• Psychedelic Medicine

Autoimmune Disorders | +30 articles

image_720-2.png



The impact of psychedelics on inflammation

by Alexander Lekhtman | LUCID News | 8 Jul 2020

This article accompanies “Eleusis Draws on Research Into Psychedelics To Develop New Medicines for Inflammation,” and details the company’s research on how psychedelic medicines could improve immunity.

A long time researcher of psychedelic substances is on the forefront of investigating how these materials may be used to reduce inflammation and support the immune system.

These investigations may provide new opportunities for scientists to develop more effective treatments for diseases impacted by inflammation such as Alzheimer’s disease and cancer.

charles-nichols.png


Charles Nichols, professor of pharmacology at LSU, is examining how the substituted amphetamine 2,5-Dimethoxy-4-iodoamphetamine, also known as DOI, affects immune function.

The psychedelic effects of DOI have been compared to LSD, but unlike that material, it is not a Schedule 1 substance in the U.S.

First synthesized by Alexander Shulgin, a radioactive iodine-125 form of DOI for PET imaging was first developed in the lab of Nichol’s father, David E. Nichols.

Charles Nichols and his research team found that by triggering the serotonin receptor 5-HT2A, DOI affects the tumor necrosis factor, a cell signaling protein or cytokine that regulates the immune cells.

Tumor necrosis factor, also known as TNF-alpha, is involved in systemic inflammation which is implicated in Alzheimer’s disease, cancer and other ailments.

Other substances that trigger the 5-HT2A serotonin receptor, including TCB-2 and LSD, have also been found to inhibit TNF-alpha, with DOI being the most active.

Nichols notes that psychedelic substances that impact serotonin receptors appear to work differently from existing treatments for inflammation. He says that presently available anti-inflammatories take three different forms: NSAIDs like ibuprofen or aspirin, corticosteroids like hydrocortisone, and biologic anti-inflammatories like antibodies.

Nichols says these drugs all have harmful side-effects that may even be fatal, and don’t treat all diseases.

“Psychedelics do not act through any of these mechanisms, but instead very potently prevent inflammation through blockade of specific cellular inflammatory pathways contributing to disease,” says Nichols. “They are not immunosuppressant in nature, and as such are predicted to have less adverse side-effects.”

In addition to triggering serotonin receptors and affecting TNF-alpha, Nicols says that DOI also affects the activity of nuclear factor-kB (NF-kB), a protein that controls cytokine and other inflammatory responses.

“There are profound effects to both prevent and treat pre-existing symptoms of asthma, and vascular inflammation associated with cardiovascular disease that we have published, and are currently working on additional disease models,” says Nichols of his work with NF-kB.

“These are complex inflammatory diseases involving more than TNF-alpha, so we believe that the anti-inflammatory effects are multifaceted and affect multiple pathways in several different tissue types.”

Cautious investigations for a range of possible treatments

Nichols is now the chair of the scientific advisory board for Eleusis Ltd., a London-based life sciences company founded in 2013. Eleusis is sponsoring Nichols’ research into a new substance called ELE-02 which is part of the ELE series of compounds.

Part of the chemical class of substituted phenethylamines, which includes LSD, the ELE compounds also bind to serotonin receptors and trigger an anti-inflammatory response without a psychoactive effect. The ELE compounds are currently being investigated by Eleusis as a possible treatment for eye inflammation.

Does research into the anti-inflammatory properties of psychedelic substances have the potential to develop treatments for diseases like COVID-19? Nichols cautions that there is presently insufficient information to determine if this might be true.

“We have found that psychedelics are not immunosuppressants, and only target subsets of pathways that together are able to treat disease pathology,” says Nichols.

“We have found that psychedelics are not therapeutic for inflammation associated with several disease models. Therefore, the therapeutic potential for psychedelics for a particular inflammatory-related condition will have to be evaluated on a disease-by-disease basis.”

Investigating DMT

Nichols says that investigations into N,N-Dimethyltryptamine, also known as DMT, also shows promise for new treatments. He says that studies show that DMT interacts with the sigma-1 receptor which impacts both the central nervous system and immune cells. "Triggering this receptor has significant impacts on immunity and inflammation," says Nichols.

attila-szabo_193px.jpg


Attila Szabo, a National Institutes of Health Distinguished Investigator, studies DMT and its derivative, 5-MeO-DMT, both naturally-occurring in the body. He examines how these substances interact with the sigma-1 receptor when stimulated by E. Coli or influenza.

Szabo found that these interactions reduced levels of inflammatory cytokines, while increasing levels of anti-inflammatory proteins. They also reduce activity of T-cells, which regulate inflammation.

Szabo notes that since the endogenous levels of these tryptamines, or those produced inside the body, are quite low in mammals, more research is needed to verify the physiological significance of these findings and how they can be applied to fight disease.

“One of the possible mechanisms is that the levels of endogenous tryptamines can be massively increased at the site of infection or inflammation. due to local cellular stress responses,” says Szabo. "These molecules may achieve concentrations sufficiently high enough to be able to influence immune or inflammatory responses.”

Like Szabo, Nichols is cautious about the need for more research. “I think the jury is still out on sigma-1 receptors,” says Nichols. “Existing investigations with DMT have not been translated, to my knowledge, to any relevant animal models of human inflammatory disease for validation. Certainly more research needs to be performed in this area to better define the potential.”

Impact of mood and motions on immunity

The psychoactive effects of psychedelics may also be relevant to immunity. Psychedelics produce altered states that can affect a person’s mood, emotional, or spiritual health in complex ways.

“It has been well known for years that the environment and experiences of an individual can influence the immune system,” says Nichols. “The immune system is highly complex, and its activity can be modulated by the brain, the gut, the food we eat, etc.”

Nichols points to the hypothalamic–pituitary–adrenal axis or HPA axis, which is a neuroendocrine system. It controls our reactions to stress and regulates many functions including mood, emotion, and the immune system. Studies have shown that MDMA and psilocybin can help alleviate disorders like PTSD, depression and anxiety, which are associated with dysfunction in the HPA axis.

According to Nichols, clinical studies investigating MDMA and psilocybin could also look at their impact on inflammation.

“It’s quite conceivable that MDMA and psilocybin-mediated reductions in stress and anxiety, and normalization of HPA axis function, would result in a strengthened immune system,” says Nichols. “Clinical studies using MDMA and psilocybin that incorporate the measurement of inflammatory biomarkers may address this.”

 
Last edited:
oxjlan.jpg



Psychedelics modulate innate and adaptive inflammatory responses through the Sigma-1R receptor

Attila Szabo, Attila Kovacs, Ede Frecska, Eva Rajnavolgyi

The orphan receptor Sigma-1R is a transmembrane chaperone protein expressed in both the central nervous system and in immune cells. It has been shown to regulate neuronal differentiation and cell survival, and mediates anti-inflammatory responses and immunosuppression in murine in vivo models.

Here we demonstrate for the first time the immunomodulatory potential of NN-DMT and 5-MeO-DMT on human moDC functions via Sigmar-1R that could be harnessed for the pharmacological treatment of autoimmune diseases and chronic inflammatory conditions of the CNS or peripheral tissues. Our findings also point out a new biological role for dimethyltryptamines, which may act as systemic endogenous regulators of inflammation and immune homeostasis through the Sigma-1R receptor.

http://publichealthwell.ie/journal/p..._complete=true
 
Last edited:
smile3-e1343309773937.jpg

Moises Velasquez-Manoff

Q&A: Parasites, modern life and immune systems gone haywire

by Brandon Keim | WIRED

Moises Velasquez-Manoff was on the road to Tijuana, where he'd meet a man who would infect him with hookworms.

So begins An Epidemic of Absence, Velasquez-Manoff's new book about a tantalizing hypothesis for a modern medical mystery: Why autoimmune diseases, in which a person's immune system attacks their own body, are becoming more common, even as infectious and parasitic diseases are beaten back. (Read an excerpt from the book)

According to Velasquez-Manoff and the scientists he writes about, it's no coincidence. A fast-growing body of research suggests that immune systems, produced by millions of years of evolution in a microbe-rich world, rely on certain exposures to calibrate themselves. Disrupt those exposures, as we have through modern medicine, food and lifestyle, and things go haywire.

Velasquez-Manoff, who has several immune-related disorders, including food allergies and alopecia, had heard about the "hookworm underground" – people who infect themselves with parasites in the hopes of restoring immune balance. Though it's something he now recommends against doing, it marked the beginning of a reportorial journey into a frontier of science and health.

Wired talked to Velasquez-Manoff about his experiences.
--------------------------------------------------------------​

Wired: What would parasites have to do with food allergies or alopecia?

Velasquez-Manoff: Parasites modulate your immune response. When you have them, the number of cells involved in tamping down inflammation increases. These cells have a bystander effect. They end up tamping down on all sorts of inflammation, not just that induced by parasites – and food allergy is an inflammation that's inappropriate. You shouldn't be throwing this huge response at what are essentially harmless pieces of plant. Your immune system has made a mistake. When you have parasites, the theory goes, your immune system is less prone to making these mistakes.

Wired: You also write that parasites and other microbes don't just keep the immune system from overreacting, but also help it know what to react to.

Velasquez-Manoff: That's how it ended up. These things are so intertwined. One way to look at it is this: Let's say you need 15 percent of all your T-cells to be regulatory T-cells. Your effector T-cells determine what to attack, and your regulatory t-cells help you stop attacking. If you're dropped into the world with those 15 percent already pre-programmed, and parasites induce another 10 percent, you end up with an immune system that doesn't work. You’ll get overwhelmed by invaders.

You’re too soft. So as an adaptation, only 5 percent of your T-regs are pre-programmed. You get the parasite exposures, you get your extra 10 percent induced, and you're at the golden level. Now if you take away the parasites, you’re left with inadequate T-regs. That’s kind of the story. We're not born with enough immune regulation; we rely on other organisms to get us to the right level, because they were always around.

Instead, you're only genetically 5 percent pre-programmed. You get the parasite exposures, you get your extra 10 percent, and you're at the golden level. We're not born with enough immune regulation; we rely on other organisms to get us to the right level, because they were always around.
Wired: There's another country, Sardinia, that has an instructive history.

Velasquez-Manoff: Sardinia was a cesspool of malaria for a long time. Part of it has to do with the hydrology of the island: Basins form in basalt rocks, and water just sits and collects in a lot of places.

The Sardinians have been invaded by wave after wave of people. They basically fled to the center of the island while various empires set up shop on the coast, and the next wave would come a few hundred years later: Carthaginians, Romans, Vandals, all the way through to Spaniards. There wasn't much genetic admixture. When they do those phylogenetic trees of the European family, Sardinians are less related to mainland Europeans than Iranians are.

Why does that matter? They developed adaptations to malaria over time. Then, after World War II, malaria is eradicated in Sardinia – and all of a sudden autoimmune diseases go crazy there. It's like the number-two place for Type I diabetes. It's near the top for multiple sclerosis. And the guy I visited there, Stefano Sotgiu, his hypothesis is that without malaria present their immune systems started going crazy.

infection_vs_autoimmunity.jpg

Inverse relationship between the incidence of infectious disease (left) and immune disease (right).

Wired: We've talked a lot about exposure to parasites and microbes in the wild, so to speak. What about the microbes we get from our mothers?

Velasquez-Manoff: We get two things. First, in the womb, you're not exposed to any microbes yet, but you get the signal of a microbial community through your mother's immune system, which is turning out to be critical. If your mother is tolerating all these microbes, then your immune system knows not to overreact.

Where it really gets interesting is the resident microbes that live in your mother. We know these clearly set the tenor of the immune function. There are certain infections that cause problems: Some studies show that if a mother has vaginitis, an imbalance in vaginal flora, the child's risk of asthma goes up. It's the inflammatory signal that's skewing the fetal immune system toward asthma.

If you look at this in the context of what's been happening in the last 150 years, there's some evidence that the internal community is more inflammatory than it used to be.

Wired: You make a case in the book that inflammation is linked to autism.

Velasquez-Manoff: There is, at this point, reams of evidence indicating two things about autism. First, that in many autistic children, the immune system is completely off-kilter. There's lots of evidence of low-grade activation in the absence of any kind of infection. You can see it in their brains, too.

One of the seminal studies in this was published in 2005 by researchers at Johns Hopkins, who got brain tissue from autistic individuals who had died, and looked at it under a microscope. They found that astrocytes, these brain cells that are part of immune function in the brain and nervous system, were enlarged and huge in autistic brains, as if they were subject to this constant signal to be active.

When you think of classical autoimmune diseases, like multiple sclerosis, you actually see degeneration in the myelin, like it's stripped off. This wasn't the same thing. It was low-grade inflammation. And if you have low-grade inflammation, nothing else works. Your immune system needs to quiet down when it's not fighting something off, but in these kids it wasn't shutting off.

The second thing is that there are some very good animal models now where they induce autism. It starts in the womb, and it's related to some sort of inflammation in the mother. There's a natural phenomenon showing this: the children of mothers who were infected with rubella virus in the 1960s were far more vulnerable to autism. The infection doesn't even need to touch the fetus. It’s the inflammatory response.

But in the last half-century, when autism has gotten out of hand, maternal infections have actually gone down. Infection is not the problem per se. It's inflammation. It turns out some of the diseases I talk about in my book – asthma, allergies, autoimmune disorders – are closely linked to autism in the sense that if the mother has them, the chance of a child having autism goes up. When there's a problem with the mother's immune system, it may cause a problem with the developing fetus.

Wired: The New York Times essay you wrote about this drew some criticism. Emily Willingham, a science writer and autism expert, said the scientific evidence for this was limited, overstated and cherry-picked. What is your response to that?

Velasquez-Manoff: My response is this: Check it out with the scientists, like this Yahoo reporter did. Call them up. See what they say about the piece. The New York Times Sunday Review fact-checks, by the way. See my source list, which, yes, was posted regrettably late after the article went up. Did I cherry pick? I think I got a whole cherry tree. A valid question, however, is whether it’s the only tree around. Did I fail to qualify and caveat? Yes. It was an op-ed. But I’m making an argument scientists are making, one that’s very well supported by the literature. (The scientist Paul Patterson apparently penned a very similar piece, but couldn’t get it published.)

One issue Emily Willingham raises that’s important: How do we know what’s cause and effect here – the immune dysfunction or the autism? That question is partly addressed by the animal models. Scientists can reproduce some of the observations in humans – brain “overgrowth,” behavioral problems, a pro-inflammatory skew, similar gene expression profiles – with prenatal inflammation. Does that mean case closed? No. But it’s a remarkable body of evidence. Don’t take my word for it. Ask the scientists.

Wired: So do we know enough about all these effects to manipulate our bacteria and parasite exposures in a way that's good for our health?

