• Log in with Facebook Log in with Twitter Log In with Google      Sign In    
  • Create Account
  LongeCity
              Advocacy & Research for Unlimited Lifespans

Photo
* * * * - 4 votes

Tickle Your Damn Amygdala's You Neurotic Fools!

amygdala visualization frontal lobes brain exercise

  • Please log in to reply
343 replies to this topic

#121 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 24 January 2020 - 11:15 PM

I would forget all that and go for Epitalon or something else that enhances memory or sleep / dream like Lions Mane, etc.

You may also which to trial Agmatine :: https://www.ncbi.nlm...les/PMC5616847/

 

Zobra you legend. So I looked into that agmatine study, and then started googling experiences of it here and elsewhere on the internet. Regardless to say the anecdotal reports were mixed but was interested because of people saying it is similar to ketamine, which has been used to help ptsd. But then I found this (post 3):  https://www.longecit...ndpost&p=621650

 

 

 

DXM from your local drugstore - cough syrup.

I used it in the Summer of 2009 to exceed all bounds for over three months every day.

One dose of only 15mg (one teaspoon syrup) or 30mg (two) just before going to bed each night.

Bounced out of bed each morning. Running with energy and vicious power all day.

The only two minor sides (personal): it dried my lungs up too much after a while and gave me the runs. I get the runs from almost everything so not likely to happen to you.

And cheap. No major tolerance increase but if it happens then a bit of NMDA antagonist will reverse it for DXM. DXM is an opiate analog, that's why.

I've got a better plan to fix you up.

All your symptoms match perfectly with low Testosterone.

Get your blood tested ASAP and report the value back here or to me by PM.

Values to test: Total Testosterone, TSH, and if possible SHBG, Free Testosterone, T3, T4.

 

So then I started googling DXM and PTSD, and found these so far:

 

https://drugs-forum.com/threads/dxm-for-rapid-treatment-of-severe-ptsd-dissociative-disorders.351706/

https://drugs-forum....my-ptsd.351092/

https://drugs-forum....iatives.229667/

 

 

 

Well, I decided to test it...

And it did help!

I wouldn't recommend it for everyone, but if you really have treatment-resistant PTSD and DON'T take SSRI's, then I think it could really help some people

 

 

sorry i havent posted lately, a lot of crap has happened lately. My dad got 2 types of cancer so life has been hectic

 
jazzyj9 said: ↑
What dosages are we talking about here? Do you have to take it daily?
 
I used around 400mgs. I discontinued use because the ptsd and dissociative amensia have improved greatly. My life is actually possible now... with my dad being sick i need to function at a higher level

 

After discontinuing use this persons ptsd and dissociative amnesia had improved greatly.

 

 

 

I tried dxm for the first time the other night. I have cptsd (my parents were drunks, my dad would smack my mom around, make sexual comments/advances towards me and molested me) as well as bipolar II. After I sobered up I felt relaxed, and I still do even though I'm supposed to be hypomanic right now which is cool. But the biggest difference is to my cptsd. I don't feel as dirty/pathetic, I don't resent my father as much and I'm not as afraid of sex as I used to be. The slightest bit of physical affection used to scare me shitless, so I'd sext a bunch of different people which would make me feel dirty and objectified. Since I used dxm I haven't experienced any of that, I've been able to flirt and cuddle which is definitely a step forward.

In short, it's helping me deal with my shit. 10/10 I'll be trying this in the future.

 

Very positive experience with cptsd and dxm here, and the trauma sounds much more horrific than why I endured.

 

 

I also have ptsd with major dissociative amnesia and ironically enough-it has helped! It forces me into facing my trauma, if that makes sense. I don't dissociate as much after I take it, not nearly as depressed if I am (I'm bipolar) at the time.

 
Now, my pdoc and I both agreed on this being an as needed medication (in my case) because I used to have a major addiction to DXM, and I also need a roughly 3rd plateau dose to experience this. There is a lot of strange things that happen when I take it now, after being diagnosed with not only ptsd but a dissociative disorder as well... basically what that means is to cope with trauma I dissociate. You would think it would make it worse, but if you truly have disorders in this realm (PTSD included) then it may help!

 

 

DXM makes me eat fear. It makes complete sense.. I'm not really sure that using drugs is so wrong or that living a life with drug habits is always a bad thing.. enjoy your life my man, get the most out of it. Good luck.

 

 

 

This is what I got up to reading before I started this post. I'm sure there are more anecdotal reports to read up, but will post this here for now. 

 

I can easily get this stuff in dry cough syrup here in the UK. I've just seen it's in night nurse, ah that stuff makes me moody the next day, but that also has Promethazine Hydrochloride 20 mg which might be the cause of that, not sure.

