• Psychedelic Medicine

NEUROSCIENCE | +80 articles

sleep-apnea-reduced-neurosceinces-public-1155x770.jpg


Ketamine normalizes hyperactivity in a key brain region of depressed patients

by Christian Rigg | PsyPost | 4 Apr 2021

There is no shortage of psychological and pharmacological therapies to combat the world’s most widespread mental health issue, major depressive disorder (MDD). However, a significant portion of the affected population fail to respond to many of these traditional therapies. For this reason, new drugs must be tested and validated. One promising candidate is ketamine –famously but somewhat improperly known as a horse tranquilizer.

However, the manner by which ketamine acts is not well known, meaning that clinicians are still circumspect regarding its use in treating MDD. Recently, researchers in New York look at how ketamine affects the subgenual anterior cingulate cortex (sgACC), a region of the brain whose hyperactivity has proven ties to MDD. The recent study, which appeared in Neuropsychopharmacology, helps bridge this gap in the literature.

In the study, 28 patients with MDD and 20 healthy controls underwent function MRI (fMRI) scans both at rest and while completing a monetary incentive-based task. The goal of the incentive task was to activate the sgACC, known to be implicated in reward anticipation.

The results of the study demonstrate a more complex relation between the sgACC and MDD than has been previously suspected. The authors describe a “double dissociation, whereby sgACC hyper-activation to positive feedback is associated with anhedonia [inability to feel happiness], whereas hyper-activation to negative feedback is associated with anxiety.”

This also enabled them to uncover what may be an important physiological distinction in the region, where the posterior region was more closely related to symptoms of anhedonia and the anterior region to anxiety.

In terms of a pharmacological treatment, ketamine was shown to operate by reducing sgACC hyperactivation to positive feedback. If this seems counterintuitive, it is important to remember that many brain centers are inhibitory by nature, meaning that the more active they are, the more strongly they inhibit other areas—thus producing, for example, a reduced response to positive feedback. Interestingly, the ketamine treatment blunted sgACC hyperactivation in response to positive feedback, but not negative feedback.

The neurological underpinnings of MDD are still not well understood. The same can be said for many of the drugs used in treating it. Rigorous clinical testing and exploratory studies like the present are thus essential in improving our understanding of both this disease and treatment options.

The study, “Ketamine normalizes subgenual cingulate cortex hyper-activity in depression“, was authored by Laurel S. Morris, Sara Costi, Aaron Tan, Emily R. Stern, Dennis S. Charney, and James W. Murrough.

 
Last edited:
DkUt3r3CSA6BCKsjwzFL_mdmaneurotoxicitycover.jpg



Is MDMA Neurotoxic?

by Dr. Ben Malcolm, Certified Psychiatric Pharmacist

Neurotoxicity resulting in low mood, anxiety, insomnia and problems with cognition is well documented in literature relating to use of ‘Ecstasy’, but what about pure MDMA?

MDMA-induced neurotoxicity use has been the subject of intense controversy and even scandal within the medical community over decades. As MDMA assisted psychotherapy (MDMA-AP) progresses toward approval as a legal therapeutic entity, the questions regarding neurotoxicity of MDMA will be clinically relevant as patients and providers consider risks and benefits of use. Moreover, clinical trials are shedding some light on thresholds for neurotoxic effects and providing high quality data to help guide safe use.

In this post we’ll give some background information on what neurotoxicity is, how MDMA may act as a neurotoxin, summarize research findings on MDMA-induced neurotoxicity, and explain use parameters that are likely to prevent development of significant neurotoxic effects.

First of all, what is neurotoxicity?

Neurotoxicity involves damage to neurons of the central or peripheral nervous system. It can be caused by several things, including drugs. One common misconception is that neurotoxicity exists as an all or nothing event - that is your neurons are either healthy or destroyed, although it is best understood as a symptom spectrum that can be reversible in some cases and irreversible in others. It can begin with small functional deficits, which gradually progress to a larger-scale functional decline or symptoms.

In the case of neurotoxicity as it relates to MDMA, this progression may present itself in an insidious manner, whether it be gradual loss of memory, behavioral and mood issues, or decreased cognitive function over repeated exposures. Of course, large overdoses can produce severe neurotoxicity rapidly. Drugs can produce neurotoxicity through many different types of mechanisms, too many to recount here, although it can be noted that a drug may have a dosing window in which no neurotoxicity is observed due to the dose being modest enough to not push a biological system to such extreme points that toxic responses occur. Even substances we consider completely benign and healthful, such as water, can become neurotoxic under particular circumstances (e.g. drinking 2 gallons in a single hour).

Therefore, our question of MDMA-induced neurotoxicity needs more nuance than ‘is it neurotoxic or not?’ The answer is “yes it is”, but it’s misleading. This is because it inevitably depends on the circumstances of administration. Instead, the better question to ask is ‘under what conditions does MDMA cause neurotoxicity?’

How does MDMA lead to neurotoxicity?

Before discussing how MDMA leads to neurotoxicity, it may be helpful to review how MDMA works. MDMA acts on several neurotransmitter systems, although primarily releases serotonin from presynaptic nerve terminals, resulting in acute depletion of serotonin stores and synaptic flooding of serotonin. This has led researchers to believe neurotoxicity is a consequence of damage to serotonergic neurocircuitry. Subsequently, evidence of neurotoxicity for MDMA is mainly quantified through 5-HT (serotonin) concentrations, activity or presence of enzymes involved with serotonin synthesis or transport (TH and SERT), and visualization of axons immunoreactive for 5-HT or SERT through imaging.

bCtUSiNgRrquyIPJtdQ5_5HT_Synapse_MDMA_MOA.png


Mechanisms of MDMA-induced neurotoxicity

Sometimes it may be as simple as the dose determining if poisonous or not. With higher doses there is a greater chance of profound serotonin depletion. Frequency of use is also probably consequential as dosing on a weekly basis (as some may be if frequenting raves often) is not enough time to allow for recovery from prior use, even in some taking doses in clinical environments. This is rather simple in that your serotonin neuron simply has a capacity for how high the concentrations can be without damage and how fast it can return to balance after use. The two may go hand in hand as frequent use can produce tolerance and result in increased doses when used.

Other explanations are more complicated. For example, MDMA is metabolized to reactive metabolites such as HHMA or HHA, which can cause cellular damage. The serotonin excess may also lead to lasting reduction in gene expression resulting in lowered expressed of 5HT or SERT. The latter hypothesis has challenges the ‘neurodegenerative’ theory of MDMA induce neurotoxicity which argues for direct damage to serotonin neurocircuits and argues instead for a genetic reason for changes in serotonin function.

It is crucial to note that MDMA’s neurotoxic potential is increased by other substances such as alcohol or amphetamine, which are commonly taken in conjunction with MDMA at nightlife events. Concurrent substances can be ingested either advertently or inadvertently as many Ecstasy tablets contain adulterants or misrepresented substances. For example, one analysis found almost half of ecstasy tablets continued < 67% MDMA and that caffeine and amphetamine were common adulterants. Other common adulterants include novel psychoactive substances such as other phenethylamine (amphetamine) based drugs. Many events sell alcohol or may limit water availability. In combination, both alcohol and MDMA exert neurotoxic effects by impairing the survival of neuronal precursors in the hippocampal dentate gyrus, an area of the brain important for neuronal generation, learning, and memory. Beyond alcohol or amphetamine(s), rave scenes can expose users to myriad other substances, such as GHB (gamma-hydroxybutyrate) or ketamine, which may also increase risk. Recently and tragically, other adulterants from diverse drug classes have began to be found on ecstasy tablets such as fentanyl. Ecstasy is recommended to be tested for presence of desired agent (MDMA) and absence of deadly adulterants (fentanyl) by harm reduction organizations. Further information and testing kits for purchase can be found at dancesafe.org

Other aspects of MDMA use that may contribute to neurotoxicity is the recreational environment and timing of administration. Users may stay up all night on MDMA and related stimulants, which could trigger a sleep deprived state. Sleep deprivation is well known to impair cognition and lower mood, thus plausibly leads to some of the observed week after effects. This issue may be minimized by timing MDMA administration such that the user can fall asleep at a regular time or with minimal disruption to habitual circadian patterns. Another factor that may play a role in toxicity is the bioenergetic stress (namely thermal stress) secondary to MDMA being used in hot environments, raising core body temperatures, and being associated with high output physical activity.

Though the jury is still out on exactly how MDMA exerts its neurotoxic effects, the bottom line is that MDMA has several underlying explanations of why it could be neurotoxic as well as evidence from recreational use environments that it can be neurotoxic under certain circumstances. There circumstances tend to work in conjunction within subcultures that use Ecstasy heavily.

iZO0azCnRMqVaSHgDA7a_MDMA_Neurotoxicity_Harm_Reduction_Graphic_.png


At what point does MDMA become neurotoxic?

The potential for MDMA to act as a neurotoxin is determined by the dose, frequency of exposure, timeline of exposure, concurrent substance use, and overall setting or context in which it is administered.

Doses of 1.7mg/kg (~125mg in a ~160lb adult) have been evaluated in clinical laboratory conditions without evidence of damage to serotonin neurocircuits or functional deficits. In the six phase II trials of MDMA-AP for PTSD conducted thus far, doses have ranged between 75-187.5mg and participants have undergone 2-3 experimental sessions spaced 3-5 weeks apart. In phase III trials, the maximum dose in any session will be 120mg as an initial dose and 60mg as a booster dose (180mg total) and all participants will undergo 3 sessions. In phase II, there were no detected changes in neurocognitive function over a 2-month period following the second and third experimental sessions. During MDMA-AP sessions participants reported a range of short-lived side effects characteristic of amphetamine use and mild serotonin excess. In the week after use fatigue peaked on day 1 and decreased towards day 6, the need for more sleep peaked on day 2 and decreased towards day 6, and low mood peaked between days 2-4 and decreased towards day 6. Notably, in 4 of the 6 phase II studies, depression scores were measured and showed greater improvements than placebo over the course of the trial. In summary, it’s clear that a course of 2-3 MDMA sessions spaced at least a month apart utilizing doses of 75-125mg followed by a booster dose of 37.5mg-75mg 2-3 hours later does not produce persistent neurotoxic effects.

In the previously mentioned study of MDMA samples, the ones that contained >67% MDMA averaged 205mg per dose, higher than the initial and booster doses in MDMA-AP combined (however weights include fillers thus is likely an overestimate of MDMA per tablet). Consumption of multiple tablets (stacking) can bring doses up to 0.5g or more throughout an evening for some users; similar to amounts consumed in all 3 MDMA-AP sessions combined. Stacking was investigated in laboratory conditions by giving 100mg of MDMA followed by another 100mg of MDMA 4 hours later. Results demonstrated that blood concentrations, cardiovascular stress, and temperature were all increased, however subjective intensity of effect was similar due to development of rapid tolerance to pleasurable effects. This may cause users to underestimate the physical stress they are placing on their body with repeated dosing.

So, what does the research tell us so far?

There are two converging streams of evidence here. One from recreation literature and Ecstasy use suggesting potential for neurotoxicity. The other from clinical environments in which MDMA is predominantly therapeutic, has a promising safety profile, and lacks evidence of clinically significant neurotoxicity despite using measures that could reasonably detect such problems. Therefore, models of dosing from clinical trials could be extrapolated and generalized into a harm reduction framework that could be applied broadly to persons who use MDMA.

While phase II trials did detect several cases of persons experiencing side effects of MDMA in the week after their session, the changes were transient and resolved after a week in almost all cases. This suggests that transient changes to mood, sleep, or cognition in the week after use is a side effect that persons should be educated about, while neurotoxicity is an adverse drug reaction associated with ‘incorrect’ or harmful administration patterns.

How can MDMA-induced neurotoxicity be avoided?

The good news is that avoiding or severely limiting the potential for significant neurotoxicity occurring with MDMA is straight-forward, not difficult, and preserves the ability for profound benefits.​
  • Limit MDMA doses to the moderate range 75-125mg initially​
  • Limit dosing in a single session to a single booster of 50% the original dose​
  • Space MDMA sessions at least one month apart​
  • Limit dosing to 3-4 times per calendar year​
  • Avoid using other drugs and alcohol with MDMA​
  • Test ‘ecstasy’ tablets for presence of MDMA prior to use (as well as absence of other drugs such as fentanyl)​
  • Limit sleep disruption due to MDMA use​
  • Plan regular breaks and sip water if exposed to hot environments (do not drink excessive amounts of water)​
  • If you notice a pattern of low mood or cognitive problems in the days after MDMA use that seems to worsen upon repeated administration or is persistent beyond a few days post-use then take an extended break from using MDMA or similar substances​
Do antidepressants or 5-HTP prevent MDMA-induced neurotoxicity?

Serotonin blocking antidepressants such as Selective Serotonin Reuptake Inhibitors (SSRIs e.g. Prozac or fluoxetine) can diminish neurotoxic potential of MDMA. However, this is a poor strategy overall because serotonin blocking antidepressants also greatly diminish the subjective experience of MDMA.

Supplements such as L-tryptophan or 5-HTP are able to boost serotonin synthesis. It may not be wise to take large doses prior to MDMA use as it could increase the risk of physical side effects of serotonin excess such as nausea, vomiting, or diarrhea. Conversely, it may be reasonable to use L-tryptophan or 5-HTP for a few days after MDMA use to aid in restoring serotonin levels.

Neither antidepressants nor serotonin precursor supplementation has adequate data suggesting benefits to recommend use. While further research and testing of interventions to reduce either week after side effects or prevent neurotoxic effects is encouraged, current clinical trial data do not support such measures as necessary.

Summary & conclusions

Neurotoxicity can manifest in many forms and severity levels and MDMA appears to be neurotoxic under certain circumstances. High doses or several doses throughout the period of ingestion, frequent use (e.g. weekly use at nightlife events), aduleration, and substance mixing all are implicated in the adverse event of MDMA neurotoxicity. Data from phase II trials of MDMA-AP do not support risks of clinically significant neurotoxicity from MDMA use as side effects were observed to be mild and limited to the week after use, while participants also saw profound improvements in clinical symptoms for PTSD. In summary, MDMA induced neurotoxicity is easy to avoid with moderate doses, adequate time between use, and pure substances without combination.

 
Last edited:
_124122299_gettyimages-460689429.jpg



Psilocybin frees up depressed brain, study*

by Philippa Roxby | BBC | 11 Apr 2022

Psilocybin, a drug found in magic mushrooms, appears to free up the brains of people with severe depression in a way that other antidepressants do not, a study has found.

The results, based on brain scans of 60 people, mean the drug could treat depression in a unique way, the researchers say.

Psychedelics are being studied to treat a range of mental health disorders.

Patients with depression are warned not to take psilocybin on their own.

A synthetic form of the drug is tested on people in trials under strict medical conditions, with psychological support from experts provided before, during and after it is taken.

Prof David Nutt, study author and head of the Imperial College London's Centre for Psychedelic Research, said the latest findings on psilocybin were "exciting" and "important".

With depression, the brain can get stuck in a rut and locked into a particular negative way of thinking, he said.

But when given psilocybin, people's brains opened up and became "more flexible and fluid" up to three weeks later.

This could be seen in increased connections between regions of the brain when patients were scanned. These patients were more likely to experience an improvement in mood months later.

Similar changes were not seen in the brains of people treated with a standard antidepressant.

"This supports our initial predictions, and confirms psilocybin could be a real alternative approach to depression treatments," Prof Nutt said.

Brain activity

Psychedelics are a type of hallucinogenic substance which affects all the senses, altering a person's thinking, sense of time and emotions.

While regular antidepressants are taken every day, psilocybin may only need to be taken once or twice to produce the same effect - but further research on more patients for longer is needed to confirm that.

The results, published in Nature Medicine, are taken from two studies. In the first, everyone received psilocybin; and in the second - a randomised controlled trial - some were given the drug while others were given a different antidepressant.

All participants also received talking therapies with registered mental health professionals. Brain scans were taken before, and then one day or three weeks after taking the therapy.

Prof Robin Carhart-Harris, senior study author, said: "We don't yet know how long the changes in brain activity seen with psilocybin therapy last, and we need to do more research to understand this."

"We do know that some people relapse, and it may be that after a while their brains revert to the rigid patterns of activity we see in depression."


Earlier findings from the studies showed a reduction in symptoms of depression with the psilocybin therapy - but the researchers were not sure how and why it worked.

Now they want to test their theory of changes in brain connectivity on other mental health illnesses, such as anorexia.

*From the article here :
 
Last edited:
neurogenesis-dmt-neurosncience.jpg



Ayahuasca stimulates the formation of new neurons

Complutense University of Madrid | Neuroscience News | 6 Nov 2020

DMT, a natural component of ayahuasca tea, promotes neurogenesis, a new study reports. Researchers found DMT was capable of activating neural stem cells and promoted the formation of new neurons.

One of the main natural components of ayahuasca tea, dimethyltryptamine (DMT), promotes neurogenesis (the formation of new neurons) according to research led by the Complutense University of Madrid (UCM).

In addition to neurons, the infusion used for shamanic purposes also induces the formation of other neural cells such as astrocytes and oligodendrocytes.

“This capacity to modulate brain plasticity suggests that it has great therapeutic potential for a wide range of psychiatric and neurological disorders, including neurodegenerative diseases,” explained José Ángel Morales, a researcher in the UCM and CIBERNED Department of Cellular Biology.

The study, published in Translational Psychiatry, reports "the results of four years of in vitro and in vivo experimentation on mice, demonstrating they exhibit a greater cognitive capacity when treated with this substance,” according to José Antonio López, a researcher in the Faculty of Psychology at the UCM and co-author of the study.

Changing the receptor eliminates the psychedelic effect

Ayahuasca is produced by mixing two plants from the Amazon: the ayahuasca vine (Banisteriopsis caapi) and the chacruna shrub (Psychotria viridis).

The DMT in ayahuasca tea binds to a type-2A serotonergic brain receptor, which enhances its psychedelic effect. In this study, "the receptor was changed to a sigma type receptor that does not have this effect, thus greatly facilitating its future administration to patients.”

In neurodegenerative diseases, it is the death of certain types of neuron that causes the symptoms of pathologies such as Alzheimer’s and Parkinson’s. Although humans have the capacity to generate new neuronal cells, this depends on several factors and is not always possible.

“The challenge is to activate our dormant capacity to form neurons and thus replace the neurons that die as a result of the disease. This study shows that DMT is capable of activating neural stem cells and forming new neurons,” concluded Morales.

 
Last edited:
LSD-2D-skeletal-formula-and-3D-models.png

How LSD affects the brain

by Joshua Krisch | The Scientist

The psychedelic drug lysergic acid diethylamide (LSD) is known for its euphoric effects, and for inducing long “trips.” Now, in two studies published last week (one in Current Biology, the other in Cell), scientists have examined how LSD produces such diverse effects and why the drug takes so long to wear off. The results of both studies suggest that LSD’s effects are mediated by the serotonin 2A receptor, and that the drug is just the right shape to bind this receptor for extended periods of time.

For the Current Biology study, 21 volunteers were given a placebo, a small dose of LSD alone, or the same dose of LSD but with kentaserin, a serotonin 2A antagonist. Study participants who took the kentaserin reported virtually the same experiences as those who took the placebo, and fMRI brain scans confirmed similar brain activities across participants in both groups. "The serotonin 2A antagonist blocked all the effects of LSD, so it was like if people didn’t take any drugs,” coauthor Katrin Preller, neuroscientist at the Zurich University Hospital in Switzerland told The Verge. “All the typical symptoms—hallucinations, everything—were gone.”

For the Cell study, researchers imaged LSD binding various serotonin receptors, observing that the drug clings to these structures in a unique way. This could explain why the effects of LSD take some time to wear off. “What we saw is that the receptor is shaped a little bit like a vase, and it has a space in the middle where the LSD binds and there’s a lid above it,” coauthor Daniel Wacker of the University of North Carolina, Chapel Hill, told The Verge. “LSD has this unique property that it actually holds onto the lid. For many other compounds like serotonin, the lid remains rather flexible. Because LSD holds onto it, it really stays in there.”

pcblsd_withskull.jpg


Psychedelic neuroimaging

Two papers—one published in PNAS on Monday (April 11) and another in Current Biology today (April 13)—report how brain activity is altered in people given doses of lysergic acid diethylamide (LSD). Using two different brain imaging approaches, the authors of both studies detailed the particular changes in activity that associate with arguably the best-known effects of LSD: visual hallucinations and an increased sense of unity with one’s surroundings—referred to in the Current Biology paper as “ego dissolution.”

“This work is very important with regards to the insights it gives us on how hallucinogens, specifically LSD, affect the brain,” said psychiatrist and behavioral scientist Charles Grob of the University of California, Los Angeles, who was not involved in either study. “Now we have a better understanding of the neurobiological substrate for the psychedelic experience.”

Research into psychedelic drugs, such as LSD and psilocybin—the hallucinogenic component in magic mushrooms—took a nosedive in the 1960s and 70s, following the banning of these substances. “Research with humans was not occurring in the United States and Europe from then on,” said Grob. By the 90s and 2000s, research on these drugs started to slowly pick up, Grob said, reflecting a “greater receptivity on the part of the regulatory agencies to allow this sort of research.”

Previous studies have suggested that LSD, in combination with psychotherapy, could be effective treatments for alcoholism, anxiety, and tobacco addiction, among other things.

Aside from their potential clinical use, the drugs also offer insight into the nature of consciousness itself, said cognitive and computational neuroscientist Anil Seth of the University of Sussex, U.K., who also did not participate in the work. “One of the main ways to try to understand the neural basis of consciousness is to try to induce different kinds of experience and see what happens in the brain,” he said, and “LSD is a very potent way of manipulating somebody’s experience of the world and of themselves.”

To examine LSD’s effects, Imperial College London’s David Nutt and Robin Carhart-Harris together with their colleagues recruited 20 healthy volunteers, all of whom had reported taking a psychedelic drug at least once before. The subjects were given injections of 75 micrograms of LSD—“equivalent to about one blotter of LSD taken orally and recreationally,” said Carhart-Harris—and then their brains were analyzed by functional magnetic resonance imaging (fMRI) and magnetoencephalographies (MEG). In addition, the volunteers were asked to rate various aspects of their experiences and to answer an Altered States of Consciousness questionnaire. As a control, the subjects’ brains were analyzed a couple of weeks before or after the LSD analyses, at which point they were given a saline placebo injection.

Connecting the participants’ ratings and questionnaire answers to the imaging data, the team found that LSD-induced visual hallucinations correlated with an increase in cerebral blood flow to the visual cortex, a pronounced increase in connectivity between the visual cortex and other brain regions, and a decrease in the power of visual cortex alpha waves, which are thought to generally inhibit neural activity. The authors suggested that this reduced alpha power may allow uninhibited visual cortex activity, which—together with the increased connectivity between normally unrelated brain regions—could help explain the subjects’ ability to visualize unusual images even with their eyes closed.

Aside from the increased connections with the visual cortex, LSD induced a global increase in connectivity across the brain, which was correlated with the participants’ sense of ego dissolution. In contrast, connections within specific systems, such as the default mode network, appeared to reduce.

“If you look at connectivity within [known] systems, what you see with both psilocybin and LSD is a weakening of connections,” said Carhart-Harris. “But when you look at the relationships between the different systems . . . they relate to each other much more.” Overall, the result appears to be a “unified, more integrated brain,” he said.

"Understanding the neurobiological basis of ego dissolution is particularly interesting, because this break down of self-referential processing may be the driver of clinical change,” said cognitive neuroscientist Fred Barrett of the Johns Hopkins School of Medicine in Baltimore who was not involved in the work. Put another way, it is this ego-dissolving, mind-expanding aspect of the psychedelic experience that seems to be associated with the drugs’ treatment successes.

"It’s possible that psychedelic drugs offer a re-equilibration of connectivity,” said Grob. “Patterns of a [psychiatric] disorder are often entrenched,” he added, “and there maybe some value in loosening things up.”