Velasquez-Manoff: The easiest recommendation is to eat well. We've known for a long time what’s good, but we didn't know how it works. We know that a high-fiber diet is better than a junk food diet, that eating a lot of fruits and veggies and whole grains is better than junk food. But now we know why: In part, it's probably because it modulates your microbiota.

But other stuff needs to be learned. Take the whole fatty acid thing. There are people who eat flax seed meal, which is rich in omega-3 fatty acids, but nothing happens. Their omega-3 levels don't go up. Some studies in mice show they only go up if the mice are also taking probiotics. You need to have the right bacteria to help absorb the fatty acids that are good for you.

Wired: And some people are going so far as to expose themselves to parasites. How did your own experience with hookworm turn out?

Velasquez-Manoff: There were definitely some changes for the good, and definitely some side effects. I had a pretty severe reaction for a few weeks. I took it in November, and then by hay fever season my nose was completely clear. My fingernails were less pitted. Hair started growing here and there – very fine, like peach fuzz, which was pretty cool, but there was nothing near remission of anything.

As I point out in the book, there was a lot of variability. That's probably what happens with living organisms you buy on the black market. The thing about the underground is that you don't know the quality of the larvae, or even if you're getting what you think you're getting. I had my parasites genetically confirmed, so I have that confidence. But I have to say, the more I got involved with it, the less I thought it was a good idea.

I think the theory has great merit. The animal models are pretty much unequivocal. But when you start thinking about yourself, or your own children, you say, 'How certain am I?' And the uncertainty undercuts the idea. No one should do what I did. Let the science provide something safe, which it will.

 
Microdosing-psychedelics-with-autoimmune-disease-pg09evwmaoso48e737mi1v3ooyr433e8x508t5i2yw.jpg



Microdosing Psychedelics with Autoimmune Diseases

Community Member "Edo" | Microdosing Institute | 4 Apr 2021

Community member Edo shares with us how microdosing psychedelics led to an unexpectedly wide range of improvements in his life – both mentally and physically. He summarizes his journey to date as follows: “It was a long search, but after some ups and downs it led to a reduction of medication use and an increased quality of life.”

Inflammatory diseases

My microdosing story is mainly a medical story. I was diagnosed with type 1 diabetes when I was 28. When I was 36 I also got scleroderma (systemic sclerosis). A rare form of rheumatism in which, in short, the skin and everything that has connective tissue, hardens. With some bad luck, the disease quickly escalates into the lungs and heart. I turned out to have an atypical form, the cause of the inflammation could not be found. I was lucky that the disease developed slowly. In a few years my skin hardened, and movement became more difficult. At a certain point, the complaints became such that I had to take immune-suppressing medication. This stopped further deterioration.​

Initial research

At first microdosing psychedelics caught my eye, because I experienced a leak in my intelligence and creativity. (Later I found out that a link is suspected between scleroderma and Alzheimer’s disease.) It would also be nice to do something about my age old problems with concentration and recurring depressions. My first concern was if I could combine it with the medication I was taking. This had become quite a collection by now. I inject insuline daily, I take immunosuppressants, pills for the thyroid and an antacid. But in my search for articles about that, I stumbled upon an article that talked about the healing effects of psychedelics on atheroscleroses, arthritis, psoriasis, type 2 diabetes, depression and Alzheimer’s disease. Multiple diseases that are related to scleroderma. The results were very promising. It was suspected that psychedelics could prevent inflammation, and possibly even repair damage already done.​

A difficult choice

I bought ALD52 which was still legal during that time [an LSD analogue similar to 1p-LSD and 1cp-LSD, MDI], but after that I remained in doubt for a long time. I worried most about whether I dared to tinker with my immune system myself; I have a chronic variant now, what if I turn it into an escalating variant. I’m not a scientist and my medical-scientific English is poor at best. I found it difficult to decide whether I would be doing the right thing. Contact with my doctors, however, yielded little. The studies were conducted by experimental pharmacists and too little was known about the subject. No one dared to say anything specific about it. Doctors become very cautious when there is no scientific consensus. Rightly so, you don’t want a gambler for a doctor, but for an individual patient everything then comes to a standstill. If scientific consensus is reached in 20 years’ time, it won’t be of much use to me. An aside: Online, via written consultations, it is easier to contact a doctor these days, but I found that when it comes to these kinds of questions, it’s better to consult in direct contact. I suspect that doctors will talk more freely. In the end, those conversations boiled down to this; there is no evidence that psychedelics can improve your situation, but there is also no evidence that it will make the problems worse. In the meantime I strongly suspected that microdosing LSD could help me. Not only to get my thinking back on track, but perhaps even to suppress the scleroderma.​

Experiments and results

It was now almost a year later and I dared to start my first experiment. Under the guidance of a coach of Microdosing Institute, I took the lowest recommended dose of ALD52, twice within one week. I didn’t really notice it that much. Only visually it looked like windows had been washed again after a long time, similar to the CBD oil I took a year earlier. But in the next two weeks, the skin on my arms started to loosen and my joints became more flexible. I could handle the chaos of our young family better, I managed to concentrate for a longer period of time and I even dared to jump on a trampoline with my stiff body without the fear of breaking in half. And this after only two microdoses. That I wanted more of. If I was going to continue with this it would be good to get some new ALD52, since I had it laying around for almost a year. Unfortunately, in the meantime, ALD52 had been declared illegal and was no longer available. I then opted for 1P-LSD. This time no drops, but a quarter of a blotter, the lowest recommended dose. Again I took the microdose twice within one week. I felt pressure on my eyes and I had energy, but it felt a bit artificial. My vision was very clear and the colours became more intense. As if it was the photographic ‘golden hour’ all day long. I felt good, but my eyes needed rest.​

Panic

A week later I had an appointment with my immunologist. This was the first meeting with a new doctor who had yet to get to know my skin. This doctor was a lot less enthusiastic about my skin than I was. During the conversation there was a very serious, dark mood and I was suddenly not so sure of my own observations. On the drive home, I suddenly felt my face tighten and my shoulders and arms harden. Never before had I experienced such a rapid aggravation of my disease. In the days that followed, panic set in. All fearful thoughts raced through me. Had I given my immune system too hard a blow and had the escalation started? Within a week I felt like I was wearing a shirt way too tight. Till two weeks later I had panic attacks and every now and then the colours became more intense again. Two weeks after the last microdose, the colours became so intense during a bike ride that I became anxious. It lasted for up to three weeks that, especially during dinner, everything around me turned a deep orange. It’s speculation, but it may have taken this long because I swallowed the quarter blotter the second time I took one, maybe some of it got stuck. Apparently I react very strongly to low doses and I had a three weeks long sort-of semi-trip. Fortunately, in the weeks that followed, the skin softened and I got calm. In the end the result was that I was more flexible and my skin was far softer than before this whole adventure.​

Psychosomatic symptoms

Although this was a very bad mistake, it did teach me a lot. Because my body suddenly changed so much after a very unpleasant visit to the doctor, it seems that my problems are partly psychosomatic. The ALD52 enhanced this effect. In this case it worked negatively, but I know it could also work in my favor.​

The journey goes on

I had experienced anxious moments, but the positive changes in my body were such that, two months later, I wanted to try again with my old drops. The problem was that I found myself drifting back into depression. Because the last time my mindset was so influential on the result, I decided to wait. A month later I was ready to start. Over a period of two months, I took four drops once a week. In the beginning I saw some intense colours again, so I brought it back to three drops. On a microdosing day I sometimes felt restless, but otherwise I didn’t notice much. Little by little, my skin became more flexible. After those two months I took a month off from microdosing and it was during this period that my skin improved even faster. I started to learn more about the world of psychedelics. The ALD52 made me a bit restless. The idea came up to give it a try with XP truffles in combination with Lion’s Mane. Chaga also caught my attention because of its claim that it could balance the immune system.​

XP truffles: finally able to sleep well and have more control over the diabetes

For two months I microdosed XP truffles according to the Fadiman protocol. In combination with a daily dose of Lion’s Mane and Chaga. I started with 0.3 grams, the lowest recommended dose of truffles. I tried to increase it twice, but that made me restless again. During those restless days I saw the world in the colours of the ‘golden hour’ again. Another side effect is that I felt some muscle pain in my neck on the microdosing day. I now know how to compensate for the sleepiness after taking the truffles with a drop of extra Lion’s Mane. Or I take them in the evening. And also with the Lion’s Mane and Chaga, I intuitively found out that I needed less than advised. The Lion’s Mane in particular made me hyperactive at the prescribed dose. Almost immediately after the first dose of truffles, I noticed improvements in my life that I wasn’t looking for beforehand.

I have been sleeping badly since I was about 13 years old. Sometimes I managed to maintain a good sleep rhythm for a few days in a row, but often I quickly lost it again. Sleep has always been a battle for me, a battle I mostly lost. When I started with the truffles I immediately slept a lot better. In the past I sometimes needed hours to fall asleep and I also needed multiple hours in the morning to really wake up (Using many cups of coffee). Now I get much more return from my sleep. I fall asleep quickly, I regularly wake up a few minutes before the alarm and really feel refreshed. That has never happened to me since I was a teenager.

In addition, my mental condition has drastically improved. A few years after I was diagnosed with scleroderma, I noticed that I was becoming less witty. In chaotic situations, I quickly became confused. I got annoyed quickly, I was always tired and became depressed for a long time. In bad moments it was like wearing a closed helmet and I was having a hard time connecting with anything that went on outside of my own mind. All this seems to have completely disappeared. (Okay, I still get annoyed sometimes, but that really is to blame on the other person…)

At the time of writing this, excluding one bad day, I am free of depression for eight months. And another unexpected success. My glucose levels calmed down. I can live-monitor the sugar level in my blood. Normally at night the glucose levels looked very messy with a lot of rises and falls. Now suddenly I saw a straight line. Also during the day I kept my glucose levels neatly within the lines. My need for insulin has decreased by about a quarter. I have become more active during the day, so I don’t know whether that decrease is directly or indirectly caused by the microdosing. The functioning of my thyroid has remained unchanged, tests showed no change and the amount of medication has remained the same.​

More ‘strange’ improvements

Nowadays I only drink 1 cup of coffee in the morning, I don’t need any more. I think I’ve replaced the coffee with the Lion’s Mane, but the energy I get from that feels much more natural and a not stressed. I have become more relaxed. I get a lot more done in a day than I used to and I no longer constantly have that rushed feeling. Strangely enough, my eyebrows are also coming back, as is the hair on my arm. These had been slowly disappearing in recent years. I suspect the Chaga is at work here. Another unexpected by-catch, a relief from light compulsive behavior. This wasn’t a real big burden in my life, but almost always after I locked the car or the house, I checked to see if it was really locked, sometimes I walked back dozens of meters to check what I already knew. I don’t do this anymore.​

Decrease in medication

I can now draw wrinkles across my skin. The hardened skin recedes further and further and my face is slowly returning to its old self. Meanwhile, I have more energy than I have had since childhood. Not hyperactive, but an energy level comparable to a healthy person. My doctor has also seen how much I’ve improved. In consultation, I reduced the immunosuppressants to a third of the original dose. If the improvements continue for another two months, I will stop taking the immunosuppressants completely.​

After a month long break from the XP truffles

During this break it became a bit more difficult to keep my glucose levels in order, I use more insulin again and the good sleep also decreased slightly. But both are still way better than what they once were. At the time of writing this there has been no deterioration of my skin, so I am going to stop taking the immunosuppressants altogether. Now I’ll have to see for some time if I really can do without.​

Looking back, half way through

It has now been a year since I first took a microdose. I searched for information from various sources, but with that in hands I acted intuitively and above all listened carefully to the feedback from my body. I’ve tried a number of different things, so I can’t always say which one caused which improvement. There is a good chance that one improvement, like a domino, caused the other. It seems that there is a psychosomatic side to my illness. Stress and depression translated directly into physical ailments. And in my life, those two were always just below the surface. The psychedelics have set me on a different path, both physically and mentally. The improvements I am experiencing are so comprehensive that I have come to believe that the psychedelics are tackling the problem at the source.

 
Last edited:
077cc28d-fbf1-4e2b-9f20-fa57bd4a1b40_ulnar_drift..jpg



Resolution of Rheumatoid Arthritis in a patient consuming psilocybin mushrooms*

by Denise S. Lin, MD | Journal of Healthcare

A 64 y/o generally healthy and physically active Caucasian male was referred by his PCP for outpatient rheumatology evaluation in March, 2014.

The patient presented to his rheumatology appointment with the complaint of polyarticular joint pain and swelling of 18 months duration. This current bout of joint pain and swelling began in his shoulders and eventually involved his hands, multiple fingers, wrists, hips, and knees. He described morning stiffness lasting about one hour. Normally, this patient was physically active and enjoyed hiking but for the past 18 months his mobility was significantly limited due to the stiffness and pain. Ibuprofen provided partial, temporary relief. He had recently received steroid injections in his shoulders which provided only transient relief.

He reported an episode about 17 years prior when he had bilateral hand and foot pain along with facial redness which resolved without treatment. Then ten years ago he developed pain and swelling in his hands and other joints which were so severe that he could not dress himself. He was treated with naproxen and it gradually resolved after about 6 months.

His past medical history was notable only for seasonal allergies and a partial laminectomy for a herniated disc in 1980. There was no prior diagnosis of autoimmune disease. He denied any recent febrile illness, travel, HIV or sexually transmitted diseases, rashes, dry eyes or mouth, vision changes, headaches, respiratory symptoms, or chest pain. His only medications were over-the counter ibuprofen, MSM (methylsulfonylmethane, a popular supplement for arthritis), fish oil, vitamin D 800 IU daily and a multivitamin. He did not smoke. He drank 2-3 glasses of wine weekly and smoked marijuana occasionally.

On exam his vital signs were within normal limits. Weight was appropriate for height. On physical exam he had mild tenderness and swelling of his right MCP (metacarpophalangeal) joint. On the left he had mild to moderate tenderness and swelling of several PIP (proximal interphalangeal) joints and the MCP joint. He was unable to make a tight fist with his left hand. Both wrists exhibited mild to moderate swelling, pain, and limitation of movement. Shoulders had mild to moderate pain and mildly limited range of motion bilaterally. He was able to move his lower extremities and had mild pain in bilateral knees. His greater trochanters had mild tenderness bilaterally.

Laboratory data: Electrolytes, CBC, liver function and renal function tests were within normal limits. His sedimentation rate and ANA were normal. C-reactive protein was elevated at 0.4 and Rheumatoid Factor was elevated at 18. A chest X-ray was unremarkable.

He was given a diagnosis of rheumatoid arthritis and offered low-dose prednisone and advised to start methotrexate. The patient declined the methotrexate but took prednisone for two weeks.