 

But I'll try a product which only has dxm in, such as this: https://www.boots.co...-100ml-10059366

 

I don't want to take crazy doses of the stuff like 400mg like that guy. But the longecity poster said taking 15-30mg before bed long term really helped him (not for ptsd) and he didn't build up a tolerance. Each 5ml dose is 7.5mg, so I would need to take a 10-20ml dose, and will do it before bed.

 

Anyway I thought I'd post this now, interesting stuff, and readily and cheaply available.

 

Oh and I forgot to post one more thing: https://www.psychcon...-mood-disorders

 

 

 

QUESTION: Can Dextromethorphan Be Used in the Treatment of Mood Disorders?

ANSWER: Dextromethorphan (DM) has long been in use as an ingredient in cough medicines. More recently, in combination with quinidine (DM/Q)—added to extend its duration of action by blocking its metabolism via CYP2D6—it has been approved as a treatment for pseudobulbar affect.

Aside from its known impact on opioid transmission, DM has a complex mode of action. Its main activity in mood disorders may be mediated by sigma-1 receptor agonism, inhibition of serotonin and norepinephrine transporters, and noncompetitive antagonism of N-methyl-D-aspartate (NMDA) glutamate receptors.1 It is uncertain if its relatively low affinity for alpha-1 norepinephrine receptors and modulation of nicotinic receptors will have a contribution in the treatment of mood disorders.

PCN_YourQuestion_250x250%402x_3.jpgAn open-label proof of concept trial in treatment-resistant major depressive disorder patients showed a demonstrable improvement in depressive symptoms after 10 weeks of adjunct treatment with DM/Q 45/10mg every 12 hours.2 Randomized phase 2 studies evaluating the efficacy of DM/Q in treatment-resistant depression are underway.

Navigating the Maze of Treatment-Resistant Depression

Furthermore, there is some preliminary evidence of benefit from DM/Q in cyclical mood disorders. A retrospective chart review focused on bipolar II and NOS patients in the depressed phase of the illness who did not respond to previous treatment. Patients continued with their previous regimen and received additional DM/Q 20/10mg once or twice a day for 90 days. Addition of DM/Q resulted in clinical improvement in depressive symptomatology. Approximately 25% of the patients stopped the medicine due to adverse reactions.3

Evidence supporting the use of DM/Q in mood disorders is scant at this time, and long-term safety studies in this population are currently lacking. Larger-scale, randomized controlled trials are necessary before one can recommend usage of this medication in mood disorders.

— Vladimir Maletic, MD, MS, Clinical Professor of Psychiatry and Behavioral Science, University of South Carolina School of Medicine, Greenville



#122 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 25 January 2020 - 12:29 AM

https://www.reddit.c...for_depression/

 

941giq0efbj31.jpg

 

https://www.reddit.c...t_dissociative/

 

 

One drug I'm not interested in is ketamine. Yes, some people found impressive antidepressant effect from that, but I found ketamine unimpressive compared to DXM.

Somehow DXM exactly hits the spot for whatever is wrong with me. Though I've also seen how when I'm in a good state to start off with, DXM doesn't really improve it much further. Actually, over the past week the best day was the one when I didn't take any DXM and spent a good chunk of time away from home with my father.

 

https://www.reddit.c...ng_others_with/

 

 

Do I see a pattern of DXM helping others with trauma issues?

renderTimingPixel.png

Starting from my very first DXM experiences, it showed me a much better state than my usual baseline. I used to think that's related to antidepressant effects, but over the last year I've come to understand that my issues have more to do with trauma than depression. It's not trauma from particular events, but trauma and neglect over a longer period, of the sort that can cause CPTSD. I have not been diagnosed with such a disorder yet.

It seems like I may also see a pattern here, of others with trauma issues finding DXM puts them in a much better state.

I also note that DXM more than any other drug helped cause seemingly beneficial long term change. It expanded my comfort zone in various ways, though I still feel trapped inside a comfort zone. It also stopped various obsessive negative thought patterns. I've seen very little posted about change which lasts long after DXM.

Still, I'm not sure if DXM helped me or hurt me. I'm worried that there was an element of escapism with this all along. Basically it seems I never really learned to process emotions outside comfort zones, and DXM helped by temporarily eliminating the stuff I don't process. I don't mean simply numbing out, but something more, like a mental state reset which removes the tension which builds up due to that lack of processing. Still, I'm not saying I'm sure I would have done better without DXM.

Last year I was disappointed by how I used DXM repeatedly to accept unacceptable situations. Partly due to this I seem to be intent on staying away from DXM now. I only used it once since early December.