*From the articles here :
 
banner-itin-CA-SF-to-LAX-via-the-coast-Big_Sur_Coast_California.jpg



Unifying Theories of Psychedelic Drug Effects


by Link Swanson

How do psychedelic drugs produce their characteristic range of acute effects in perception, emotion, cognition, and sense of self? How do these effects relate to the clinical efficacy of psychedelic-assisted therapies? Efforts to understand psychedelic phenomena date back more than a century in Western science. In this article I review theories of psychedelic drug effects and highlight key concepts which have endured over the last 125 years of psychedelic science. First, I describe the subjective phenomenology of acute psychedelic effects using the best available data. Next, I review late 19th-century and early 20th-century theories, model psychoses theory, filtration theory, and psychoanalytic theory, and highlight their shared features. I then briefly review recent findings on the neuropharmacology and neurophysiology of psychedelic drugs in humans. Finally, I describe recent theories of psychedelic drug effects which leverage 21st-century cognitive neuroscience frameworks, entropic brain theory, integrated information theory, and predictive processing, and point out key shared features that link back to earlier theories. I identify an abstract principle which cuts across many theories past and present: psychedelic drugs perturb universal brain processes that normally serve to constrain neural systems central to perception, emotion, cognition, and sense of self. I conclude that making an explicit effort to investigate the principles and mechanisms of psychedelic drug effects is a uniquely powerful way to iteratively develop and test unifying theories of brain function.

Introduction

Lysergic acid diethylamide (LSD), N,N-dimethyltryptamine (DMT), psilocybin, and mescaline, the classic psychedelic drugs, can produce a broad range of effects in perception, emotion, cognition, and sense of self. How do they do this? Western science began its first wave of systematic investigations into the unique effects of mescaline 125 years ago. By the 1950s, rising interest in mescaline research was expanded to include drugs like DMT, LSD, and psilocybin in a second wave of psychedelic science. Because of their dramatic effect on the character and contents of subjective awareness, psychedelic drugs magnified the gaps in our scientific understanding of how brain chemistry relates to subjective experience. Huxley commented that our understanding circa 1954 was absurdly inadequate and amounted to a mere clue that he hoped would soon develop into a more robust understanding. Meanwhile the clue is being systematically followed; the sleuths, biochemists, psychiatrists, psychologists, are on the trail. A third wave of psychedelic science has recently emerged with its own set of sleuths on the trail, sleuths who now wield an arsenal of 21st-century scientific methodologies and are uncovering new sets of clues.

Existing theoretical hurdles span five major gaps in understanding. The first gap is that we do not have an account of how psychedelic drugs can produce such a broad diversity of subjective effects. LSD, for example, can produce subtle intensifications in perception, or it can completely dissolve all sense of space, time, and self. What accounts for this atypical diversity?

The second gap is that we do not understand how pharmacological interactions at neuronal receptors and resulting physiological changes in the neuron lead to large-scale changes in the activity of neural populations, or changes in brain network connectivity, or at the systems-level of global brain dynamics. What are the causal links in the multi-level pharmaco-neurophysiological chain?

The third gap is that we do not know how psychedelic drug-induced changes in brain activity, at any level of description. map onto the acute subjective phenomenological changes in perception, emotion, cognition, and sense of self. This kind of question is not unique to psychedelic drugs but our current understanding of psychedelic drug effects clearly magnifies the disconnect between brain science and subjective experience.

Fourth, there is a gap in our understanding of the relationships between psychedelic effects and symptoms of psychoses, such as perceptual distortion, hallucination, or altered self-reference. What is the relationship between psychedelic effects and symptoms of chronic psychotic disorders?

Fifth and finally, there is a gap in our clinical understanding of the process by which psychedelic-assisted therapies improve mental health. Which psychedelic drug effects (in the brain or in subjective experience) enable clinical improvement? How?

Scientific efforts to understand diverse natural phenomena aim to produce a single theory that can account for many phenomena using a minimal set of principles. Such theories are sometimes called unifying theories. Not everyone agrees on the meaning of unification or unifying theory in science. Morrison observed that, although theory unification is a messy process which may not have discernible universal characteristics, historically successful unifying scientific theories tend to have two common features: (1) a formalized framework (quantitative mathematical descriptions of the phenomena) and (2) unifying principles (abstract concepts that unite diverse phenomena). On this conception, then, a unifying theory of psychedelic drug effects would offer a single formalized (mathematical or computational) framework capable of describing diverse psychedelic phenomena using a minimal set of unifying principles. Unfortunately, the survey of literature in this review does not locate an existing unifying theory of psychedelic drug effects. It does, however, highlight enduring abstract principles that recur across more than a century of theoretical efforts. Furthermore, it reviews recent formalized frameworks which, although currently heterogeneous and divergent, hint at the possibility of a quantitative groundwork for a future unifying theory.

The field of cognitive neuroscience offers formalized frameworks and general principles designed to track and model the neural correlates of perception, emotion, cognition, and consciousness. These broad frameworks span major levels of description in the brain and attempt to map them onto behavioral and phenomenological data. Corlett et al. argue that until this is done our understanding of how the pharmacology links to the symptoms will remain incomplete. Montague et al. argue that computational psychiatry can remedy the lack of appropriate intermediate levels of description that bind ideas articulated at the molecular level to those expressed at the level of descriptive clinical entities. Seth argues that computational and theoretical approaches can facilitate a transition from correlation to explanation in consciousness science, and explains how a recent LSD, psilocybin, and ketamine study was motivated by a need to elucidate descriptions at intermediate levels somewhere between pharmacology and phenomenology: We know there is a pharmacological link, we know there is a change in experience, and we know there is a clinical impact. But the middle bit... what are these drugs doing to the global activity of the brain..., that is the gap we are trying to fill with this study. Taken together, the above quotations point to an emerging sense that cognitive neuroscience frameworks can address gaps in our understanding of psychedelic drug effects.

In this article I review theories of psychedelic drug effects. First, making an effort to clearly define the target explananda, I review the acute subjective phenomenological properties of psychedelic effects as well as long-term clinical outcomes from psychedelic-assisted therapies. Second, I review theories from first-wave and second-wave psychedelic science, model psychoses theory, filtration theory, and psychoanalytic theory, and identify core features of these theories. Third, I review findings from recent neurophysiological research in humans under psychedelic drugs. Finally, I review select 21st-century theories of psychedelic effects that have been developed within cognitive neuroscience frameworks; namely, entropic brain theory, integrated information theory, and predictive processing. My analysis of recent theoretical efforts highlights certain features, first conceptualized in 19th- and 20th-century theories, which remain relevant in their ability to capture both the phenomenological and neurophysiological dynamics of psychedelic effects. I describe how these enduring theoretical features are now being operationalized into formalized frameworks and could serve as potential unifying principles for describing diverse psychedelic phenomena.

Psychedelic Drug Effects

There are dozens of molecules known to cause psychedelic-like effects. This review focuses only on a limited set of drugs dubbed classical hallucinogens or classic psychedelics which are: LSD, DMT, psilocybin, and mescaline. Importantly, there are qualitative inter-drug differences between the effects of the four classic psychedelic drugs. Drug dosage is a primary factor in predicting the types of effects that will occur. Effects unfold temporally over a drug session; onset effects are distinct from peak effects and some effects have a higher probability of occurring at specific timepoints over the total duration of drug effects. Furthermore, effects are influenced by non-drug factors traditionally referred to as set and setting, such as personality, pre-dose mood, drug session environment, and external stimuli, Figure 1.

fphar-09-00172-g001.jpg

FIGURE 1
‘Extra-pharmacological’ factors that can determine psychedelic drug effects. “Trait factors may be biological [e.g., receptor polymorphisms or psychological in nature [e.g., personality or suggestibility. The pre-state refers to such things as anticipatory anxiety, expectations and assumptions (which account for so-called ‘placebo’ and ‘nocebo’ effects), and readiness to surrender resistances and ‘let go’ to the drug effects. In the context of psychedelic research, the pre-state is traditionally referred to as the ‘set’. State refers to the acute subjective and biological quality of the drug experience and may be measured via subjective rating scales or brain imaging. Dose relates to the drug dosage—which may be a critical determinant of state—as well as long-term outcomes. Environment relates to the various environmental influences. In the context of psychedelic research this is traditionally referred to as ‘setting’. We recognize that the environment can be influential at all stages of the process of change associated with drug action. The long-term outcomes may include such things as symptoms of a specific psychiatric condition such as depression—measured using a standard rating scale as well as relatively pathology-independent factors such as personality and outlook”.

The above variables, while crucial, do not completely prohibit meaningful characterization of general psychedelic effects, as numerous regularities, patterns, and structure can still be identified. Indeed, common psychedelic effects can be reliably measured using validated psychometric instruments consisting of self-report questionnaires and rating scales though some of these rating scales may be in need of further validation using modern statistical techniques. Items from these rating scales are wrapped in scare quotes in the following discussion in an effort to characterize the subjective phenomenology of psychedelic effects from a first-person perspective. An example of rating scale results is given in Figure 2.

fphar-09-00172-g002.jpg

FIGURE 2
Subjective rating scale items selected after psilocybin (blue) and placebo (red). “Items were completed using a visual analog scale format, with a bottom anchor of ‘no, not more than usually’ and a top anchor of ‘yes, much more than usually’ for every item, with the exception of ‘I felt entirely normal,’ which had bottom and top anchors of ‘No, I experienced a different state altogether’ and ‘Yes, I felt just as I normally do,’ respectively. Shown are the mean ratings for 15 participants plus the positive SEMs. All items marked with an asterisk were scored significantly higher after psilocybin than placebo infusion at a Bonferroni-corrected significance level of p < 0.0022.

Perceptual Effects

Perceptual effects occur along a dose-dependent range from subtle to drastic. The range of different perceptual effects includes perceptual intensification, distortion, illusion, mental imagery, elementary hallucination, and complex hallucination. Intensifications of color saturation, texture definition, contours, light intensity, sound intensity, timbre variation, and other perceptual characteristics are common. The external world is experienced as if in higher resolution, seemingly more crisp and detailed, often accompanied by a distinct sense of clarity or freshness in the environment. Sense of meaning in percepts is altered, e.g. Things around me had a new strange meaning for me, or, Objects around me engaged me emotionally much more than usual.

Perceptual distortions and illusions are extremely common, e.g. Things looked strange or My sense of size and space was distorted, or, Edges appeared warped or I saw movement in things that were not actually moving. Textures undulate in rhythmic movements, object boundaries warp and pulsate, and the apparent sizes and shapes of objects can shift rapidly. Controlled psychophysical studies have measured various alterations in motion perception, object completion, and binocular rivalry.

In what are known as elementary hallucinations, e.g. I saw geometric patterns, the visual field can become permeated with intricate tapestries of brightly colored, flowing latticework and other geometric visuospatial form constants. In complex hallucinations visual scenes can present elaborate structural motifs, landscapes, cities, galaxies, plants, animals, and human (and non-human) beings. Complex hallucinations typically succeed elementary hallucinations and are more likely at higher doses, especially under DMT. Both elementary and complex hallucinations are more commonly reported behind closed eyelids (closed eye visuals, or CEVs) but can dose-dependently occur in full light with eyes open (open eye visuals; OEVs). CEVs are often described as vivid mental imagery. Under psychedelic drugs, mental imagery becomes augmented and intensified, e.g. My imagination was extremely vivid, and is intimately linked with emotional and cognitive effects. Sometimes sensible film-like scenes appear, but very often the visions consist of scenes quite indescribable in ordinary language, and bearing a close resemblance to the paintings and sculptures of the surrealistic school. Psychedelic mental imagery can be modulated by both verbal and musical auditory stimuli. Synaesthesia has been reported, especially visual phenomena driven by auditory stimuli, e.g. Sounds influenced the things I saw, but classification of these effects as true synaesthesia is actively debated.

Somatosensory perception can be drastically altered, e.g. I felt unusual bodily sensations including body image, size, shape, and location. Sense of time and causal sequence can lose their usual linear cause-effect structure making it difficult to track the transitions between moments.

Overall the perceptual effects of psychedelics are extremely varied, multimodal, and easily modulated by external stimuli. Perceptual effects are tightly linked with emotional and cognitive effects.

Emotional Effects

Emotional psychedelic effects are characterized by a general intensification of feelings, increased (conscious) access to emotions, and a broadening in the overall range of emotions felt over the duration of the drug session. Psychedelics can induce unique states of euphoria characterized by involuntary grinning, uncontrollable laughter, silliness, giddiness, playfulness, and exuberance. Negatively experience emotions, e.g. I felt afraid or I felt suspicious and paranoid, are often accompanied by a general sense of losing control, e.g. I feared losing control of my mind. However, the majority of emotional psychedelic effects in supportive contexts are experienced as positive. Both LSD and psilocybin can bias emotion toward positive responses to social and environmental stimuli. Spontaneous feelings of awe, wonder, bliss, joy, fun, excitement (and yes, peace and love) are also consistent themes across experimental and anecdotal reports. In supportive environments, classic psychedelic drugs can promote feelings of trust, empathy, bonding, closeness, tenderness, forgiveness, acceptance, and connectedness. Emotional effects can be modulated by all types of external stimuli, especially music.

Cognitive Effects

Precise characterization of cognitive psychedelic effects has proven enigmatic and paradoxical. Acute changes in the normal flow of linear thinking, e.g. My thinking was muddled or My thoughts wandered freely, are extremely common. This is reflected in reduced performance on standardized measures of working memory and directed attention; however, reductions in performance have been shown to occur less often in individuals with extensive past experience with the drugs effects. Crucially, cognitive impairments related to acute psychedelic effects are dose-dependent. Extremely low doses, known as microdoses, have been anecdotally associated with improvements in cognitive performance, a claim that urgently requires empirical verification through controlled research. Theoretical attempts to account for the reported effects of microdosing have yet to emerge in the literature
and therefore present an important opportunity to future theoretical endeavors.

Certain cognitive traits associated with creativity can increase under psychedelics such as divergent thinking use of unlikely language patterns or word associations, expansion of semantic activation, and attribution of meaning to perceptual stimuli, especially musical stimuli. Primary-process thinking, a widely validated psychological construct associated with creativity, is characterized phenomenologically by image fusion; unlikely combinations or events; sudden shifts or transformations of images; and contradictory or illogical actions, feelings, or thoughts. Psilocybin and LSD have been shown to increase primary-process thinking as well as the subjective bizarreness and dreamlike nature of mental imagery associated with verbal stimuli. Cognitive flexibility (or loosening of cognition) and optimism can remain for up to 2 weeks after the main acute drug effects have dissipated. Furthermore, long-term increases in creative problem-solving ability and personality trait openness have been measured after just one psychedelic experience.

Ego Effects and Ego Dissolution Experiences

Kluever observed that under peyote the line of demarcation drawn between object and subject in normal state seemed to be changed. The body, the ego, became objective in a certain way, and the objects became subjective. Similar observations continued throughout first-wave and second-wave psychedelic science. Importantly, effects on sense of self and ego occur along a dose-dependent range spanning from subtle to drastic. Subtle effects are described as a softening of ego with increased insight into ones own habitual patterns of thought, behavior, personal problems, and past experiences; effects which were utilized in psycholytic psychotherapy. Drastic ego-effects, known as ego dissolution, are described as the dissolution of the sense of self and the loss of boundaries between self and world, e.g. I felt like I was merging with my surroundings, or All notion of self and identity dissolved away, or I lost all sense of ego or I experienced a loss of separation from my environment, or I felt at one with the universe. These descriptions resemble non-drug mystical-type experiences; however, the extent of overlap here remains an open question. Ego dissolution is more likely to occur at higher doses. Furthermore, certain psychedelic drugs cause ego dissolution experience more reliably than others; psilocybin, for example, was found to produce full ego dissolution more reliably compared with LSD. Ego dissolution experiences can be driven and modulated by external stimuli, most notably music. Interestingly, subjects who experienced complete ego dissolution in psychedelic-assisted therapy were more likely to evidence positive clinical outcomes as well as long-term changes in life outlook and the personality trait openness.

Clinical Efficacy and Long-Term Effects

Mescaline-assisted therapies showed promising results during first-wave psychedelic science, and this trend continued through second-wave psychedelic research on LSD-assisted therapies. Recent studies have produced significant evidence for the therapeutic utility of psychedelic drugs in treating a wide range of mental health issues, including anxiety and depression, obsessive-compulsive disorder, and addiction to alcohol and tobacco. In many clinical studies, ego-dissolution experience has correlated with positive clinical outcomes.

Remarkably, as mentioned above, a single psychedelic experience can increase optimism for at least 2 weeks after the session and can produce lasting changes in personality trait openness. A study of regular (weekly) ayahuasca users showed improved cognitive functioning and increased positive personality traits compared with matched controls. Interestingly, these outcomes may expand beyond sanctioned clinical use, as illicit users of classic psychedelic drugs within the general population self-report positive long-term benefits from their psychedelic experiences, are statistically less likely to evidence psychological distress and suicidality, and show an overall lower occurrence of mental health problems in general.

Summary

The above evidence demonstrates the broad diversity of acute subjective effects that classic psychedelic drugs can produce in perceptual, emotional, and cognitive domains. Unique changes in sense of self, ego, body image, and personal meaning are particularly salient themes. How do these molecules produce such dramatic effects? What are the relationships between acute perceptual, emotional, cognitive, and self-related effects? What is the link between acute effects and long-term changes in mental health, personality, and behavior? Theories addressing these questions emerged as soon as Western science recognized the need for a scientific understanding of psychedelic drug effects beginning in the late 19th century.

19th and 20th Century Theories of Psychedelic Drug Effects

The effects described above are what captured the interest of first-wave and second-wave psychedelic scientists, and the theories they developed in their investigations have two central themes. The first theme is the observation that psychedelic effects share descriptive elements with symptoms of psychoses, such as hallucination, altered self-reference, and perceptual distortions. This theme forms the basis of model psychoses theory and is what motivated the adoption of the term psychotomimetic drugs. The second theme is the observation that psychedelic drugs seem to expand the total range of contents presented subjectively in our perceptual, emotional, cognitive, and self-referential experience. This theme forms the basis of filtration theory and is what motivated the adoption of the term psychedelic drugs. A third theoretical account uses psychoanalytic theory to address the expanded range of mental phenomena produced by psychedelic drugs as well as the shared descriptive elements with symptoms of psychoses. The next section reviews these themes along with their historically associated theories before tracing their evolution into third-wave (21st-century) psychedelic science.

Model Psychoses Theory

When Lewin discovered the peyote cactus, his reports caught the attention of adventurous 19th-century scientists like Prentiss and Morgan, Mitchell, and Ellis, who promptly obtained samples and began consuming the cactus and observing its effects on themselves. When Heffter isolated mescaline from the peyote cactus and Spaeth paved the way for laboratory synthesis, scientists began systematically dosing themselves with mescaline and publishing their findings in medical journals. Kluever, intrigued by the approach of Knauer and Maloney, ingested peyote at the University of Minnesota Psychological Laboratory and, after the effects had taken hold, completed standard psychophysical measures. Kluever argued that systematic investigations into the neural mechanisms of mescaline effects would help neurology elucidate more general questions of the psychology and pathology of perception. However, it was the pathology aspect, not the general psychology questions, which became the dominant focus of ensuing mescaline research paradigms.

Model psychoses theory began long before any of the classic psychedelic drugs became known to Western science. Moreau (1845) linked hashish effects with mental illness, and Kraepelin founded pharmacopsychology by dosing himself and his students with various psychoactive drugs in the laboratory of Wilhelm Wundt. These scientists hoped to study psychotic symptoms using drugs to induce model psychoses (1) in themselves, to gain first-person knowledge of the phenomenology of psychotic symptoms by administering to one another such substances as will produce in us transitory psychoses, and (2) in normal research subjects, allowing for laboratory behavioral observations on how the symptoms emerge and dissipate. Kraepelin and colleagues attempted to model psychoses using many drugs, e.g. tea, alcohol, morphine, trional, bromide, and other drugs, yet Kraepelins pupils Knauer and Maloney argued that these drugs unfortunately produce mental states which have little similarities to actual insanities and argued instead that mescaline was unique in its ability to truly model psychoses. The dramatic subjective effects of mescaline invigorated the model psychoses paradigm. Growing demand for the ideal chemical agent for model psychoses eventually motivated Sandoz Pharmaceuticals to bring LSD to market in the 1940s.

Importantly, model psychoses theory was not initially a theory of drug effects; it was an idealistic paradigm for researching psychoses that was already in use before Western science discovered classic psychedelic drugs. Nonetheless, it seeded the idea that psychedelic effects themselves could be explained in terms of psychopathology and motivated a search for common neural correlates. The founding figures of neuropharmacology were driven by questions regarding the relationship between psychoactive drugs and endogenous neurochemicals. The putative psychoses-mimicking effects of LSD and mescaline inspired the idea that psychotic symptoms might be caused by a hypothetical endotoxin or some yet-unknown endogenous neurochemical gone out of balance. The discovery that LSD can antagonize serotonin led to the hypothesis that the effects of LSD are serotonergic and simultaneously to the historic hypothesis6 that serotonin might play a role in regulating mental function.

At the 1955 Second Conference on Neuropharmacology, the whole class of drugs was dubbed psychotomimetic. Interestingly, the word mimetic means to imitate, mimic, or exhibit mimicry, which is the act of appearing as something else, for example, when one species mimics the appearance or behavior of another. Psychotomimetic drug effects, on this literal reading of the term, would merely mimic or imitate, appear as if they are, psychoses. However, to mimic is not to model. A model intends to capture important structural or functional principles of the entity or phenomena that it models. A mimic, by contrast, merely creates the illusion that it possesses the properties it mimics. Thus, the term psychotomimetic implies that the effects of these drugs merely resemble psychoses but do not share functional or structural properties in their underlying biology or phenomenology. Nonetheless, LSD and mescaline were used as models to investigate psychotic symptoms. Yet the scientific utility of drug models hinges on our understanding of the mechanisms underpinning the drugs effects; we still need a theory of how psychotomimetic drugs work. A subtle explanation, explananda circularity can come into play here, in which psychoses are explained using drug models yet the drug effects are explained using theories of psychoses. Further complicating the matter is the clear difference between acutely induced drug effects and the gradual development of a chronic mental illness. This cluster of conceptual challenges poured fuel on the flaming debates about the merits of drug-induced model psychoses, which in 1957 had already smoldered for nearly 50 years. An additional conceptual challenge was the fact that mescaline had for years shown promise in treating psychopathologies, and LSD was gaining popularity for pharmaceutically enhanced psychotherapy. Model psychoses theory needed to explain how it was the case that drugs putatively capable of inducing psychotic symptoms could simultaneously be capable of treating them, what Osmond termed, the hair of the dog problem. In fact, to this day, the apparent paradox by which the same compound can be both a model of, and yet a treatment for, psychopathology has never been properly addressed. Taken together, the above cluster of conceptual challenges drove Osmond to doubt his own prior work on model psychoses, and he declared psychotomimetic an outmoded term, arguing that the effects of these drugs could not be captured wholly in terms of psychopathology. If mimicking mental illness were the main characteristic of these agents, psychotomimetics would indeed be a suitable generic term. It is true that they do so, but they do much more.

Filtration Theory

Osmond argued that the psychotomimetic class of drugs needed a more appropriate name. My choice, because it is clear, euphonious, and uncontaminated by other associations, is psychedelic, mind-manifesting. But how exactly should we understand psychedelic effects as mind-manifesting? Osmonds nomenclature legacy was directly influenced by his friend Aldous Huxley, who described the core idea to Osmond in the following personal letter dated April 10, 1953:

Dear Dr. Osmond,

It looks as though the most satisfactory working hypothesis about the human mind must follow, to some extent, the Bergsonian model, in which the brain with its associated normal self, acts as a utilitarian device for limiting, and making selections from, the enormous possible world of consciousness, and for canalizing experience into biologically profitable channels. Disease, mescaline, emotional shock, aesthetic experience and mystical enlightenment have the power, each in its different way and in varying degrees, to inhibit the function of the normal self and its ordinary brain activity, thus permitting the other world to rise into consciousness.

Yours sincerely,

Aldous Huxley


Huxleys letter can help unpack the intended mind-manifesting etymology of Osmonds new term psychedelic. Huxley saw the biological function of the brain as a device engaged in a continuous process of elimination and inhibition to sustain the normal self of everyday waking experience to maximize adaptive fit. Huxleys choice metaphor for visualizing this was the cerebral reducing valve, Figure 3.

fphar-09-00172-g003.jpg

FIGURE 3
Aldous Huxley’s “cerebral reducing valve.” On the ‘inlet’ (right) side of the cerebral reducing valve is a vast ocean of all possible perceptual, emotional, and cognitive experiences. On the ‘outlet’ (left) side is our moment-to-moment stream of experience in normal waking life. Mechanisms inside the valve ‘reduce’ the character and contents of experience, ‘canalizing’ the ocean of possible experience into a more limited stream of waking consciousness aimed at maximum biological utility.