Shortly after that he consumed some psilocybin mushrooms and was surprised to observe that his joint pain and swelling completely resolved for about 24 hours. In addition, his seasonal allergy symptoms also completely resolved. When he had another flare of joint pain several years later, he took four doses of psilocybin mushrooms of 2 grams each over a two-week period and again had complete resolution of his symptoms. That was one year prior to the writing of this paper. Subsequently he has been using psilocybin mushrooms periodically and he has not experienced any signs or symptoms of rheumatoid arthritis since then.​

Rheumatoid arthritis

Rheumatoid arthritis is a chronic, inflammatory joint disease with a prevalence rate between 0.5 and 1% in developing countries. It can occur at any age but the peak incidence is in the 6th decade and it is 2-3 times more prevalent in women than in men.

The causes of rheumatoid arthritis are not fully understood but do seem to involve an interplay between genetic and environmental factors. It is possible that single nucleotide polymorphism (SNP) genetic variations in the serotonin 5HT2A receptor could increase the risk for RA.

External factors such as cigarette smoking, infection or physical trauma may serve as triggers in predisposed individuals which then sets off an autoimmune response. Once triggered, the immune system directs an attack on the synovial membrane in multiple joints. Endothelial activation and neovascularization occur early on followed by infiltration of the synovial membrane with immune cells, including T-cells, B-cells and monocytes. Hyperplasia of the synovial lining occurs as cells such as fibroblasts and macrophages accumulate there. This activated synovium, or "pannus," then invades and damages the bone near the cartilage-bone junction. Pro-inflammatory cytokines, including TNF-alpha and IL-6 are released by the immune cells within the synovial membrane. These cytokines continue to propagate an inflammatory processwhich involves TNF-alpha, NF-κB, prostaglandins and matric metalloproteases leading to pain, swelling and degradation and damage of bone and cartilage.

The typical clinical presentation of RA involves pain and swelling in the joints of the hands and feet and a complaint of morning stiffness. The wrists, MCP (metacarpophalangeal), MTP (metatarsal phalangeal) and PIP (proximal interphalangeal) joints are commonly involved. Larger joints, such as ankles, knees, elbows, and shoulders may also be involved [40]. Laboratory testing may include RF (Rheumatoid Factor), ACPAs (anticitrullinated peptide antibodies), erythrocyte sedimentation rate, ANA (antinuclear antibody) and C-reactive protein. Laboratory tests may be helpful but are not diagnostic and physicians must look at the entire clinical picture to make the diagnosis.​

Guidelines for the treatment of RA emphasize an integrated approach that usually includes corticosteroids and NSAIDS [44]. However, the mainstay of treatment of RA is with a group of drugs called disease modifying antirheumatic drugs, (DMARDs) which slow the progression of joint damage. There are a variety of DMARDs, including methotrexate which suppresses DNA synthesis and replication. Other drugs have been designed to antagonize inflammatory mediators. For example, the TNF Inhibitors suppress the actions of the pro-inflammatory cytokine, TNF-alpha. TNF Inhibitors are reported to lead to sustained remission in about a third of patients.

Without treatment, the disease leads to irreversible joint damage and significant disability with 40% unable to work within 10 years of disease onset. The economic burden of rheumatoid arthritis is substantial, with total annual healthcare costs about 3 times higher than those without a rheumatoid arthritis diagnosis. In 2005 the total annual healthcare costs, (including direct and indirect costs) for rheumatoid arthritis patients overall was estimated at about $19.3 billion. While a large proportion of the cost is attributable to medication, costs associated with medical co-morbidity are also substantial. In the 2018 study by Chen, a group of 115,857 individuals in the Medicare system with RA was compared to a matched non-RA cohort. In this study medical co-morbidities were found to be significantly higher in those with rheumatoid arthritis diagnoses. For example, 76.4% had cardiovascular disease, compared to 44.8% without RA.​

Serotonin, TNF and inflammation

While serotonin is widely known as a neurotransmitter produced in the brain, the vast majority of serotonin is synthesized and stored in the intestinal mucosa. Outside of the brain, serotonin has numerous important functions, including roles in vasoconstriction/vasodilation, hemostasis, blood pressure and other cardiac functions as well as in the gastrointestinal system. In addition, serotonin is involved with pain perception at both the central and peripheral nervous system levels.

Serotonin is also an important modulator of inflammatory processes. Serotonin receptors and machinery for the synthesis of serotonin are found in cells of the immune system, including T-cells, macrophages, and mast cells. Serotonin is involved in modulating the inflammatory response by influencing the production of TNF-alpha and some pro-inflammatory interleukins.

In most animal studies, serotonin appears to promote inflammation. Depletion of serotonin generally leads to reductions in inflammatory responses and restoration of serotonin leads to increases in the severity of inflammatory diseases.

Given that serotonin plays an important role in inflammation it may follow that serotonergic antidepressant medications might also affect inflammatory processes. There have been a few investigations of antidepressants in inflammatory autoimmune disorders, and they do appear to provide benefit beyond their effect on mood. For example, duloxetine was found to improve the course of irritable bowel syndrome in humans. In a mouse model of multiple sclerosis, the antidepressant clomipramine ameliorated clinical signs of acute and chronic phases of the disease. In a rheumatoid arthritis model in rats, paroxetine protected their joints from inflammation and destruction via inhibition of T-lymphocyte infiltration.​

The serotonin 5HT2A receptor

Serotonin exerts its myriad effects via 7 different serotonin receptor families, all of which have subtypes (for a total of 14 different variations). The 5HT3 receptor is the only ligand-gated ion channel receptor, while all the others are G-protein-coupled receptors (GPCRs). GCPRs are proteins which span the cell-membrane. Once an agonist binds to this type of receptor a conformational change occurs in the receptor, a cascade of events follows which eventually signals specific genes to be expressed and the subsequent production of proteins which lead to biological actions.

The 5HT2A receptor subtype is a focus in psychedelic research because psychedelic agents such as psilocybin have a high affinity for this receptor, where they act as agonists.

In brain, the 5HT2A receptor is intimately involved with complex cognitive behaviors, such as working memory. Outside the brain, 5HT2A receptors are found in a wide array of tissues and perform a variety of functions. Serotonin 5HT2A receptors are also found in endothelial cells and in vascular smooth muscle.

Importantly, serotonin 5HT2A receptors are also found on a several differenttypes of immune cells, including monocytes, and T-lymphocytes where they are involved in the mediation of immune-inflammatory processes. Thus, the 5HT2A receptor appears to be a link between serotonin, serotonin agonists, such as psilocybin, and inflammatory autoimmune disease, such as rheumatoid arthritis.​

Potent anti-inflammatory effects of 5HT2A receptor agonism

Several elegant studies have greatly advanced the understanding of the immune-inflammatory functions of the 5HT2A receptor. The psychedelic agent R-DOI (R-2,4 dimethoxy-4-iodoamphetamine) is a highly selective agonist at the 5HT2A receptor which makes it very useful for studying the functions of this receptor. In a study on rat aorta smooth muscle cells, and in a subsequent study with mice, activation of 5HT2A receptors by R-DOI powerfully and potently inhibited TNF-alpha-mediated inflammatory events, including gene expression for ICAM-1, VCAM-1 and the pro-inflammatory cytokine, IL-6. In addition, this research was able to demonstrate that the anti-inflammatory effects were specific to the 5HT2A receptor, and not any other serotonin receptors. Using a mouse model of asthma, mice who were pretreated with nebulized R-DOI and then exposed to a potent allergen demonstrated normal airway morphology (e.g. no evidence of asthma) while mice who were not pre-treated with the psychedelic did develop hyperresponsive airways, pulmonary inflammation, excess mucus and eosinophilia consistent with an acute asthma episode. Of note, the doses of the psychedelic agent used to prevent the inflammatory response was below the threshold for behavioral effects, which, indicates that a pronounced anti-inflammatory effect may be achieved at doses below the dose that would cause hallucinogenic effects.

Inflammation is not only seen in acute immune reactions to allergies butis also well-established as an etiological factor in chronic diseases, such as coronary artery disease. A recent study looked at the effect of selective 5HT2A receptor activation in mice who were fed a high fat, Western-type atherogenic diet, which is known to promote systemic inflammation and immune cell dysregulation eventually leading to coronary artery disease. While the control group demonstrated increased expression of inflammatory markers as expected, (R)-DOI treatment prevented this inflammatory response, showing that 5-HT2 receptor activation prevents diet-induced vascular inflammation.​

In summary, the profound anti-inflammatory effects of the psychedelic R-DOI has been demonstrated in both acute and chronic disease models in animals. In these models, the underlying molecular mechanisms of inflammation TNF-alpha mediated, which have been demonstrated in the pathophysiology atherosclerosis, rheumatoid arthritis, psoriasis, type II diabetes, depression, schizophrenia, and Alzheimer's disease. In simplistic terms, the process starts with the activation of immune cells, such as mast cells, T-lymphocytes, and monocytes. These cells contain various serotonin receptors, including the 5HT2A receptor which is known to be especially important in inflammation. Serotonin, by attaching to the 5HT2A receptor is one way these cells can be activated. These cells then release a variety of proinflammatory cytokines, including TNF. TNF proceeds to trigger subsequent events which lead to the production of more inflammatory mediators. However, when the psychedelic agent R-DOI, a high affinity agonist at the serotonin receptor binds to these cells, the proinflammatory response is suppressed.

Functional selectivity and the 5HT2A receptor

It turns out that different agents attaching to the same receptor can elicit divergent responses in a process known as functional selectivity. When serotonin attaches to the 5HT2A receptor specific signal transduction pathways are initiated. This process includes activation of the NF-κB Subunit p65 which then translocates into the cell nucleus where it causes gene expression to make proinflammatory cytokines. When a psychedelic 5HT2A agonist attaches to the receptor, the process starts the same but NF-κB Subunit p65 translocation into the nucleus appears to be blocked. In simpler terms, serotonin primarily stabilizes the receptor in a conformation that recruits proinflammatory pathways, whereas psychedelics stabilize the receptor in a slightly different conformation that prevents inflammatory signaling pathways.​

Summary

In the rheumatoid arthritis case presented here the patient declined methotrexate and serendipitously found that taking psilocybin mushrooms resolved his RA symptoms. At this writing he has been symptom free for over one year. He continues to take moderate doses of psilocybin mushrooms approximately once a month. RA is a progressive inflammatory, autoimmune disease which is likely caused by interactions between an environmental trigger and a genetic predisposition (possibly a SNP in the gene coding for the 5HT2A receptor). The pathophysiology of RA includes activation of immune cells which produces a cascade of TNF-alpha-mediated events. TNF is the target of a class of disease-modifying drugs developed to slow the disease course of RA, and which appear to do so in about 1/3 of patients [44]. Immune cells involved in the inflammatory process have been found to contain serotonin 5HT2A receptors, which when bound to serotonin appear to promote TNF-induced inflammation, yet when bound to a psychedelic agonist, such as R-DOI strongly inhibit TNF-induced inflammation. While psilocybin itself has not yet been investigated as a modifier of inflammation, it is plausible that it could behave like R-DOI in suppressing inflammation because it too is a high-affinity, psychedelic agonist at the serotonin 5HT2A receptor. If this is the case it could explain why this patient's RA has remained in remission despite not taking a DMARD.​

Discussion

There are many limitations inherent in a single case report and many questions remain unanswered. For one, there is no way to be sure if the patient experienced relief due to the anti-inflammatory effect of psilocybin's 5HT2 agonism, the placebo effect, or because his emotional state was enhanced by the mood altering effect of the mushrooms.​

Assuming the former, then next question might be: To what extent is the "psychedelic" nature of psilocybin relevant to the anti-inflammatory effects? Certainly in the psychiatric research, those participants who met criteria for having had a mystical experience had a greater chance of having resolution or significant improvement of depression or anxiet. If it turns out that all therapeutic effects of psychedelic agents depend upon experiencing such a "non-ordinary" state of consciousness it would ultimately limit the use of these agents to those people who have been carefully screened and determined to be psychologically resilient and without a history of psychosis.

Evidence so far, however, suggests that a mystical or hallucinatory experience may not be necessary to achieve a clinically relevant anti-inflammatory effect. As noted previously, rodent experiments with the highly selective 5HT2A receptor agonist DOI demonstrate potent anti-inflammatory actions at doses below the threshold for perceptual effects. Further, a Mclean Hospital survey of cluster headache suffers found that 42% of participants who self-treated their cluster headaches with LSD or psilocybin reported complete or partial relief at doses that were sub-hallucinogenic [80]. While the mechanism of action of psychedelic agents in cluster headaches is unknown (and unverified) this survey does at least hint that clinically significant beneficial effects might occur at doses that are too low to produce psychological effects.​

A third, unanswered question relates to the fact that psilocybin appears to bind to several other serotonin receptor subtypes besides 5HT2A, including 1D, 2B, 2C, 5, 6 and 7. If so, could the anti-inflammatory effects be attributable to a receptor or receptors which do not cause psychological effects? The functions of all the serotonin receptor subtypes have not been fully elucidated. However, anti-inflammatory effects in the cerebral vasculature are seen with activation of 5HT1B and 5HT1D receptors as exemplified by the migraine drug sumatriptan [82]. It is then conceivable that psilocybin or psilocin binding at the 5HT1D receptor could contribute to some of the anti-inflammatory effect, but the contribution would likely be very small as experiments with the highly selective 2A receptor agonist DOI elicit a dramatic anti-inflammatory response without any involvement of the 5HT1D receptor.​

Finally, how important is it that this patient was consuming whole mushrooms, not pure psilocybin? Mushrooms contain a large variety of nutrients and other molecules which may inhibit TNF-alpha, inflammatory interleukins, prostaglandins or act as free-radical scavengers. Therefore, it is possible that some of those components could be contributing to his disease remission. Edible mushroom constituents with anti-inflammatory properties include saccharides, fatty acids, phenols, indoles, antioxidant vitamins, terpenoids, glutathione and minerals. For example, lycopene, a common mushroom constituent has been found to inhibit NF-κB signaling. Research on edible mushrooms has demonstrated a variety of anti-inflammatory actions.​

While many more questions are yet to be explored the fact remains that rheumatoid arthritis is associated with significant human suffering, disability and a heavy economic burden. There is a need for additional cost-effective and safe treatments. Growing evidence indicates that psilocybin mushrooms modulate the immune-inflammatory process in a beneficial manner and do so after only a single dose. Given its anti-inflammatory effects, along with the reassuring safety profile and low toxicity the therapeutic potential of psilocybin mushrooms in inflammatory diseases warrants further study.