 

https://www.reddit.c..._saved_my_life/

 

 

I felt like a sensation in my mind, like a loading of some sort. In my head I described it as "cascading", like neurons loading and building and building a world in front of me. It also felt in a weird way like when a person starts yelling and their voice gets more and more electronic and modulated and slides up and up in pitch . I hope that makes sense to someone. I walked outside and suddenly realized I lived on a planet. I knew that as a fact before but it was as if I had a deep understanding for the first time. I remember seeing patterns that I didn't see before, like a row of tiles would look like a pattern. I continued seeing more patterns, not like insane patterns but normal everday ones.

The next day I went to work and marveled at how easy it all felt in comparsion to before. I also felt like was...it's hard to describe but the feeling of realizing you've just felt gross and you don't know you did and now you don't. Clothes felt better when they would also feel awkward before. I remember thinking about concepts like dying and gravity and humans. I had this idea that I'm this intelligent hominoid and I live on a planet with all these other humans.

I remember watching a movie and felt like I was watching a movie for real the first time even thought I've seen probably hundreds of movies. Things felt dangerous and I could feel the plot and characters and people etc. I could tell the other people had emotions and feelings and I could suddenly read people's gestures and facial expressions where I couldn't see before.

 

And some other threads:

 

https://www.reddit.c...e_for_ketamine/

 

 

Well I've read a more round view of the substance:

 

Firstly people take 300mg-450mg+ doses to trip balls on it, people get addicted to it, people ruin their lives over it.

Secondly people have said those trips have caused them PTSD and traumas.

 

It's an interesting substance non the less and might have potential. But it's something to tread very carefully. 

 

I use to take night nurse (not as an addict), and only realized this stuff has dxm in. I use to be hella moody the next day, and wonder if this is realted to dxm and my emotional flashbacks, or the Promethazine Hydrochloride made me moody the next day. TBH I never measured night nurse, I just took 2 caps of the liquid, or 2 caps of the pills. 

 

I can see 3 experiments with dxm (at low doses):

 

1. Trying Robitussin Dry Cough medicine which only has dxm as the active ingredient, and at a measured dose of 15mg-30mg (10-20ml) before bed would be the first interesting experiment, and see what happens the next day (like that longecity members post).

 

2. Do the same as 1 but tickle my amygdalas while in bed falling asleep.

 

3. Or just tickle my amygdalas to cause an emotional flashback and dissociation to occur on purpose, and once recognized, then taking it, and see if it helps alleviate or rewire the flashbacks. i.e. do it in the moment

 

 

The good thing is I take a range of supplements/vitamins regularly to help keep my body and mind more healthy than the average person does (like most of us do here), and I only want to experiment with normal doses, so I'm not too worried about addiction or damage. However addiction and damage are real possibilities, and so would need to tread with caution. But another problem would be detoxing too fast (due to the supplements) and taking a normal dose might not have the desired effects I'm looking for (if the desired effect exists with this drug and doses). 

 

Ideally experiment 3 would be the best situation if dxm works as I hope. I.e. I only take it on a need to take basis (when going through an emotional flashback and disassociation).

 

Anyway interesting stuff.

 

 

 

 



sponsored ad

  • Advert
Advertisements help to support the work of this non-profit organisation. To go ad-free join as a Member.

#123 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 25 January 2020 - 01:07 AM

On another note I'm still experimenting with hypnosis, and have had some success in some areas, and not so much in others. But l-theanine seems to be my trancing supplement of choice atm. But I am happy to say my recently developed addictions to weed (over the past year) and alcohol (over the past 6 months) have completely vanished. I still have that thing where if I force myself to drink alcohol, I'll get to a certain point where I will start binge drinking (I've always been like this even though I could go months without alcohol at a time). However recently weed and drinking started becoming automatic behaviours, but especially weed. I could go the whole day without thinking about it, but then in the evening, I would automatically seek to get some. Anyway those automatic behaviours have gone. But I have used the experience to modify my hypnosis script to become addicted to trance (this addiction only occurs when I start listening to a hypnosis file, and ends when the file finishes). But the point is if I can cause this automatic behaviour/addiction to occur like it did with wanting to smoke weed (triggered only in the evenings), it might rewire my brain/subconscious to force me into the trance state as an automatic behaviour (I can only hope).

 

Here is what get's repeated (compounded) 5 times as the first hypnosis suggestion in my script:

 

 

Whenever I mention hypnosis, I'm talking about being in a hypnotic trance. And you find hypnosis so pleasurable and addictive, you find hypnosis so pleasurable and addictive. Hypnosis feels so good, so pleasurable, so wonderful, and so addictive.  And this addiction to hypnosis only starts when you listen to self hypnosis files, and then stops when all the files end. And being in hypnosis feels so pleasurable, feels so wonderful, feels so amazing, and feels stronger than any other addiction or drug you've experienced in your life. And this addiction forces your subconscious mind to automatically enter into hypnosis when listening to self hypnosis files, just like other addictions cause automatic behaviours where resistance is futile.