What I have called the cerebral reducing valve is a normal brain function that limits our mental processes to an awareness, most of the time, of what is biologically useful. Huxley argued that this normal brain function emerges developmentally during the course of psychological maturity, so for a period during childhood, before the cerebral reducing valve has fully developed, there is this capacity to live in a kind of visionary world. Once the valve is fully developed, however, normal waking life becomes restricted to a world fabricated by our everyday, biologically useful and socially conditioned perceptions, thoughts and feelings.

Huxley borrowed the core idea from 19th-century filtration theory accounts of various mental phenomena: According to filtration theorists, consciousness is ordinarily kept narrow by biological and psychological selection processes that exclude a great deal of subconscious material. Filtration theorists include founding figures of psychopharmacology, psychology, and parapsychology, along with early 20th-century philosophers Bergson. Bergson applied his own filtration framework to drug effects in his brief response to James, glowing descriptions of what it is like to inhale nitrous oxide. James peculiar state of mind, explained Bergson, should be thought of as a latent potential of the brain/mind, which nitrous oxide simply brought about materially, by an inhibition of what inhibited it, by the removing of an obstacle; and this effect was the wholly negative one produced by the drug. Huxley picked up Bergsons line of thinking and eventually convinced Osmond that it was important to reflect this principle in scientific descriptions of the effects of LSD and mescaline. Smythies also subscribed to this idea, stating that mescaline may be supposed to inhibit that function in the brain which specifically inhibits the mescaline phenomena from developing in the sensory fields.

Thus, Osmonds proposed name-change, psychedelic, was intended to capture the spirit of filtration theory. In this new descriptive model, psyche (mind) delic (manifesting) drugs manifest the mind by inhibiting certain brain processes which normally maintain their own inhibitory constraints on our perceptions, emotions, thoughts, and sense of self. Osmond and Huxley both found this principle highly applicable to their own direct first-person knowledge of what it is like to experience the effects of mescaline and LSD, the expanded range of feelings, intensification of perceptual stimuli, vivid vision-like mental imagery, unusual thoughts, and expanding (or dissolving) sense of self and identity.

Osmond argued that his mind-manifesting description had further theoretical virtues that could address the conceptual challenges of model psychoses theory and improve our understanding of (1) the diverse range of psychedelic effects, (2) their relationship to psychotic symptoms, and (3) their role in psychedelic-assisted therapies. First, the pharmacological disruption of hypothetical inhibitory brain mechanisms that normally attenuate internal and external stimuli suggested that the kinds of effects produced by the drug would depend on the kinds of stimuli in the system, which is consistent with the diverse range of effects on multiple perceptual modalities, emotional experience, and cognition.

Second, the brains selective filtration mechanisms, while evolutionarily adaptive and biologically useful, could develop pathological characteristics in two fundamentally distinct ways. First, a chronically overactive filter limits too much of the mind, causing a rigid, dull, neurotic life in which mental contents become overly restricted to those enumerated in the Sears-Roebuck catalog which constitutes the conventionally real world. Second, a chronically underactive or leaky filter places too few constraints on the mind and allows too much Mind at Large to enter conscious awareness, potentially resulting in perceptual instability, cognitive confusion, or hallucination. This picture helped Huxley and Osmond understand the relationship between psychedelic phenomena and psychotic phenomena: temporarily opening the cerebral reducing valve with psychedelics could produce mental phenomena that resembled symptoms of chronic natural psychoses precisely because both were the result of (acute or chronic) reductions in brain filtration mechanisms.

Third and finally, filtration theory addressed the paradoxical hair of the dog issue, e.g. why drugs that mimic psychoses can aid psychotherapy, which, as described in the previous section, was a conceptual challenge for model psychoses theory. The solution to the paradox was in the filtration theory idea that psychedelic drugs temporarily disable brain filtration mechanisms, which could allow patients and therapists to work outside of the patients everyday (pathological) inhibitory mechanisms. Thus, filtration theory offered a way to understand psychedelic effects that was consistent with both their psychotomimetic properties and their therapeutic utility.

Osmond and Huxley argued that filtration theory concepts were fully consistent with the subjective phenomenology, psychotomimetic capability, and therapeutic efficacy of psychedelic drugs. However, it remains unclear exactly what it is that the brain is filtering and consequently what it is that emerges when the filter is pharmacologically perturbed by a psychedelic drug. According to Huxley, LSD and mescaline inhibit the function of the normal self and its ordinary brain activity, thus permitting the other world to rise into consciousness. Huxley spoke of the brain as a device that filters the world and when the filter is removed we experience more of reality. Osmonds mind-manifesting (psyche) (delic) name, by contrast, suggests that these drugs permit latent aspects of mind to rise into conscious awareness. So which is it? Do psychedelic drugs manifest latent aspects of mind or of world? How we answer this question will crucially determine our ontological and epistemological conclusions regarding the nature of psychedelic experience. Huxley and Osmond did not make this clear. Huxley seems to favor the position that psychedelic experience reveals a wider ontological reality and grants epistemic access to greater truth. Osmonds view, on which these drugs reveal normally hidden aspects of mind, seems less radical, more compatible with materialist science, and less epistemically and ontologically committed. Still, if mind provides us with access to world, then lifting restrictions on mind could in principle expand our access to world. This important point resurfaces in section Predictive Processing below.

Psychoanalytic Theory

Freud developed an elaborate theoretical account of mental phenomena which, like filtration theory, placed great emphasis on inhibition mechanisms in the nervous system. Freud divided the psyche into two fundamentally distinct modes of activity: the primary process and the secondary process. In the primary process, the exchange of neuronal energy is freely mobile and its psychological dynamics are characterized by disorder, vagueness, conceptual paradox, symbolic imagery, intense emotions, and animistic thinking. In the secondary process, by contrast, the exchange of neuronal energy is bound and its psychological dynamics are characterized by order, precision, conceptual consistency, controlled emotions, and rational thinking. Freud hypothesized that the secondary process is maintained by an organizing neural mass called the ego which contains and exerts control over the primary process by binding primary process activity into its own pattern of activity. Freud hypothesized that secondary process neural organization, sustained by the ego, is required for certain aspects of perceptual processing, directed attention, reality-testing, sense of linear time, and higher cognitive processes. When Freuds ego is suppressed, such as during dream sleep, wider worlds of experience can emerge, but secondary process functions are lost. The secondary process and its supporting neural organizing pattern, the ego, emerges during ontogenetic development and solidifies with adult maturity: A unity comparable to the ego cannot exist from the start; the ego has to be developed. Furthermore, pathological characteristics can emerge when Freuds ego restricts either too much or too little of the primary process.

Freud himself was apparently uninterested in psychedelic drugs and instead emphasized dreams as the royal road to a knowledge of the unconscious activities of the mind. Nonetheless, psychedelic drugs produce dreamlike visions and modes of cognition that feature symbolic imagery, conceptual paradox, and other hallmark characteristics of the primary process. How did other psychoanalytic theorists describe psychedelic drug effects? The core idea is that psychedelic drugs interfere with the structural integrity of the ego and thereby reduce its ability to suppress the primary process and support the secondary process. This frees the primary process which then spills into conscious awareness, resulting in perceptual instability, wildly vivid imagination, emotional intensity, conceptual paradox, and loss of usual self-boundaries. Due in part to the close resemblance between psychedelic effects and primary process phenomena, psychoanalytic theory became the framework of choice during the mid 20th-century boom in psychedelic therapy. Psychedelic ego effects, which range from a subtle loosening to a complete dissolution of ego boundaries, were found to be great tools in psychotherapy because of their capacity to perturb ego and allow primary process phenomena to emerge.

But how do psychedelic drugs disrupt the structure of the ego? Freud hypothesized that the organizational structure of ego rests upon a basic perceptual schematic of the body and its surrounding environment. Perceptual signals are continuously bound and integrated into the somatic boundaries of the ego. Savage speculated that the LSDs perceptual effects and ego effects are tightly linked. LSD acts by altering perception. Continuous correct perception is necessary to maintain ego feeling and ego boundaries. Perception determines our ego boundaries. Disturbances in perception caused by LSD make it impossible for the ego to integrate the evidence of the senses and to coordinate its activities... Klee expanded Savages insights into a set of hypotheses aimed at elucidating the neurobiological mechanisms of a Freudian stimulus barrier and its dissolution under LSD:

Such barriers would presumably consist of processes limiting the spread of excitation between different functional areas of the brain. The indications are that LSD, in some manner, breaks down these stimulus barriers of which Freud spoke. Nor is this merely a figure of speech. There is some reason to suspect that integrative mechanisms within the central nervous system (CNS) which handle inflowing stimuli are no longer able to limit the spread of excitation in the usual ways. We might speculate that LSD allows greater energy exchanges between certain systems than normally occurs, without necessarily raising the general level of excitation of all cortical and subcortical structures.

Freud hypothesized that ego is sustained by a delicate balance of neuronal energy which critically depends on integrative mechanisms to process inflowing sensory stimuli and to bind neural excitation into functional structures within the brain. Psychedelic drugs, according to Savage and Klee, perturb integrative mechanisms that normally bind and shape endogenous and exogenous excitation into the structure of the ego. As we will see below, Klee?s ideas strongly anticipate many neurophysiological findings and theoretical themes from 21st-century psychedelic science.

Summary

From the above analysis of first-wave and second-wave theories I have identified four recurring theoretical features which could potentially serve as unifying principles. One feature is the hypothesis that psychedelic drugs inhibit a core brain mechanism that normally functions to reduce or filter or constrain mental phenomena into an evolutionarily adaptive container. A second feature is the hypothesis that this core brain mechanism can behave pathologically, either in the direction of too much, or too little, constraint imposed on perception, emotion, cognition, and sense of self. A third feature is the hypothesis that psychedelic phenomena and symptoms of chronic psychoses share descriptive elements because they both involve situations of relatively unconstrained mental processes. A fourth feature is the hypothesis that psychedelic drugs have therapeutic utility via their ability to temporarily inhibit these inhibitory brain mechanisms. But how are these inhibitory mechanisms realized in the brain?

Neuropharmacology and Neurophysiological Correlates of Psychedelic Drug Effects

Klee recognized that his above hypotheses, inspired by psychoanalytic theory and LSD effects, required neurophysiological evidence. As far as I am aware, however, adequate neurophysiological evidence is lacking. The long awaited millennium in which biochemical, physiological, and psychological processes can be freely correlated still seems a great distance off. What clues have recent investigations uncovered?

A psychedelic drug molecule impacts a neuron by binding to and altering the conformation of receptors on the surface of the neuron. The receptor interaction most implicated in producing classic psychedelic drug effects is agonist or partial agonist activity at serotonin (5-HT) receptor type 2A (5-HT2A). A molecules propensity for 5-HT2A affinity and agonist activity predicts its potential for (and potency of) subjective psychedelic effects. When a psychedelic drugs 5-HT2A agonist activity is intentionally blocked using 5-HT2A antagonist drugs, e.g. ketanserin, the subjective effects are blocked or attenuated in humans under psilocybin, LSD, and ayahuasca. Importantly, while the above evidence makes it clear that 5-HT2A activation is a necessary mediator of the hallmark subjective effects of classic psychedelic drugs, this does not entail that 5-HT2A activation is the sole neurochemical cause of all subjective effects. For example, 5-HT2A activation might trigger neurochemical modulations downstream, e.g. changes in glutamate transmission, which could also play causal roles in producing psychedelic effects. Moreover, most psychedelic drug molecules activate other receptors in addition to 5-HT2A, and these activations may importantly contribute to the overall profile of subjective effects even if 5-HT2A activation is required for their effects to occur.

How does psychedelic drug-induced 5-HT2A receptor agonism change the behavior of the host neuron? Generally, 5-HT2A activation has a depolarizing effect on the neuron, making it more excitable (more likely to fire). Importantly, this does not necessarily entail that 5-HT2A activation will have an overall excitatory effect throughout the brain, particularly if the excitation occurs in inhibitory neurons. This important consideration (captured by the adage one neurons excitation is another neurons inhibition) should be kept in mind when tracing causal links in the pharmaco-neurophysiology of psychedelic drug effects.

In mammalian brains, neurons tend to fire together in synchronized rhythms known as temporal oscillations (brain waves). MEG and EEG equipment measure the electromagnetic disturbances produced by the temporal oscillations of large neural populations and these measurements can be quantified according to their amplitude (power) and frequency (timing). Specific combinations of frequency and amplitude can be correlated with distinct brain states, including waking resting state, various attentional tasks, anesthesia, REM sleep, and deep sleep. In what ways do temporal oscillations change under psychedelic drugs? MEG and EEG studies consistently show reductions in oscillatory power across a broad frequency range under ayahuasca, psilocybin, and LSD. Reductions in the power of alpha-band oscillations, localized mainly to parietal and occipital cortex, have been correlated with intensity of subjective visual effects, e.g. I saw geometric patterns, or, My imagination was extremely vivid, under psilocybin and ayahuasca. Under LSD, reductions in alpha power still correlated with intensity of subjective visual effects but associated alpha reductions were more widely distributed throughout the brain. Furthermore, ego-dissolution effects and mystical-type experiences, e.g. I experienced a disintegration of my self, or ego, or, The experience had a supernatural quality, have been correlated with reductions in alpha power localized to anterior and posterior cingulate cortices and the parahippocampal regions under psilocybin and throughout the brain under LSD.

The concept of functional connectivity rests upon fMRI brain imaging observations that reveal temporal correlations of activity occurring in spatially remote regions of the brain which form highly structured patterns (brain networks). Imaging of brains during perceptual or cognitive task performance reveals patterns of functional connectivity known as functional networks, e.g. control network, dorsal attention network, ventral attention network, visual network, auditory network, and so on. Imaging brains in taskless resting conditions reveals resting-state functional connectivity (RSFC) and structured patterns of RSFC known as resting state networks. One particular RSN, the default mode network, increases activity in the absence of tasks and decreases activity during task performance. DMN activity is strong during internally directed cognition and a variety of other metacognitive functions. DMN activation in normal waking states exhibits inverse coupling or anticorrelation with the activation of task-positive functional networks, meaning that DMN and functional networks are often mutually exclusive; one deactivates as the other activates and vice versa.

In what ways does brain network connectivity change under psychedelic drugs? First, functional connectivity between key hub areas, mPFC and PCC, is reduced. Second, the strength or oscillatory power of the DMN is weakened and its intrinsic functional connectivity becomes disintegrated as its component nodes become decoupled under psilocybin, ayahuasca, and LSD. Third, brain networks that normally show anticorrelation become active simultaneously under psychedelic drugs. This situation, which can be described as increased between-network functional connectivity, occurs under psilocybin, ayahuasca and especially LSD. Fourth and finally, the overall repertoire of explored functional connectivity motifs is substantially expanded and its informational dynamics become more diverse and entropic compared with normal waking states. Notably, the magnitude of occurrence of the above four neurodynamical themes correlates with subjective intensity of psychedelic effects during the drug session. Furthermore, visual cortex is activated during eyes-closed psychedelic visual imagery and under LSD, the early visual system behaves as if it were receiving spatially localized visual information, as V1-V3 RSFC is activated in a retinotopic fashion.

Taken together, the recently discovered neurophysiological correlates of subjective psychedelic effects present an important puzzle for 21st-century neuroscience. A key clue is that 5-HT2A receptor agonism leads to desynchronization of oscillatory activity, disintegration of intrinsic integrity in the DMN and related brain networks, and an overall brain dynamic characterized by increased between-network global functional connectivity, expanded signal diversity, and a larger repertoire of structured neurophysiological activation patterns. Crucially, these characteristic traits of psychedelic brain activity have been correlated with the phenomenological dynamics and intensity of subjective psychedelic effects.

21st-Century Theories of Psychedelic Drug Effects

How should we understand the growing body of clues emerging from investigations into the neurodynamics of psychedelic effects? What are the principles that link these thematic patterns of psychedelic brain activity (or inactivity) to their associated phenomenological effects? Recent theoretical efforts to understand psychedelic drug effects have taken advantage of existing frameworks from cognitive neuroscience designed to track the key neurodynamic principles of human perception, emotion, cognition, and consciousness. The overall picture that emerges from these efforts shares core principles with filtration and psychoanalytic accounts of the late 19th and early 20th century. Briefly, normal waking perception and cognition are hypothesized to rest upon brain mechanisms which serve to suppress entropy and uncertainty by placing various constraints on perceptual and cognitive systems. In a selecting and limiting fashion, neurobiological constraint mechanisms support stability and predictability in the contents of conscious awareness in the interest of adaptability, survival, and evolutionary fitness. The core hypothesis of recent cognitive neuroscience theories of psychedelic effects is that these drugs interfere with the integrity of neurobiological information-processing constraint mechanisms. The net effect of this is that the range of possibilities in perception, emotion, and cognition is dose-dependently expanded. From this core hypothesis, cognitive neuroscience frameworks are utilized to describe and operationalize the quantitative neurodynamics of key psychedelic phenomena; namely, the diversity of effects across many mental processes, the elements in common with symptoms of psychoses, and the way in which temporarily removing neurobiological
constraints is therapeutically beneficial.

This section is organized according to the broad theoretical frameworks informing recent theoretical neuroscience of psychedelic effects: entropic brain theory, integrated information theory, and predictive processing.

Entropic Brain Theory

Entropic Brain Theory links the phenomenology and neurophysiology of psychedelic effects by characterizing both in terms of the quantitative notions of entropy and uncertainty. Entropy is a quantitative index of a systems (physical) disorder or randomness which can simultaneously describe its (informational) uncertainty. EBT proposes that the quality of any conscious state depends on the systems entropy measured via key parameters of brain function. Their hypothesis states that hallmark psychedelic effects, e.g. perceptual destabilization, cognitive flexibility, ego dissolution) can be mapped directly onto elevated levels of entropy/uncertainty measured in brain activity, e.g. widened repertoire of functional connectivity patterns, reduced anticorrelation of brain networks, and desynchronization of RSN activity. More specifically, EBT characterizes the difference between psychedelic states and normal waking states in terms of how the underlying brain dynamics are positioned on a scale between the two extremes of order and disorder, a concept known as self-organized criticality. A system with high order (low entropy) exhibits dynamics that resemble petrification and are relatively inflexible but more stable, while a system with low order (high entropy) exhibits dynamics that resemble formlessness and are more flexible but less stable. The notion of criticality describes the transition zone in which the brain remains poised between order and disorder. Physical systems at criticality exhibit increased transient metastable states, increased sensitivity to perturbation, and increased propensity for cascading avalanches of metastable activity. Importantly, EBT points out that these characteristics are consistent with psychedelic phenomenology, e.g. hypersensitivity to external stimuli, broadened range of experiences, or rapidly shifting perceptual and mental contents. Furthermore, EBT uses the notion of criticality to characterize the difference between psychedelic states and normal waking states as it describes cognition in adult modern humans as near critical but sub-critical, meaning that its dynamics are poised in a position between the two extremes of formlessness and petrification where there is an optimal balance between order and flexibility. EBT hypothesizes that psychedelic drugs interfere with entropy-suppression brain mechanisms which normally sustain sub-critical brain dynamics, thus bringing the brain, closer to criticality in the psychedelic state.

Entropic Brain Theory further characterizes psychedelic neurodynamics using a neo-psychoanalytic framework proposed in an earlier paper by Carhart-Harris and Friston where they recast some central Freudian ideas in a mechanistic and biologically informed fashion. Freuds primary process (renamed primary consciousness) is hypothesized to be a high-entropy brain dynamic which operates at criticality, while Freuds secondary process (renamed secondary consciousness) is hypothesized to involve a lower-entropy brain state which sustains a sub-critical dynamic via a key neurobiological entropy-suppression mechanism, the ego, which exerts an organizing influence in order to constrain the criticality-like dynamic of primary consciousness. EBT argues that these ego functions have a signature neural footprint; namely, the DMNs intrinsic functional connectivity and DMN coupling of medial temporal lobes (MTLs) in particular. Furthermore, EBT argues that DMN/ego develops ontogenetically in adult humans and plays an adaptive role in which it sustains secondary consciousness and associated metacognitive abilities along with an integrated sense of self.

Importantly, this hypothesis maps onto the subjective phenomenology of psychedelic effects, particularly ego dissolution. As psychedelics weaken the oscillatory power and intrinsic functional connectivity of the DMN, the normally constrained activity of subordinate DMN nodes, MTLs in particular, becomes freely mobile, allowing the emergence of more uncertain (higher entropy) primary consciousness. This view, based on Freudian metapsychology, is also consistent with filtration accounts, like those of Bergson and Huxley, who hypothesized that psychedelic drug effects are the result of a pharmacological inhibition of inhibitory brain mechanisms. EBT recasts these theoretical features using the quantitative terms of physical entropy and informational uncertainty as measured via the repertoire of functional connectivity motifs that form and fragment across time. In normal waking states, the DMN constrains the activity of its cortical and subcortical nodes and prohibits simultaneous co-activation with TPNs. By interfering with DMN integration, psychedelics permit a larger repertoire of brain activity, a wider variety of explored functional connectivity motifs, co-activation of normally mutually exclusive brain networks, increased levels of between-network
functional connectivity, and an overall more diverse set of neural interactions.

Carhart-Harris et al. point out a number of implications of EBT. First, they map the feelings of uncertainty that often accompany psychedelic effects onto the fact that a more entropic brain dynamic is the information-theoretic equivalent to a more uncertain brain dynamic. Thus, according to the entropic brain hypothesis, just as normally robust principles about the brain lose definition in primary states, so confidence is lost in how the world is and who one is as a personality.

Second, like Huxleys cerebral reducing valve and Freuds ego, EBT argues that the DMNs organizational stronghold over brain activity can be both an evolutionary advantage and a source of pathology. It is argued that this entropy-suppressing function of the human brain serves to promote realism, foresight, careful reflection and an ability to recognize and overcome wishful and paranoid fantasies. Equally however, it could be seen as exerting a limiting or narrowing influence on consciousness. Carhart-Harris et al. point out that neuroimaging studies have implicated increased DMN activity and RSFC with various aspects of depressive rumination, trait neuroticism, and depression. The suggestion is that increased DMN activity and connectivity in mild depression promotes concerted introspection and an especially diligent style of reality-testing. However, what may be gained in mild depression (i.e., accurate reality testing) may be offset by a reciprocal decrease in flexible or divergent thinking (and positive mood).

Third, consistent with both psychoanalytic and filtration theory, is the notion that psychedelic drugs capacity to temporarily weaken, collapse, or disintegrate the normal ego/DMN stronghold underpins their therapeutic utility. Specifically, it is proposed that psychedelics work by dismantling reinforced patterns of negative thought and behavior by breaking down the stable spatiotemporal patterns of brain activity upon which they rest.

Fourth and finally, EBT sheds light on the shared descriptive elements between psychedelic effects and psychotic symptoms by characterizing both in terms of elevated levels of entropy and uncertainty in brain activity which lead to a regression into primary consciousness. The collapse of the organizing effect of DMN coupling and anticorrelation patterns, according to EBT, point to system-level mechanics of the psychedelic state as an exemplar of a regressive style of cognition that can also be observed in REM sleep and early psychosis.

Thus, EBT formulates all four of the theoretical features identified in filtration and psychoanalytic accounts, but does so using 21st-century empirical data plugged into the quantitative concepts of entropy, uncertainty, criticality, and functional connectivity. EBT hints at possible ways to close the gaps in understanding by offering quantitative concepts that link phenomenology to brain activity and pathogenesis to therapeutic mechanisms.

Integrated Information Theory

Integrated Information Theory (IIT) is a general theoretical framework which describes the relationship between consciousness and its physical substrates. While EBT is already loosely consistent with the core principles of IIT, Gallimore demonstrates how EBTs hypotheses can be operationalized using the technical concepts of the IIT framework. Using EBT and recent neuroimaging data as a foundation, Gallimore develops an IIT-based model of psychedelic effects. Consistent with EBT, this IIT-based model describes the brains continual challenge of minimizing entropy while retaining flexibility. Gallimore formally restates this problem using IIT parameters: brains attempt to optimize the give-and-take dynamic between cause-effect information and cognitive flexibility. In IIT, a (neural) system generates cause-effect information when the mechanisms which make up its current state constrain the set of states which could casually precede or follow the current state. In other words, each mechanistic state of the brain: (1) limits the set of past states which could have causally given rise to it, and (2) limits the set of future states which can causally follow from it. Thus, each current state of the mechanisms within a neural system (or subsystem) has an associated cause-effect repertoire which specifies a certain amount of cause-effect information as a function of how stringently it constrains the unconstrained state repertoire of all possible system states. Increasing the entropy within a cause-effect repertoire will in effect constrain the system less stringently as the causal possibilities are expanded in both temporal directions as the system moves closer to its unconstrained repertoire of all possible states. Moreover, increasing the entropy within a cause-effect repertoire equivalently increases the uncertainty associated with its past (and future) causal interactions. Using this IIT-based framework, Gallimore argues that, compared with normal waking states, psychedelic brain states exhibit higher entropy, higher cognitive flexibility, but lower cause-effect information, Figure 4.

fphar-09-00172-g004.jpg

FIGURE 4
“Increasing neural entropy elevates cognitive flexibility at the expense of a decrease in the cause-effect information specified by individual mechanisms.”