*From the article (including references) here :
 
Last edited:
logo-retina-m.png



Psilocybin and Autoimmune Disorders*

ELEUSINIA PSILO RETREAT | 1 May 2022

Psilocybin may be the future answer to age related neurological issues like Alzheimer’s and other dementias. The current landscape for Alzheimer’s treatment is quite bleak. There are currently no successful therapies that alter the course of the disease. The success rate for clinical trials is arguably ZERO percent. But magic mushrooms and some similar psychedelics have unique properties that set them apart. They cross the blood brain barrier to stimulate neurogenesis, provoke neuroplastic changes, and reduce neuroinflammation.

Neuroplasticity and neurogenesis

Neurogenesis is the process of forming new neurons while neuroplasticity is the process of forming new connections between them. Engaging in exercise and novel activities stimulate neuroplasticity and neurogenesis. This is the reason doctors and neuroscientists encourage staying active and participating in learning new skills such as ballroom dancing in our older years. Keep moving, keep learning, and hopefully our brains will get the memo and keep up.

Neuroinflammation

Current science indicates that neuroinflammation plays a big part in Alzheimer’s and other forms of dementia. In fact, some studies have shown that specific medications that target the inflammation associated with autoimmune conditions like rheumatoid arthritis or psoriasis also have had a favorable effect on the progress of neurodegenerative diseases. These medications can dramatically inhibit the production of TNF, a protein your body produces to signal inflammation. So why aren’t we all taking them preemptively to reduce the chances of Alzheimer’s? Well, these medications usually have quite severe side effects that affect the immune system.
Magic mushrooms and inflammation

So, what part do the magic mushrooms play here? Well, the classic psychedelics work directly to stimulate certain serotonin receptors (5HT2A). These receptors are directly involved in regulating the pro-inflammatory cytokines that play a significant role in neuroinflammation. While psilocybin itself has not yet been investigated as a modifier of inflammation, it is plausible that it could behave like other similar psychedelics used commonly in research such as such as R-DOI in suppressing inflammation because of the affinity for the same receptor. More info on the anti-inflammatory properties of psilocybin can be found here.

I do not really want to get too technical, but to sum it up, magic mushrooms target inflammation in a way that resembles that of the medications used to treat autoimmune diseases like RA, psoriasis, and colitis, but without the immunosuppressive side effects. Data seems to suggest that reducing neuroinflammation can improve outcomes, but most pharmaceuticals that can reduce inflammation like this come with a steep cost to your immune system. The information regarding favorable trends in results regarding Alzheimer’s is mainly based on data from that involved using these medications for other purposes, such as rheumatoid arthritis. According to this Washington Post article, Pfizer even had some PR difficulties after they declined to investigate some initially promising results with treatment with ENBREL, one of their RA drugs.

Why is this not common knowledge?

Why isn’t this common knowledge? And why haven’t there been multiple studies and scientists lining up to collect their Nobel prizes for “curing” Alzheimer’s? First of all, it isn’t exactly a cure. It is what it is- a substance that has the potential to reduce neuroinflammation. Neuro inflammation is complex and difficult to test, and the part it plays in neurodegenerative disease is equally complex. But using magic mushrooms does not come with the same risk of side effects that these other pharmaceuticals have.

In fact magic mushrooms, are extremely safe, so you may find yourself wondering- why not? If taking small amounts of magic mushrooms is harmless and has the potential to reduce the neuroinflammation that can lead to Alzheimer’s or other forms of dementia, why not go for it? There is also the factor of lack of profitability. Psilocybin grows easily and cheaply at home from spores that are legal to purchase in most states. There is just little incentive for research in this area.

Magic mushrooms and age

Many older people think of psychedelics as the territory of the young and reckless. They might even have fond memories of experience with them from a “misspent youth”. It is unfortunate that this attitude is so common, because these effects are often wasted on the young and we are set to benefit most from them in our older years. What a strange twist of events that the same substance that was so popular for recreational purposes, then demonized in the “war on drugs”, is turning out to be such a powerful tool for brain health.

Implementation

The studies suggest that a light dose can achieve the desired effect on neuroinflammation. An intense psychedelic trip is not necessary to achieve the benefits. Do I think it is a good idea to live a little and try a real trip at least once? Absolutely. But you can enjoy the potential anti-inflammatory benefits without necessarily subjecting yourself regularly to an intense psychedelic experience. Small doses are sufficient, though

I would avoid the frequency of microdosing, especially until more studies are published regarding safety. A minidose feels no more intense than a glass of wine. If you want to give an intense psychedelic experience a try, I suggest checking out this article we have on managing the experience.

So how can you start incorporating psilocybin into your routine? With magic mushrooms being decriminalized in various states and cities around the US, it will become easier to find legal commercial suppliers. But if waiting around does not sound appealing, you can visit one of our retreats. At the retreat, we teach you to comfortably manage an intense psychedelic experience and discuss the various dosing schedules for different applications. We also have a full day comprehensive mushroom growing class so that you can grow your own mushrooms in the future.

Is it a good idea to take psilocybin mushrooms with advancing Alzheimer’s or other dementia? Maybe not. It is a can be a very confusing and destabilizing experience and this is probably not a good experience for someone who is already struggling with confusion. The best time to take advantage of the anti-inflammatory effects of psilocybin is earlier as part of a prevention plan.​

*From the article here :
 
Last edited:
10.png

Charles D. Nichols, PhD

Psychedelic Research Targets Systemic Inflammation

by Nate Seltenrich | PROJECT CBD | 4 May 2021

Can scientists convert a super-hallucinogen into a wonder drug?

Medical scientists have caught on to the fact that cannabinoids help fight unwanted inflammation that contributes to many diseases. But the next anti-inflammatory wonder drug could come from an even more unexpected source: psychedelics.

A team at the LSU Health Sciences Center in New Orleans is now working to refine one of the most potent anti-inflammatory molecules known to science, a synthetic psychedelic similar to LSD called DOI (2,5-Dimethoxy-4-iodoamphetamine) that’s widely used for the study of serotonin receptors in the brain.

LSU researchers have shown that DOI is highly effective at preventing and treating widespread inflammation in mice.

Led by professor of pharmacology Charles Nichols in concert with life science company Eleusis, where Nichols is scientific founder and director of molecular pharmacology, the team’s current work revolves around reducing the behavioral effects of DOI, which are reported by recreational users to include an even longer trip than LSD of up to 30 hours, with aftereffects that can last for days.

Nichols said he has fashioned a variant of DOI, code-named ELE-02 by the company, that produces the same anti-inflammatory effect but two-thirds less behavioral effect. ELE-02 should be entering clinical trials in the form of an eyedropper for treating ocular inflammation within the next year, he said.

“Even though we don’t expect the amounts of drug to have any behavioral effects, giving it to the eye is going to reduce the exposure to the brain, so that will further minimize any risk of somebody tripping if they are giving themselves some eyedrop with our drug in it,” Nichols said.

5-HT2A Receptor Activation

Nichols and Eleusis are in the process of developing third- and fourth-generation psychedelic-derived anti-inflammatories with even less behavioral effect than ELE-02 that could eventually be sold, likely in pill form, for the treatment of systemic inflammation and conditions like arthritis and asthma.

Such molecules would be considered standard pharmaceuticals rather than psychedelics, even though they are derived from DOI and target the same cell receptor in the brain – Serotonin 5-HT2A – that produces hallucinogenic effects.

Interestingly, the behavioral, neuroplastogenic, and anti-inflammatory properties of psychedelic drugs all appear to stem from this single receptor, a subject of growing interest within the neuroscience field. The difference is that slightly different molecules, perhaps by as little as a single atom, can exert unique downstream effects by recruiting different enzymes to the cell.

“When you activate [5-HT2A], it signals to the inside of the cell that it’s been activated with a ligand, and that recruits probably a dozen or so, if not more, different enzymes, to do different things to the cell to change its physiology,” he explained. “And what we’ve shown is that the two major pathways that have been linked to behavioral changes do not correlate with the anti-inflammatory ability.”

For example, DOI has both strong behavioral and anti-inflammatory effects. ELE-02 has strong anti-inflammatory but modest behavioral effects. LSD is an extremely potent psychedelic but only partially effective as an anti-inflammatory, at least compared to DOI and psilocin, a byproduct of psilocybin.

Meanwhile, the endogenous hallucinogen N, N-dimethyltryptamine (DMT), another “classic” psychedelic that activates the 5-HT2A receptor, produces profound subjective effects yet appears to have no efficacy as an anti-inflammatory.

And the neurotransmitter serotonin, which binds to 15 different 5-HT receptor subtypes, produces no hallucinogenic effects and actually promotes inflammation.

In their search for the perfect anti-inflammatory drug, Nichols and his colleagues are working to continue isolating anti-inflammatory pathways from behavioral pathways, then tune the latter down or all the way out by further modifying the composition and configuration of the original DOI molecule.

“The goal is to have a non-psychedelic drug that somebody could take that would activate the 5-HT2A receptor to treat something like arthritis,” Nichols said. “Right now it’s really in the early stages of that, we’re just trying to understand mechanisms. Once we identify what the enzyme is that we’re recruiting to the receptor, that’s going to help us better design drugs.”

LSD for Alzheimer’s

Nichols and Eleusis are also engaged in other research projects involving classic psychedelics. One is centered on using psilocybin to treat depression, a condition that (like addiction and post-traumatic stress disorder) is associated with inflammation.

Additionally, the company is investigating the use of low doses of LSD to treat Alzheimer’s Disease. It recently completed a successful Phase 1 trial of safety and tolerability in 48 older healthy volunteers.

Scientists believe that neuroinflammation plays a role in Alzheimer’s.

"But LSD holds promise as a treatment for the disease because of much more than just its anti-inflammatory effect," Nichols said.

“LSD targets nearly all serotonin receptors, and dopamine receptors, among others, for activation,” he said. “Activation of several of these receptors have been shown by us and others using different chemicals individually to have benefits such as enhancing memory, maintaining cell health … and reducing stress-related markers. We think that these multiple positive effects from several different receptors may act synergistically together to be able to slow the neurodegeneration and cognitive symptoms of Alzheimer’s Disease.”

Although LSD would be administered at sub-hallucinogenic doses to Alzheimer’s patients, its potential psychoactivity could be construed as a safety risk by the FDA, Nichols acknowledged. That’s less of a concern with ELE-02, and shouldn’t be an issue with subsequent generations of 5-HT2A anti-inflammatories possessing low or no behavioral activity that are now being developed in Nichols’ lab.

“With anti-inflammatories…it’s much more of a traditional pathway,” Nichols said. “We could go to the FDA and show that there’s no adverse behavioral effects, we have efficacy, we have safety. For the new molecules that we’re making, they’re not scheduled [by the Drug Enforcement Administration]. At that point the FDA will decide, well then what do we do with it? Let’s just make it Schedule 4, and now you’ve got a new anti-inflammatory on the market.”

 
Last edited:
Histamine-may-be-a-missing-link-connecting-inflammation-and-depression-0--1536x828.jpg


Inflammation and Depression*

by Luana Steffen | Intelligent Living | 18 Nov 2021

For several decades scientists have seen a distinct association between inflammation and depression. Various autoimmune conditions, including inflammatory bowel disease, have been steadily linked with higher rates of mood disorders, while chronic, low-grade inflammation has been linked with lower dopamine levels and motivation. However, there is still significant debate over what kind of causal mechanism could directly connect inflammation with depression.

Histamine

A new robust study conducted by an international team of scientists has discovered one possible missing link clarifying how inflammation leads to depression. Through a series of mouse experiments, the team found evidence that inflammation-induced histamine activity can inhibit the release of serotonin in the brain. Thus, the findings suggest histamine may play a significant role in triggering depression.

The scientists started by making a new kind of microelectrode that can be implanted in the hippocampus of mice to measure serotonin levels, a critical mood-regulating neurotransmitter, in real-time. Serotonin is often focused on as a therapeutic target for depression.

Next, the team injected the mice with a toxin that triggers inflammatory responses to investigate how inflammation impacts serotonin activity. Within only minutes, serotonin levels were seen plummeting in the animals’ hippocampus. Since the toxin used in the experiment cannot cross the blood-brain barrier, the researchers were confident that inflammation was responsible for the serotonin reduction.

Further investigations led to the researchers discovering that increased histamine levels in the brain, produced as part of the animals’ inflammatory response, caused the reduction in serotonin levels by “acting on inhibitory histamine H3 heteroreceptors on serotonin terminals.”

The next phase of the experiment involved administering the animals’ escitalopram, a common anti-depressant drug belonging to a family known as selective serotonin reuptake inhibitors (SSRIs). However, serotonin levels did not increase in the animals treated with the inflammatory toxin SSRI administration. Instead, the anti-depressant’s effect was blunted since it hindered the brain’s ability to clear histamine, interfering with the regulation of serotonin levels.

Finally, the researchers treated the animals with histamine-reducing drugs alongside the anti-depressant to confirm this potential mechanism. And sure enough, this combination of drugs saw serotonin levels increase in the inflammation-induced mice.

Parastoo Hashemi, the study’s lead author, said:
Our work shines a spotlight on histamine as a potential key player in depression. This, and its interactions with the ‘feel-good molecule’ serotonin, may thus be a crucial new avenue in improving serotonin-based treatments for depression.

People who have or know someone with allergies and hay fever are familiar with histamines. These molecules play a significant role in the body’s immune response to foreign pathogens and influence sleep cycles, sexual function, and blood pressure.

Standard anti-histamines that are available over-the-counter for allergies work by hindering the ability of histamine molecules to bind with specific histamine receptors. Therefore, it is essential to note that these common anti-histamines for allergies do not affect this newly discovered mechanism.

Instead, in this study, the scientists used drugs that completely stifle histamine levels across an entire body. Therefore, since histamine is vital for various bodily functions, it is not feasible to translate these findings to humans just yet.

In addition, this research, published in the journal JNeurosci, was only conducted on animals. That said, more work is needed to confirm whether the exact mechanism is at play in humans. Nevertheless, the team suggests that it is possible this inflammation-induced histamine mechanism could help shed light on some of the inconsistencies in SSRI anti-depressant effectiveness from patient to patient.

However, suppose it can be validated in humans, and a pharmacological target is identified. In that case, new depression treatments can be developed, including ones that boost the efficacy of currently available SSRI drugs by preventing histamine from disrupting serotonin levels.
Inflammation is a whole-body response and is therefore hugely complex. Depression is similarly complex, and the chemicals involved are affected in myriad ways by both genetic and environmental factors. Thus, we need to look at more complex models of depression behaviors in both mice and humans to get a fuller picture of both histamine and serotonin’s roles in depression.

Gut microbiota

Unhealthy gut microbiota can lead to several conditions, including inflammation and depression. For example, a 2019 study found that two different gut bacteria are depleted in people with depression, regardless of anti-depressant treatments. They discovered this from the first population-level study on the link between gut bacteria and mental health, precisely quality of life and depression.