 

Hope it works, but I've never been able to trigger myself into hypnosis (like some people can) with all the previous suggestions I've used over the years, so it's a long shot. But theanine is definitey helping with trancing. 

 

Now having gotten rid of my most destructive behaviours with hypnosis, I thought I could focus on many other goals such as diet, exercise, or whatever. But hypnosis is something where some things work fast, while other things take time to drill into your subconscious. Take this along with the fact I can't get always trance to hypnosis, and you could be talking about months or even years. So I decided to prioritize on one goal. The frontal lobes pop, amygdala tickling. All my other goals I would say, aren't as hard as the frontal lobes pop. People turn their lives around with diet, exercise, and such all the time without hypnosis. But how many get that magical higher reality of emotions in their lifetime, maybe by the off chance once or twice. So if I spend this year drilling into my subconscious (using hypnosis) one thing, which is to recreate the frontal lobes pop I experienced before, then by the end of the year I might have experienced another one or many of them (we can hope).

 

And so this hypnotic suggestion has been compounded 11 times:

 

 

And every single second of every day, your subconscious knows what feelings emotions and processes were required for your past frontal lobes pop, and will use your past frontal lobes pop experience to create new ones. That's right, every single second of every day, your subconscious will use your past frontal lobes pop experience to create new ones. This is a new subconscious routine being implanted in your subconscious, and your subconscious knows how to do this, and does it.

 

Now the last thing I want to do is summarize my journey since we're hitting the 2 year mark of since this thread was created from then to now:

 

1. My OCD has largely been reduced to the point where I don't notice it at all.

2. My anhedonia has been reduced to where life is more funner than before.

3. My supplement regime has been updated to cover all my essential nutrients, and some extra additions such as NAC I believe has helped with both points 1 and 2.

4. My emotional flashbacks from when I first started experiencing them to now has reduced in intensity and duration. 

5. I experience synchronicity often, though it's more subtle than the obvious connections when I experienced them back when I tried amygdala tickling a little bit back when I was 18.

6. I guess I feel overall more content and positive in life.

7. I often hear clicks in my head, even when ceasing amygdala tickling practice (though they occur extremely more frequently when I practice).

 

I will say though I had to stop practicing amygdala tickling regularly because of the emotional flashbacks, and I came back more down to reality when I stopped tickling. When I was tickling you felt more automatic, I even started to question my own free will.

 

Lastly I want to say, more a note to myself, but I believe the pushing method, as oppose to the visualization method, has merit to it.



#124 zorba990

  • Guest
  • 1,601 posts
  • 315

Posted 25 January 2020 - 02:30 AM

You may wish to explore HDAC inhibitors like tributyrin ir black seed oil. Ref.

http://www.longecity...ation-and-more/

https://www.reddit.c...ibitors_geared/

Amygdala tickling worked better for me than hypnosis but combining them worked even better. Its quite hard to get through your own stuff without a guide though.

#125 zorba990

  • Guest
  • 1,601 posts
  • 315

Posted 26 January 2020 - 03:39 AM

Methylene blue for fear extinction:
https://www.ncbi.nlm...pubmed/25018057

Effects of post-session administration of methylene blue on fear extinction and contextual memory in adults with claustrophobia.

Telch MJ, Bruchey AK, Rosenfield D, Cobb AR, Smits J, Pahl S, Gonzalez-Lima F.
Abstract
OBJECTIVE:
Preclinical studies have shown that low-dose methylene blue increases mitochondrial cytochrome oxidase activity in the brain and improves memory retention after learning tasks, including fear extinction. The authors report on the first controlled experiment to examine the memory-enhancing effects of posttraining methylene blue administration on retention of fear extinction and contextual memory following fear extinction training.
METHOD:
Adult participants displaying marked claustrophobic fear were randomly assigned to double-blind administration of 260 mg of methylene blue (N=23) or administration of placebo (N=19) immediately following six 5-minute extinction trials in an enclosed chamber. Retesting occurred 1 month later to assess fear renewal as indexed by peak fear during exposure to a nontraining chamber, with the prediction that the effects of methylene blue would vary as a function of fear reduction achieved during extinction training. Incidental contextual memory was assessed 1 and 30 days after training to assess the cognitive-enhancing effects of methylene blue independent of its effects on fear attenuation.
RESULTS:
Consistent with predictions, participants displaying low end fear posttraining showed significantly less fear at the 1-month follow-up if they received methylene blue posttraining compared with placebo. In contrast, participants displaying moderate to high levels of posttraining fear tended to fare worse at the follow-up if they received methylene blue posttraining. Methylene blue's enhancement of contextual memory was unrelated to initial or posttraining claustrophobic fear.
CONCLUSIONS:
Methylene blue enhances memory and the retention of fear extinction when administered after a successful exposure session but may have a deleterious effect on extinction when administered after an unsuccessful exposure session.