Neuroimaging data suggests that human brains exhibit a larger overall repertoire of neurophysiological states under psychedelic drugs, exploring a greater diversity of states in a more random fashion. For example, in normal waking states, DMN activity rules out the activity of TPNs, and vice versa, due to their relatively strict anticorrelation patterns. Brain network anticorrelation generates cause-effect information because it places constraints on the possible causal interactions within and between brain mechanisms; for example, DMN-TPN anticorrelation patterns rule out the DMN activity in the presence of activated TPNs. However, psychedelic drugs dissolve DMN-TPN (and other) network anticorrelation patterns, which permits simultaneous activation of brain networks which are normally mutually exclusive. The cause-effect repertoire of brain mechanisms thus shifts closer to the unconstrained repertoire of all possible past and future states. This has the effect of increasing the probability of certain states from zero or, at least, from a very low probability. Therefore the subjective contents perception and cognition become more diverse, more unusual, and less predictable. This increases flexibility but decreases precision and control as the subjective boundaries which normally demarcate distinct cognitive concepts and perceptual objects dissolve. Gallimore leverages IIT in an attempt unify these phenomena under a formalized framework.

However, as Gallimore notes, this model does not explain how neural entropy is increased by (psychedelic drugs), but predicts consequences of the entropy increase revealed by functional imaging data. How do psychedelic drugs increase neural entropy?

Predictive Processing

The first modern brain imaging measurements in humans under psilocybin yielded somewhat unexpected results: reductions in oscillatory power (MEG) and cerebral blood flow (fMRI) correlated with the intensity of subjective psychedelic effects. In their discussion, the authors suggest that their findings, although surprising through the lens of commonly held beliefs about how brain activity maps to subjective phenomenology, may actually be consistent with a theory of brain function known as the free energy principle.

In one model of global brain function based on the free-energy principle, activity in deep-layer projection neurons encodes top-down inferences about the world. Speculatively, if deep-layer pyramidal cells were to become hyperexcitable during the psychedelic state, information processing would be biased in the direction of inference, such that implicit models of the world become spontaneously manifest, intruding into consciousness without prior invitation from sensory data. This could explain many of the subjective effects of psychedelics.

What is FEP? In this view, the brain is an inference machine that actively predicts and explains its sensations. Central to this hypothesis is a probabilistic model that can generate predictions, against which sensory samples are tested to update beliefs about their causes. FEP is a formulation of a broader conceptual framework emerging in cognitive neuroscience known as predictive processing. PP has links to bayesian brain hypothesis, predictive coding, and earlier theories of perception and cognition dating back to Helmholtz, who was inspired by Kant. At the turn of the 21st century, the ideas of Helmholtz catalyzed innovations in machine learning, new understandings of cortical organization, and theories of how perception works.

PP subsumes key elements from these efforts to describe a universal principle of brain function captured by the idea of prediction error minimization. What does it mean to say that the brain works to minimize its own prediction error? Higher-level areas of the nervous system (i.e., higher-order cortical structures) generate top-down synaptic predictions aimed at matching the expected bottom-up synaptic activity at lower-level areas, all the way down to input activity at sense organs. Top-down signals encode a kind of best guess about the most likely (hidden) causes of bodily sensations. In this multi-level hierarchical cascade of neural activity, high-level areas attempt to explain the states of levels below via synaptic attempts to inhibit lower-level activity, high-level areas tell lower levels to shut up. But lower levels will not shut up until they receive top-down feedback (inference) signals that adequately fit (explain) the bottom-up (evidence) signals. Mismatches between synaptic expectation and synaptic evidence generate prediction error signals which carry the news by propagating the surprise upward to be explained away by yet higher levels of hierarchical cortical processing anatomy. This recurrent neural processing scheme approximates (empirical) Bayesian inference as the brain continually maps measured bodily effects to different sets of possible causes and attempts to select the set of possible causes that can best explain away the measured bodily effects.

Crucially, the sets of possible causes must be narrowed in order for the system to settle on an explanation). Prior constraints which allow the system to narrow the hypothesis space are known as inductive biases or priors. Efforts in Bayesian statistics and machine learning have demonstrated that improvements in inductive capabilities occur when priors are linked in a multi-level hierarchy, with not just a single level of hypotheses to explain the data, but multiple levels: hypothesis spaces of hypothesis spaces, with priors on priors. Certain priors in the hierarchy, known as hyperpriors or overhypotheses are more abstract and allow the system to rule out large swaths of possibilities, drastically narrowing the hypothesis space, making explanation more tractable. For example, the brute constraints of space and time act as hyperpriors, e.g. prior knowledge that there is only one object (one cause of sensory input) in one place, at a given scale, at a given moment, or the fact that we can only perform one action at a time, choosing the left turn or the right but never both at once.

Thus, PP states that brains are neural generative models built from linked hierarchies of priors where higher levels continuously attempt to guess and explain activity at lower levels. The entire process can be characterized as the agents attempt to optimize its own internal model of the sensorium (and the world) over multiple spatial and temporal scales. Interestingly, PP holds that our perceptions of external objects recruit the same synaptic pathways that enable our capacity for mental imagery, dreaming, and hallucination. The brains ability to simulate its own virtual reality using internal (generative) models of the worlds causal structure is thus crucial to its ability to perceive the external world. A fruitful way of looking at the human brain, therefore, is as a system which, even in ordinary waking states, constantly hallucinates at the world, as a system that constantly lets its internal autonomous simulational dynamics collide with the ongoing flow of sensory input, vigorously dreaming at the world and thereby generating the content of phenomenal experience.

How do psychedelic molecules perturb predictive processing? If normal perception is a kind of controlled hallucination where top-down simulation is constrained by bottom-up sensory input colliding with priors upon priors, then, as the above quotation from Muthukumaraswamy et al. suggests, psychedelic drugs essentially cause perception to be less controlled hallucination. The idea is that psychedelic drugs perturb the (learned and innate) prior constraints on internal generative models. Via their 5-HT2A agonism, psychedelic drugs cause hyperexcitation in layer V pyramidal neurons, which might cause endogenous simulations to run wild so that awareness becomes more imaginative, dreamlike, and hallucinatory. This hypothesis could in principle still be consistent with observed reductions in brain activity under psychedelics; recall from above that, in PP schemes, the higher-level areas explain away lower-level excitation by suppressing it with top-down inhibitory signals. Here, explaining away just means countering excitatory bottom-up inputs to a prediction error neuron with inhibitory synaptic inputs that are driven by top-down predictions.

How does PP tie into filtration theories and psychoanalytic accounts? Carhart-Harris et al. link Huxley with Friston to interpret their initially surprising fMRI scans of humans under psilocybin. One objection to this linkage might be that Huxley often describes psychedelic opening of the cerebral reducing valve as revealing more of the world. At first glance this seems at odds with the above PP account of psychedelic effects, which describes psychedelic drugs causing rampant internal simulations of reality, not revealing more of the external world. However, this apparent tension might be resolved in light of active inference, a key principle of FEP. Active inference shows how internal models do not merely generate top-down (inference) signals but also shape the sampling and accumulation of bottom-up sensory (evidence) signals. In short, the agent will selectively sample the sensory inputs that it expects. This is known as active inference. An intuitive example of this process (when it is raised into consciousness) would be feeling our way in darkness: we anticipate what we might touch next and then try to confirm those expectations. The principle of active inference hints at a resolution to the apparent tensions between Osmonds mind-manifesting model and Huxleys world-manifesting model. Psychedelics manifest mind by perturbing prior constraints on internal generative models, thereby expanding the possibilities in our inner world of feelings, thoughts, and mental imagery. Importantly, this could also manifest normally ignored aspects of world by altering active inference, which would in effect expand the sampling of sensory data to include samples that are normally routinely explained away. Potentially, this understanding goes some way in explaining the perception-hallucination continuum of psychedelic drug effects, as it shows how perceptual intensifications, on the one hand, and distortions and hallucinations, on the other hand, could both be caused by a synaptic disruption of hierarchically linked priors in internal generative models.

The brief speculative remark by Muthukumaraswamy et al. is not the only PP-based account of psychedelic drug effects. The PP framework describes a recurrent back-and-forth give-and-take between colliding top-down and bottom-up signals, where internal models serve to shape experience and experience serves to build internal models, so this leaves room for rival PP-based accounts that diverge regarding where exactly the psychedelic drug perturbs the system. For example, increased top-down activity could be the result of pharmacological hyperactivation of top-down synaptic transmission; yet equally plausible is the hypothesis that increased top-down activity is a compensatory response to pharmacological attenuations or distortions of bottom-up signal.

For example, Corlett et al. hypothesize that LSD hallucinations result from noisy, unpredictable bottom-up signaling in the context of preserved and perhaps enhanced top-down processing. In contrast to the PP-based account outlined above, which focuses on changes to top-down signals, the strategy of Corlett et al. is to map various psychedelic effects to disturbances of top-down and/or bottom-up signals. The issue of what is primary and what is compensatory illustrates the vast possibilities in the hypothesis space of PP-based accounts.

While most PP-based accounts point to changes in top-down signaling, even within this hypothesis space there are contrasting conceptions of exactly how psychedelic molecules perturb top-down processing. Briefly, these differing hypotheses include: (1) hyperactivation or heavier weighting of top-down signaling, (2) reduced influence of signals from higher cortical areas, (3) interference with multisensory integration processes and PP-based binding of sensory signals, and (4) changes in the composition and level of detail specified by top-down signals.

Carhart-Harris and Friston argue that the Freudian conception of ego, with its organizing influence over the primary process, is consistent with PP descriptions of higher-level cortical structures predicting and suppressing the excitation in lower levels in the hierarchy (i.e., limbic regions). Freud hypothesized that the secondary process binds, integrates, and organizes the lower and more chaotic neural activity of the primary process into the broader and more cohesive composite structure of the ego.

Carhart-Harris and Friston argue that when large-scale intrinsic networks become dis-integrated, the activity at lower levels can no longer be explained away (suppressed) by certain higher-level systems, causing conscious awareness to take on hallmark characteristics of the primary process. In normal adult waking states, networks based in higher-level areas can successfully predict and explain (suppress and control) the activity of lower level areas. In non-ordinary states, this function may be perturbed, e.g. in the case of hallucinogenic drugs, through actions at modulatory post-synaptic receptors), compromising the hierarchical organization and suppressive capacity of the intrinsic networks.

Similar PP-based theories of psychedelic ego dissolution have been proposed without invoking Freud. PP posits that the brain explains self-generated stimuli by attributing its causes to a coherent and persisting entity (i.e., the self), much like how it predicts and explains external stimuli by attributing their causes to coherent and persisting external objects. Letheby and Gerrans use the PP framework to recast the psychoanalysis-based theories of LSD ego effects proposed by Savage and Klee described in Section Psychoanalytic Theory. The core idea is that psychedelic drugs interfere with processes that bind and integrate stimuli according to probabilistic estimates of how relevant the stimuli are to the organisms (self) goals. Letheby and Gerrans point out that ego dissolution under psychedelic drugs is correlated with the desynchronization (reductions in intrinsic functional connectivity) of brain networks implicated in one aspect or another of self-representation, specifically the salience network (SLN) and the DMN. This causes an unbinding of stimuli that are normally processed according to self-binding multisensory integration mechanisms. Attention is no longer guided exclusively by adaptive and egocentric goals and agendas; salience attribution is no longer bound to personal concern. This description echoes Huxleys cerebral reducing valve in which the brain with its associated normal self, acts as a utilitarian device for limiting, and making selections from, the enormous possible world of consciousness, and for canalizing experience into biologically profitable channels. Letheby and Gerrans PP-based account elucidates how psychedelic drugs could perturb the brains associated normal self preventing the usual self-binding of internal and external stimuli.

Pink-Hashkes et al. argue that under psychedelic drugs top-down predictions in affected brain areas break up and decompose into many more overly detailed predictions due to hyper activation of 5-HT2A receptors in layer V pyramidal neurons. Pink-Hashkes et al. state that when internal generative models are described as categorical probability distributions rather than Gaussian densities, the state space granularity (how detailed are the generative models and the predictions that follow from them) is crucial. Categorical predictions that are less detailed will explain more bottom-up data (because they cover more ground) and thus produce less prediction error. Categorical predictions that are more detailed, by contrast, will carry less precision and thus potentially generate more prediction error. Pink-Hashkes et al. propose that psychedelic drugs cause brain structures at certain levels of the cortical hierarchy to issue more detailed decomposed predictions that fit less data than the usual broad prediction. They argue that many psychedelic effects stem from the brains attempts to compensate for these decomposed top-down predictions as it responds to the increase in prediction errors that result from overly detailed predictions.

In summary, the current state of PP-based theories of psychedelic effects reveals a divergent mix of heterogeneous ideas and conflicting hypotheses. Do psychedelic molecules perturb top-down (feedback) signaling, or bottom-up (feedforward) signaling, or both? Do the subjective phenomenological effects result from direct neuropharmacological changes or compensatory mechanisms responding to pharmacological perturbations? Yet there seems to be a core intuition that transcends the conceptual variance here: psychedelic drugs somehow interfere with established priors that normally constrain the brain?s internal generative models.

Predictive processing-based accounts, consistent with EBT and IIT (and filtration and psychoanalytic accounts), propose that psychedelic drugs disrupt neural mechanisms (priors on internal generative models) which normally constrain perception and cognition. Perturbing priors causes subjective phenomenology to present a wider range of experiences with increased risk of perceptual instability and excessive cognitive flexibility. As prior constraints on self and world are loosened, the likelihood of psychosis-like phenomena increases. At the same time, novel thinking is increased and the brain becomes more malleable and conducive to therapeutic cognitive and behavioral change.

Conclusion

The four key features identified in filtration and psychoanalytic accounts from the late 19th and early 20th century continue to operate in 21st-century cognitive neuroscience:

(1) psychedelic drugs produce their characteristic diversity of effects because they perturb adaptive mechanisms which normally constrain perception, emotion, cognition, and self-reference,

(2) these adaptive mechanisms can develop pathologies rooted in either too much or too little constraint,

(3) psychedelic effects appear to share elements with psychotic symptoms because both involve weakened constraints, and

(4) psychedelic drugs are therapeutically useful precisely because they offer a way to temporarily inhibit these adaptive constraints.

It is on these four points that EBT, IIT, and PP seem consistent with each other and with earlier filtration and psychoanalytic accounts. EBT and IIT describe psychedelic brain dynamics and link them to phenomenological dynamics, while PP describes informational principles and plausible neural information exchanges which might underlie the larger-scale dynamics described by EBT and IIT. Certain descriptions of neural entropy-suppression mechanisms (EBT), cause-effect information constraints (IIT), or prediction-error minimization strategies (PP, FEP) are loosely consistent with Freuds ego, and Huxleys cerebral reducing valve.

In surveying the literature for this review I can confidently conclude that 21st-century psychedelic science has yet to approach a unifying theory linking the diverse range of phenomenological effects with pharmacology and neurophysiology while tying these to clinical efficacy. However, the historically necessary ingredients for successful theory unification, formalized frameworks and unifying principles, seem to be taking shape. Formal models are an integral part of 21st-century neuroscience where they help to describe natural principles in the brain and aid explanation and understanding. Here I have reviewed a handful of formalized frameworks, EBT, IIT, PP, which are just beginning to be used to account for psychedelic effects. I have also highlighted the fact that all of the accounts reviewed here, from the 19th-century to the 21st-century, propose that psychedelic drugs inhibit neurophysiological constraints in order to produce their diverse phenomenological, psychotomimetic, and therapeutic effects.

Why should we pursue a unified theory of psychedelic drug effects at all? To date, theories of brain function and mind in general have resisted the kind of unification that has occurred in other areas of science. Because the human brain has evolved disparate and complex layers under diverse environmental circumstances, many doubt the possibility of and debate the merits of seeking grand unified theories (GUTs) of brain function. There is every reason to think that there can be no grand unified theory of brain function because there is every reason to think that an organ as complex as the brain functions according to diverse principles. Indeed, Anderson and Chemero caution that we should be skeptical of any GUT of brain function, and charge that PP in particular, when taken as a unified theory as outlined by Clark, threatens metaphysical disaster.

Given these understandable critical reservations about seeking after GUTs of brain function (and therefore any truly unifying theory of psychedelic drug effects), it is perhaps safer to aspire for theories that feature broad explanatory frameworks and offer conceptual breadth allowing us to paint the big picture. PP and FEP, at the very least, offer a broad explanatory framework that emcompasses a large swath of perceptual and cognitive phenomena. Psychedelic drugs offer a unique way to iteratively develop and test such big-picture explanatory frameworks: these molecules can be used to probe the links between neurochemistry and neural computation across multiple layers of neuroanatomy and phenomenology. Meeting the challenge of predicting and explaining psychedelic drug effects is the ultimate acid test for any unified theory of brain function.

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5853825/
 
Last edited:
neuropsych-m.jpg



Special Report on Neuroscience: Demystifying Psychedelics

by Randall Willis | Exploring Drug Discovery and Development | 26 Mar 2021

Late last year, University of Basel’s Felix Müller and colleagues offered a case report of a 39-year-old wife and mother admitted to their care with severe treatment-resistant depression and complex personality disorder.

She first reported symptoms as a teenager, including feelings of worthlessness and suicidal ideations, that evolved to pseudo-hallucinations (e.g., seeing snakes), panic attacks, and compulsive thoughts. She didn’t seek treatment until she was 22, when her partner committed suicide, and was first hospitalized at age 30 after attempting suicide herself by drug intoxication.

“Over the last years, the patient had been treated with several psychiatric drugs, including antidepressants of different types (escitalopram, sertraline, fluoxetine, duloxetine, moclobemide, reboxetine, trazodone, mirtazapine, vortioxetine, nortriptyline), mood stabilizers (lithium, lamotrigine, valproate), antipsychotics (aripiprazole, quetiapine, olanzapine), and stimulants (modafinil, methylphenidate, atomoxetine),” the authors wrote. “She also had been using benzodiazepines on a regular basis, mostly to cope with fears of contamination.”

She had been prescribed and did not experience long-term relief from 20 or more different drugs in just a few years.

Given her medical history, the woman was started on the psychedelic drug MDMA, which offered only temporary improvement that relapsed quickly. She was then started on weekly, low-dose LSD.

Almost immediately, her mood elevated and over time, she felt calmer and more stable and her suicidal thoughts diminished. Eventually, treatment was continued in an outpatient setting.

Hopefully, the benefits for this woman continue, but beyond this single case, there is growing evidence of a need not only for new treatments for mental health disorders, but for better treatments.

Psychedelics like LSD, MDMA, psilocybin, and others may fill this need, but there is much yet to understand beyond case studies, anecdotes, and old clinical studies.

soul_ship-14408f7e-c7ce-436a-a263-98789d4ae5e6.jpg


Failing status quo

“I think it really begins with the unmet needs,” says Roger McIntyre, head of University Health Network’s Mood Disorders Psychopharmacology Unit and the Brain and Cognition Discovery Foundation. “There is a tremendous demand for better treatments for mental illness.”

To hammer home his point, he suggests that up to one in four people in society will be affected by a mental illness at some time in their life.

The condition that has received the most attention, in terms of psychedelics research, is depression, he adds, but he also highlights conditions like PTSD and drug and alcohol use disorders.

“Those three alone account for sizeable distress, not just to people who are affected, but also to society at large with respect to the impairment in their function,” he explains.

Diamond Therapeutics founder and CEO Judy Blumstock echoes these sentiments with numbers from a BIO Industry analysis of venture capital spending in the United States from 2007 to 2016.

“You can see that, by far, cancer gets the most funding,” she says. “But if you look at the psychiatric illnesses, the prevalence is far greater. We're looking at something like 15 million versus 45 million, respectively—three times greater prevalence—and yet, literally, [psychiatric illnesses receive] one-twelfth the amount of funding.”

And this relatively anemic funding occurs despite the direct costs to the US healthcare system, she continues, with psychiatric illness at roughly $170 billion versus cancer at about $80 billion.

For McIntyre, a second reason for the renewed interest in psychedelics is simply the age of the current innovations in psychiatric pharmacology—the fact that most of the current drugs reflect treatments developed in the 1950s.

“The mid-1950s was the introduction, for example, of some of the antipsychotics, the antidepressants, some of the pills for anxiety, and the list grows on from there,” he says.

"And even with these drugs," McIntyre presses, "it is a glass half-full, glass half-empty scenario."

“For about 20 percent or so of people who have a mental illness like depression, just as an example, they do very well with the conventional treatments,”
he says. “The symptoms are under control, if not fully eliminated. They get back to work. They get their livelihood back with their family, their kids.”

But that leaves a huge gap of people for whom the current crop of medicines largely don’t work.

“The symptoms continue to be incredibly distressing,”
he continues. “They are not able to function in their personal lives, their family lives, their work lives.”

"And even if the drugs do work,"
he adds, "they often take a considerable time to kick in. It might take four to eight weeks for the maximum therapeutic effect to kick in."

McIntyre then adds another complication. "The rates of suicide, which he says have not decreased in Canada and have actually increased in the United States over the last three decades," he says.

“Suicide is not a mental illness,” he stresses, “but suicide is most often associated with mental illness, like depression. So, it stands to reason that if you treat the mental illness that should help suicide.”

He is quick to acknowledge other modalities of treatment, such as counseling and talk therapy, but suggests that they are not filling the unmet need gap either.

But with six decades of experience, why are the success rates so low?

Psychedelic-trees.jpg


Working in the dark

One of the inherent challenges of psychiatric illness is that they have largely been diagnosed based on symptoms rather than quantifiable molecular markers, as is the evolving case with other neurological conditions like Alzheimer’s disease or Parkinson’s disease.

“One of the ways that psychiatric diagnosis is made is through what's called DSM-5, the fifth in a series of diagnostic and statistics manuals,” says Lyle Oberg, cofounder and CEO of Mynd Life Sciences.

In depression’s case, he continues, if the patient has five of the eight symptoms and they're debilitated for more than two weeks, they are diagnosed with depression.

“There's no test,” Oberg continues. “If you exhibit these symptoms, you have depression.”

This complicates prescribing, he presses, offering the example of the SSRIs, which first entered the market in the 1980s with Eli Lilly’s blockbuster Prozac.

“You've got various SSRIs—just sons of Prozac—and when you combine that with the DSM-5 diagnosis, what you get is basically a trial-and-error type of approach to medicine,” he says. “Someone goes in, they have the classic 5/8 symptoms, they’re diagnosed as depressed, and the doctor says here try this.”

The patient tries the treatment and they come back after three or four months because it didn’t work. Or it did work, but it took that long to know.

“Either way, it's been three or four months that they have been in depression,” Oberg presses. “They've had horrible symptoms, and in many cases, it just doesn't work. And there's no telling exactly why it doesn't work. You're just going by symptoms.”

Not to trivialize the challenge, but Diamond’s head of pipeline development Jeffrey Sprouse recounts with some humor his time working on SSRIs.

“I remember using fluoxetine in the laboratory before the age of Prozac,” he explains. “I was at Pfizer when its SSRI hit the market, and at Lundbeck when their SSRI hit the market. So, I've been trailing this for quite a while.”

“The joke that we always told each other was that all of these drugs would ride their wave of sales and use, fall into generic forms and then fall out of favor,”
he recalls, “and we still wouldn't know how they worked.”

There's always been some working hypotheses, he is quick to add. But the science was challenging, and it was never clear why it took SSRIs so long to work.

“They raise levels of serotonin quickly on the first dose, within minutes,” Sprouse offers. “So, why does it take so long for the therapeutic effects to kick in?”

Part of the challenge to understand the mechanisms of these drugs was the technology available at the time, suggests Joseph Araujo, chief scientific officer and director of Mindset Pharma, as well as president and CEO of InterVivo Solutions.