Therefore, what you eat is significant. A good diet full of fiber and varied fruits and vegetables is vital for a healthy gut microbiome. Aside from fighting depression, making the right food choices can also help control histamine levels and combat inflammation.

Foods to control histamine levels

A healthy diet comprises moderate histamine levels. However, some foods that are high in histamine can trigger inflammatory reactions and other negative symptoms.

Histamine-rich foods to avoid:​
  • Alcohol and other fermented beverages;​
  • Fermented foods and dairy products, like yogurt and sauerkraut;​
  • Dried fruits;​
  • Avocados;​
  • Eggplant;​
  • Spinach;​
  • Processed or smoked meats;​
  • Shellfish;​
  • Aged cheese​

Foods that activate histamine release in the body:​

  • Alcohol;​
  • Bananas;​
  • Tomatoes;​
  • Wheat germ;​
  • Beans;​
  • Papaya;​
  • Citrus fruits;​
  • Nuts, specifically walnuts, cashews, and peanuts;​
  • Food dyes and other additives.​
People who have histamine intolerance should incorporate low-histamine foods into their diet to help reduce negative symptoms. However, keep in mind that a histamine-free diet does not exist.​

Some of the best foods to eat that are low in histamine:​

  • Fresh meat and freshly caught fish;​
  • Non-citrus fruits;​
  • Eggs;​
  • Gluten-free grains, like rice and quinoa;​
  • Dairy substitutes, such as almond milk and coconut milk;​
  • Fresh vegetables except for spinach, avocados, tomatoes, and eggplant;​
  • Cooking oils, like olive oil.​
Foods to fight inflammation

Aside from depression, inflammation can result in many harmful health conditions, like cancer, heart disease, and obesity. What you eat plays a big part – it can either reduce inflammation or cause it.

A minor inflammation is natural because it’s part of the body’s immune response necessary for healing. However, when it’s out of control, that’s when serious health issues occur. So, here are some tips and a list of the best things to eat that will keep your inflammation levels to a minimum.

First: Eliminate foods and beverages that cause inflammation, including high-sugar, processed, and fatty foods (like doughnuts, fried chicken, and soda, for example).

Second: Incorporate foods that curb inflammation. These include:​
  • Oily fish- such as anchovies, herring, mackerel, salmon, sardines, and tuna.​
  • Whole grains- Avoid refined white bread, rice, cereal, and pasta. Instead, consume whole grains.​
  • Dark leafy greens- such as broccoli, brussels sprouts, kale, spinach, and swiss chard.​
  • Avocados.​
  • Nuts.​
  • Peppers.​
  • Tomatoes.​
  • Beets.​
  • Turmeric.​
  • Garlic and onions.​
  • Extra virgin olive oil.​
  • Parsley.​
  • Berries.​
  • Green tea and Matcha.​
  • Dark chocolate and cocoa.​
Some foods help relieve tension more than others, while the lack of some stress-fighting foods can make you feel worse (like anxiety, depression, and panic attacks).

Of course, occasional bouts of stress are usual. But chronic stress isn’t, and it can lead to severe health problems not only mentally but physically too (like heart disease). So, to help, here’s a list of some of the best things to eat that will keep your stress levels to a minimum.

Green Leafy Vegetables: Spinach and other leafy vegetables contain a vitamin called folate (also known as folic acid or Vitamin B9) that reduces the risk of depression symptoms.

Sweet Potatoes: Sweet potatoes are a great source of whole, nutrient-rich carbs, vitamin C, and potassium, helping lower stress hormone cortisol levels. Chronic stress can lead to cortisol dysfunction, resulting in pain, inflammation, and other adverse effects.

Artichokes: Artichokes are high in magnesium, potassium, vitamins C, and vitamin K, all of which are essential for a healthy stress response. On top of that, they’re especially rich in prebiotics, a type of fiber that feeds the good bacteria in your gut.

Artichokes contain a concentrated amount of prebiotics like fructooligosaccharides (FOSs), which have been shown to reduce stress levels in animal studies. Another study demonstrated that people who consumed five or more grams of prebiotics daily experienced improved depression and anxiety symptoms.

Broccoli: Eating broccoli or other cruciferous vegetables can reduce your risk of mental health disorders as well as certain cancers and heart disease. Broccoli can be considered a superfood with concentrated nutrients, including stress-fighting magnesium, folate, and vitamin C.

Another excellent thing broccoli provides is a high amount of sulforaphane – a sulfur compound with neuroprotective properties. In addition, it may offer anti-depressant and calming effects – and vitamin B6, which is linked to a lower risk of depression and anxiety in women if taken in higher amounts. One cup (184 grams) of the vegetable (cooked) has more than 20% of the daily value for vitamin B6.

Blueberries: Blueberries are high in flavonoid antioxidants, which provide powerful neuroprotective and anti-inflammatory effects. Eating flavonoid-rich foods may protect against depression and improve your mood.

Organ Meats: The heart, liver, and kidneys of animals are an excellent source of B vitamins – especially B6, B12, folate, and riboflavin. These vitamins are essential for stress control because they produce neurotransmitters like serotonin and dopamine that regulate mood.

Turkey Breast: Turkey breast contains the amino acid tryptophan, which helps produce serotonin – the chemical that regulates feelings of well-being, happiness, and hunger. Other foods high in tryptophan include beans, lentils, nuts, seeds, oats, tofu, fish, and eggs.

Fatty Fish: Fatty fish like herring, mackerel, salmon, and sardines are high in mood-enhancing omega-3 fats and vitamin D. Omega-3s are so essential for brain health that low omega-3 intake may cause increased anxiety and depression. Similarly, low levels of Vitamin D can result in an increased risk of anxiety and depression.

Dark Chocolate– small indulgence of dark chocolate (70% cocoa or more) may have the power to regulate your stress levels because it can reduce stress hormones like cortisol.

Chamomile Tea– The medicinal herb Chamomile has been used as a natural stress reducer since ancient times. It can be consumed to promote restful sleep and reduce depression and anxiety symptoms.

Psychedelics

In 2019, Johns Hopkins researchers surveyed the anti-depressant qualities of DMT – a short-acting psychedelic. They reported that it resulted in extraordinarily positive improvements in self-reported depression and anxiety when given in a ceremonial group setting.

Another study, conducted last year, revealed preliminary evidence that psychedelic drugs can improve mental health by forcing individuals to confront their issues and become more accepting of distressing experiences. The research adds to a growing body of work that suggests using psilocybin substances can result in long-term improvements in depressive symptoms.

According to the study’s author Richard Zeifman, a Ph.D. student at Ryerson University, psychedelic therapy is promising as a novel treatment for a variety of mental health conditions, including:​
  • Anguish, associated with a life-threatening illness.​
  • Major depressive disorder.​
  • Substance use disorders.​
Zeifman added:
Our results provide further support for the negative mental health effects associated with avoidance. This can be summed up with a saying that is often used in the context of psychedelic therapy, that ‘The only way out is through.’

Besides confronting your problems, other simple ways to avoid depression include:​
  • Reducing alcohol and drug use.​
  • Quitting nicotine.​
  • Exercising regularly.​
  • Cutting back on social media.​
  • Staying away from toxic people.​
  • Reducing stress.​
  • Minimizing your daily chores.​
  • Getting plenty of rest.​

*From the article here :
 
Last edited:
oslo-universitet_1280.jpg

Institute of Clinical Medicine, University of Oslo

Psychedelics as a novel approach to treating autoimmune conditions

Caitlin Thompson, Attila Szabo | December 2020

With a rise in the incidence of autoimmune diseases (AiD), health care providers continue to seek out more efficacious treatment approaches for the AiD patient population. Classic serotonergic psychedelics have recently been gaining public and professional interest as novel interventions to a number of mental health afflictions. Psychedelics have also been shown to be able to modulate immune functions, however, while there has been great interest to researching into their psychotherapeutic applications, there has so far been very little exploration into the potential to treat inflammatory and immune-related diseases with these compounds. A handful of studies from a variety of fields suggest that psychedelics do indeed have effects in the body that may attenuate the outcome of AiD. This literature review explores existing evidence that psychedelic compounds may offer a potential novel application in the treatment of pathologies related to autoimmunity. We propose that psychedelics hold the potential to attenuate or even resolve autoimmunity by targeting psychosomatic origins, maladaptive chronic stress responses, inflammatory pathways, immune modulation and enteric microbiome populations.

1. Introduction

The incidence of autoimmune diseases (AiD) has been steadily increasing in the last decades, as reported by The National Institute of Health (NIH) and other sources. There are currently over 100 defined autoimmune-related diseases affecting roughly 50 million Americans, or 20 % of the population according to the American Autoimmune Related Diseases Association (AARDA). The NIH also calculates that over $100 billion in health care costs is spent annually in the United States on treating those with autoimmune diseases, compared to cancer which has an estimated cost of $50 billion annually. It is conservatively estimated that 80 % of those affected by AiD are women. There are several theories for why women are more susceptible to AiDs, but known reasons remain unclear. Different theories for the increased prevalence of AiDs include overuse of antibiotics, increase in environmental toxin exposure, increased caesarian births, reduced breast feeding, improvement of diagnostic tools, increased awareness of AiDs and increased societal stressors.

Autoimmunity is defined as the loss of tolerance to self-antigens in which the immune system attacks healthy tissues in an individual. While there are different characteristics of expression and tissues affected amongst the autoimmune disorder types, there are many shared mechanisms of action and potential origins. Many diseases that were initially considered to be unrelated to autoimmunity are now being reexplored as autoimmune-related, especially in the field of psychiatry. This includes major depressive disorder (MDD), schizophrenia, Parkinson’s disease, Alzheimer’s disease and Amyotrophic Lateral Sclerosis.

While the causes of AiDs are controversial, they appear to be multifactorial with many facets that contribute to their development such as genetic predisposition, environmental triggers, and psychosocial stress. There is strong evidence that traumatic experiences such as Adverse Childhood Events (ACEs) and prolonged stress may contribute to the development of an AiD. AiDs often encompass a variety of symptoms and features. Common characteristics observed among the various types of AiD include increased vascular and epithelial (e.g. intestinal) permeability, chronic inflammation, the presence of chronic infections, dysregulated Hypothalamic-pituitary-adrenal axis (HPA axis), mitochondrial dysfunction, and microbiome dysbiosis. Symptoms can manifest as chronic fatigue, allergies, psychiatric and mood disorders, pain, rashes, GI distress, poor cognition and more.

Currently, biologics and immunosuppressive drugs are the primary treatment tools for autoimmune disorders along with non-steroidal anti-inflammatory drugs, steroids and antidepressants. However, immunosuppressant and biologic medications designed to resemble antibodies, receptors and other immunological factors can result in an immune compromised state, putting the affected population at risk for serious infections or immune-related illnesses. Although existing biological drug approaches show evidence for producing positive treatment outcomes in some individuals, the increased risks for infection, malignancy and cardiovascular conditions, as well as contradicting results in efficacy studies have prompted interest in exploring other approaches to treating AiDs.

2. The immunopharmacology of psychedelics

2.1. The link between autoimmunity and mental disorders


The comorbidity between autoimmune conditions and mental and mood disorders, such as MDD, anxiety, schizophrenia, and bipolar disorder has become apparent in the last two decades. There is a higher risk of developing clinical depression or mood disorders if one has been diagnosed with an autoimmune condition. While there is certainly argument that the burden of having an autoimmune condition could contribute to MDD, researchers suggest that depression and anxiety symptoms could perhaps be a result of autoimmune mechanisms and resulting inflammation occurring in the nervous system, or via dysregulated inflammatory cytokine loops between peripheral and brain-resident immune cells. Potentially by their immunomodulatory activity and in part through the mobilization of cell-intrinsic neuroprotective mechanisms, psychedelics may represent a promising intervention for autoimmune-related depression and other mental illness.

2.2. Pharmacology of psychedelics substances

Psychedelic compounds have recently made their way back into mainstream medical research. Psychedelic compounds such as LSD, psilocybin, DMT, ibogaine and mescaline have been of great interest based on their apparent high efficacy for treating mental afflictions and their impressive safety profile. Psychedelic drugs were popularized in the 1960s by researchers such as Timothy Leary, Ram Dass, Terrence McKenna and other enthusiasts. Between the 1960’s and the 1980’s a number of scientific studies were being conducted on psychedelic substances including biochemical, pharmacological and psychological studies. Therapists were experimenting with using psychedelics as adjuncts to various psychotherapy methods. Due to radical activism and drug propaganda, psychedelics were scheduled as Schedule I substances by the Controlled Substances Act in 1970 and were made illegal to consume, possess or distribute. Contradictory to this scheduling decision, research currently suggests that class psychedelic compounds do not show evidence of addiction potential and exhibit anti-addiction properties while also appearing to be generally physiologically safe, non-toxic and exhibiting minimal individual and societal harms. This prohibition resulted in an abrupt halt in research on psychedelics. In the early 90’s there was the start of a new renaissance in psychedelic research with the work of Risk Strassman on intravenous DMT administration in humans. Psychedelic research currently is a rapidly accelerating field in science, psychology and medicine.

“Classic” psychedelics generally refer to LSD, psilocybin, mescaline, ibogaine, DMT, and 5-MeO-DMT, and are typically defined by their indole-containing molecular structure as illustrated in Fig. 1. Non-classic psychedelics such as ketamine and MDMA may have some similar receptor activity and hallucinogenic properties, but lack this fundamental indole structure. Classic psychedelics typically and predominately exhibit agonistic activity at various serotonin (5-hydroxytryptamine, 5-HT) receptors, with the most commonly shared feature being their 5-HT2A receptor agonist binding. The classic psychedelics vary in their receptor binding properties but overall show binding affinity for a variety of receptors including 5-HTR subtypes. LSD [Fig. 1.2] is known to bind to 5-HT1A, 1B, 1D, 1E, 2A, 2B, 2C, 5A, 6, and 7 as well as dopaminergic receptors subtypes D1 and D2. Psilocin, the active metabolite of psilocybin [Fig. 1.3], binds to 5-HT1A, 1D, 2A, 2C but does not bind to dopaminergic receptors. Mescaline exhibits the highest binding affinity to the 5-HT2A and 2C receptors.

1-s2.0-S0165247820303977-gr1.jpg

Fig. 1. Structure of classic psychedelic compounds. Many classic psychedelics share an indole structure
within their molecular makeup, often characterizing them as serotonin analogues and agonists.

