3. What are some of the other anti-purinergic compounds you've found?
 

dreampop

Senior Member
Messages
296
I feel like I just got an education. But seriously, this is very well written, and you make complicated issues palatable for someone whose brain is a mess. Would treatments that increase CD39+ expression be an alternative to purinergic antagonism?
 

Jonathan Edwards

"Gibberish"
Messages
5,256
Interesting idea. @Jonathan Edwards' model of B cell autoimmunity is conceptually similar I believe (immune signals that harm the body while simultaneously driving further immune response and thereby creating a vicious cycle of autoimmunity).

I agree. An anti-CD39 autoantibody would be a nice concept here.

My one thought about sharpening the analysis would be to remove all reference to 'pro-inflammatory'. The reason for this is that we are now dealing with cellular responses at a level of detail that makes 'inflammation' too clunky a concept to be of any help. When I worked on the cellular mechanisms for RA I started making progress when I threw out vague terms like inflammation and just stuck to pathways. 'Pro-inflammatory' is a bit like 'go-faster' as applied to the red stripes people used to put on their cheap cars in the 1960s. It makes you feel you mean something but doesn't actually do anything. And we have no real evidence for any inflammation in ME anyway.

So a co-stimulus given by a T cell to a B cell to survive and proliferate is a meaningful idea, but if the B cell produces the right sort of antibody it will prevent inflammation by removing microbes. In most autoimmune diseases the antibodies do not cause inflammation - they have specific effects on tissue function.
 

bertiedog

Senior Member
Messages
1,738
Location
South East England, UK
Can somebody explain how this theory would fit with the fact that as a group ME sufferers are split into those that get very frequent viruses/infections and the other group who just don't.

My immune function is always on the low side with below the range white blood cell count and I do struggle fighting endless viruses which always seem to be almost identical in the way they start and finish. On the other hand there are many here who never seem to get anything. I also have an autoimmune condition, i.e. Hash's so that would seem to complicate things further? Just trying to understand this interesting theory.

Pam
 

Murph

:)
Messages
1,799
Big thanks to @necessary8 for this terrific analysis! So clearly written. You have a gift! Which is not to understate the hard work that also clearly went into this. Kudos to you.

A question: if "rampant eATP signaling is at the core of ME/CFS", how do we explain this:

"Davis has found that pyruvate, a substance which bypasses glycolysis, and ATP – a signaling molecule (outside the cell) – makes ME/CFS patients cells look healthy again"

https://www.healthrising.org/blog/2...-explored-open-medicine-foundation-symposium/

You suggest Davis injected the cells with ATP but I got the sense he was testing Naviaux's theory, and bathing them in it. Is there any clarity on this?

You can also see the result in the blue line in this chart, which Davis presented at the recent Stanford symposium:

Screen Shot 2017-10-30 at 11.27.01 PM.png


The terminology 'exposed to' to my mind suggests eATP not intracellular ATP. But I will happily be wrong because I'd really rather the theory held together. eATP as friend not foe would certainly be a curve ball.

Of course we don't know much about what the impedance is measuring nor whether in vitro and in vivo results are comparable so it need not be the end of the purinergic signalling theory. Especially because suramin reduces the impedance difference between the cells of healthy controls and sufferers, Ron claims.

I can't wait to see the next parts of your analysis and if you find an endothelial link I'd be keen to learn more about it. My quick search of pubmed for "cd39 + endothelial" certainly threw up a few interesting results.
 
Last edited:

necessary8

Senior Member
Messages
134
Glad you guys like it : ) Thanks for all the kind words.

It must've taken hours
I wish. The writing and revision portion itself was a week of putting all my energy into this.

1. Is there an existing readily available assay that can measure serum eATP (or a closely related biomaker?)
Okay, imagine a tissue, with a bunch of cells next to each other, and with capilaries connecting them to the bloodstream. When a cell releases a substance, and that substance acts on the cell itself, this is called autocrine signaling. When the substance acts on nearby cells in the tissue, this is called paracrine signaling. When the substance enters the bloodstream, travels a large distance in the body, and then acts on a completely different set of cells in different tissues, this is called endocrine signaling.
eATP has been shown to exhibit autocrine and paracrine signaling, but not endocrine. So serum levels of it might be quite useless.
In theory, a larger number of ATP molecules released overall in the tissues of the body, could, maybe, possibly, translate higher levels in the blood also. But it might not.
And yes, immune cells are found in the blood too. But the processes mediated with eATP that i described earlier, take place mostly in the tissues (macrophage/dendritic cell activation) and in lymph nodes (T cell activation and clonal expansion)
eATP amounts directly outside of T cells in the blood might be some indicator here, but they might not be. They also would be mostly present in what's called "the unstirred water layer" right outside the cell, but I don't know if it's possible to prepare the cells for a test like that without disrupting this layer.