#126 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 28 January 2020 - 02:51 AM

Ok so I just made a big connection (and remade another), very important ones.

 

So I’ve been taking some dxm for a few days only at 15mg a day the last 2 days and 7.5mg today. And even this low dose, caused me to become dissociative and very moody (most reports of dissociation are from much higher doses).

 

Now this explains, that when before I understood emotional flashbacks, why I would get them the next day and have a fight with my wife after I used night nurse. I always kept saying to her (before I understood I was experiencing emotional flashbacks), that every time I seem to take night nurse, I end up really moody the next day. I always thought it was just the anti histamine in it making me feel shit the next day, not knowing anything about dxm that was in it at the time.

 

Anyway get this. I was feeling dissociative most of the day, and kinda moody, and was easily becoming negative about everything, and then me and the misses have a fight and I go to bed cause I think I’m tired and need to lower my dose of dxm because it’s making me moody (hence why 7.5mg today before bed). Anyway for the first time in a while I do my temple massage with my knuckles for a long while, trying to find any tender spots and then just massaging them.

 

After about 10 minutes of messaging my dissociation has completely gone and all the negative (moody) emotions, completely.

 

Now there are 3 times I remember that stick out how effective the massage is. When I first discovered it by accident when I was writing in a diary and going through and emotional flashback and got rid of it. The second time was when I was in my car and feeling very dissociative and did it when I parked up and it got rid of dissociation (first time I discovered it could do this for dissociation), this experience is jotted down on the 4th page of this thread just look for the word massage. And then today.

 

The funny thing is back when I first discovered this technique before I knew what emotional flashbacks were, I had no idea of dissociation. Since then I begun to recognise dissociation, in most cases it’s very slight, it’s like your vision gets blurry. However I first realised I was dissociating when I put some hypnotic suggestions in my script that whenever I drove my car (post hypnotic reinforcement) my subconscious would find traumas in the background and replace them with forgiveness and love, also at this time when driving my right eyelid would regularly flutter. And I was dissociating bigly when I started messing with NAC, Sarcosine, and increasing my salt intake (sodium chloride). And I think I was tickling a lot. Anyway whatever it was, it was the first time I recognised I was experiencing dissociation, because it was much stronger, and one of the strong dissociation  times was the driving time I was able to get rid of it with a temple massage.

 

Now I realize dissociation and emotional flashbacks are the same thing, or are occurring at the same time. When I’m dissociating, even slightly, blurry vision, I feel moody, even if I don’t realize I’m experience a minor emotional flashback. And with the 2nd time I learnt I could get rid off dissociation with a temple massage because my vision would return back clearer. And then the first time I was able to get rid of a emotional flashback. And now today I was able to get rid off both the dissociation and all negative feelings (minor building up emotional flashback). I’ve just realised I literally have had the tool all along to avoid emotional flashbacks completely.

 

im writing from my phone, and going a bit in circles, but I needed to make this post because I’d been sitting on the answers all this time.

 

disscocitation = emotional flashbacks

got rid of emotional flashbacks twice with a temple massage

got rid of dissociation twice with a temple massage

but really I got rid of all 3 of them all 3 times, I just didn’t understand what they were and what was happening to me until now

 

now you may ask surely it must have not been that effective since you stopped tickling due to the emotional flashbacks. Well I’ll be honest, I didn’t apply it regularly, it was something I discovered and worked, now 3 separate occasions. The thing when I was dissociating slightly and becoming moody, going through a minor emotional flashback which is building up, is I kinda don’t recognise it. I mean if you think about it when you’re experiencing a strong emotional flashback, your current reality and emotions are out of whack on an extreme level, you can be crying, cursing, raging, and feeling very emotional, this is dissociation on the extreme. In fact I had a major emotional flashback last Tuesday which had been building up behind me for awhile even though I wasn’t tickling anymore, then it all came out through talking with a close relative on the phone and my wife beside me, and all that time I was playing FIFA 20 on the PS4 for the entire duration of the phone call 58 minutes. My automatic behaviour (or another me) was on Fifa, and surprisingly I was scoring goals and winning matches, while my awareness was on the phone call.

 

anyway anyway anyway, I’m just writing my thoughts down while it’s fresh. But I don’t think I need to take dxm at all anymore, since I made the connection at the low dose, and I don’t feel with it on it and I get insomnia at these low doses. But today’s and realising my previous experiences and new connections brings me renewed hope, that it might be completely possible to bypass all my emotional flashbacks from tickling, if I start doing the knuckle massaging the temples thing as soon as I recognise dissociative behaviour (blurry type vision), as I was able to get rid of it today and all negative emotions (hours of this feeling gone in 10 minutes).