“Before molecular pharmacology, the assays that were available were phenotypic if we're talking about animal models, where you're looking for patterns in behavior,” he offers. “And a lot of the most promising psychiatric drugs were developed prior to molecular pharmacology.”

Without knowing or by making assumptions on the molecular causes of diseases, he continues, one really limits their bandwidth in developing new drugs.

Given the historical prevalence of clinical data and the relative paucity of molecular understanding with psychedelics, Araujo describes the current interest and approaches as something of a “back to the future” scenario.

“We have the advantage that we now know about the serotonin 5-HT2A target,” he enthuses. “When psilocybin was studied previously, nobody knew about serotonin receptors or which serotonin receptor was important So, it's really a nice circular pattern happening in drug discovery, and I think good timing for it as well, with all of the increased neuropsychiatric needs.”

“The nice thing about 2021 is that there are so many ways to accelerate research,”
says Oberg. “Even from the point of view of knockout mice—that in itself is massive.”

“Before CRISPR, you couldn't do that,”
he stresses. “So that saved five or 10 years of a drug’s identification.”

In recent years, animal models of psychiatric illness and mood disorders have been called into question with suggestions that, at best, behavioral changes seen in rodents and other organisms are poor proxies for human mental processing.

Gabriella Gobbi, head of the Neurobiological Psychiatry Unit at McGill University, has heard these complaints and sees things quite differently. Given her 30 years of experience in animal research and treating patients, she suggests that the drugs that work in patients also work in behavioral animal models and in electrophysiology.

“The main problem of drug discovery in mental health is the lack of scientists and clinicians who have a background to translate basic research into clinical trials,” she explains. “While translational medicine is less challenging in other areas of medicine, in mental health we have to deal with the brain, which is a complex system, and not an organ.”

“Consequently,”
she presses, “the interpretation of data from behavioral animal models, neurobiology, electrophysiology, and molecular biology requires a lot of knowledge and expertise and capacity of integration.”

Likewise, reflecting on his experience, Araujo suggests that there are clear consistencies between changing animal behavior and 5-HT2A agonists, offering head twitch in mice and wet dog shakes in rats as examples when trying to understand if an animal is having a psychedelic experience.

“Ultimately, one of the bigger challenges is to try and understand whether that behavioral effect is due to what you think it's due to, in this case, potentially a hallucinogenic type of experience that maybe the animal is perceiving or maybe not perceiving,” he offers. “As we move along, we can look at some more sophisticated animal models and begin to better understand what those drugs are doing, and maybe benchmark what has been done in the literature in the past.”

He points to drug discrimination experiments, where you train animals to a dose level that you would anticipate has a hallucinogenic-like effect, and then test other compounds to see if the animal responds similarly to those compounds.

“You can also begin to get into models that assess things like cognitive improvements,” he adds, “maybe in a micro-dose situation where you might see some cognitive benefit.”

For Sprouse, it is perhaps less about leveraging new technologies so much as being able to access technologies that existed but were perhaps previously outside of the neuropsychopharmacologist’s price point.

“I was brought up in drug discovery when everything was high-throughput and large-scale,” he recounts. “I think what's changed more recently is the ability to probe fewer molecules and fewer endpoints to actually get to an answer.”

Thus, while acknowledging the shortcomings of early research into SSRIs, he is hopeful looking forward.

“I think, if we were to do this all over again starting today, we'd have the tools to understand them better,” says Sprouse. “But because of their history, we're not spending all that much time in trying to figure it out anymore. I think we'll do a better job with the psychedelics.”

GettyImages-471918252-e1588096013123.jpg


Diamond in the rough

Timing has been a vital factor not just in terms of technological and molecular advances, however, as Blumstock discovered in trying to launch Diamond Therapeutics back in 2006.

Her interest in the field of psychedelics was first piqued by an article in The Economist called “The God Pill,” and from there she found the work of University of Arizona’s Francisco Moreno, who demonstrated some efficacy of psilocybin in a small group of patients with obsessive-compulsive disorder.

Given her experience in the capital markets and her then current position at MaRS Innovation (now Toronto Innovation Acceleration Partners), 2006/2007 seemed like a good time to explore this market with the launch of Diamond. She was wrong.

“It was just not the right time,” Blumstock acknowledges in retrospect. “But something was nevertheless burning away at me. Why can't we do this?”

“This is something that should be done,”
she recalls. “And then finally, in late 2017, it seemed like now was the right time to launch Diamond properly.”

Following that initial enthusiasm, the company focused its efforts on psilocybin.

“With psilocybin, there's a great deal of research out there already, and it seems like an ideal starting point for drug discovery and drug development, with at least one obvious liability, which is the hallucinogenic side of it,” she explains.

“When we started the company, we had active debate about psilocybin as a known molecule versus an NCE [new chemical entity] program, which is more of a typical pharma program,” she continues. “And we just decided we wanted to have our cake and eat it, too.”

“We just determined that psilocybin was such an amazing gift to be given, that we understand so much about it already,”
she presses. “So, we are trying to explore both and understand as much as we can about psilocybin in the hope that it guides us to novel drugs.”

“Psilocybin obviously is of tremendous importance to Diamond,”
adds Sprouse. “Even though it's a small molecule—a simple molecule—it's going to have multiple actions, and we know from this huge body of usage that there's some therapeutic value there.”

“But we also want to think about other molecules, because I think we're going to learn a lot along the way,”
he offers. “We're going to learn a lot about patient response and the ideal patient. The ability to essentially map drug to patient is something that we're going to give very serious thought to.”

Sprouse also acknowledges the commercial and intellectual property reasons to press for NCEs, but for him at least, this consideration is secondary.

“The thing that is most motivating is the thinking that small differences that you might observe in a molecule you're patterning using psilocybin as a parent may end up being huge in terms of patient benefit,” he suggests.

Similarly recognizing the potential of chemical diversity is Mindset Pharma.

src.adapt.960.high.shulgin_060414.1401920536489.jpg


Changing mindset

Like Diamond, Mindset is focused on psilocybin as a starting point.

“Using a whole range of different strategies, we've created a number of different families of new drugs with interesting characteristics that could make them really well suited to a number of different use cases and indications,” explains company CEO James Lanthier.

"And a key part of developing those compound families," he continues, "is the company’s focus on developing synthetic processes to generate not only psilocybin and myriad derivatives, but also completely novel compounds." As he explains, "there is a major need in the market for a more efficient, lower-cost method to produce GMP-grade psilocybin."

“We're really focused on preclinical development, and from a business strategy standpoint with all the capital rushing into the space and the regulatory momentum, we felt like there was going to be a real demand for next-generation drugs that were really more optimized and that could enjoy full patent protection,”
Lanthier explains. “In comparison, a lot of the development efforts by groups in the space had focused on classic drugs and a lot of those on formulations of the classic drugs.”

“We knew when we initially started that in order to differentiate Mindset’s psychedelics program from competitors in the field, we had to be creative and let the science drive the company’s innovations to create novel, patentable NCEs and thereby a solid IP [intellectual property] portfolio,”
echoes Mindset chief scientific adviser Malik Slassi. “From the beginning, this was the first thing we did. We had to come up with novel patentable small-molecule psychedelic drug candidates.”

Araujo elaborates on the company’s three families of compounds, which reflect Mindset’s decision to let the science drive the clinical indication rather than focus initially on a psychiatric target.

“The first [family] can be divided into deuterated compounds and prodrugs, which we feel could rapidly be developed to clinical candidates and really present as quick next-generation psilocybin-like compounds,” he explains. “They're going to be very similar to psilocybin with respect to drug characteristics while potentially reducing some of the metabolic liability.”

The second family works from a different chemical scaffold with a restricted side chain, he continues, adding that in-vitro and in-vivo tests suggest many of the compounds have a greater effect size than psilocybin.

“We're trying to optimize those for shorter durations of action, which we think would be really well suited for a psychedelic-assisted psychotherapy,” he offers.

Mindset’s least mature category, he presses, includes compounds with lower effect size at the 5-HT2A receptor, but with extended half-lives, suggesting potential for a much longer duration of action.

“Those compounds, we think, fit an ideal profile for potential micro-dosing applications, particularly in patient populations that might be compromised, or where you don't want them to potentially experience hallucinogenic-type effects—juvenile ADHD and geriatric Alzheimer's disease patients come to mind,” he says.

Slassi reinforces that having such a diversity of candidates really allows the company to stay open to indications beyond depression and anxiety disorders. And the growing volumes of in-vitro and in-vivo data as well as the IP are attracting interest from Big Pharma and biotechs.

Not everyone in this field, however, sees 5-HT2A as their primary target.

2B25EC4700000578-3370542-image-a-22_1450800312717.jpg


A new model in mind

Mynd Life Sciences is taking a different approach to psychedelics development in mental health. Rather than focus on the impact of compounds like psilocybin on serotonin receptors, the company is instead pursuing links between depression and inflammation.

The inflammatory model has gained traction over the past few years. Indeed, earlier studies of anti-TNF-based therapies commonly given modulate autoimmune disorders such as rheumatoid arthritis and psoriasis have shown therapeutic benefits when aimed at depression.

“There was a correlation between rheumatoid arthritis and depression,” says Oberg, “and people used to think well, ‘I'm depressed because I have rheumatoid arthritis.’

“In actual fact, what they're now seeing is that it's the same process that causes the depression as causes the rheumatoid arthritis. And that process is looking more and more like a chronic inflammatory process.”


A large-scale study of patients in the United States, he further explains, identified the presence of a common gene: the human mycogene.

“This gene is the switch that turns the body from a pro-inflammatory state into an anti-inflammatory state,” Oberg says. “It turns macrophages from M1 to M2, and it's a trigger gene.”

In a study published in 2013, Timothy Powell and colleagues at King’s College London performed a transcriptomic analysis of the inflammatory cytokine pathway in patients receiving the SSRI escitalopram for major depressive disorder. They found that the pathway regulator ABCF1 (human mycogene) was upregulated following SSRI administration and that the effect was larger in patients classified as responders than in non-responders.

Following data such as this, Mynd is using psilocybin as a starting point and to date, has generated 38 analogues, which it hopes to winnow down to three or four candidates for preclinical screening and optimal anti-inflammatory activity.

Further facilitating their understanding of the disease pathology, the identification of a gene target allows them to engineer knockout rodents to determine what effect, if any, the presence or absence of the gene has on animal behavior and response to treatment.

hallucinations-drug-abuse_f_600x250.jpg


'Easy dose it'

Much as with any drug development program, the goal is to improve efficacy while minimizing side effects. Thus, as is seen in much of the clinical development of cannabinoids, one could easily imagine a desire to remove the psychoactive or hallucinatory components of psychedelics while retaining psychiatric efficacy.

There is a debate, however, as to whether—at least in some cases—the psychedelic experience is part of the healing.

“I keep hearing that mysticism experiences are critical to the therapeutic effect, that you have to have a trip to have the therapeutic effect, there has to be some type of out of body experience or whatever,” recalls McIntyre. “I don't know that's true.”

That said, he understands how a patient might confound a psychedelic experience with improvement in depressive symptoms. And it could be that the two are linked, but in biological terms rather than psychological terms; that is, the “trip” may be symptomatic of the neural rewiring that leads to psychiatric benefit.

“The theoretical and prevailing view of psychedelics is that what we're effectively doing is disconnecting and reconnecting the circuits,” McIntyre explains. “And when you disconnect/reconnect the circuits in the brain, that process critical to the benefit may also be what's subserving the psychedelic experience.”

He offers a comparison with a faulty computer. Remove and replace some wires to reset the computer, but he questions what the computer “experiences” as its screen flashes and components power off and on.

From a pharmacological perspective, Sprouse wonders if the question isn’t moot, suggesting that he sees no merit in fixating solely on the psychedelic dose. Instead, he wants to understand the dose response curve.

In practical terms, different compounds almost assuredly require different doses for different individuals in different indications.

“Maybe there's the need for a psychedelic effect in one patient group, but not another,” he says. “I think there's some richness here that warrants examination.”

Efforts to avoid the psychedelic effect have led many to explore the concept of micro-dosing, a term of which Sprouse is not fond. Typically, it is considered one-tenth the minimum dose that induces the psychedelic experience.

“The real challenge is there are very few studies that have been well designed that have really examined micro-dosing,” says Araujo. “There's a lot of anecdotal information that's really difficult to draw conclusions from.”

That said, he notes that some groups have initiated placebo-controlled clinical trials evaluating micro-dosing.

Diamond, for one, will shortly publish results from their efforts to evaluate low-dose ketamine and low-dose psilocybin in animal models. And the company recently initiated a collaboration with McGill’s Gobbi, who has been studying low-dose LSD.

In late January, Gobbi and colleagues published their effort to understand the molecular basis of low-dose LSD’s influence on social behavior, which mimics the increased empathy seen in human subjects.

“The key finding of this paper was that low doses of LSD enhance social behavior in mice,” explains Gobbi. “By binding the 5-HT2A receptor, LSD indirectly enhances the AMPA neurotransmission by also promoting mTOR complex phosphorylation and plasticity in the glutamatergic neurons of the prefrontal cortex.”

The findings are critical to the use of LSD to treat psychiatric illnesses because at higher doses, the compound becomes a dopaminergic agonist and likely increases the risk of psychotic-like symptoms.

“Moreover, it suggests that drug discovery research should focus on designing highly selective 5-HT2A agonists with a wider therapeutic window and higher safety profile,” she adds.

For the most part, Araujo echoes Sprouse’s openness to the possibilities.

“What about a molecule that doesn't necessarily activate the receptor to the same extent?” he asks. “Is that a micro-dose effect at a higher dose?”

“Again, I think we'll have to follow the science, understand what the effect is, and what the right dose is for the right compound,”
he explains. “Ultimately, I think, there are going to be patients for whom, for whatever reason, a macro-dose or a large dose or a hallucinogenic experience might not be the right approach, and so there needs to be more than one option for different patients.”

Any hope of answering these questions, however, will come from a deeper understanding of the science behind these compounds and their effects.

“In my career, I've seen a number of very promising medicines—not in the psychedelic area but in the more conventional pharmacotherapeutic area—that have come along, and these drugs unfortunately did not demonstrate superiority over the placebo in the clinical trial,” he recounts. “And there's two interpretations of that.”

One is simply that the drug did not work as expected, which is unfortunate but just a part of doing business. The other possibility, he suggests, is that the drug did work, but that the trials weren’t designed properly.

This is where McIntyre sees an opportunity to move forward from the studies of the past.

“If you really want to understand the efficacy and the safety, you have to have adequate studies,” he says. “And what I've noticed about the psychedelic space in general, there is heterogeneity in the way these studies are being done.”

“I'm all for heterogeneity and creativity, but sometimes you just want to keep it simple,”
he suggests. “My concern is that if these drugs are really going to help people, we could spoil the broth so to speak, because we're not adhering to good clinical practice research parameters.”

In the absence of such a considered approach, there is a risk that we will end up with even more interesting data, but no better understanding of the potential for psychedelics in mental health.

Gateway drugs

Ultimately, the success of the renaissance of psychedelics research will hinge on the clinical outcomes and improvements in patients’ lives. Many, however, point to other factors that helped launch this renewed interest, with cannabis leading the way.

“When you take a look at the people involved in the psychedelic research, a lot of them came from cannabis,” explains Lyle Oberg, founder and CEO of Mynd Life Sciences. “You have a lot of people that made money in the cannabis industry in Canada who have now migrated into the psychedelic area, which in itself makes a large amount of research money available.”

“People saw that cannabis worked,”
he adds. “They saw the analogies between psychedelics and cannabis in that both of them were working. It became a very easy stretch to take a look at cannabis and psychedelics.”

“The success of cannabis in Canada maybe allowed people to look at these compounds with a more liberal view,”
echoes Judy Blumstock, founder and CEO of Diamond Therapeutics, addressing the rapid growth of drug discovery companies exploring the psychedelic space north of the US border. “But I think there's very strong interest in the US, as well, and worldwide.”

Joseph Araujo, chief scientific officer and director at Mindset Pharma as well as president and CEO of InterVivo Solutions, recognizes a Canadian effect in terms of both social and regulatory acceptance of cannabis.

“What's happened with cannabis, I think, has really paved the path forward for looking at controlled drugs in unique ways and with the goal of medicinal applications,” he says.

“Interestingly, the hallucinogens are Schedule Three in Canada, whereas in most of the world, they’re Schedule One,” he adds. “So that might be another influence factor that has made maybe research easier in Canada.”

For his part, Blumstock’s colleague and head of pipeline development Jeffrey Sprouse also sees inspiration beyond the cannabis experience.

“I think there's a story that you can tell with the cannabinoids, with cannabis, but I actually prefer the story that you could construct with ketamine,” he says.

“I've been interested in psychiatry targets for years—antidepressants, anxiolytics, etc.—and once you get beyond the SSRIs, it gets very quiet,” he explains. “It is this desert and without really much hope of improvement.”

And then ketamine came along.

“Research done right around 2000 by a group at Yale led by Rob Berman uncovered this amazing response in depressed patients,” Sprouse recounts. “Not only was it durable and a significant effect, but it was one that occurred rather quickly, which was always the Holy Grail of antidepressive research.”

Unlike SSRIs, he says, which are useful but can take upwards of 12 weeks to see any effect, ketamine offered its effects rapidly and once patients moved through the K-hole, they began to feel much better.

“That reset the table for everybody,” he enthuses. “Suddenly, there was a direction to move, a different mechanism, one that you could link to downstream pathways from serotonin.”

McGill University’s Gabriella Gobbi, who recently signed a collaborative research agreement with Diamond Therapeutics, agrees with the importance of this discovery.

“The approval of the derivate of ketamine, which belongs to the non-classical hallucinogens category, for the treatment-resistant depression encouraged the exploration of other hallucinogenic drugs in mental health,” she says.

"Suddenly," Sprouse continues, "the treasure trove of anecdotal information about psychedelics takes on a whole new significance, so why not broaden the thinking beyond ketamine to other psychedelics?"

“Some of the research that Diamond has sponsored is more or less focused on commonalities between ketamine-like molecules, the dissociative anaesthetics, with the psychedelics like psilocybin and LSD,”
he explains.

So, the old concerns that cannabis was a gateway to other drugs may have been true; just not in the way everyone thought.

 
Last edited:
2-spines.fw_-1024x512.png



Psilocybin and Neurogenesis: What are the long-term effects of magic mushrooms on your brain?*

by Patrick Smith | EntheoNation

Neurogenesis and neuroplasticity are two terms you might have heard before – especially if you’ve been learning about psychedelics.

Our brains have the potential to change and grow over the course of our lives, and if they are more “plastic” in later life we are less likely to suffer from depression, and we may find it easier to learn new things. Psychedelics have been shown to have the potential to boost this neuroplasticity in our brains.

Psilocybin, the main psychedelic ingredient of magic mushrooms, has an important link to neurogenesis and neuroplasticity. An important study shows that psilocybin can influence neuroplasticity in rodent brains – but the direction of the effect depends on dosage.

Since microdosing with magic mushrooms is one of the easiest ways to bring psilocybin into your daily life, could the neuroplastic effects of magic mushrooms be most effective and accessible in small doses? We investigate here…

What are neurogenesis and neuroplasticity?

Neurogenesis
is the specific term for the growth of new brain cells – but it falls under the umbrella of neuroplasticity. Neuroplasticity is the term used to describe pretty much any change in the way brain cells (neurons) connect to each other, grow, and communicate.

There are six main forms of neuroplasticity (including neurogenesis):​
  1. Changes in the size of individual neurons​
  2. Changes in the number of connections between neurons​
  3. Changes in where neurons connect to each other​
  4. The growth of brand new neurons (neurogenesis)​
  5. Changes in the strength of the connections between neurons (also known as synaptic plasticity)​
  6. Changes in the response of neurons to signals​
Although all brains are capable, to some extent, of neuroplasticity – most of the changes in the brain happen in childhood. Our adult brains stay pretty stable. This is why boosts in neuroplasticity in adulthood are special, and usually have pretty healing effects!

Neurogenesis%20Article.jpg


What are the benefits of neurogenesis and neuroplasticity?

Although research into neuroplasticity is still in its infancy, we know some of the benefits of increases in neuroplasticity in adult mammals.

Loads of evidence suggests that neuroplasticity in one particular area of the brain, the hippocampus, is crucial for learning and memory. When both humans and rodents are learning tasks, or remembering patterns, the hippocampus is very active – and synaptic plasticity (changes in the strength of the connections between neurons) seems to be a crucial part of this.

There’s also a strong connection between neuroplasticity and the fight against depression. People with depression are more likely to have signs of reduced numbers of neurons and connections between neurons across different areas of the brain. This is thought to be how some of the most common antidepressants work; by boosting neuroplasticity.

In general, the research into neuroplasticity suggests that having a brain capable of changing its structure and activity may help you adapt more quickly to new information, and be more flexible – skills that are crucial in psychological wellbeing.

Pursuits such as exercise and meditation have been proven to boost neuroplasticity, and changing to a diet that includes less sugar and fat, and superfoods like walnuts and blueberries, has been shown to increase neuroplasticity in rodents.

Paul-Stamets-2020-1280x720-1.jpg

Paul Stamets

Psilocybin, neurogenesis and neuroplasticity

As well as changes to your lifestyle, psychedelics can also influence neuroplasticity. Studies using both psilocybin and other common psychedelics have shown how they can change the connectivity and growth of neurons.

LSD and DMT have been shown to help rat neurons create more complex connections between each other, and the psychoactive components of the ayahuasca vine have been shown to boost the growth of mouse neurons.8,9 But the results with psilocybin aren’t as clear cut…

One study in mice found that small doses (0.1mg/kg) of psilocybin increased the growth of new brain cells in the hippocampus, while high doses (1mg/kg) reduced this growth.10 Although we don’t know for sure that something similar is happening in humans, it suggests that dosage could be really important if you’re using psychedelics to boost neuroplasticity!

However, while small doses of psilocybin may be better at boosting the growth of brain cells, we know that larger doses are effective at changing the way neurons communicate. Studies show that typical tripping doses of psilocybin allows neurons to connect with each other along unusual pathways,11 and then reset themselves into a more stable and healthy configuration after the trip.12 This kind of neuroplasticity is much more about the patterns of connection throughout the brain, rather than the growth of new cells in particular areas – but it can be particularly important for the fight against depression.

How to use psilocybin to boost your neuroplasticity

From the research we have so far, it seems that small doses of psilocybin are more likely to boost neurogenesis (the growth of new brain cells), while larger doses have a greater effect on the connectivity between brain cells. While both of these types of neurogenesis and neuroplasticity could likely impart benefits such as increased creativity, reduced depressive symptoms, and boosted psychological flexibility, it’s hard to know which is better.

There is plenty of evidence that shows a single moderate or high dose of psilocybin can have therapeutic effects, especially in the treatment of depression and anxiety. So if you’re looking for an immediate and powerful boost to neuroplasticity, a magic mushroom retreat or ceremony where larger doses are used might be your best option.

Conversely, there is barely any research into the effects of microdosing with magic mushrooms, so we don’t know for sure the role of neurogenesis or neuroplasticity with small doses. Taking what we can from the study on mice, we know that doses closer to a microdose (around 1-2mg of psilocybin) seem to boost neurogenesis more than larger doses – it’s just hard to know what this means for humans, and whether a microdosing regimen will be overall more beneficial for neuroplasticity than a single large dose.

Despite the lack of evidence, what we do know is how to amplify any benefits to neuroplasticity if you’re microdosing with magic mushrooms…

How to microdose with magic mushrooms for neuroplasticity

If you’re new to microdosing with magic mushrooms, check out our Seeker’s Guide for an introduction. But if you want to microdose with magic mushrooms specifically for neuroplasticity, you should also try to combine your microdosing with some additional practices.

There’s a lot of evidence connecting wellbeing practices with neuroplasticity, so to maximize any neuroplasticity benefits you should incorporate these things as much as you can:​
  • Yoga and exercise. Increases in neuroplasticity could help you learn new exercise routines faster than normal, and help them remain a permanent fixture on your calendar.​
  • Meditation or regular affirmation (prayer). You may find meditation easier to learn if magic mushrooms are giving you a boost in neuroplasticity, and meditation has also been proven to increase neuroplasticity in itself.​
  • Healthy dieting. Microdosing may be the best time to try out a new, healthier diet, as you may find it easier to stick to – and switching away from high-fat high-sugar diets has been shown to increase neuroplasticity.​
  • Therapy or counseling. If a boost in neuroplasticity is helping improve your psychological flexibility, now may be the perfect time to get professional guidance in how to best apply any increased capacity for self-reflection.​
Making these pursuits part of your microdosing routine could not only help to maximize any boost in neuroplasticity that psilocybin gives you, but the effects of microdosing could also help you adapt to these lifestyle changes much faster than normal! Many people report that microdosing helps them enjoy exercise more, or actually get joy from changing to a healthier diet.