DMT [Fig. 1.4] and its psychopharmacologically more potent analogue 5-MeO-DMT [Fig. 1.5] bind to the 5-HT1A, 1B, 1D, 2A, 2B, 2C, 5A, 6 and 7 receptors, as well a number of iontropic and metabotropic glutamate receptors, dopamine, acetylcholine, Trace-Amine-Associated Receptors (TAAR), and the opioid-like receptor sigma-1 (Sig1R). DMT is found in trace amounts in most plant, animal (including humans) and fungal organisms with some plants, such as the Acacia and Mimosa genera, containing more substantial amounts. It is one of the components in the traditional Amazonian brew ayahuasca, which combines DMT-containing plants, such as Psychotria viridis with the Monoamine Oxidase Inhibitor (MAOI)-containing Banisteriopsis caapi (ayahuasca) vine to potentiate a psychoactive effect from the oral administration of DMT Fig. 2. The ayahuasca vine contains harmala alkaloids, such as harmine, harmaline and tetra-harmaline that act as MAOIs in the ayahuasca brew. Typically, the DMT contained in the ayahuasca brew is considered the psychoactive constituent, however a ritual called Natemamu does exist in the Shuar culture in which only the B. caapi vine is used, suggesting that at certain doses, the MAOIs from the ayahuasca vine on its own can create a hallucinatory experience.

Fig. 2

Fig. 2. Naturally occuring psychedelics. (2.1) Banisteriopsis caapi, also known as the ayahuasca vine, contains monoamine-oxidase inhibitors which, when combined with a DMT-containing plant, such as Psychotria viridis, make up the psychoactive brew known as ayahuasca. (2.2) Incilius alvarius, commonly known as the Colorado River Toad, excretes 5-MeO-DMT in its defensive secretions. (2.3) Lophophora williamsii, or Peyote, has been used as a sacrament in traditional Native American ceremonies for thousands of years. (2.4) Psilocybe cubensis is a common species of psilocybin mushrooms. Images sourced from Wikipedia.

5-MeO-DMT is also found in many plants and animals including in the Amazonian shamanic snuff called “yopo” (Anadenanthera peregrinaa), mammalian tissues and within the defensive secretion of the Colorado River Toad (Incilius alvarius) Fig. 2. Because of the degradation of the DMT and 5-MeO-DMT molecules by MAO in the digestive tract, the most common way of ingesting these substances is through inhalation via vaporization. There is a long history of mixing DMT with MAOIs in various South American traditions, however, there does not appear to be any traditional practices mixing 5-MeO-DMT with MAOIs as it appears to be potentially dangerous due to higher 5-HT binding affinity and disparate metabolic degradation pathways.

Some “not-so-classic” psychedelic compounds, such as ketamine and ibogaine have complex pharmacology that is not 5-HT dominant. Ketamine [Fig. 1.6] exhibits non-competitive NMDA receptor antagonism, while displaying monoaminergic release as well as cholinergic, adrenergic, μ- and δ-opioid receptor binding. Ibogaine [Fig. 1.7], an alkaloid from the Tabernanthe iboga shrub, has a highly promiscuous binding affinity for the 5-HT and dopamine (DA) transporters, non-competitive antagonism for several types of nicotinic AChR, NMDA Glu, μ-, δ- and к- opioid, Sig1R and Sig2 receptors. MDMA [Fig. 1.8], while generally referred to as an empathogen rather than a true psychedelic, releases presynaptic vesicular monoamines 5-HT, DA and norepinephrine (NE) while inhibiting their reuptake into the presynaptic cell, flooding the synapse. It does however have some agonistic binding affinity to 5-HT2A receptors like classic psychedelics, as well as muscarinic receptor 1, and the H1 histamine receptor.

Although at this point there has been increased exploration into the use of psychedelics as tools for the treatment of MDD, post-traumatic-stress-disorder (PTSD), addiction, eating disorders, anxiety and other mental disorders, there has been very little investigation into the immune modulating and anti-inflammatory effects of psychedelics. Biochemical and anecdotal evidence suggests that psychedelics could provide a novel treatment approach to AiDs and other immune and inflammatory-related diseases. It is likely that psychedelics can attenuate autoimmunity via a number of different mechanisms. Psychedelics may directly and indirectly target a number of physiological factors and resulting dysfunctions in AiDs. This review covers the current evidence that psychedelics may offer therapeutic solutions and potential modalities for the attenuation of AiDs.

2.3. Inflammation and immune modulation

In AiD, rampant chronic inflammation is a keystone feature. Elevated levels of cytokines and their dysfunctional regulation involving interleukin(IL)-6, IL-1β, IL-17, tumor necrosis factor-α (TNF)-α, IL-12, interferon(IFN)-γ and others are a shared feature observed in many AiDs including Rheumatoid Arthritis (RA), Systemic Lupus Erythematosus (SLE), systemic sclerosis, and Sjögren's syndrome. Lipopolysaccharide (LPS) is an inflammatory component of the cell wall of Gram-negative bacteria that can induce inflammation and excessive oxidative stress that may play a role in elevated inflammatory biomarkers seen in those with AiD diseases. Those with AiD display auto-antibodies and yet also display immunodeficiency. Immune modulation appears to be a feasible target in approaching the treatment of AiD.

While the area of research is quite underdeveloped, there is some evidence that psychedelic compounds have anti-inflammatory and immune-modulating effects Table 1. For instance, LSD displays the ability to suppress the proliferation of B-lymphocytes, as well as the production of the pro-inflammatory cytokines IL-2, IL-4, and IL-6 in in vitro splenic lymphocytes derived from 6−8 year old female B6C3F1 rats at concentrations of LSD between 1–100 μM. The same study observed that administered doses of LSD between 0.001−0.1 μM in rat lymphocytes increased the number of Natural Killer (NK) cells, while concentrations of 100 μM suppressed NK cell production. In another study, the substituted amphetamine DOI, was shown to reduce TNF-α levels in 10 week old adult male C57BL/6 J mice via agonism of the 5-HT2A receptor in doses ranging from 0.01 μM/kg to 10μM/kg. Furthermore, in a recent study the same group expanded the previously observed anti-inflammatory effects of 5-HT2A agonism to additional psychedelics in a rat model of lung inflammation.

Table 1. Receptor modulatory and physiological effects of psychedelics.
�​
Classic psychedelics​
Receptors of interest in Autoimmune Disease​
Resulting effect​
References​
LSD​
5-HT2A (partial agonism)​
TNFα, IL-6, IL-2​
TrK-B (agonism)​
^BDNF, ^ mTOR​
Psilocybin​
Trk-B (agonism)​
^BDNF, ^ mTOR​
DMT & 5-MeO-DMT​
Sig1R (agonism)​
^IL-10, IL-1β, IL-6, TNFα CXCL8/IL-8​
5-HT2A (agonism)​
^BDNF, ^ mTOR​
Trk-B (agonism)​
Glu excitotoxicity​
ROS damage​
Cell apoptosis​
mescaline (peyote)​
Trk-B (agonism)​
^BDNF, ^ mTOR​
penicillin resistant staphylococcus bacteria​
ayahuasca (DMT & Harmala alkaloids)​
5-HT2A​
Cortisol​
Sig1R​
^BDNF, ^ mTOR​
Trk-B​


�​
Non-classic psychedelics​
Receptors of interest in Autoimmune Disease​
Resulting effect​
References​
Ketamine​
NMDA Glu (antagonism)​
^BDNF, ^ mTOR, Glu excitotoxicity​
IL-6, TNFα​
MDMA (in assisted psychotherapy)​
VMAT, SERT​
Amygdala response​
HPA-axis dysregulation​
Harmine/Harmaline​
MAO (inhibitor)​
^5-HT, NF-kB, HSV-2 ^NK cell numbers/activity, Glu excitotoxicity​
Ibogaine/Noribogaine (metabolite)​
5-HT2A (partial agonism)​
^BDNF, ^ mTOR​
Sig1R (agonism)​
TrK-B (agonism)​

One human study where ayahuasca was given to healthy volunteers recorded blood levels of lymphocytes and found a decrease in CD4 and CD3 cells, and an increase in NK cells. This was compared to controls and subjects treated with D-amphetamine. Another, more recent human study reported significant decrease in C-reactive protein levels in the blood in both healthy and depressed volunteers, but no significant reduction in IL-6 blood levels. In addition, in a human intramuscular ketamine study, levels of proinflammatory markers, including C-reactive protein (CRP), IL-6, and TNF-α, were examined prior to initiation of test infusions, 40 min post-infusion, 240 min post-infusion, and on day 3 and day 7 post-fusion in patients who received 0.5 mg/kg or 0.2 mg/ kg of R/S-ketamine hydrochloride. Acute reductions of IL-6 and TNF-α were found in the 0.5 mg/kg group between 40 min. and 240 min post-infusion, but not in day 3 or day 7. Reductions in pro-inflammatory cytokine levels were directly correlated with depression scores, except on days 3 and 7 when depression scores improved despite cytokine levels returning to pre-infusion baselines. Interestingly, a different study obtained contradicting results showing an increase in IL-6 post-infusion and no correlation of cytokine levels with anti-depression outcome, suggesting that more studies need to be conducted to understand the complex systemic immune effects of ketamine and its metabolites.

There is substantial literature on the 5-HT system and its complex inflammation and immune-regulating abilities in tissue specific manners. Functional studies showed that 5-HT modulates the release of IL-1beta, IL-6, IL-8/CXCL8, IL-12p40 and TNF-α, while it has no effect on the production of IL-18 and IFN-gamma in LPS-stimulated human blood monocytes. 5-HT can also modulate human macrophage polarization and dendritic cell functions, and can contribute to the maintenance of an anti-inflammatory state via 5-HT2B and 5-HT7 receptor binding. Given the role of the 5-HT system in immune-modulation and inflammatory properties, it is highly likely that there are undiscovered immune and inflammatory effects from exposure to psychedelic compounds due to their serotonergic activity. In the previously mentioned study where DOI was observed to reduce TNF-α in mice, in groups where the 5-HT2A antagonist drug M100907 was administered, the reduction in TNF-α was not observed supporting the theory that the 5-HT2A receptor and the 5-HT system as a whole may be fundamental to creating downstream immunological and inflammatory-regulating effects.

In addition to the 5-HT receptors, DMT and 5-MeO-DMT also have high binding affinity for the Sig1R. The Sig1R plays a fundamental role in the regulation of different cellular mechanisms such as mitochondrial function, apoptosis, proliferation, and neuroprotection. Sig1R also modulates inflammatory and immune responses by regulating the activation of the transcription factors nuclear factor kappa B (NF-кB) and several mitogen-activated protein kinases (MAPKs). Both NF-кB and MAPKs are important regulators of gene transcription involving immune responses and the production of inflammatory cytokines.

Abnormal Sig1R functions have been implicated in a number of psychiatric and inflammatory-related conditions such as MDD, Alzheimer’s disease, Parkinson’s disease, cardiovascular disease, immune reactions, and proliferation of cancer cells. Activation of the Sig1R is also pivotal in facilitating stress responses. Dysregulated NF-кB and MAPK activity patterns have been implicated in a number of AiD pathologies. One study showed evidence of suppression of pro-inflammatory NF-кB cytokine production by administration of harmine, a component of the ayahuasca vine, via intraperitoneal injection at doses of 25 and 50 mg/kg, in response to treatment with LPS in Male Kunming mice to induce acute kidney injury. Another study found that harmine inhibited tumor necrosis factor-α (TNF-α)- and LPS-induced NF-κB transactivity and nuclear translocation in mouse macrophage RAW 264.7 cells when treated with concentration between 2.5–25 μM. 5-MeO-DMT was also found to inhibit the NF-κB signaling pathway in human cerebral organoids exposed to concentrations between 23 nM to 7.11 μM, and examined for proteomic analysis via mass spectrometry. In other in vitro and animal in vivo studies, DMT and 5-MeO-DMT have been shown to increase anti-inflammatory IL-10 while decreasing the levels of the pro-inflammatory IL-1β, IL-6, TNF-α, and the chemokine CXCL8/IL-8 in human monocyte-derived dendritic cells at concentrations of 100 μM. DMT also exhibited considerable neuroprotective and anti-neuroinflammatory effects in a rat model of stroke where male Wistar rats received an intra-peritoneal (IP) bolus of 1 mg/kg-body weight (bw) DMT followed by a maintenance dose of 2 mg/Kg-bw/h delivered over 24 h after induced transient middle cerebral occlusion.

Furthermore, inhaled 5-MeO-DMT has recently been reported to decrease salivary IL-6 levels in a study involving a small group of human subjects. There results suggest that serotonergic psychedelics may emerge as potential candidates in the treatment of autoinflammatory and autoimmune conditions Table 1. Since pharmaceutical grade DMT and other tryptamine analogs designed for human clinical trials are already available or will soon enter the market, testing the in vivo physiological effects of serotonergic psychedelics in humans is now closer than ever before.

2.4. Trauma and emotional effects

A number of studies have suggested that significant or prolonged stress, especially in early childhood, contributes to the development of a number of diseases including AiD. The severity of childhood trauma can be clinically assessed by scoring ACEs (Adverse Childhood Experiences). Many studies have found strong correlations between ACEs and the statistical risk of developing an AiD including studies looking at associations between childhood stress and RA, SLE, and fibromyalgia.

The downstream physiological detriment due to stress and vagal nerve disruption is well documented in the literature and appears to play a significant role in the development of an AiD. Psychological stress and psychoemotional trauma can severely compromise the immune system leading to increased risk of chronic infections and gut dysbiosis. Psychosocial stress increases inflammatory cytokines, oxidative stress, glutamate excitotoxicity, inhibits effective digestion and nutrient absorption and contributes to HPA axis dysfunction. Stress has been found to increase intestinal permeability which can lead to food sensitivities and LPS-induced inflammation due to gut-blood transepithelial bacterial translocation (“Leaky Gut syndrome”). All of these inflictions are hallmark characteristics of patients with AiD, further supporting the hypothesis that stress may play a significant role in AiDs.

The psychedelic compounds psilocybin, LSD, MDMA, and ayahuasca are currently being explored for their promising potential to assist in treating trauma-derived illnesses such as PTSD, depression, and addiction. The research with MDMA assisted psychotherapy for PTSD is currently the furthest along in its drug development and approval process by the FDA than any other psychedelic-related compound with psilocybin, cannabis and ayahuasca following closely behind. MDMA assisted psychotherapy has received breakthrough status by the FDA, accelerating its entry into mainstream medical practice. The MDMA therapy for PTSD protocol developed by MAPS, consisting of two MDMA-assisted therapy sessions with the patient receiving 125 mg of MDMA orally in conjunction with non-directive psychotherapy, along with preparatory and post non-MDMA therapy sessions, has so far exceeded all existing treatments for PTSD in its symptom remission statistics. Although the classic psychedelics have not been clinically established as strongly for PTSD as MDMA has, there is evidence that they also hold potential for stress and trauma-oriented applications. There are several human and non-human primate studies that suggest that the DMT-containing ayahuasca brew has modulatory effects on salivary cortisol response and plasma cortisol in both healthy normals and in individuals with treatment-resistant MDD. Thus, potential neuroendocrine-immune modulation is one of the recently proposed mechanism for the rapid anti-depressant effects of ayahuasca in those with MDD.