 

i have no idea if it will work on the really big ones, or if leave it too late that it builds up to big. But I’m pretty sure the first time I discovered it, that was one big one I felt coming on and got rid of it.

 

so it’s time to get back on tickling my amygdalas (which will cause emotional flashbacks), and see if I can bust through all my emotional flashbacks/dissociation with this simple and overlooked technique. I just have to force myself to do it on every single one to tell if this really works.

 

oh I forgot to mention that last bug emotional flashback which had been building up for days and I finally broke last Tuesday. That like the first ones I experienced, actually felt like a release afterward. As you may have read, my later emotional flashbacks I wasn’t sure if I was healing afterward at all or just getting worse. I’m not saying all emotional flashbacks are healing afterward, but that last Sure felt like it in the days that followed.


Edited by Jesus is King, 28 January 2020 - 02:54 AM.


#127 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 31 January 2020 - 07:52 PM

OK I don't think I can implement the temple massage every time, just because the muscles being sore around that area. And yes emotional flashbacks are back since I've started tickling, and not sure a massage can get rid of them every time. 

 

But anyway I started looking into peoples experiences with CBD oil for cptsd, and a few people on reddit said it helped them a little bit, and then found an research paper it might help alleviate some PTSD symptoms, so I got some today. But something I come across with all this CBD stuff, was Anandamide, the bodies natural thing it products for cannaboid receptors.

 

But while there I found this: https://en.wikipedia...and_degradation

 

 

Paracetamol (called acetaminophen in the US and Canada) is metabolically combined with arachidonic acid by FAAH to form AM404.[23] This metabolite of paracetamol is a potent agonist at the TRPV1 vanilloid receptor, a weak agonist at both CB1 and CB2 receptors, and an inhibitor of anandamide reuptake. As a result, anandamide levels in the body and brain are elevated. In this fashion, paracetamol acts as a pro-drug for a cannabimimetic metabolite. This action may be partially or fully responsible for the analgesic effects of paracetamol.[24][25]

 

So a couple hours later I google PTSD and paracetamol, and find this study: https://www.research...onditioned_Rats

 

 

Background: The endogenous cannabinoid system (eCB) has been shown to play an important role in the extinction of aversive memories. AM404, the metabolite of paracetamol, was shown to enhance eCB signaling. Objective: The present study was designed to pre-clinically evaluate the effectiveness of paracetamol against Post-traumatic stress disorder (PTSD). Methods: Contextual Fear conditioning (CFC) was used as animal model of PTSD. The behavioral and biochemical indicators of anxiety were also measured using elevated plus maze (EPM) and corticosterone levels, respectively. Results: Our data showed that the paracetamol treatment (100, 250 and 500 mg/kg) caused significant dose dependent decrease in animal's freezing behavior in fear conditioned rats. The EPM data revealed that the treated rats spent more time in the open arm of elevated plus maze. However, the corticosterone levels remained unaltered. Conclusion: The present study provide robust evidence that the paracetamol, having established safety in humans, holds promise against PTSD and merits further investigation as potential lead compound in anti-PTSD drug development program.

 

So maybe it's time to start experimenting with paracetamol.


Edited by Jesus is King, 31 January 2020 - 07:53 PM.


#128 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 31 January 2020 - 07:57 PM

Acetaminophen Reduces Social Pain: Behavioral and Neural Evidence
 
Abstract

 

Pain, whether caused by physical injury or social rejection, is an inevitable part of life. These two types of pain—physical and social—may rely on some of the same behavioral and neural mechanisms that register pain-related affect. To the extent that these pain processes overlap, acetaminophen, a physical pain suppressant that acts through central (rather than peripheral) neural mechanisms, may also reduce behavioral and neural responses to social rejection. In two experiments, participants took acetaminophen or placebo daily for 3 weeks. Doses of acetaminophen reduced reports of social pain on a daily basis (Experiment 1). We used functional magnetic resonance imaging to measure participants’ brain activity (Experiment 2), and found that acetaminophen reduced neural responses to social rejection in brain regions previously associated with distress caused by social pain and the affective component of physical pain (dorsal anterior cingulate cortex, anterior insula). Thus, acetaminophen reduces behavioral and neural responses associated with the pain of social rejection, demonstrating substantial overlap between social and physical pain.