But there is something else we can check pretty easily, and that is the state of the pannexin/connexin hemichannels. You put a dye of with correctly sized molecules outside of the cells, and use a microscope to see if the dye flows into the cells. The faster the dye flows, the more hemichannels are open.
(this is a bit different than the luciferase assay mentioned by Jaime. luciferase catalyzes the direct reaction of luciferin with ATP to produce light, measuring the quantity of ATP by the intensity of the light)
Maybe this test would show us some results in ME/CFS cells copared to controls. (It might still not, because of the reasons I mentioned earlier)

If hypoxia is one cause of eATP release might amerliorating that through hyperbaric oxygen therapy be one of the mechanisms through which that treatment improves some ME patients?
Probably no. I don't know that much about hypoxia, but as it's a fairly well-studied phenomenon, I imagine that if it occured in ME/CFS we would have biomarkers by now.

What are some of the other anti-purinergic compounds you've found?
The most comprehensive list was hidden in plain sight, on wikipedia's page about purinergic signaling.

Would treatments that increase CD39+ expression be an alternative to purinergic antagonism?
Maybe, but it's not quite that simple. CD39 is expressed by many cell types in different quantities, maintaining a set of delicate balances between ATP hydrolysis and release, that make the immune system (and other systems) work. We need to figure out *if*, *where*, and *how* CD39 activity is impaired, and then we can think about fixing it. If we meddle with CD39 expression without this knowledge, we might unbalance the immune system even more.
That, and also purinoreceptors are just a much easier and direct target from pharmacological point of view.

Can somebody explain how this theory would fit with the fact that as a group ME sufferers are split into those that get very frequent viruses/infections and the other group who just don't.
I don't have a satisfyingly elegant answer for this, but I wanted to point out that a large amount of flu/cold symptoms are actually the inflammatory response of the body. As I've mentioned earlier, classic inflammatory response requires co-stimulation by both pathogens (detected by activation of TLRs and TCRs), as well as eATP. If we have overactive eATP signaling, it would make sense for our immune system to activate more readily in response to a virus. The "don't get colds at all" group is more tricky, but it's possible that some sort of immune exhaustion or compensatory immune suppression is making the inflammatory response not activate at all when it should. The presence of those might be dependent on genetics, or some environmental factors.

A question: if "rampant eATP signaling is at the core of ME/CFS", how do we explain this:

"Davis has found that pyruvate, a substance which bypasses glycolysis, and ATP – a signaling molecule (outside the cell) – makes ME/CFS patients cells look healthy again"
This is the main question I have for Ron about his assay. I earlier assumed that the ATP and pyruvate was injected into the cells, because Ron talked about making the technology to do that, and because he said it circumvented the possible block in glycolysis, which would make sense only if the ATP/pyruvate was in the cell.
But I started having doubts about that assumption, so I want to ask him about it. I already asked Jaime earlier, but she doesn't know.

If the ATP was introduced outside of the cells, there is still a way to make this all work, depending on what concentration it was. The pannexin/connexin hemichannels are basically just holes in the cell membrane. There is no fancy one-directional transport here. Under normal conditions, ATP has orders of magnitude higher concentrations in the cells than outside, so when the channels open, it flows out. But if in his tests Ron introduced extracellular concetrations of ATP higher than the intracellular ones, the ATP would actually flow into the cell, making the cell have more energy for operating its sodium-potasium pump, to pump out the excess salt, therefore behaving like a healthy cell and not rising in impedance.
This can be tested by introducing ATP along with a direct hemichannel blocker, like carbenoxolone, which should result in the increased impedance signal not disappearing, despite the addition of ATP, if this model is correct.
 

A.B.

Senior Member
Messages
3,780
The "don't get colds at all" group is more tricky, but it's possible that some sort of immune exhaustion or compensatory immune suppression is making the inflammatory response not activate at all when it should. The presence of those might be dependent on genetics, or some environmental factors.