Given that trauma and chronic sympathetic activation are prevalent among those with AiD, the therapeutic outcome of psychedelic-assisted psychotherapy could resolve or improve stressful psychological states that cause or contribute to physiological outcomes seen in the majority of AiD patients.

2.5. Glutamate excitotoxicity

Those with autoimmune conditions, particularly those affecting the nervous system, are more likely to have increased glutamate (Glu) binding sensitivity and post-synaptic glutamate levels which can result in cytoxicity to the nerve cell. The term glutamate excitotoxicity is used to describe the cytotoxic effects of excessive and uncontrolled release and binding of glutamate to post-synaptic neurons. Hypersensitization of Glu receptors due to cytokine-mediated transcriptional changes that affect depolarization gradients potentially play a role in this glutamate-induced cytotoxic phenomenon. Excitotoxicity appears to be implicated in a number of brain related diseases, including multiple sclerosis, depression, addiction and neurodegenerative diseases. During glutamate-excitotoxicity, excessive stimulation of the Glu receptors resulting in action potentials to be propagated through the neuron in an uncontrolled manner. This in turn activates the influx of Ca2+ ions into the cytoplasm where they may induce mitochondrial respiration and the release of reactive oxygen species (ROS). Under normal circumstances, the generated ROS are cleared from the cell and transmembrane ion gradients are restored quickly after glutamate transmission. However, in activation-induced excitotoxic cascades, the permeability transition pore (PTP) is opened and mitochondrial dysfunction occurs due to increased oxidative stress, abnormal ATP synthase activity and inhibited mitochondrial respiration. Chronic excitation of the Glu receptors can therefore contribute to oxidative stress, which may result in damage to neural structures or apoptotic cell death. Excitotoxicity from excessive Glu receptor binding may result in changes in cortical, amygdala, and hippocampal density leading to atrophy and shrinkage of brain regions.

While not a classic psychedelic compound, the dissociative anesthetic drug ketamine displays an ability to protect against glutamate-mediated excitotoxicity by non-competitively blocking the NMDA glutamate receptor. It is unclear if inhibition of the NMDA Glu receptor is the fundamental mechanism for the neuroprotective properties or if this, in turn, redirects glutamate to bind to other Glu receptors such as AMPA, Kainate and metabotropic Glu receptors. It appears that blocking the NMDA GluR, while flooding AMPA and metabotropic Glu receptors, paradoxically enhances Glu transmission in the prefrontal cortex in a way that is synaptogenic and potentially restorative for appropriate Glu receptor sensitivity. This could potentially be one mechanism for how ketamine delivers such rapid anti-depressant effects. In theory, blocking or correcting an excitotoxic effect from glutamate may alleviate the oxidative stress and tissue damage to cells by reducing the ROS released via action potentials.

Sig1R agonists such as dimemorfan, and dipentylammonium have shown neuroprotective mediation of glutamate excitotoxicity. Furthermore, DMT has been shown to possess potent neuroprotective effects via the Sig1R in both human in vitro and animal in vivo studies. It may be possible that the psychedelic Sig1R agonists DMT and 5-MeO-DMT are also capable of mediating glutamate levels and receptor activity via indirect transcriptional effects on cytokine production. Harmine, a MAOI contained in the ayahuasca vine has been found to increase Excitatory Amino Acid Transporter 2 (EAAT2) glutamate pump expression in the central nervous system, therefore raising the possibility of reducing glutamate toxicity in vivo.

5-HT receptor activation also has modulatory effects on downstream Glu release and binding activity, in particular through activity at the 5-HT1A, 5-HT1B, 5-HT2, 5-HT3 and 5-HT6 receptors in various brain regions. Downstream Glu effects from 5-HT activity of psychedelic drugs appears to play a role in the neurogenic properties of psychedelic compounds. It is possible that classic psychedelics that act on the 5-HT system may have indirect mitigation of pathological glutamate transmission and receptor sensitivity.

2.6. BDNF and neuroplasticity

The classic psychedelics psilocybin, DMT, 5-MeO-DMT, LSD, the psychedelic ritual brew ayahuasca, as well as the non-classic psychedelics ketamine have shown potential in interacting with neurogenic pathways, such as Tropomyosin receptor kinase B (Trk-B) and the mammalian target of rapamycin (mTOR), in an equivalent manner to the neurotrophin protein brain-derived neurotrophic factor (BDNF), and in influencing neuroplasticity in in vitro and in vivo models of Drosophila melanogaster, rats and humans Table 1. There are conflicting results from studies measuring BDNF gene expression and protein levels in serum with some studies showing evidence of enhanced BDNF gene expression as a result of exposure to psychedelics (LSD) in in vivo rat models, while others show no increase in gene expression but increase in BDNF protein levels in vitro and in vivo (serum) in rats. In studies, where the Trk-B selective antagonist ANA-12 was administered, the synaptogenic and neurogenic properties of psychedelics and BDNF are completely blocked. Similarly, treating rat cortical neurons with the drug rapamycin to inhibit mTOR also blocked the psychedelic-induced neuritogenesis suggesting that BDNF and psychedelics possess a shared mechanism for promoting neuritogenesis through an mTOR-related process of protein synthesis in synaptogenesis. It appears that this pathway is directly related to 5-HT2A receptor agonist activity of classic psychedelics, given that when ketanserin, a 5-HT2A antagonist, is administered the neuroplastic effects of DMT, LSD, and DOI are abrogated. Studies on ketamine’s effect on BDNF levels show increase of BDNF protein levels in the hippocampus but not an increase of BDNF mRNA. The increased BDNF levels were not sustained past 24 h after treatment, implying that BDNF interaction at TrK-B receptor sites prompts intracellular signaling that is responsible for the synaptic plasticity and anti-depressant effects.

One study that explored the neurogenic properties of indole-containing psychedelic compounds and ketamine in Drosophila and rat in vitro cortical neuron cell cultures used concentrations of each substance at 10 nM as the upper limit of each substance, with the exception of DMT, where 90 nM was used to simulate levels naturally occurring in rat brains. In in vivo rat models using Sprague-Dawley rats 10 mg/kg and 1 mg/kg doses of DMT were administered via injection and each dosage category produced similar responses in increasing density of dendritic spines. The results demonstrated that psychedelics can influence neuronal structure in vertebrates (Sprague-Dawley rats) and invertebrates (Drosophila) in both in vitro and in vivo models suggesting an evolutionarily conserved mechanism. In a similar manner, in human studies a single-dose of ayahuasca increased BDNF serum levels in both depressed and control subjects. Human brain imaging studies have also shown grey matter density changes as thinning in the posterior cingulate cortex (PCC) and thickening in the anterior cingulate cortex (ACC) in long-term ayahuasca users suggesting modulated neural plasticity and structural changes as a result of regular ayahuasca consumption.

BDNF is fundamental for facilitating the repair, maintenance and survival of neurons. Through its Trk-B-dependent signaling, BDNF prompts the growth of new synaptic spines, dendrites and new whole neurons. BDNF signaling is fundamental in dictating brain tissue density and the formation and conservation of neural networks. Low expression of BDNF mRNA in brain, and protein levels in serum and plasma in humans have been linked to MDD, anxiety, schizophrenia, and neurodegenerative diseases. Given the prevalence of MDD and glutamate excitotoxicity in patients with autoimmune diseases, it is possible that the BDNF-upregulating qualities of psychedelics could attenuate the inflammatory and cytotoxic pro-oxidative effects of glutamate excitotoxity in autoimmune disease, resulting in increased survivability of neurons that may be susceptible to ROS damage.

2.7. Microbial aspects of the physiological effects of psychedelics

Chronic infections with bacteria, fungi and viruses are common in those with AiDs. Studies suggest that infection by the Epstein-Barr virus and other herpes family viruses are prevalent in those with AiD. There is also evidence that Borrelia burgdorferi, the bacteria that causes Lyme disease, is also a common infectious agent in AiDs. Many patients with AiD display evidence of increased gut permeability, small intestinal bacterial overgrowth, and other signs of gut dysbiosis, which can be classified as chronic low-grade infections.

Constituents in psychedelic plants such as the ayahuasca vine (Banisteriopsis caapi) and Peyote cactus (Lophophora williamsii) display specific antimicrobial effects. Harmine and harmaline are beta-carboline alkaloids with MAOI properties in the B. caapi vine. Studies have demonstrated invitro anti-viral activity against the Herpes Virus Simplex 2 (HSV2) virus, a common infection in those with AiD, in HeLa, Vero, and Hec-1-A cells. The same alkaloids also appear to have antifungal properties which could perhaps be advantageous in an individual with a filamentous fungi overgrowth. Peyote extract has also shown broad antibacterial activity invitro, in particular against 18 different penicillin-resistant strains of Staphylococcus aureus. There may be mild antibiotic effects of some psychedelic plants which could potentially yield notable results in managing chronic or acute infections in AiD patients.

While there is evidence to suggest that psychedelic plants may have antimicrobial activity against pathogenic organisms, there has been no investigation of the effects on commensal bacteria and psychedelic compound exposure. With increasing interest in the roles of commensal gut ecology in health and disease, there has been more investigation into the psychoactive effects and complex host-organism interactions with bacterial colonies in the gastrointestinal tract. Microbiome bacterial populations appear to have play a significant role in a number of immune responses and have been found to influence the presence of self-antigens and AiD.

Gut bacteria have been discovered to possess the ability to manipulate neurotransmitter activity and are capable of producing serotonin, dopamine, acetylcholine, GABA, and more. Many of these neurotransmitters are derived from essential amino acids such as tryptophan, phenylalanine, and glutamine. Commensal gut bacteria possess highly conserved metabolic pathways for processing amino acids for a variety of uses. Many of the classic psychedelics are also biologically synthesized from tryptophan and share an analogous structure to serotonin and other endogenous tryptamine molecules. It is tempting to speculate that gut bacteria may produce these tryptamine metabolic products to use as signaling molecules meaning that bacteria are likely to not only produce these molecules, but also receive information from them as signal inputs. One study found that spore-forming endogenous bacteria species from Turicibacteraceae, Clostridiales, Lachnospiraceae and Ruminococcaceae were enriched in abundance when 5-HT was orally administered to wild type rats suggesting that the secretion of 5-HT from gut bacteria and the host intestine may prompt an advantage towards thriving colonies and promote the bacteria’s own community membership in the mammalian intestine, rather than 5-HT simply being a metabolic byproduct. It is very possible that the psychedelic serotonin analogues psilocybin, DMT, and 5-MeO-DMT may interact with bacterial receptors such as those found in Turicibacter sanguinis, a gut bacterium that expresses a neurotransmitter sodium symporter-related protein with sequence and structural homology to mammalian SERT. It is unclear if this SERT orthologue is solely responsible for the mechanism of transporting 5-HT into bacterial cells, but spore-forming gut bacteria do show the ability to transport 5-HT through some mechanism of action. If serotonergic psychedelic compounds were found to interact with these SERT-homologous bacterial proteins or other bacterial 5-HT uptake pathways in a way that alters the behavior of the bacterial colonies, this could have implications on gut microbe ecology and thus AiD disease outcome.

Whether there are direct pharmacological effects of psychedelics on bacteria is currently unknown, however the emotional and psychological benefits of psychedelics may indirectly alter microbial communities in the GI tract via induced changes in vagal nerve tone, stress response, and enteric environment. Several studies have explored the effects of chronic stress and resulting conditions like PTSD on microbiome pattern associations. The psychological and neurological benefits from a psychedelic experience may create biochemical cascades in systemic physiology, specifically within the HPA axis and the enteric nervous system, which may downstream influence the ecology of microbial populations.

As mentioned previously, LPS is a cell wall component of Gram-negative bacteria. LPS is known to induce inflammation and is used in research as an inflammatory trigger in animal models to simulate Parkinson’s, Alzheimer’s, MDD and general inflammatory conditions. In patients with increased gut-permeability, LPS fragments can leak past the intestinal epithelial barrier and cause inflammation while in circulation. Models using LPS-induced inflammation were used in some of the previously noted human invitro, and rat studies on the anti-inflammatory effects of psychedelics. This research may be particularly relevant to AiD as it suggests psychedelics may be helpful in mitigating inflammation caused by LPS due to increased intestinal permeability and chronic gut dysbiosis.

2.8. Mitigating the risks of psychedelic side-effects in therapeutic settings

Given the powerful psychoactive effects of psychedelic compounds, their use in medicine and psychiatry should not be taken lightly. Currently most psychedelic compounds are schedule I compounds meaning they have the highest categorical ranking of illegality in the Unites States with Schedule I stating that a substance has no acknowledged medical use, and is considered to have high-abuse potential.

Most psychological harms associated with the illegal use of psychedelics appear to be a result of poorly controlled environmental factors such as set and setting, as well as questionable sources of the drugs from the black market. In clinical administration of psychedelics, there is documented very low chance of psychological harm to properly screened patients provided with integrative support. One population study suggests that psychedelics do not appear to linked to long term mental health problems or suicidality. One 2010 analysis of psilocybin studies done between 1999 and 2008 looked at the experiences of 110 patients. Negative experiences were not common and seemed to be dose-dependent where higher doses of psilocybin were associated with higher rates of adverse reactions. All of the short-term adverse reactions were “successfully managed through interpersonal support” and did not require taking medications and seemed to have no lasting effects, based on follow-up interviews.

Hallucinogen Persisting Perception Disorder (HPPD) is a controversial neurocognitive condition where the patient reports persisting sensory effects after using a psychedelic substance. Also known as “flashbacks” the patient may experience brief re-occurrences/episodes of alterations in perception, mood, and/or consciousness, as previously experienced during a hallucinogenic intoxication. These symptoms can occur on a spectrum from occasional to near-constant. There is little conclusion about how common or debilitating these “flashback” experiences may be. Some studies suggest that they are less likely to occur in those who have taken a psychedelic in a clinical setting. Based on the phenomena existing in psychedelic-naïve patients and inconclusive data from studies that have examined the disorder, a causal relationship between persistent perceptual symptoms and use of psychedelics remains unproven.

Some precautions that may reduce harm to those receiving psychedelic medicine in clinical environments may include screening for a history of schizophrenia or psychosis, comfortable therapeutic set and settings that reduce the risk of panic or distress, interpersonal support provided by the clinicians, pharmaceutical-grade drugs, and low, subperceptual dosing, also known as micro-dosing.