 
 

 


Over-the-Counter Relief From Pains and Pleasures Alike: Acetaminophen Blunts Evaluation Sensitivity to Both Negative and Positive Stimuli
 
Abstract

 

Acetaminophen, an effective and popular over-the-counter pain reliever (e.g., the active ingredient in Tylenol), has recently been shown to blunt individuals’ reactivity to a range of negative stimuli in addition to physical pain. Because accumulating research has shown that individuals’ reactivity to both negative and positive stimuli can be influenced by a single factor (an idea known as differential susceptibility), we conducted two experiments testing whether acetaminophen blunted individuals’ evaluations of and emotional reactions to both negative and positive images from the International Affective Picture System. Participants who took acetaminophen evaluated unpleasant stimuli less negatively and pleasant stimuli less positively, compared with participants who took a placebo. Participants in the acetaminophen condition also rated both negative and positive stimuli as less emotionally arousing than did participants in the placebo condition (Studies 1 and 2), whereas nonevaluative ratings (extent of color saturation in each image; Study 2) were not affected by drug condition. These findings suggest that acetaminophen has a general blunting effect on individuals’ evaluative and emotional processing, irrespective of negative or positive valence.

 
 

 



#129 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 31 January 2020 - 08:05 PM

Neural hyperactivity in the amygdala induced by chronic treatment of rats with analgesics may elucidate the mechanisms underlying psychiatric comorbidities associated with medication-overuse headache
Abstract
Background

Patients with medication-overuse headache suffer not only from chronic headache, but often from psychiatric comorbidities, such as anxiety and depression. The mechanisms underlying these comorbidities are unclear, but the amygdala is likely to be involved in their pathogenesis. To investigate the mechanisms underlying the comorbidities we used elevated plus maze and open field tests to assess anxiety-like behavior in rats chronically treated with analgesics. We measured the electrical properties of neurons in the amygdala, and examined the cortical spreading depression (CSD)-evoked expression of Fos in the trigeminal nucleus caudalis (TNC) and amygdala of rats chronically treated with analgesics. CSD, an analog of aura, evokes Fos expression in the TNC of rodents suggesting trigeminal nociception, considered to be a model of migraine.

Results

Increased anxiety-like behavior was seen both in elevated plus maze and open field tests in a model of medication overuse produced in male rats by chronic treatment with aspirin or acetaminophen. The time spent in the open arms of the maze by aspirin- or acetaminophen-treated rats (53 ± 36.1 and 37 ± 29.5 s, respectively) was significantly shorter than that spent by saline-treated vehicle control rats (138 ± 22.6 s, P < 0.001). Chronic treatment with the analgesics increased the excitability of neurons in the central nucleus of the amygdala as indicated by their more negative threshold for action potential generation (–54.6 ± 5.01 mV for aspirin-treated, –55.2 ± 0.97 mV for acetaminophen-treated, and –31.50 ± 5.34 mV for saline-treated rats, P < 0.001). Chronic treatment with analgesics increased the CSD-evoked expression of Fos in the TNC and amygdala [18 ± 10.2 Fos-immunoreactive (IR) neurons per slide in the amygdala of rats treated with aspirin, 11 ± 5.4 IR neurons per slide in rats treated with acetaminophen, and 4 ± 3.7 IR neurons per slide in saline-treated control rats, P < 0.001].

Conclusions

Chronic treatment with analgesics can increase the excitability of neurons in the amygdala, which could underlie the anxiety seen in patients with medication-overuse headache.

 


#130 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 31 January 2020 - 08:08 PM

Anxiety‐ and activity‐related effects of paracetamol on healthy and neuropathic rats
Abstract

Paracetamol has recently been suggested to affect emotion processing in addition to alleviating pain in humans. We investigated in adult male Hannover–Wistar rats whether acute intraperitoneally administrated paracetamol affects behavior in tests measuring anxiety, mood, motor activity, and memory. Unoperated rats received saline or a low (50 mg/kg) or high (300 mg/kg) dose of paracetamol, while rats with a spared nerve injury (SNI) model of neuropathy and sham‐operated rats received saline or the low dose of paracetamol. Rats were tested on open‐field (OFT), elevated plus‐maze (EPM), light‐dark box (LDB), novel‐object recognition (NOR), sucrose preference, rotarod, and monofilament tests. In unoperated rats, both the low and high dose of paracetamol reduced line crossings, and grooming time in the OFT, and novel preference in NOR. The high dose of paracetamol increased the time spent in the closed arm in EPM, reduced the number of rearings and leanings in OFT, the time spent in the light box in LDB, and sucrose preference. Paracetamol had no significant effect on the rotarod test measuring motor activity. The low dose of paracetamol suppressed mechanical pain hypersensitivity in SNI rats, without influencing pain behavior in sham‐operated rats. Saline‐ but not paracetamol‐treated SNI rats spent more time than sham‐operated rats in the closed arm in the EPM test. Together the results suggest that a high dose of paracetamol increases anxiety‐like and anhedonic behavior, and impairs recognition memory in unoperated controls, while in neuropathy, a low dose of paracetamol reduces nerve injury‐associated anxiety probably by reducing neuropathic pain.