As always, when microdosing with magic mushrooms for neuroplasticity you should be keeping a journal to help monitor any effects and find your perfect dose. You should stop if you feel any negative effects, and even if you are feeling great during your regimen you should never microdose for more than three months at a time.

*From the article here :
 
Last edited:
macnider-bg.png

UNC-Chapel Hill School of Medicine

Psilocybin’s rewiring of the brain, study

by Barbara E. Bauer, MS | Psychedelic Science Review | 22 Jul 2021

Changes in structural neuroplasticity start within 24 hours, are enduring, and may not be solely dependent on 5-HT2A.

The ability of psychedelics to alter structural and functional neuroplasticity in the brain is a significant area of exploration for labs around the world. Researchers are teasing apart the fine details of the mechanisms by which compounds like psilocybin (the prodrug of psilocin found in magic mushrooms) can relieve anxiety, depression, and other symptoms while maintaining their effects for weeks after administration. Scientists are continuing to learn how psilocybin brings on its therapeutic effects by altering the structure and function of brain cells.

A psilocybin study recently published by Dr. Lingxiao Shao and her research team in bioRxiv1 is catching the attention of prominent psychedelic researchers. For example, Dr. Bryan Roth of the Department of Pharmacology at UNC-Chapel Hill School of Medicine said on Twitter, “This is potentially a landmark paper in #psychedelics research!”

Unanswered questions about psilocybin and neuroplasticity

Dr. Lingxiao Shao’s research team set out to fill two critical gaps regarding psilocybin that they identified in the literature: 1) No one had demonstrated that psilocybin changes structural plasticity at the cellular level in a mammalian brain, and 2) it is unknown how long it takes for psilocybin to elicit its neuronal effects in vivo.

Shao et al. used techniques including two-photon microscopy and the head twitch response (HTR) to observe, among other things, the effects of one dose of pure psilocybin on dendritic spine growth in layer 5 pyramidal neurons in the mouse medial frontal cortex. They also utilized the 5-HT2A receptor antagonist ketanserin to test whether the receptor is involved in the brain structural plasticity brought on by psilocybin.

Study results

Using five different doses of psilocybin in the HTR test, Shao et al. corroborated the work of Halberstadt et al.2 and Sherwood et al.,3 finding a sharp rise in HTRs at a dose of 1 mg/kg. They also observed that the number of HTRs peaked 6-8 minutes after administration. The HTRs gradually declined and stopped in about 2 hours. The researchers used the 1 mg/kg psilocybin or saline (control) dose in subsequent experiments on structural plasticity. This paper contains a treasure trove of fascinating findings and corroborations of other work. Below are just some of the observations reported by Shao et al.

Increased dendritic spine density, head width, and growth rate

The research team tracked the size and growth of “1,820 dendritic spines on 161 branches from 12 animals including 6 males and 6 females” before and after a 1 mg/kg intraperitoneal (i.p.) dose of psilocybin or saline. Statistical analysis found significant changes in the dendritic spine density and head width over time (Figure 1).​
  • +7 increase in spine density on Day 1​
  • +12 increase on Day 7​
  • Main effect of treatment, P = 0.011​
  • +11 increase in spine head width on Day 1​
  • +5 increase on Day 7​
  • Main effect of treatment, P = 0.013​

Dr. Shao and her team wondered if the increased spine density meant that more of them were forming or if the rate of their elimination was slowed by psilocybin. So, they calculated the turnover rate of the dendritic spines and, from that, made the following observations from the experimental data (Figure 2).​
  • Psilocybin caused an 8+/-2% increase in the spine formation rate in female mice.​
  • Psilocybin caused a 4+/-2% increase in spine formation rate in males.​
  • The data showed no changes in the rate of spine elimination.​

Interestingly, the peak rate of spine formation occurred shortly after the mice received the single psilocybin dose. The formation then steadily decreased over time, eventually returning to equilibrium with the spine elimination rate. The authors concluded...
These data therefore support the view that the long-term increase in spine density is due to an initial boost of enhanced spine formation.

Some dendritic spines persisted one month after psilocybin

Shao et al. kept watching the newly formed dendritic spines for several weeks. They found about half of them were still stable by Day 7 after psilocybin administration (Figure 3). From this, they commented, “This suggests that a portion of the new dendritic spines produced by psilocybin would become functional synapses.” They added...
Intriguingly, select individual dendritic branches appeared to retain all the new spines, while other branches lost them almost completely, suggesting heterogeneity and potentially responsive and nonresponsive subpopulations of pyramidal neurons.

Further, they looked for the spines formed after psilocybin administration on Day 1 in four mice after 34 days had passed (Figure 3). They found that 34+/-10% of the spines persisted in females and 37+/-12% in males. Shao et al. commented that these results correlate with clinical trial findings that the effects of psilocybin are long-lasting.

Ketanserin revealed more details about 5-HT2A

In another aspect of the paper, Shao et al. investigated if the 5-HT2A receptor is related to psilocybin’s effects on structural plasticity in the mouse brain. They pretreated mice with 1 mg/kg ketanserin i.p. ten minutes before dosing them with 1 mg/kg psilocybin or saline and doing HTR testing. As expected, ketanserin stopped the HTR completely.

Next, the research team watched “1,443 dendritic spines on 120 branches from 8 animals including 4 males and 4 females” pretreated with ketanserin and dosed with psilocybin as described above. Using the two-photon imaging, they observed that dendritic spine density was no longer statistically significant under ketanserin. However, they did see increases in the width of spine heads, spine protrusion length, and spine formation rate on Day 1. The authors summarized the findings by saying...
Our results demonstrate that while a moderate knockdown of 5-HT2A receptor function eliminates head-twitch responses, it is not sufficient to abolish the psilocybin-induced structural remodeling in mice.

However, the authors noted that “our results do not rule out the involvement of 5-HT2A receptors because this dose of ketanserin only blocks ~30% of 5-HT2A receptors in rodents and the unaffected receptors might be enough to drive the dendritic remodeling.”

Differences between sexes

There is another interesting aspect of psilocybin pharmacology that this study brings to light. Dr. Shao and her team looked for sex differences in the data. As mentioned earlier, female mice exhibited twice the rate of spine formation after psilocybin administration than males (+8 percent vs. +4 percent). Also, the data showed that one month after a single dose of psilocybin, females retained 34% of the new dendritic spines they grew while males retained 37% (Shao et al. captured more sex-related data which will be featured in an upcoming Psychedelic Science Review article). These similarities and differences between the sexes present intriguing new questions for psychedelic researchers to answer.

Summary

In this recent work, Shao et al. demonstrated “that a single dose of psilocybin evokes growth of dendritic spines in the medial frontal cortex of the mouse.” Integrating the findings from other studies, they explain why this study is important for understanding the therapeutic effects of psilocybin at the cellular level. Also, from their HTR and ketanserin findings, they proposed that “Future studies with region- and cell-type-specific knockout of serotonin receptor subtypes are needed to produce more decisive evidence on the role of 5-HT2A and other receptors in mediating the effects of psilocybin on dendritic plasticity.”

Finally, Shao et al. noted the following regarding their findings on the time course of psilocybin effects.
By showing that the time course for psilocybin-induced structural remodeling is rapid and persistent in vivo, our study suggests that synaptic rewiring may be a mechanism shared by compounds with rapid antidepressant effects.

Regarding the different effects Shao et al. observed in male and female mice, Psychedelic Science Review has published several articles on women and psychedelics including “Psychedelics and Women’s Health,” “Female Hormones, 5-HT2A Receptors, and Psychedelics,” and “2005 Literature Review Proposes Serotonin Mediates the Effects of Estrogen.” The unique effects of psychedelic drugs on women present a whole new world for research. It is reasonable to postulate that a women’s unique physiology requires different therapeutic formulations than what works for men. Is there a uniquely female entourage effect with psychedelics?

Shao et al. cautioned that, although humans and mice are both mammals, mouse brains have fundamental differences from human brains, including receptors. Thus, it is unclear at this time if these results can be extrapolated to humans.

Barbara Bauer, MS

Barb is Editor and one of the founders of Psychedelic Science Review. Her goal is making accurate and concise psychedelic science research assessable so that researchers and private citizens can make informed decisions.

*From the article here :
 
Last edited:
mescaline-like-psychedelic-alters-synaptic-plasticity-356210-1280x720.webp



DOI found to alter synaptic plasticity*

Virginia Tech | TECHNOLOGY NETWORKS | 25 Nov 2021

A new study suggests that a mescaline-like psychedelic called DOI can cause alterations to epigenomic markers and synaptic plasticity in the mouse brain after a single dose.

One in five U.S. adults will experience a mental illness in their lifetime, according to the National Alliance of Mental Health. But standard treatments can be slow to work and cause side effects.

To find better solutions, a Virginia Tech researcher has joined a renaissance of research on a long-banned class of drugs that could combat several forms of mental illness and, in mice, have achieved long-lasting results from just one dose.

Using a process his lab developed in 2015, Chang Lu, the Fred W. Bull Professor of Chemical Engineering in the College of Engineering, is helping his Virginia Commonwealth University collaborators study the epigenomic effects of psychedelics.

Their findings give insight into how psychedelic substances like psilocybin, mescaline, LSD, and similar drugs may relieve symptoms of addiction, anxiety, depression, and post-traumatic stress disorder. The drugs appear to work faster and last longer than current medications — all with fewer side effects.

The project hinged on Lu’s genomic analysis. His process allows researchers to use very small samples of tissue, down to hundreds to thousands of cells, and draw meaningful conclusions from them. Older processes require much larger sample sizes, so Lu’s approach enables the studies using just a small quantity of material from a specific region of a mouse brain.

And looking at the effects of psychedelics on brain tissues is especially important.

Researchers can do human clinical trials with the substances, taking blood and urine samples and observing behaviors, Lu said. “But the thing is, the behavioral data will tell you the result, but it doesn’t tell you why it works in a certain way,” he said.

But looking at molecular changes in animal models, such as the brains of mice, allows scientists to peer into what Lu calls the black box of neuroscience to understand the biological processes at work. While the brains of mice are very different from human brains, Lu said there are enough similarities to make valid comparisons between the two.

VCU pharmacologist Javier González-Maeso has made a career of studying psychedelics, which had been banned after recreational use of the drugs was popularized in the 1960s. But in recent years, regulators have begun allowing research on the drugs to proceed.

In work by other researchers, primarily on psilocybin, a substance found in more than 200 species of fungi, González-Maeso said psychedelics have shown promise in alleviating major depression and anxiety disorders. “They induce profound effects in perception,” he said. “But I was interested in how these drugs actually induce behavioral effects in mice.”

To explore the genomic basis of those effects, he teamed up with Lu.

In the joint Virginia Tech - VCU study, González-Maeso’s team used 2,5-dimethoxy-4-iodoamphetamine, or DOI, a drug similar to LSD, administering it to mice that had been trained to fear certain triggers. Lu’s lab then analyzed brain samples for changes in the epigenome and the gene expression. They discovered that the epigenomic variations were generally more long-lasting than the changes in gene expression, thus more likely to link with the long-term effects of a psychedelic.

After one dose of DOI, the mice that had reacted to fear triggers no longer responded to them with anxious behaviors. Their brains also showed effects, even after the substance was no longer detectable in the tissues, Lu said. The findings were published in the October issue of Cell Reports.

It’s a hopeful development for those who suffer from mental illness and the people who love them. In fact, it wasn’t just the science that drew Lu to the project.

For him, it’s also personal.

"My older brother has had schizophrenia for the last 30 years, basically. So I've always been intrigued by mental health,” Lu said. “And then once I found that our approach can be applied to look at processes like that — that's why I decided to do research in the field of brain neuroscience."

González-Maeso said research on psychedelics is still in its early stages, and there’s much work to be done before treatments derived from them could be widely available.

Reference:

Revenga M de la F, Zhu B, Guevara CA, et al. Prolonged epigenomic and synaptic plasticity alterations following single exposure to a psychedelic in mice.
Cell Reports. 2021;37(3). doi:10.1016/j.celrep.2021.109836

 
Last edited:
ba0f343239d0e2d4ce6f0d6ad04202c9.jpg



How psychedelics bind to the 5-HT2A serotonin receptor

University of North Carolina | Science News | 17 Sep 2020

For the first time, scientists solved the high-resolution structure of these compounds when they are actively bound to the 5-HT2A serotonin receptor on the surface of brain cells. This discovery is already leading to the exploration of more precise compounds that could eliminate hallucinations but still have strong therapeutic effects. Psilocybin - the psychedelic compound in mushrooms - has already been granted breakthrough status by the FDA to treat depression.

Psychedelic drugs such as LSD, psilocybin, and mescaline cause severe and often long-lasting hallucinations, but they show great potential in treating serious psychiatric conditions, such as major depressive disorder. To fully investigate this potential, scientists need to know how these drugs interact with brain cells at the molecular level to cause their dramatic biological effects. Scientists at UNC-Chapel Hill and Stanford have just taken a big step in that direction.

For the first time, scientists in the UNC lab of Bryan L. Roth, MD, PhD, and the Stanford lab of Georgios Skiniotis, PhD, solved the high-resolution structure of these compounds when they are actively bound to the 5-HT2A serotonin receptor (HTR2A) on the surface of brain cells.

This discovery, published in Cell, is already leading to the exploration of more precise compounds that could eliminate hallucinations but still have strong therapeutic effects. Also, scientists could effectively alter the chemical composition of drugs such as LSD and psilocybin -- the psychedelic compound in mushrooms that has been granted breakthrough status by the FDA to treat depression.

"Millions of people have taken these drugs recreationally, and now they are emerging as therapeutic agents," said co-senior author Bryan L. Roth, MD, PhD, the Michael Hooker Distinguished Professor of Pharmacology at the University of North Carolina School of Medicine. "Gaining this first glimpse of how they act at the molecular level is really important, a key to understanding how they work. Given the remarkable efficacy of psilocybin for depression (in Phase II trials), we are confident our findings will accelerate the discovery of fast-acting antidepressants and potentially new drugs to treat other conditions, such as severe anxiety and substance use disorder."

Scientists believe that activation of HTR2A, which is expressed at very high levels in the human cerebral cortex, is key to the effects of hallucinogenic drugs. "When activated, the receptors cause neurons to fire in an asynchronous and disorganized fashion, putting noise into the brain's system," said Roth, who holds a joint faculty appointment at the UNC Eshelman School of Pharmacy. "We think this is the reason these drugs cause a psychedelic experience. But it isn't at all clear how these drugs exert their therapeutic actions."

In the current study, Roth's lab collaborated with Skiniotis, a structural biologist at the Stanford University School of Medicine. "A combination of several different advances allowed us to do this research," Skiniotis said. "One of these is better, more homogeneous preparations of the receptor proteins. Another is the evolution of cryo-electron microscopy technology, which allows us to view very large complexes without having to crystalize them."

Roth credits co-first author Kuglae Kim, PhD, a postdoctoral fellow in his lab, for steadfastly exploring various experimental methods to purify and stabilize the very delicate serotonin receptors.​

"Kuglae was amazing," Roth said. "I'm not exaggerating when I say what he accomplished is among the most difficult things to do. Over three years in a deliberate, iterative, creative process, he was able to modify the serotonin protein slightly so that we could get sufficient quantities of a stable protein to study."

The research team used Kim's work to reveal the first X-ray crystallography structure of LSD bound to HTR2A. Importantly, Stanford investigators then used cryo-EM to uncover images of a prototypical hallucinogen, called 25-CN-NBOH, bound together with the entire receptor complex, including the effector protein Gαq. In the brain, this complex controls the release of neurotransmitters and influences many biological and neurological processes.

The cryo-EM image is like a map of the complex, which Kim used to illustrate the exact structure of HTR2A at the level of amino acids -- the basic building blocks of proteins such as serotonin receptors.

Roth, a psychiatrist and biochemist, leads the Psychoactive Drug Screening Program, funded by the National Institute of Mental Health. This gives his lab access to hallucinogenic drugs for research purposes. Normally, these compounds are difficult to study in the lab because they are regulated by the Drug Enforcement Agency as Schedule 1 drugs.

Roth and colleagues are now applying their findings to structure-based drug discovery for new therapeutics. One of the goals is to discover potential candidates that may be able offer therapeutic benefit without the psychedelic effects.

"The more we understand about how these drugs bind to the receptors, the better we'll understand their signaling properties," Skiniotis says. "This work doesn't give us the whole picture yet, but it's a fairly large piece of the puzzle."

*From the article here :
 

ysm_EricWang_photoeditor.jpg


Psilocybin spurs growth of neural connections lost in depression*

by Bill Hathaway | YaleNews | 5 Jul 2021

The psychedelic drug psilocybin, a naturally occurring compound found in some mushrooms, has been studied as a potential treatment for depression for years. But exactly how it works in the brain and how long beneficial results might last is still unclear.

In a new study, Yale researchers show that a single dose of psilocybin given to mice prompted an immediate and long-lasting increase in connections between neurons. The findings are published July 5 in the journal Neuron.

“We not only saw a 10% increase in the number of neuronal connections, but also they were on average about 10% larger, so the connections were stronger as well,” said Yale’s Alex Kwan, associate professor of psychiatry and of neuroscience and senior author of the paper.

Previous laboratory experiments had shown promise that psilocybin, as well as the anesthetic ketamine, can decrease depression. The new Yale research found that these compounds increase the density of dendritic spines, small protrusions found on nerve cells which aid in the transmission of information between neurons. Chronic stress and depression are known to reduce the number of these neuronal connections.

Using a laser-scanning microscope, Kwan and first author Ling-Xiao Shao, a postdoctoral associate in the Yale School of Medicine, imaged dendritic spines in high resolution and tracked them for multiple days in living mice. They found increases in the number of dendritic spines and in their size within 24 hours of administration of psilocybin. These changes were still present a month later. Also, mice subjected to stress showed behavioral improvements and increased neurotransmitter activity after being given psilocybin.

For some people, psilocybin, an active compound in “magic mushrooms,” can produce a profound mystical experience. The psychedelic was a staple of religious ceremonies among indigenous populations of the New World and is also a popular recreational drug.

"It may be the novel psychological effects of psilocybin itself that spurs the growth of neuronal connections," Kwan said.

“It was a real surprise to see such enduring changes from just one dose of psilocybin,” he said. “These new connections may be the structural changes the brain uses to store new experiences.”

*From the article here :
 
Last edited:
2B25EC4700000578-3370542-image-a-22_1450800312717.jpg



Homological scaffolds of brain functional networks

Petri, Expert, Turkheimer, Carhart-Harris, Nutt, Hellyer, Vaccarino

Networks, as efficient representations of complex systems, have appealed to scientists for a long time and now permeate many areas of science, including neuroimaging. Traditionally, the structure of complex networks has been studied through their statistical properties and metrics concerned with node and link properties, e.g. degree-distribution, node centrality and modularity. Here, we study the characteristics of functional brain networks at the mesoscopic level from a novel perspective that highlights the role of inhomogeneities in the fabric of functional connections. This can be done by focusing on the features of a set of topological objects—homological cycles—associated with the weighted functional network. We leverage the detected topological information to define the homological scaffolds, a new set of objects designed to represent compactly the homological features of the correlation network and simultaneously make their homological properties amenable to networks theoretical methods. As a proof of principle, we apply these tools to compare resting-state functional brain activity in 15 healthy volunteers after intravenous infusion of placebo and psilocybin—the main psychoactive component of magic mushrooms. The results show that the homological structure of the brain's functional patterns undergoes a dramatic change post-psilocybin, characterized by the appearance of many transient structures of low stability and of a small number of persistent ones that are not observed in the case of placebo.

Motivation

The understanding of global brain organization and its large-scale integration remains a challenge for modern neurosciences. Network theory is an elegant framework to approach these questions, thanks to its simplicity and versatility. Indeed, in recent years, networks have become a prominent tool to analyse and understand neuroimaging data coming from very diverse sources, such as functional magnetic resonance imaging (fMRI), electroencephalography and magnetoencephalography, also showing potential for clinical applications.

A natural way of approaching these datasets is to devise a measure of dynamical similarity between the microscopic constituents and interpret it as the strength of the link between those elements. In the case of brain functional activity, this often implies the use of similarity measures such as (partial) correlations or coherence, which generally yield fully connected, weighted and possibly signed adjacency matrices. Despite the fact that most network metrics can be extended to the weighted case, the combined effect of complete connectedness and edge weights makes the interpretation of functional networks significantly harder and motivates the widespread use of ad hoc thresholding methods. However, neglecting weak links incurs the dangers of a trade-off between information completeness and clarity. In fact, it risks overlooking the role that weak links might have, as shown for example in the cases of resting-state dynamics, cognitive control and correlated network states.

In order to overcome these limits, Rubinov & Sporn recently introduced a set of generalized network and community metrics for functional networks that among others were used to uncover the contrasting dynamics underlying recollection and the physiology of functional hubs.

In this paper, we present an alternative route to the analysis of brain functional networks. We focus on the combined structure of connections and weights as captured by the homology of the network.

Discussion

In this paper, we first described a variation of persistent homology that allows us to deal with weighted and signed networks. We then introduced two new objects, the homological scaffolds, to go beyond the picture given by persistent homology to represent and summarize information about individual links. The homological scaffolds represent a new measure of topological importance of edges in the original system in terms of how frequently they are part of the generators of the persistent homology groups and how persistent are the generators to which they belong to. We applied this method to an fMRI dataset comprising a group of subjects injected with a placebo and another injected with psilocybin.

By focusing on the second homology group H1, we found that the stability of mesoscopic association cycles is reduced by the action of psilocybin, as shown by the difference in the probability density function of the generators of H1.

It is here that the importance of the insight given by the homological scaffolds in the persistent homology procedure becomes apparent. A simple reading of this result would be that the effect of psilocybin is to relax the constraints on brain function, ascribing cognition a more flexible quality, but when looking at the edge level, the picture becomes more complex. The analysis of the homological scaffolds reveals the existence of a set of edges that are predominant in terms of their persistence although they are statistically part of the same number of cycles in the two conditions. In other words, these functional connections support cycles that are especially stable and are only present in the psychedelic state. This further implies that the brain does not simply become a random system after psilocybin injection, but instead retains some organizational features, albeit different from the normal state, as suggested by the first part of the analysis. Further work is required to identify the exact functional significance of these edges. Nonetheless, it is interesting to look at the community structure of the persistence homological scaffolds in figure 6.

rsif20140873f06.jpg

Figure 6.

Simplified visualization of the persistence homological scaffolds. The persistence homological scaffolds Inline Formula (a) and Inline Formula (b) are shown for comparison. For ease of visualization, only the links heavier than 80 are shown. In both networks, colours represent communities obtained by modularity optimization on the placebo persistence scaffold using the Louvain method [50] and are used to show the departure of the psilocybin connectivity structure from the placebo baseline. The width of the links is proportional to their weight and the size of the nodes is proportional to their strength. Note that the proportion of heavy links between communities is much higher (and very different) in the psilocybin group, suggesting greater integration. A labelled version of the two scaffolds is available as GEXF graph files as the electronic supplementary material.

The two pictures are simplified cartoons of the placebo (figure 6a) and psilocybin (figure 6b) scaffolds. In figure 6a,b, the nodes are organized and coloured according to their community membership in the placebo scaffold. This is done in order to highlight the striking difference in connectivity structure in the two cases. When considering the edges in the tail of the distribution, weight greater than or equal to 80, only 29 of the 374 edges present in the truncated psilocybin scaffold are shared with the truncated placebo scaffold (165 edges). Of these 374 edges, 217 are between placebo communities and are observed to mostly connect cortical regions. This supports our idea that psilocybin disrupts the normal organization of the brain with the emergence of strong, topologically long-range functional connections that are not present in a normal state.

The two key results of the analysis of the homological scaffolds can therefore be summarized as follows (i) there is an increased integration between cortical regions in the psilocybin state and (ii) this integration is supported by a persistent scaffold of a set of edges that support cross modular connectivity probably as a result of the stimulation of the 5HT2A receptors in the cortex.