3. Discussion and perspectives

There is vast opportunity to explore the effects of psychedelics on the immune system, in particular, autoimmunity. Systematic screening for various autoantibodies, inflammatory biomarkers, and for the expression of autoimmune-related genes in response to psychedelic treatments could provide intriguing observations and lead to more focused investigations.

The effects of psychedelic compounds on enteric bacterial behavior is of particular interest. Studies could be conducted examining the effects on bacterial growth and metabolism as a result of exposure to psychedelic compounds. In vitro and in vivo studies may hold many answers and even more questions about if and how serotonergic psychedelic compounds may be transported into bacterial cells and influence microbiome colonization and species composition. Studies could examine the effect of administration of psychedelics on enteric epithelial tissue integrity in the gut, or effects on LPS-induced cytotoxicity, etc.

Although results from preclinical studies on the immunomodulatory and anti-inflammatory effects are promising, the field of psychedelic research in biomedicine is still in its infancy. Current hypotheses regarding signaling mechanisms and systemic immune effects of psychedelics are based on a very limited amount of factual data. Only until we better understand the genetic and pathophysiological bases for AiD can there be appropriately and rigorously designed human clinical studies that consider psychedelics as potential therapeutic agents.

While there is a need for deeper investigation, there appears to be enough evidence of direct and indirect effects from psychedelic compounds that may benefit those with AiD to merit further exploration on the topic. Given the complexity of factors that contribute to AiD, a multi-facetted approach may be appropriate to address the multiple features and causes of AiD simultaneously. Unlike many current conventional treatment methods, it appears that psychedelics may potentially offer an efficacious strategy for relieving and perhaps even resolving autoimmunity by targeting psychospiritual origins, maladaptive chronic stress responses, inflammatory pathways, immune modulation and enteric microbiome populations. The evidence presented in this paper provides support to the idea that there is untapped potential of exploring the use of psychedelics within this specific disease category. Given the limitation of efficacious treatment options for AiDs, and the physiological safety of psychedelic substances, it is likely a research topic worth pursuing. There is hope that the collection of this evidence may guide or inspire others to pioneer such studies. Overall, this direction of research is virtually unexplored and underdeveloped, providing a wealth of opportunities to discover novel applications for psychedelics in the field of immunology.

 
800px-Shulgin_sasha_2011_hanna_jon.jpg



DMT: Biochemical swiss army knife in neuroinflammation and neuroprotection?*

Attila Szabo, Ede Frecska | 25 May 2022

The inflammatory theory of many neuropsychiatric illnesses has become an emerging trend in modern medicine. Various immune mechanisms – mainly via the activity of microglia – may contribute to the etiology and symptomatology of diseases, such as schizophrenia, bipolar disorder, depression, or Alzheimer's disease. Unwanted and excess inflammation is most typically the result of dysregulated innate immune responses. Recognition of self-derived damage-associated molecular patterns (DAMPs) or pathogen-associated molecular pattern molecules (PAMPs) is usually leading to the activation of tissue resident immune cells including macrophages (microglia) and dendritic cells. They act as ‘gatekeepers’ continuously monitoring the tissue microenvironment for potential ‘danger signals’ by means of their pattern recognition receptors, such as Toll-like receptors or RIG-I-like receptors.

Once a DAMP or PAMP has been recognized by a pattern recognition receptor various downstream signaling pathways are initiated, which eventually leads to the secretion of inflammatory cytokines and many other soluble factors important in the elimination of invading microbes. Pattern recognition receptors couple to nuclear factor kappaB (NF-kB), the master transcription regulator of inflammatory cytokines and chemokines. Macrophages and dendritic cells are also capable of antigen-presentation so they can initiate adaptive immune responses by priming naive T-cells. During inflammation of the central nervous system, polarization towards the T helper 1 and 17 subsets is especially important as these T cells play a major role in the development of chronic inflammation and brain tissue damage in infectious diseases and autoimmunity.

It has been known for decades that immunomodulation through serotonin/5-hydroxytryptamine receptors (5-HTRs) has the potential to regulate inflammation and prevent damage of the nervous tissue. Recently another receptor has been added to the greater picture: the orphan receptor sigma-1 (Sig-1R). 5-HTRs and Sig-1R have been shown to be expressed ubiquitously in higher vertebrate tissues and mediate various processes, including the regulation of cognition and behavior, body temperature, as well as immune functions. Both 5-HTRs and the Sig-1R use G protein-coupled (GPCR) pathways thereby modulating a plethora of cellular functions, such as cytokine/neurotransmitter release, proliferation, differentiation, and apoptosis.

The molecular chaperone Sig-1R is located at the endoplasmic reticulum-mitochondrion interface and has an important role in the fine-tuning of cellular metabolism and energetics under stressful conditions. At the MAM, Sig-1Rs are involved in the regulation and mobilization of calcium from endoplasmic reticulum stores. Neuroprotection by Sig-1R activation can be attained by preventing elevations of intracellular calcium-mediated cell death signaling. Based on its central localization and function, pivotal physiological activities of the Sig-1R have been described such as indispensable role in neuronal differentiation, neuronal signaling, cellular survival in hypoxia, resistance against oxidative stress, and mitigating unfolded protein response.

Tryptaminergic trace amines (e.g. DMT) as well as neurosteroids are endogenous ligands of the Sig-1R. Tryptamines are naturally occurring monoamine alkaloids sharing a common biochemical – tryptamine – backbone. DMT was shown to be endogenously present in the human brain and in other tissues of the body, however the exact physiological role of this tryptamine has not been identified yet. It has been shown that, besides its affinity for the Sig-1R, DMT also acts as an agonist at numerous serotonin receptors, such as 5-HT1A, 5-HT2A, and 5-HT2C. This wide-spectrum agonist activity may allow DMT to modulate several physiological processes and regulate inflammation through the Sig-1R and 5-HTRs.

Indeed, DMT has been found to modulate immune responses through the Sig-1R under various conditions. These include the suppression of inflammation by blocking inflammatory cytokine and chemokine release of dendritic cells, as well as inhibiting the activation of Th1 and Th17 subsets. The biochemical background of this extensive ability lies in the possible cross-talk of the GPCR-coupled downstream signaling of 5-HTRs/Sig-1R and other inflammatory pathways in immune cells, as well as the fine-tuning of cytokine feedback loops in peripheral tissues. Thus, in neuroinflammation, two major scenarios are possible:

i) The modulation of cytokine production by brain resident microglia that implies a negative feedback regulation of inflammation via the induction of the release of anti-inflammatory IL-10 and TGFB occurring subsequent of both 5-HTR and Sig-1R activation;

ii) The direct/indirect control of NF-kB signaling and possibly other pathways involved in inflammation through intracellular kinases, adaptor proteins, etc. This way, the activation of 5-HTRs and Sig-1R may also interfere with the chemokine, inflammatory cytokine signaling of immune cells through intracellular mechanisms.

Most of the receptors that are involved in psychedelic effects belong to the GPCR family or interact with GPCRs. The role of 5-HTR/Sig-1R GPCR-coupled signals in the intracellular regulation and orchestration of NF-kB and MAPK pathways may be of particular importance regarding the complex neuroimmunological effects of DMT.

The above outlined picture suggests a direct control of NF-kB transcriptional regulation of chemokines, pro-inflammatory and anti-inflammatory cytokines, which may render DMT as a potentially useful therapeutic tool in a broad range of chronic inflammatory and autoimmune diseases, and pathological conditions connected to increased unfolded protein responseincluding but not restricted torheumatoid arthritis, multiple sclerosis, amyotrophic lateral sclerosis (ALS), Alzheimer's and Parkinson's disease, etc. However, the powerful sychedelic property of DMT poses an important problem that must be addressed in future drug design.

Protective and neuroregenerative effects of Sig-1R agonists have been reported in several in vitro and in vivo studies. The selective Sig-1R agonists 2-1 phenylcyclohexanecarboxylate and cutamesine have been shown to strongly promote neuroprotective mechanisms and significantly increase neuronal cell survival and regeneration under various conditions, such as traumas, autoimmunity, and neurodegenerative disorders. Specific Sig-1R stimulation has also been found to greatly increase the levels of the glial cell-derived neurotrophic factor GDNF that promotes neuronal cell survival and differentiation.

The neuroregenerative potential of DMT through the Sig-1R has been suggested earlier as multiple biochemical and physiological mechanisms exist, which facilitate the transportation and binding of DMT to the Sig-1R in the mammalian brain. Thus DMT, as a natural, endogenous agonist at both the Sig-1R and 5-HTRs, is hypothesized to be an unique, many-faced pharmacological entity, which has many important roles in the immunoregulatory processes of peripheral and brain tissues, as well as involved in the promotion and induction of neuroregeneration in the mammalian nervous system.

*From the article here :
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4828992/
 
Last edited:
95207.jpg



Chronic inflammation is dangerous. You may not even know you have it.

by Marlene Cimons | Washington Post | 9 Jun 2022

People often learn they have it when by developing an autoimmune disease. But the ailment might also play a role with heart disease, cancer, other disorders.

Most of us think of inflammation as the redness and swelling that follow a wound, infection or injury, such as an ankle sprain, or from overdoing a sport, “tennis elbow,” for example. This is “acute” inflammation, a beneficial immune system response that encourages healing, and usually disappears once the injury improves.

But chronic inflammation is less obvious and often more insidious.

Chronic inflammation begins without an apparent cause — and doesn’t stop. The immune system becomes activated, but the inflammatory response isn’t intermittent, as it is during an acute injury or infection. Rather, it stays on all the time at a low level.

Experts think this may be the result of an infection that doesn’t resolve, an abnormal immune reaction or such lifestyle factors as obesity, poor sleep or exposure to environmental toxins. Over time, the condition can, among other things, damage DNA and lead to heart disease, cancer and other serious disorders.

“Unlike acute inflammation, which benefits health by promoting healing and recovery, chronic inflammation is characterized by persistent increases in inflammatory proteins all throughout the body and can damage health and promote several major diseases,” says George Slavich, associate professor of psychiatry and biobehavioral sciences at UCLA, referring to small proteins called cytokines that the immune system releases at the site of an injury to promote recovery.

“People typically don’t know that they have chronic inflammation until it’s too late,” he says.

Individuals often learn they have chronic inflammation when they develop an autoimmune disease, such as Crohn’s disease, lupus, or Type 1 diabetes, since inflammation is a hallmark of autoimmune disorders. But experts believe chronic inflammation also plays a role in developing heart disease, cancer, kidney disease, nonalcoholic fatty liver disease, neurodegenerative disorders, cognitive decline and mental health illnesses, such as depression, post-traumatic stress disorder and schizophrenia.

Scientists are still learning about why chronic inflammation is so dangerous and how it contributes to disease. Meanwhile, they suggest actions people can take to reduce their risk, specifically by changing certain behaviors.

Numerous factors appear to raise the risk of chronic inflammation, among them social isolation, psychological stress, disturbed sleep, chronic infections, physical inactivity, poor diet, obesity and exposure to air pollutants, hazardous waste products, industrial chemicals and tobacco smoke.

Experts believe individuals can reduce their risk by adopting lifestyle changes, including eating a healthy diet, improving sleep, exercising regularly, quitting smoking and finding ways to decrease stress and exposure to environmental pollutants.

“Diet is one of the key factors that influences inflammation in the body,” Slavich says. “Whereas fried foods, red meat, sodas, and white bread and pastries that have refined carbohydrates tend to increase inflammation, fruits, nuts, green leafy vegetables, tomatoes and olive oil tend to reduce inflammation. Therefore, while diet is not the only factor that can be targeted to improve immune health, it is an important one.”

Scientists think chronic inflammation causes oxidative stress in the body, which is an imbalance between the production of dangerous free radicals, molecules that harm healthy tissue in the body, and antioxidants, substances that clean up waste products and neutralize them. This can damage DNA as well as proteins and fatty tissue, which in turn accelerates biological aging.

“Chronic inflammation is involved in not just a few select disorders but a wide variety of very serious physical and mental health conditions,” says Slavich, senior author of a paper signed by scientists from 22 institutions urging greater prevention, early diagnosis and treatment of severe chronic inflammation. “Indeed, chronic inflammatory diseases are the most significant cause of death in the world today, with more than 50 percent of all deaths being attributable to inflammation-related diseases.”

Researchers still don’t understand the exact mechanisms of how certain behaviors influence chronic inflammation, although a few examples are clear. In heart disease, for example, cigarette smoking and air pollution irritate the arteries, which stimulates inflammation.

“The ‘damage accumulation’ theory is a possibility, but the reality is that we do not know whether inflammation is causing these health and functional problems, or whether it’s an indication that some other process is evolving that undermines health,” says Luigi Ferrucci, scientific director of the National Institute on Aging. “The evidence is clearer for cardiovascular disease, since it has been demonstrated that blocking inflammation with specific drugs prevents cardiovascular events. For the other outcomes, it’s still uncertain.”

Chronic inflammation can contribute to cognitive decline and mental health disorders by boosting age-related immune system deterioration, known as immunosenescence, and by promoting vascular and brain aging, which, in combination, degrade neural and cognitive function, experts say.

“Chronic inflammation can also cause threat sensitivity and hypervigilance, which gives rise to anxiety disorders and PTSD, as well as fatigue and social-behavioral withdrawal, which are key symptoms of depression,” Slavich says.

Scientists say more research is needed to identify biomarkers or other substances that suggest the presence of chronic inflammation.

There are probably hundreds of these potential diagnostic tools produced by the immune system, but they remain unidentified, Slavich says.

The most widely used test measures levels of C-reactive protein (CRP) in the blood. CRP, a substance produced by the liver, rises when chronic inflammation is present, although the standard CRP test is nonspecific — that is, it indicates inflammation, but cannot pinpoint exactly where it is. A second, more sensitive test (hs-CRP) suggests a higher risk of heart attack, although it too can be imprecise.

Some doctors screen for CRP as part of routine physical exams and also among people at risk for heart disease and autoimmune conditions. Experts think wider screening could identify more patients. “This isn’t a bad idea,” Ferrucci says.

Another test — this one more specific to heart disease — screens for myeloperoxidase, or MPO, an enzyme released by white blood cells that kills harmful bacteria in inflamed blood vessels. Increases in MPO can be dangerous, causing further damage to arterial walls, which encourages the formation of clots. These, in turn, can block blood flow, leading to heart attack and stroke. MPO also reduces the effectiveness of HDL, the “good” cholesterol, and removes nitric oxide, which is important for the regulation of healthy blood flow.

The good news, however, is that people worried about developing chronic inflammation can take affirmative steps to prevent it.

“If we make people aware of these risk factors, our hope is that individuals will reduce the factors that apply to them,” Slavich says.

 
Top