An immune suppression so strong the immune system does not noticably react would presumably lead to a swift death.

I think what's happening is that patients are more resistant to colds and flus. The metabolic changes could create unfavorable conditions for infections to take hold.
 

necessary8

Senior Member
Messages
134
An immune suppression so strong the immune system does not noticably react would presumably lead to a swift death.

I think what's happening is that patients are more resistant to colds and flus. The metabolic changes could create unfavorable conditions for infections to take hold.
Yeah, I thought about this also. I'm not sure if it would lead to a swift death, but I a viral infection without the inflammatory response should probably have some dire consequences for the body. So yeah, your version makes more sense, especially in the context of Naviaux's CDR theory.
 

necessary8

Senior Member
Messages
134
My one thought about sharpening the analysis would be to remove all reference to 'pro-inflammatory'. The reason for this is that we are now dealing with cellular responses at a level of detail that makes 'inflammation' too clunky a concept to be of any help.
This is true, and for the most part, I try to stick to the pathways as well. In this piece, when I use "pro-inflammatory", it just means "contributing to the activation of a cell/pathway responsible for pathogen extermination". It's just a bit of a mouthful to say every time, and I wanted my post to be understandable to people without advanced biochemical knowledge. It is still a generalization, and as you suggest, a "sharper" analysis (or one operating on a lower abstraction level) would stray away from this concept, but that will come more in Part 2. (Probably.)
 
Last edited:

Hip

Senior Member
Messages
17,820
A fantastic theory @necessary8! Reading it has certainly added a wider context and deeper understanding of Naviaux ideas; but moreover, it provides a possible framework for understanding what might be the fundamental pathophysiology of ME/CFS. Thanks for your efforts in researching this, thinking it through, and writing it up in such a clear way.

Just in case there are others who, like me, due to brain fog are struggling to get to grips with your theory, I have made some summary points here:



Key Points in the ATP Signaling Theory of ME/CFS by Necessary8

ATP is normally contained within a cell, but spills out into the extracellular spaces when the cell is ruptured — for example, in circumstances when a cell is infected by a virus and is ruptured by viral lysis. So the appearance of extracellular ATP outside the cell serves as a good danger signal, as it flags up that cells may have been damaged or infected by a virus.

Intact cells located nearby a ruptured cell spilling its ATP can further amply this extracellular ATP danger signal by means of these cells' P2 receptors, such as P2X receptor (and to a lesser extent P2Y and P1), which sense the presence of extracellular ATP, and respond by getting the intact cells to release even more ATP into the extracellular spaces (P2X does this by triggering a mechanism based on the protein Panx1, which creates large pores in the intact cell, which in turn releases more ATP from inside the cell into the extracellular spaces). P2X thus acts as a positive feedback loop, whereby the presence of extracellular ATP from a ruptured cell will trigger the release of even more extracellular ATP from adjacent healthy cells.

Extracellular ATP is an important co-factor for nearly all aspects of immune activation. Thus any ATP that is released into the extracellular spaces you can think of as a pro-inflammatory signal; and without this extracellular ATP, you cannot adequately activate many facets of the immune response.

In order to prevent a runaway activation of the immune response from the P2X positive feedback loop, there is also a counter-balancing negative feedback loop which mops up the extracellular ATP. This moping up of extracellular ATP is performed by CD39, a molecule which along with CD73 is found in especially high amounts on T-regs. CD39 and CD73 also reduce immune activation by producing adenosine, which has anti-inflammatory effects in many immune cells. So CD39 and CD73 are the anti-inflammatory counter-balance to the pro-inflammatory extracellular ATP.

The hypothesis that @necessary8 posits is that CD39 function may be impaired in ME/CFS, leading to unregulated immune activation and inflammation from extracellular ATP. Three possible ways that CD39 may become impaired are suggested:

(1) Not enough T-regs that express CD39 and CD73 are being produced.

(2) The T-regs that express CD39 are not being activated (in mice, CD39 is only switched on when the T-reg cell is activated).

(3) Inhibition of CD39 activity, which might be caused by an anti-CD39 autoantibody. In other words, the immune system may be mounting an autoimmune attack on its own stop button, CD39.

Another possibility that @necessary8 posits is that CD39 may be working fine, but that the P2 receptors are being activated by some unknown agonist in the blood other than ATP. Or that there is some factor in the blood up-regulates P2 receptor expression in cells. These would also lead to immune activation and inflammation.