We can speculate on the implications of such an organization. One possible by-product of this greater communication across the whole brain is the phenomenon of synaesthesia which is often reported in conjunction with the psychedelic state. Synaesthesia is described as an inducer-concurrent pairing, where the inducer could be a grapheme or a visual stimulus that generates a secondary sensory output—like a colour for example. Drug-induced synaesthesia often leads to chain of associations, pointing to dynamic causes rather than fixed structural ones as may be the case for acquired synaesthesia. Broadly consistent with this, it has been reported that subjects under the influence of psilocybin have objectively worse colour perception performance despite subjectively intensified colour experience.

To summarize, we presented a new method to analyse fully connected, weighted and signed networks and applied it to a unique fMRI dataset of subjects under the influence of mushrooms. We find that the psychedelic state is associated with a less constrained and more intercommunicative mode of brain function, which is consistent with descriptions of the nature of consciousness in the psychedelic state.

 
tingri-everest-base-camp-trek.jpg



Psychedelics for Brain Injury: A Mini-Review

Shariq Mansoor Khan(1), Gregory T. Carter(2), Sunil K. Aggarwal(3), Julie Holland(4)

1Washington University School of Medicine in St. Louis, St. Louis, MO, United States
2Department of Physical Medicine & Rehabilitation, St. Luke's Rehabilitation Institute, Spokane, WA, United States
3Advanced Integrative Medical Sciences Institute, Seattle, WA, United States
4Private Practitioner, New York, NY, United States

Objective: Stroke and traumatic brain injury (TBI) are among the leading causes of disability. Even after engaging in rehabilitation, nearly half of patients with severe TBI requiring hospitalization are left with major disability. Despite decades of investigation, pharmacologic treatment of brain injury is still a field in its infancy. Recent clinical trials have begun into the use of psychedelic therapeutics for treatment of brain injury. This brief review aims to summarize the current state of the science's relevance to neurorehabilitation, and may act as a resource for those seeking to understand the precedence for these ongoing clinical trials.
Methods: Narrative mini-review of studies published related to psychedelic therapeutics and brain injury.
Results: Recent in vitro, in vivo, and case report studies suggest psychedelic pharmacotherapies may influence the future of brain injury treatment through modulation of neuroinflammation, hippocampal neurogenesis, neuroplasticity, and brain complexity.
Conclusions: Historical data on the safety of some of these substances could serve in effect as phase 0 and phase I studies. Further phase II trials will illuminate how these drugs may treat brain injury, particularly TBI and reperfusion injury from stroke.

Introduction

Despite millennia of historical use around the world, research into medical uses of psychedelic drugs has been stymied for years by stigma (1). “Classical” psychedelic drugs refer to the most well studied and culturally significant psychedelics, including mescaline, lysergic acid diethylamide (LSD), psilocybin, and dimethyltryptamine (DMT). Though having a wide range of molecular structures and target receptors, psychedelics are unified by their ability to produce marked alterations in sensory perception, consciousness, distortion of time, and perception of reality. Evidence suggests that activation of 5-HT2A receptors (a class of excitatory receptors of serotonin or 5-HT), is the common mechanism for the psychological experience of classical psychedelics, though these compounds are known to act at other receptors (2).

Currently, however, psychedelics are experiencing a scientific renaissance due to advances in research methodology and changes in the regulation of these substances. Trials of psilocybin for disorders of consciousness, and DMT for stroke, are in discussion to begin in coming years (3, 4). In vitro and in vivo studies suggest psychedelics may influence the future of brain injury treatment in both the acute and chronic phases through a variety of mechanisms including modulation of neuroinflammation, neuroplasticity, hippocampal neurogenesis, and increases in brain complexity.​

Methods

We conducted a literature search of articles relevant to psychedelic therapeutics for brain injury using PubMed. The search terms are available in the Appendix. Bibliographies of the main review papers were also used to detect other relevant articles. Studies were selected with emphasis to their relevance to psychedelics' purported neuroregenerative and neuroprotective potential, rather than their psychotherapeutic properties. A narrative mini-review format was employed with the intention of providing a brief overview for readers seeking to understand the scientific basis for anticipated clinical trials.​

Neuroinflammation

Within the brain, depression, addiction, Alzheimer's, and Parkinson's all appear to be linked to neuroinflammatory states (57). There are currently three main classes of anti-inflammatory drugs: non-steroidal anti-inflammatory drugs (NSAIDs), steroids such as prednisone, and biologics which act like sponges to “soak up” inflammatory cytokines (1). Psychedelics may represent a fourth class of anti-inflammatory drug.

Neuroinflammation after stroke is responsible for both infarct expansion as well as remodeling and repair (8, 9). Modulation of this inflammation is currently a target for new therapies. The inflammatory response to ischemic stroke is thought to derive largely from reperfusion injury (10). In general, there are no conventional medical therapies addressing reperfusion injury after stroke, with the exception of edavarone in Japan, which is only modestly effective (11).

After stroke, immune cells invade the injured tissue, interacting with microglia and neurons (8). Modulating this inflammatory response, particularly through tumor necrosis factor (TNF), interleukin (IL)-1, IL-6, and IL-10, may be the next frontier in stroke recovery (8, 12). However, it should be expected that the cytokine response to brain injury has both beneficial and harmful effects to the recovering patient. In contrast to steroids, which cause generalized systemic immunosuppression, psychedelics produce a unique pattern of cytokine expression favoring anti-allergic conditions (13, 14). In other words, psychedelics may target many of the pathologic immune responses without exposing the body to the risks of total immune suppression (e.g., serious infection) or potential side effects of existing biologics (e.g., malignancy and cardiovascular disease). Instead, careful regulation of the inflammatory response, rather than blunt reduction of the response, or “single-target” approaches, is critical to improved outcomes.

Mizuma and Yenari (10) argue that immunomodulatory therapies have previously not shown efficacy in clinical trials because they largely took place before the use of revascularization techniques. Because reperfusion injury is thought to be the inciting event for much of the neuroinflammatory response, they argue that combining revascularization with immunomodulation may hold promise for stroke treatment. Indeed, Jickling et al.'s (12) review of neutrophil modulation as treatment for stroke notes the contrast seen in the efficacy of these treatments in animal models is dependent on whether reperfusion is induced.

Classical psychedelics act principally on the 5-hydroxytryptamine receptors (5-HTRs) to produce their psychological effects, specifically the 5-HT2a receptor (5). These same receptors are well-known to have the potential to regulate inflammation within the central nervous system and peripherally (15). In fact, the 5-HT2a receptor is the most widely expressed serotonin receptor throughout the human body (1). It is present on nearly all tissue and cell types, including all major immune-related cell types (1). However, the highest density of 5-HT2a receptors is found in the brain (1). Though peripheral immunomodulation has been documented with other psychedelics like lysergic acid diethylamide (LSD) (16), 3,4-methylenedioxy-methamphetamine (MDMA) (17), and 2,5-dimethoxy-4-iodoamphetamine (DOI) (13), N,N-Dimethyltryptamine (DMT) has been especially well-studied with regards to its effects on neuroinflammation and reperfusion injury.

DMT is a psychedelic which is endogenously produced by the human brain, particularly the pineal gland, likely in local concentrations comparable to other monoamine neurotransmitters (18). Though it has been known for at least 40 years that DMT is an endogenously produced hallucinogen, its physiologic function remains elusive. The lack of consensus may be due in part to a paradigm in which the scientific community has assumed DMT can only be, or primarily acts as, a hallucinogen, which keeps research focus on its psychologic effects at 5-HTRs, rather than its non-hallucinogenic effects (9). Recent experiments have shown that an additional receptor, S1R, is critical in the immunomodulating response of DMT.

S1R dysfunction is known to be involved in a wide range of neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis (ALS), and TBI (1921). S1R agonists produce neuroprotective effects via regulation of intracellular calcium, reducing expression of pro-apoptotic genes, and inhibiting anti-apoptotic activity by the Bcl-2 gene (22). Specifically, DMT's action at S1Rs has been shown in murine models to modulate inflammation by reducing IL-1b, IL-6, TNFa, and IL-8, while increasing the secretion of the anti-inflammatory cytokine IL-10, and by inhibiting activation of Th1 (T Helper cell type 1) and Th17 (T Helper cell type 17) subsets (20). In mice, S1R is known to play an important role in the endoplasmic reticulum stress response, including oxidative stress, probably by up-regulating antioxidants, quinine oxidoreductase 1 and superoxide dismutase (SOD) (23). Being present in reactive astrocytes, microglia, and neurons, it has also been shown to regulate neuritic outgrowth, myelination, synaptogenesis, and neuro-regeneration (22, 24). It should be noted that S1R is now known to have multiple endogenous agonists, such as progesterone (25). However, with no clear alternative explanation, it may be reasonable to hypothesize that DMT has some, or all, of these neuroprotective and anti-inflammatory physiologic functions as well. The only clinical trial of a selective S1R agonist for treatment of stroke showed statistically significant functional recovery in post-hoc analysis of moderately-to-severely affected patients (26).

DMT has been shown to reduce ischemic brain injury after middle cerebral artery occlusion (MCAO) through S1R dependent activity in murine models (27). Nardai et al. (27) documented a significant reduction of the infarct core volume in rats at 24 h post MCAO with administration of DMT, as well as enhanced functional regeneration of the affected limb at 30 days post MCAO (27). These results are consistent with other in vitro studies showing cytoprotective effects of DMT against reperfusion injury (28). Furthermore, similar effects have been documented in human-specific models including human cerebral organoids. Dakic et al. (22) were the first to document that a closely related congener of DMT, 5-MeO-DMT, found in high concentrations of the toxin of Incilius alvarius (Colorado River Toad), favorably altered the cerebral proteome with regard to factors involved with plasticity and neuroprotection including long-term potentiation, apoptosis, morphogenesis/maturation of dendritic spines, and T-lymphocyte differentiation, while inhibiting factors involved in neurodegeneration and cell death. The authors note that other dimethyltryptamines may have similar effects, but their results show that the mechanisms may be different and that each deserves careful study. Previous studies in monolayer neuronal cultures did not show the same effects, suggesting a more complex circuitry is required for these effects (22).​

Hippocampal Neurogenesis

TBI and stroke alter hippocampal neurogenesis in murine models (29, 30). Though hippocampal neurogenesis is recognized as an important component of cognitive recovery from TBI and stroke, there is not a direct correlation between increased neurogenesis and recovery. Complicating factors include the nature of the injury, the timing of intervention, how the cells integrate into the hippocampal circuits, and whether the target of intervention is either increased neuronal proliferation or increased survival. While hippocampal neurogenesis after TBI is implicated in improved cognition, relief from depressed mood, and encoding of episodic memory, it is also associated with pro-epileptogenic changes and spatial memory impairment (29, 31, 32).

Though many factors are implicated in hippocampal neurogenesis, one of the most important is 5HTR stimulation (33, 34). Acute administration of psilocybin to mice alters hippocampal neurogenesis in a non-linear fashion (35). Low doses lead to increased neurogenesis while higher doses inhibit it. However, increased neurogenesis has also been seen when high dose psilocybin was administered once-per-week, avoiding the issue of rapid tolerance buildup via 5HTR downregulation (33). Targeting hippocampal neurogenesis for treatment of brain injury and other psychiatric and neurologic disorders is an emerging area of research (36).​

Neuroplasticity

Novel interventional approaches hold promise to improve functional outcomes after brain injury by inducing neural plasticity. However, like targeting inflammation and hippocampal neurogenesis, targeting neuroplasticity is a double-edged sword. Ischemia induced plasticity may be responsible for recovery of function, but also drive complications such as epilepsy and memory disturbance. A full discussion on ischemia induced plasticity is available elsewhere (37).

Functional recovery due to post-stroke plasticity is currently most effectively recruited through intensive physical therapy (37). However, both non-invasive techniques for inducing neuroplasticity such as transcranial magnetic stimulation and direct current stimulation, as well as invasive techniques such as deep brain stimulation, show promising results for stroke recovery (37).

The search for pharmacologic agents which stimulate neuroplasticity after brain injury, including amphetamines, dopaminergic, serotonergic, noradrenergic, and cholinergic agents, has thus far been inconclusive. To our knowledge, the effect of psychedelics on neuroplasticity has not been tested in brains subjected to injury such as stroke. However, recent reports demonstrate that psychedelics promote both structural and functional neuroplasticity in non-injured brains. The persistent symptom improvement in psychiatric disorders with administration of psychedelics has been proposed to be driven by this neuroplastic adaptation (2). Ly et al. (38) found that some psychedelics were more efficacious (e.g., MDMA) or more potent (e.g., LSD) than ketamine in promoting plasticity. These results were demonstrated in vivo in both non-human vertebrates and invertebrates, suggesting that these mechanisms are evolutionarily conserved.

One hypothesized mechanism for psychedelics' beneficial effect on neuroplasticity in brain injury is via Brain-derived neurotrophic factor (BDNF), which is well-known to be implicated in both neuritogenesis and spinogenesis (39). In Ly et al.'s (38) experiments, they found that many psychedelics rivaled administration of pure BDNF in their ability to induce plasticity. At least one randomized control trial has demonstrated increases in serum BDNF in volunteers with administration of a DMT containing tisane, ayahuasca, which persisted for 48 h (40). Importantly, ayahuasca, a traditional South American ceremonial brew, contains not only DMT, but also β-carbolines which act as reversible monoamine oxidase inhibitors (MAOIs) potentiating and prolonging the effects of DMT (40). However, it should be noted that many other non-psychedelic drugs have failed to alter the course of brain injury through induction of BDNF.

However, there is also evidence to suggest that S1R is an important part of the plasticity response to stroke. For example, Ruscher et al. (41) found that delivery of a synthetic S1R agonist enhanced plasticity-mediated recovery of lost sensorimotor function in rats, even when initiation of therapy was started up to 2 days after MCAO, possibly expanding the currently very narrow therapeutic window for acute stroke.

Mirror visual-feedback (MVF) has been shown to enhance key features of neuroplasticity including cross-modal cortical reorganization and learning (42). In patients <12 months post stroke, MVF therapy enhances functional recovery of lower limbs and hands (43, 44). Psilocybin combined with mirror visual-feedback has been shown in a case report to have a dose-dependent reduction in phantom limb associated pain, hypothesized to be due to facilitating MVF's ability to “unlearn” paralysis via 5HTR-dependent changes in neuroplasticity (42). Clinical trials for use of psilocybin in phantom limb pain are currently underway (45).​

Increase in Brain Complexity

Disorders of consciousness (DOC) can arise from a variety of brain injuries including trauma, hypoglycemia, anoxia, and stroke. Between 4 and 38% of stroke patients will experience a DOC (46). Though many therapies have been proposed for patients with DOC, including pharmacologic (e.g., amantadine, D-amphetamine, levodopa, modafinil, and zolpidem), invasive and non-invasive stimulation (e.g., transcranial direct current stimulation), benefits for functional recovery are usually modest and current evidence supporting their use is inconsistent (4750). Other stimulants acting on dopamine including apomorphine may hold promise (51).
Scott and Carhart-Harris have proposed an experimental protocol for testing the capacity of psilocybin to increase conscious awareness in patients with DOC (52). They hypothesize psychedelics increase brain activity complexity and conscious content, in contrast to current stimulant drugs that increase arousal. However, to our knowledge there are no studies of brain complexity measures in DOC patients given stimulants. Their hypothesis relies on findings that psilocybin increases brain complexity, and that these particular measures of complexity, namely perturbational complexity index (53) and the closely related Lempel-Ziv complexity (LZC), reliably predict conscious level. They argue that psychedelic-related elevations in LZC reflect an increased richness of conscious experience, and that targeting increases in conscious content, rather than arousal, as with stimulants, may be key to increasing conscious awareness in DOC patients.

In human subjects, increases in LZC have been observed in excess of those seen in normal wakefulness with administration of psilocybin, LSD, and ketamine at psychedelic doses (54). This increase in complexity has also been demonstrated via other measures including electroencephalogram (EEG) and functional magnetic resonance imaging (fMRI) (55).

Psychedelics are thought to increase brain complexity primarily through 5HTRs (53). As previously discussed, 5HTR agonism is associated with increased neuroplasticity, while antagonism is associated with reduced cognitive flexibility and increased slow-wave sleep and sedation (56, 57). These receptors are most densely expressed in the cortical areas belonging to the default-mode network (DMN) (52). The DMN is known to be implicated in conscious processing as well as the subjective experience of psychedelic states (52). Strength of DMN connectivity is significantly decreased in stroke patients with DOCs, and is highly correlated with Glasgow Coma Scale scores (58). Secondarily, 5HTRs have been shown to play an important role in the control of thalamo-frontal connectivity, known to be important for consciousness (59, 60).​

Conclusions

Psychedelics may play a future role in treatment of brain injury through a variety of mechanisms. Though these are a novel class of drugs deserving close study, more data are necessary to prove their efficacy for treatment of brain injury, as historically many compounds have seemed promising in vitro, including likely hundreds of compounds thought to facilitate neuroplasticity and neuroprotection, but have not borne out in clinical trials (61, 62). There is already mixed evidence to suggest the use of non-classical psychedelics ketamine (63, 64), as well as tetrahydrocannabinol (THC) (65, 66) and cannabidiol (67, 68), as neuroprotectants after TBI and stroke. Presence of THC on urine drug screen is associated with decreased mortality in adult patients sustaining TBI (65). However, randomized control trials of a non-psychoactive cannabinoid analog, dexanabinol, administered once after TBI has failed to show benefit over placebo in increasing Glasgow Coma Scores at 6 months post-injury (69). One explanation for this discrepancy may be the lack of “entourage” effect whereby various cannabinoids and cannabis phytochemicals are clinically more efficacious when working synergistically compared to administration of a single cannabinoid in isolation (70), though the existence of such an effect with cannabis is contentious with mixed evidence for its existence (71). Of note, an “entourage” effect has been demonstrated with psychedelic mushrooms in which whole mushroom extracts are on the order of 10 times more potent than purified psilocin administered alone in neurobehavioral rat models (72).

Another possible explanation is that the subjective hallucinogenic effects are necessary for some, or all, of psychedelics' therapeutic effects. This remains a fundamental research question and strong evidence exists on both sides of the debate (73). Few human trials on the therapeutic effects of psychedelics at sub-behavioral “micro-” doses have been completed, but results from animal and cell studies for their use as anti-inflammatories are promising (5). In contrast, studies on the psychological effects of micro-dosing psychedelics seem to be explained by placebo effect (74, 75). Extrapolating from animal studies, the doses required at least for the anti-inflammatory effects of psychedelics are predicted to be magnitudes less than threshold for hallucinations (13). Furthermore, recent studies treating rat models of asthma with 2,5-dimethoxyphenethylamine (2C-H) which is a non-hallucinogenic 5-HT2a agonist structurally related to MDMA, suggest that psychedelics' behavioral and anti-inflammatory effects may have separate, but related, underlying mechanisms (76). A non-hallucinogenic analog of ibogaine has been shown to induce structural neuroplasticity, as well as reduce depression, alcohol-, and heroin-seeking behavior in rodents, similarly to its hallucinogenic variant (77). In contrast, Brouwer and Carhart-Harris have introduced a construct known as the “pivotal mental state” to explain the evolutionary function that 5-HT2a receptor agonism has in inducing neurologic hyper-plasticity and psychological adaptation when the body is exposed to subjective acute stress (e.g., hallucinations, intense spiritual experience, psychosis, trauma) (78). They hypothesize that “psychedelics hijack a system that has evolved to mediate rapid and deep learning” to provide a psychological “fresh start” or “rebirth.” It is likely that some, but not all, of the psychological therapeutic benefits of psychedelics is dependent on the subjective experience (73), but whether the same can be said for their use in brain injury is unclear.

Classical psychedelics have millennia of historical use, do not have significant risk of dependence, and are safe to use under close medical supervision (79, 80). Though there should be caution in over-interpreting the relevance of the aforementioned animal studies' relevance to human pathological states, this historical data should serve in effect as phase 0 and phase I studies (9). However, presumably much of this historical data is based on intermittent, infrequent dosing, so trial safety data may need to be repeated with continuous, regular doses. Further phase II trials will illuminate how these drugs may treat brain injury, particularly TBI and reperfusion injury from stroke.

Further study and design of non-psychoactive analogs may answer fundamental questions regarding the interplay of hallucinations with other properties of psychedelic therapy, as well as facilitate more practical use in acute hospital settings. The subjective hallucinogenic effects of psychedelics, with clinical psychological support, may also prove valuable in fostering recovery from brain injury from a trauma recovery rehabilitation psychology standpoint due to their anti-PTSD and anti-depression effects that are now well-documented (81, 82). Depression and PTSD significantly hinder recovery from stroke and TBI (83, 84). Psychedelics' use in this area may be more immediately clinically relevant in this area than in the neuroprotection and neuro-regeneration, where comparatively less is known. Further research is needed to determine the optimal timing after injury and route of administration for neuroprotective effects. All the compounds discussed in this paper have reasonable oral absorption. Route of administration would impact the degree and timeliness of the pharmacological effects yet there are not enough studies specifically on this aspect to make recommendations. The emerging use of intranasal route of administration may afford rapid induction into the central nervous system and could be a promising future option (85).

 
microdosers-of-lsd-and-magic-mushrooms-are-wiser-and-more-creative-311419-1280x720.webp



Single dose of psilocybin can remodel connections in the brain*

by Ruairi J Mackenzie | Technology Networks | 5 Jul 2021

Psilocybin, a psychedelic compound that can be derived from over 200 species of mushroom, can remodel connections in the mouse brain. That is the conclusion of a new study that examined structural changes in the brain that might explain psilocybin’s enduring antidepressant effects.​

10% increase in connections

The study was led by researchers at Yale University. Senior author Alex Kwan, associate professor of psychiatry and neuroscience, said in a press release, "We not only saw a 10% increase in the number of neuronal connections, but also they were on average about 10% larger, so the connections were stronger as well.”

The antidepressant effects of psilocybin and its active component, psilocin, have been studied extensively since the compound was granted Breakthrough Therapy designation for the treatment of depression by the US Food and Drug Administration in 2019. This kickstarted numerous clinical trials into the drug’s therapeutic effects, but the question of how these potential effects might be achieved in the brain remains unanswered.​

The plastic brain

Kwan and his team sought to investigate whether alterations to dendrites, microscopic brain connections that help integrate signals sent from other nerve cells, might be involved. Dendrites are highly plastic, growing and shrinking in response to neurobiological changes. In depression, human studies show that dendritic number and size in the prefrontal cortex reduces. Kwan’s team wanted to see whether psilocybin administration to mice would influence their dendritic density.

To find the answer, Kwan’s team used a powerful imaging technique called two-photon microscopy to look at the density of synapses in the mouse prefrontal cortex. The researchers were able to longitudinally image the brain using this technique, meaning they could examine how dendritic structure changed within the first 24 hours after psilocybin administration, and then repeatedly in the first week after dosing. This was key to determining the strength and endurance of the dendritic connections formed. The researchers showed that roughly 50% of the connections that formed immediately after dosing remained intact a week later. A final analysis 34 days later showed that roughly a third of connections were still intact.

These findings are key to teasing out one of the central mysteries of psilocybin research – why a compound that produces very short-term behavioral changes should produce an antidepressant effect that last far longer.​

Serotonin signaling

To examine this discrepancy further, the team dosed the mice with a compound called ketanserin, which blocks signaling through serotonin 5-HT2A receptors, thought to be responsible for the psychedelic trips experienced when taking magic mushrooms. Mice can’t tell us whether they are undergoing profound psychedelic experiences, but they do exhibit specific head-twitching behaviors immediately after taking psilocybin, which is used as an indicator of serotonin receptor stimulation. The researchers noted that while ketanserin stopped this behavior, it did not have an impact on dendritic changes. This may suggest, the researchers say, that the psychedelic and antidepressant effects of psilocybin could be separated.

More research will need to be conducted to work out whether specific brain areas or cell types are especially important in psilocybin’s antidepressant effects, but these findings illustrate that psychedelics like psilocybin, and the already-approved novel antidepressant ketamine, may work in a similar way. “Synaptic rewiring may be a mechanism shared by compounds with rapid antidepressant effects,” write the authors. Summing up, Kwan commented, "It was a real surprise to see such enduring changes from just one dose of psilocybin," he said. "These new connections may be the structural changes the brain uses to store new experiences."

*From the article here :
 
Last edited:
default-1280x720.webp


The Brain on LSD*

Technology Networks | 12 Apr 2016

New scans show how LSD affects the brain.

In a series of experiments, scientists have gained a glimpse into how the psychedelic compound affects brain activity. The team administered LSD (Lysergic acid diethylamide) to 20 healthy volunteers in a specialist research centre and used various leading-edge and complementary brain scanning techniques to visualise how LSD alters the way the brain works.

The findings, published in Proceedings of the National Academy of Sciences (PNAS), reveal what happens in the brain when people experience the complex visual hallucinations that are often associated with LSD state. They also shed light on the brain changes that underlie the profound altered state of consciousness the drug can produce.

A major finding of the research is the discovery of what happens in the brain when people experience complex dreamlike hallucinations under LSD. Under normal conditions, information from our eyes is processed in a part of the brain at the back of the head called the visual cortex. However, when the volunteers took LSD, many additional brain areas - not just the visual cortex - contributed to visual processing.

Dr Robin Carhart-Harris, from the Department of Medicine at Imperial, who led the research, explained: "We observed brain changes under LSD that suggested our volunteers were 'seeing with their eyes shut' - albeit they were seeing things from their imagination rather than from the outside world. We saw that many more areas of the brain than normal were contributing to visual processing under LSD - even though the volunteers' eyes were closed. Furthermore, the size of this effect correlated with volunteers' ratings of complex, dreamlike visions. "

32538.jpg


The study also revealed what happens in the brain when people report a fundamental change in the quality of their consciousness under LSD. Dr Carhart-Harris explained: "Normally our brain consists of independent networks that perform separate specialised functions, such as vision, movement and hearing - as well as more complex things like attention. However, under LSD the separateness of these networks breaks down and instead you see a more integrated or unified brain."

"Our results suggest that this effect underlies the profound altered state of consciousness that people often describe during an LSD experience. It is also related to what people sometimes call 'ego-dissolution', which means the normal sense of self is broken down and replaced by a sense of reconnection with themselves, others and the natural world. This experience is sometimes framed in a religious or spiritual way - and seems to be associated with improvements in well-being after the drug's effects have subsided."


Dr Carhart-Harris added: "Our brains become more constrained and compartmentalised as we develop from infancy into adulthood, and we may become more focused and rigid in our thinking as we mature. In many ways, the brain in the LSD state resembles the state our brains were in when we were infants: free and unconstrained. This also makes sense when we consider the hyper-emotional and imaginative nature of an infant's mind."

In addition to these findings, research from the same group, part of the Beckley/Imperial Research Programme, revealed that listening to music while taking LSD triggered interesting changes in brain signalling that were associated with eyes-closed visions.

In a study published in the journal European Neuropsychopharmacology, the researchers found altered visual cortex activity under the drug, and that the combination of LSD and music caused this region to receive more information from an area of the brain called the parahippocampus. The parahippocampus is involved in mental imagery and personal memory, and the more it communicated with the visual cortex, the more people reported experiencing complex visions, such as seeing scenes from their lives.

PhD student Mendel Kaelen from the Department of Medicine at Imperial, who was lead author of the music paper, said: "This is the first time we have witnessed the interaction of a psychedelic compound and music with the brain's biology. The Beckley/Imperial Research Programme hope these collective findings may pave the way for these compounds being one day used to treat psychiatric disorders. They could be particularly useful in conditions where negative thought patterns have become entrenched, say the scientists, such as in depression or addiction."

Mendel Kaelen added: "A major focus for future research is how we can use the knowledge gained from our current research to develop more effective therapeutic approaches for treatments such as depression; for example, music-listening and LSD may be a powerful therapeutic combination if provided in the right way."

Beckley-LSD-research.jpg


Professor David Nutt, the senior researcher on the study and Edmond J Safra Chair in Neuropsychopharmacology at Imperial, said: "Scientists have waited 50 years for this moment - the revealing of how LSD alters our brain biology. For the first time we can really see what's happening in the brain during the psychedelic state, and can better understand why LSD had such a profound impact on self-awareness in users and on music and art. This could have great implications for psychiatry, and helping patients overcome conditions such as depression."

Amanda Feilding, Director of the Beckley Foundation, said: "We are finally unveiling the brain mechanisms underlying the potential of LSD, not only to heal, but also to deepen our understanding of consciousness itself." The research involved 20 healthy volunteers - each of whom received both LSD and placebo - and all of whom were deemed psychologically and physically healthy. All the volunteers had previously taken some type of psychedelic drug. During carefully controlled and supervised experiments in a specialist research centre, each volunteer received an injection of either 75 micrograms of LSD, or placebo.

Their brains were then scanned using various techniques including fMRI and magneto encephalography (MEG). These enabled the researchers to study activity within the whole of the brain by monitoring blood flow and electrical activity. "Neural correlates of the LSD experience revealed by multimodal neuroimaging," by Robin L. Carhart-Harris et al., is published in the Proceedings of the National Academy of Sciences April 2016. "LSD modulates music-induced imagery via changes in parahippocampal connectivity," by Mendel Kaelen et al. is published in European Neuropsychopharmacology.

*From the article here :
 
Last edited:
LN-dtm.jpg



Psychedelics forge connections between neurons

by Cici Zhang | C&EN

About one-third of people with depression do not find relief from their symptoms with current drugs for the disease. In recent years, psychiatrists have become interested in using low doses of the anesthetic ketamine as an antidepressant because it often works in patients who don’t respond to conventional drugs and it acts fast—having an effect within hours instead of weeks or even months. But the drug has undesirable side effects. It can produce out-of-body feelings and hallucinations, and it has the potential for abuse. Scientists would like to understand how ketamine works as an antidepressant so they can design new molecules that lack the negative aspects of the anesthetic.

Now, a study reports that other psychedelic compounds have similar effects on neurons as ketamine does: They promote the growth of connections between neurons. The work suggests new chemical scaffolds that could mimic ketamine’s antidepressant properties, says lead author David Olson of the University of California, Davis.

Previous research had shown that ketamine can rapidly grow synapses—the connections between neurons—in brain areas that regulate emotion and mood, possibly accounting for the drug’s fast-acting therapeutic effects. In the current study, the UC Davis team found that several psychedelic compounds—for example, N,N-dimethyltryptamine (DMT), 2,5-dimethoxy-4-iodoamphetamine (DOI), and lysergic acid diethylamide (LSD)—increased the number of synaptic connections in the brains of rats and fruit flies, as well as in cultured neurons from the animals. As with ketamine, the effects of the drugs were long lasting. When the researchers injected rats with DMT, which is found in an Amazonian herbal tea called ayahuasca, they still observed synaptic changes 24 hours later, well after the animals had cleared the drug from their bodies, Olson says. The team previously had demonstrated that DMT produced antidepressant-like effects and stopped behaviors that resemble posttraumatic stress disorder in rats.

In the new study, the scientists also determined that these other psychedelics promote synaptic growth through a similar signaling pathway involving the protein mTOR that ketamine does. Ronald Duman, a neuroscientist at Yale University who studies ketamine, says that the findings are “interesting and important” because they could explain how these agents might treat mood disorders. While ketamine activates the mTOR pathway by blocking N-methyl-D-aspartate receptors on neurons, the psychedelic compounds do so via activating a different target, the 5-HT2A receptor, which according to Duman suggests a novel approach for treating depression.

griffiths-ted.jpeg


Roland Griffiths, a neuroscientist at Johns Hopkins University School of Medicine agrees that the new study is important. “Almost nothing is known about the neural mechanisms underlying the ability of psychedelics to produce enduring change in moods, attitudes, and behavior”he says.

But David Feifel, a professor emeritus of psychiatry at University of California, San Diego, who has used ketamine to treat patients with depression, says more work is needed before the reported mechanisms can be identified as those responsible for these drugs’ antidepressant properties. The next step, he says, would be to block the pathway outlined in this study and then see if that prevents the drugs from producing an antidepressant effect in animals.

 
Last edited:
Enhanced-MRI-scan-of-the-brain-by-Wellcome-Images-Creative-Commons.jpg


Cannabinol found to protect aging brain cells*

Salk Institute | 25 Jan 2022

Decades of research on medical cannabis has focused on the compounds THC and CBD in clinical applications. But less is known about the therapeutic properties of cannabinol (CBN). Now, a new study by Salk scientists shows how CBN can protect nerve cells from oxidative damage, a major pathway to cell death.

The findings, published online in the journal Free Radical Biology and Medicine, suggest CBN has the potential for treating age-related neurodegenerative diseases, like Alzheimer’s.

“We’ve found that cannabinol protects neurons from oxidative stress and cell death, two of the major contributors to Alzheimer’s,” says senior author Pamela Maher, a research professor and head of Salk’s Cellular Neurobiology Laboratory. “This discovery could one day lead to the development of new therapeutics for treating this disease and other neurodegenerative disorders, like Parkinson’s disease.”

Derived from the cannabis plant, CBN is molecularly similar to THC, but is not psychoactive. It’s also less heavily regulated by the FDA. Previous research by Maher’s lab found that CBN had neuroprotective properties, but it wasn’t clear how it worked. Now, this new study explains the mechanism through which CBN protects brain cells from damage and death.

Maher’s team looked at the process of oxytosis, also called ferroptosis, which is thought to occur in the aging brain. Growing evidence suggests that oxytosis may be a cause of Alzheimer’s disease. Oxytosis can be triggered by the gradual loss of an antioxidant called glutathione, causing neural cell damage and death via lipid oxidation. In the study, the scientists treated nerve cells with CBN, and then introduced an agent to stimulate oxidative damage.

They further found that the CBN worked by protecting mitochondria, the cell’s powerhouses, within the neurons. In damaged cells, oxidation caused the mitochondria to curl up like donuts—a change that’s also been seen in aging cells taken from the brains of people with Alzheimer’s disease. Treating cells with CBN prevented the mitochondria from curling up and kept them functioning well.

To confirm the interaction between CBN and mitochondria, researchers then replicated the experiment in nerve cells that had the mitochondria removed. In these cells, CBN no longer demonstrated its protective effect.

“We were able to directly show that maintenance of mitochondrial function was specifically required for the protective effects of the compound,” Maher said.

In another key finding, researchers showed that CBN did not activate cannabinoid receptors, which are required for cannabinoids to produce a psychoactive response. Thus, CBN therapeutics would work without causing the individual to become “high.”

“CBN is not a controlled substance like THC, the psychotropic compound in cannabis, and evidence has shown that CBN is safe in animals and humans. And because CBN works independently of cannabinoid receptors, CBN could also work in a wide variety of cells with ample therapeutic potential,” says first author Zhibin Liang, a postdoctoral fellow in the Maher lab.

In addition to Alzheimer’s, the findings have implications for other neurodegenerative diseases, such as Parkinson’s, which is also linked to glutathione loss. “Mitochondrial dysfunction is implicated in changes in various tissues, not just in the brain and aging, so the fact that this compound is able to maintain mitochondrial function suggests it could have more benefits beyond the context of Alzheimer’s disease,” Maher said.

Maher adds that the study shows the need for further research into CBN and other lesser-studied cannabinoids. As a next step, Maher’s team is working to see if they can reproduce their results in a preclinical mouse model.

Other authors on the study are David Soriano-Castell, Devin Kepchia, Antonio Currais and David Schubert from Salk; and Brendan Duggan from the University of California San Diego.

*From the article here :
 
Last edited:
Ibogaine_Neuroplasticity.jpg


Ibogaine’s Unique Impact on Neuroplasticity

by Katie Stone | Psychable

The idea that the human brain can change its physical structure and function in response to changes in environmental input has been around since the late 19th century, but it was only recently that technology allowed us to measure these changes objectively.

It was even more recently when we learned that certain psychedelics support neurogenesis (the growth of new brain cells) and neuroplasticity (the ability for your brain to create new neural pathways and connections). Ibogaine, a psychedelic medicine derived from the iboga shrub native to Gabon, is one of those psychedelics that may be quite beneficial in this regard.

Ibogaine is most well-known for its ability to address opiate withdrawal. While the exact mechanism by which it accomplishes that is unknown, research is finding that ibogaine interacts with the nervous system by activating numerous neurotransmitters that create a wide range of effects in the brain, including neuroplasticity.

Before we get into the specifics about ibogaine’s impacts on neuroplasticity, we’ll dive a little deeper into the science behind it.

What is Neuroplasticity?

For much of the history of psychology, researchers were under the impression that the adult personality is established around age thirty and has very little room for change. There was even a belief that one had a limited number of brain cells and that these cells could not be replaced – a hypothesis that has been disproven.

The discovery of neuroplasticity has changed this view. We now know that the brain can evolve throughout life in response to internal factors and external events or stressors. It is understood that the brain can change throughout life in response to experience, both positive and negative.

There are three significant proteins we will talk about that mediate neuroplasticity in the brain. The first is BDNF (brain-derived neurotrophic factor), the second protein is called GDNF (glial cell-line derived neurotrophic factor), and the third is NGF (nerve growth factor). Ibogaine acts on receptors that modulate the release of all three of these proteins — which makes ibogaine different from other psychedelic compounds.

These proteins are known as neurotrophic factors, and they work together to promote survival, growth, and maintenance of the central nervous system (CNS). It has long been known that these proteins are essential during childhood development, but they have recently been recognized as critical to the adult brain as well.

BDNF and GDNF have also been shown to play a role in modulating addiction behaviors in animal trials.

Ibogaine and BDNF

BDNF is a protein that is released in response to neuronal activity. It stimulates the growth, survival, and differentiation of neurons as well as their connections with each other. These connection points are called synapses. Classical psychedelics like psilocybin increase BDNF by acting on the same receptor as serotonin, the neurotransmitter that regulates mood. Low levels of BDNF have been linked to diseases like Alzheimer’s, Parkinson’s, and Huntington’s disease.

Ibogaine and GDNF

GDNF is a protein that is responsible for maintaining the health and survival of dopaminergic and motor neurons in the brain. GDNF may also stimulate the release of dopamine and other neurotransmitters in synapses, affecting synaptic plasticity. GDNF also plays a role in neurodegeneration, which is thought to be connected to Parkinson’s disease, specifically at the dopamine receptor. As far as we know, ibogaine is the only compound that can stimulate the release of GDNF naturally.

Ibogaine and NGF

NGF works to maintain the health of nerve cells called neurons. NGF is essential for learning and memory, playing a vital role in the preservation of brain cells. It has also been shown to stimulate new neuron growth, which can be beneficial during recovery after a stroke or traumatic brain injury to replace damaged tissue.

Ibogaine Supports Neuroplasticity

Neuroplasticity refers to “the ability of neural networks in the brain to change through growth and reorganization.” One way that brain cells grow is through a process called dendritic branching. Dendrites are the branching roots of the nerve cell that connect to other nerve cells, creating spaces of connection called synapses where neurotransmitters interact and can be taken up into the cell for transport to the appropriate channels of activation.

Once ibogaine is metabolized in the liver into noribogaine, noribogaine then binds to specific receptors and begins to active neurotransmitters that stimulate the release of neurotrophic factors. So while ibogaine is the starter compound, once it has been ingested and metabolized, noribogaine is leading to the resulting neurogenesis.

In one study involving rats, ibogaine was found to catalyze neuroplasticity in a dose-dependent manner. The researchers demonstrated that ibogaine alters the cellular transcription levels of both GDNF and BDNF. This means that ibogaine regulates the rate at which the neurons produce and release these compounds.

The researchers administered ibogaine to rats in a time and dose-dependent manner, analyzing the various neurotrophic factors present in the brain post-mortem. After checking the rats’ brains for variations across dose, time, brain region, and concentration of neurotrophic factors, researchers suggest there could be potential for ibogaine to support neurodegenerative diseases such as Parkinson’s.

It is already well established in the literature that an increase in serotonin transmission results in an increase in BDNF expression. It is further well established that ibogaine and its metabolite noribogaine increase serotonin transmission because both compounds are serotonin-reuptake inhibitors, meaning that they help keep serotonin available for interaction in the synapses.

Ibogaine also interacts with NGF, which may be involved in specific areas of the brain connected to substance-seeking behavior. However,t this protein is less studied than BDNF and GDNF. There is evidence that NGF levels are decreased in the brains of alcohol-treated mice, specifically in the hippocampus and hypothalamus. These conditions were also apparent in the serum of chronic heroin and cocaine users, which might suggest a common relationship between NGF and addictive behavior.

Ultimately, this study revealed that ibogaine administration impacts the expression of three neurotrophic factors: BDNF, GDNF, and NGF, resulting in the production of neuroplasticity. This verifies another study that classified ibogaine as a “psychoplastogen” for its ability to rapidly promote neurogenesis. While these are all promising results, until we have clinical trials to further investigate these effects in humans, we will not know the full impacts of ibogaine on neuroplasticity.

 
Panorama_of_Kings_Parade_in_Cambridge%2C_UK%2C_at_St._Mary%27s.jpg

Cambridge
The Neuroscience of Psychedelic Visuals

Dr. James Cooke
| Reality Sandwich

The terms ‘psychedelic’ can be applied not only to a class of chemical, but to a whole style of visual art, inspired by the visions that these chemicals produce. Why do these substances produce similar visual experiences across different individuals and where do these experiences come from? From breathing walls to melting hands, from symbolic imagery to entity encounters, psychedelic visions can tell us something about who and what we are.

Normal Perception

During everyday waking consciousness we typically perceive a stable world of objects. This feels like a completely passive process–just open your eyes and there’s the world, no effort required. In reality, the felt simplicity of this experience masks the highly complex processing going on in your brain that allows you to see. Your retina is not a clear window through which your soul looks out onto the world. Instead it is a fleshly surface similar to the rest of your body, a piece of meat that blocks the light from the outside world, rather than letting it in.

What actually happens when we see is that the patterns of light are transformed into electrochemical signals that are sent down the optic nerve to the brain. The brain, sitting in your pitch-black skull, learns to actively build models of the objects out there in the world that these electrochemical clues relate to. Your normal perception can be understood as a controlled hallucination, kept in check by the data coming in through your senses and by your expectations of the world around you. However, this balance between the internal creativity of the models in your brain and the extent to which they are kept in check can be altered.

Distortions

Psychedelics interact with brain cells to alter their activity in ways that disrupt the normal process of perception. At low doses of a psychedelic, the visual world becomes distorted in reliable ways. Perhaps you experience trails of light following your hands as you move them, or you perceive the walls to be breathing. This can be understood as the result of a temporary impairment in the Default Mode Network (DMN).

The DMN is a group of brain areas that builds up expectations about the world and uses them to keep our perception in check. When its ability to do this is reduced, our internal models of the visual world are free to make guesses about what we’re seeing. We may overestimate the distance of the wall then correct for our error based on the sensory data. Give the newfound flexibility offered to the brain areas involved in this process, they may underestimate, then overestimate. This process can continue on and on, resulting in a pendulating interpretation of the wall’s distance and the perception of the walls breathing. The same goes for the precise position of one’s hand in space over time.

Hallucinations of Interpretation

We’ve all seen shapes in the clouds or perhaps faces in the bark of a tree. Here we are using ambiguous sensory input and are finding internal models in our brains that roughly match the pattern. When on a psychedelic this process goes into overdrive and can result in us mistaking a flower for a lizard or a bowl of spaghetti for a bowl of worms. Why do we tend to see natural shapes like animals and not artificial ones? Why lizards and worms and not buildings and airplanes?

Our visual system evolved to recognize the patterns of the natural world that are relevant to survival and so, when given to opportunity to play, our visual systems show us these natural forms that we were built to detect. We have templates for snakes and spiders deeply programmed into us for example, something that simply isn’t the case for modern dangerous objects like cars and guns.

Our hallucinations can be thought of as what our brains expect to see, something that may also account for why we perceive colors to be highly saturated in the psychedelic state, as we’re experiencing the more pure template of the color than we typically experience in daily life. Understanding the basis of hallucination in our evolutionarily programmed expectation of the visual world also offers a way of understanding why eyes, serpents, and insects are so common in higher-dose psychedelic experiences, as these are highly biologically relevant patterns for us as a species.

Tripping AIs

The models in our brain that underlie perception are constructed by networks of brain cells. The vast computational power of such networks has resulted in their being imitated in modern Artificial Intelligence systems. Artificial Neural Networks such as Google’s Deep Dream can be trained to recognize different images, resulting in the construction of models in the neworks in a way that approximates learning in the brain. When we dream or hallucinate, the contents of these models become active with no sensory input to keep them in check. The same can be done with the artificial Neural Networks. When it is tasked with generating rather than recognizing images, they produce distinctly psychedelic visuals. The fact that this is the case provides striking evidence that certain psychedelic visuals are generated by the models in your brain being pushed into “create” mode.

Geometric Hallucinations

At higher doses geometric patterns can be observed, especially when one closes one’s eyes, thereby excluding the sensory data that would otherwise keep one’s models in check. The patterns of the natural word are geometric in essence, all patterns are. Geometric hallucinations can reveal certain common patterns that are fundamental to our experience of the world. Psychedelics are not the only way to shift the visual brain into a mode where it displays their core patterns. When one “sees stars” after being hit on the head, the visuals being perceived are called phosphenes. Phosphenes can also be seen when pressure is placed on the eyeball. These geometric patterns are routinely experienced in the psychedelic state and have been observed in ancient cave art, leading to the suggestion that early cave art represents the earliest attempts of our species to carry back the experiences of the psychedelic state.

Ancient Imagery

At high doses of classical psychedelics, people routinely experience ancient imagery exemplified in the art of cultures as found in ancient Egypt and Mesoamerica. Mesoamerican cultures are known to have ingested psychedelic mushrooms and it has been suggested that religion in ancient Egypt for a time revolved around the consumption of such mushroom, based on the similarity of depictions of Egyptian crowns to different stages of the development of this type of mushroom. The psychoactive blue lotus is also believed to have been consumed ritually in ancient Egypt. From our contemporary perspective, the styles depicted in the artworks feel as if they originated in these cultures. In reality the imagery of the psychedelic experience may have come first and the art later.

Interdimensional Travel

Another experience that can be had at high doses, especially with DMT, is the experience of visiting another “dimension”. In such an experience the person typically feels as if they have left their body behind and have been transported to another world. Understanding that our perception of the world around us is not the passive experience of a true picture of the world but instead is a controlled hallucination generated by our minds allows us to explain such experiences. As in a dream, the brain is pushed into a creative mode where it is especially decoupled from the sensory data coming from the world around. Certain contents, such as snakes and eyes, can be readily explained by the idea that they reflect deeply programmed visual patterns that are relevant to survival.

Not all of the contents fit neatly into this interpretation however, with visions of technology being particularly difficult to account for. One speculative explanation for such visions is that they reflect geometric hallucinations varying in three dimensions of space and in time.

Tunnels

When having the experience of moving to another dimension it is common to experience passage down a tunnel. It has been suggested by “the Godfather of LSD”, Stan Grof, that the tunnel experience is a memory of the experience of birth, although the fact that those born by cesarean section can still experience tunnels seems to rule this explanation out. Neurobiologist Jack Cowan has argued that spontaneous activity moving across the visual cortex might translate into the experience of concentric rings, as a result of the way that the retina maps onto the surface of the cortex.

Alternate Explanations

Dramatic psychedelic experiences can feel like they reflect actual experiences of pre-existing phenomena that exist outside oneself, rather than being generated from within. This shouldn’t be a surprise as our perception always presents us with the feeling of an objective reality outside of ourselves, even though it is a controlled hallucination created inside us.

The nature of these experiences has led some scientists and philosophers to suggest that the material world is secondary to another reality that we interact with in these states, this ultimate reality varying from a spiritual realm, other dimensions, baseline reality in which our current world is merely a simulation, or a universal consciousness that underpins reality itself. The challenge of these perspectives, other than overturning the current prevailing paradigm, is to explain why low doses produce effects that are so readily explained by our understanding of how normal perception functions in the brain, and at high doses why these other worlds are show patterns that are biologically relevant to us, such as human eyes and natural imagery.

Seeing Under the Hood

Psychedelic experiences can allow us to delve into our deepest personal programming and beyond into our deepest evolutionary programming, bequeathed to us by countless ancestors. Understanding the origin of psychedelic visions as coming from within does not reduce their meaning. We get the opportunity to explore ancient patterns on which our sense of meaning itself is scaffolded, we may confront aspects of Jung’s collective unconscious or realize how our visions connect us in time to our unimaginably long past as an evolved creature. Understanding how such experiences are generated, while interesting, is not necessary to derive pleasure and benefit from them. The value really is in the experience itself, whatever is going on behind the scenes.

 
Top