In terms of treatment possibilities, compounds that inhibit Panx1 or the P2 receptors may reduce the immune activation driven by extracellular ATP and insufficient CD39. @necessary8 points out that the food dye Brilliant Blue FCF is a Panx1 inhibitor, and of course suramin, the drug proposed by Dr Naviaux, antagonizes P2X7 and P2Y2.

I also found some other P2X7 and P2Y2 antagonists listed below, including the dye Brilliant Blue G, cassic acid from rhubarb, and the flavonoid supplements kaempferol and tangeretin.



Panx1 Antagonists:
  • Brilliant Blue FCF food dye is a selective inhibitor of the ATP release channel Panx1, with an IC50 of 0.27 μM. Ref: 1
P2X7 Antagonists:
  • Brilliant Blue G (aka: Coomassie Brilliant Blue) dye antagonizes P2X7 receptor, with an IC50 of 200 nM (= 0.2 μM). Ref: 1 Brilliant Blue G is not used as a food dye, but is a close relative of the food dye Brilliant Blue FCF. Some medical applications for Brilliant Blue G are discussed here.
  • Lidocaine (one of Dr Jay Goldstein's ME/CFS treatments) antagonizes P2X7 receptor, with an IC50 282 μM. Ref: 1
  • Rhein / cassic acid (found in rhubarb) antagonizes P2X7 receptor, with an IC50 of 1.31 μM. Ref: 1
  • Calcium, magnesium, zinc, copper ions antagonize the P2X7 receptor, with IC50 values of 2,900, 500, 11 and 0.5 μM respectively. Ref: 1 Note: the lower the IC50, the more potent the antagonism. Interesting how magnesium injections are a classic ME/CFS treatment.
  • Puerarin (from the herb kudzu, Pueraria lobata) antagonizes P2X7 receptors. Ref: 1 This comes from @Jesse2233's thread.
  • Rheedia longifolia methanol extract antagonizes P2X7 receptor, with an IC50 of 1.31 μg/ml. Ref: 1
  • Emodin antagonizes P2X7 receptor, with an IC50 of 200 to 500 nM (= 0.2 to 0.5 μM). Ref: 1
  • Colchicine inhibits P2X7 receptor-associated pore opening at EC50 = 290 to 540 μM. Ref: 1 Prof Jose Montoya uses colchicine as an ME/CFS treatment.
P2Y2 Antagonists:
  • Kaempferol and tangeretin (flavonoid supplements) antagonize P2Y2 receptor, with IC50 values 6.6 and 12 μM respectively. Apigenin, quercetin and rutin have IC50 values of > 25 μM. For reference, for P2Y2 the suramin IC50 = 31 μM. Ref: 1



EDIT: I calculated in this post that many of the above inhibitors although they work in vitro will not be effective in vivo when you take them orally. Though high dose magnesium injections (or high dose transdermal magnesium cream) may be a viable P2X7 receptor inhibitor (high dose magnesium is an old and well-known helpful treatment for ME/CFS).

I calculated in this post that oral Brilliant Blue FCF might have some useful Panx1 antagonism in vitro.
 
Last edited:

necessary8

Senior Member
Messages
134
Thanks for the summary, @Hip

The only thing I want to correct you on, because this is pretty important, is that while Brilliant Blue FCF is a food dye, Brilliant Blue G is not. It is a textile dye, found for example in blue jeans. It is not approved for use in food. (I'm like 90% sure of this, if you find a direct source stating that it is approved for ingestion, link it please) So don't eat that one.

As for FCF, could you help me calculate the necessary dose, based on the IC50 in my source? You're probably better at that than me. I couldnt find its oral bioavalibility (kinda figured, since its not a drug), but let's assume a 100% at first and work from there.

We should also probably move to Jesse's thread with this, to keep this one about the theories and mechanisms of ME/CFS
 

Hip

Senior Member
Messages
17,820
Thanks @necessary8, I've corrected my post regarding Brilliant Blue G not actually being a food dye; though I read here it is a close relative of the food dye Brilliant Blue FCF (aka: Blue No 1).

I am currently looking into the pharmacokinetics of these Panx1, P2X7 and P2Y2 inhibitors, to see if sensible and safe oral doses can achieve the IC50 values given in the studies. I will post the details shortly on Jesse's thread.