 


#131 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 31 January 2020 - 08:11 PM

Pain Medication Use Among Patients With Posttraumatic Stress Disorder
Abstract

The relationship of analgesic medication use with posttraumatic stress disorder (PTSD) diagnosis was investigated among a sample of 173 African Americans presenting for routine outpatient visits at an urban mental health center. Seventy-eight (43.5%) of the sample met DSM-IV PTSD criteria. Those with PTSD had significantly higher use of analgesic medication (both opiate and non-opiate), as compared with non-PTSD patients. PTSD symptoms, as measured by the Posttraumatic Symptom Scale, were significantly higher in subjects who were prescribed analgesics. The authors conclude that there may be a relationship between PTSD and use of pain medications warranting further examination of the endogenous opiate system in the pathophysiology of PTSD.

Posttraumatic stress disorder (PTSD) and chronic pain disorder are highly comorbid.1,2 The literature indicates a high degree of co-occurrence between pain and PTSD, regardless of whether the pain is being assessed in patients with PTSD or PTSD is being assessed in patients with chronic pain. Also, they may interact in such a way as to negatively affect the course and outcome of treatment of either disorder.3 The high comorbidity between these disorders has been postulated as being due to either shared vulnerability or mutual maintenance.1,2

PTSD symptoms are associated with greater reporting of physical health problems and symptoms, and also are strongly associated with current pain, overall pain ratings, and pain-related disability.1,4-8 Evidence suggests that PTSD symptoms and pain frequently co-occur on an acute level, yet this association also appears to hold in cases where pain persists beyond the acute phase.1 In one of the first studies looking at the co-occurrence of PTSD and chronic pain, White and Faustman9 reported that 1 in 5 military veterans with PTSD developed chronic pain. Beckham and colleagues4 investigated chronic pain patterns in Vietnam veterans with PTSD and observed that 80% reported chronic pain. McFarlane and co-authors10 investigated the reporting of physical complaints in a sample of firefighters with and without PTSD and found statistically higher rates of musculoskeletal pain (primarily in the back) in those with PTSD, as compared with 21% of those without PTSD.

Also, studies have shown that PTSD symptoms tend to be elevated in patients with chronic pain and fibromyalgia.8,11,12 It appears that between 10% and 50% of patients receiving tertiary-care treatment for chronic pain and related conditions have symptoms that meet criteria for PTSD, as compared with approximately 8% of the population in general.1 For example, approximately 10% of patients referred to a pain clinic met criteria for PTSD.13 The prevalence of PTSD increased when the pain problem directly resulted from a traumatic event.3 Rates of PTSD in patients for which pain is secondary to a motor vehicle accident range from 30% to 50%.3,14-16 PTSD symptoms in individuals who experience work-related injury were 34.7%.17 High rates of PTSD (45%) were reported in hospitalized burn patients 12 months post-injury.18 PTSD-like symptoms were found to be more prevalent in fibromyalgia-syndrome patients.8,11 PTSD subjects also reported more pain, lower quality of life, and more functional impairment, and suffered more psychological distress than PTSD patients not having fibromyalgia syndrome.8 These findings indicate that pain symptoms and chronic pain are prevalent in patients with PTSD and that PTSD symptoms are common in patients with chronic pain.

Despite this relatively large literature on the co-occurrence of pain symptoms and PTSD, we know of no data demonstrating actual differences in pain medication use among those with PTSD versus those without. If such a difference exists, it would be critical to demonstrate this potentially more objective measure, given the complex issues of symptom reporting. Furthermore, it raises potential questions about the role of analgesic use as self-medication in patients with PTSD.

There is evidence that endogenous opioids play a role during the human stress response;19 the endogenous opiate system has therefore been implicated in psychobiological models of PTSD-maintenance.20,21 Although experimental data on opioid mechanisms in PTSD are scarce, a number of investigators have suggested that abnormalities in the endogenous opioid system may be important in PTSD symptomatology.22-24 It has been shown that PTSD patients exhibit lower pain thresholds, lower β-endorphin levels, and decreased production and release of methionineenkephalin and, possibly, stress-induced analgesia.19 Beyond the obvious pain-reducing qualities of opiates, these medications have direct and potent inhibitory effects on neurological systems known to be important for the development of PTSD. For example, Saxe et al.23 showed that morphine administration reduced PTSD symptoms over a 6-month period in children with burn-related pain.

Because of the high degree of comorbidity between chronic pain disorders and PTSD, we hypothesized that pain medication would be prescribed more for patients with PTSD than those without PTSD among an outpatient sample. An additional prediction is that the symptom clusters of numbing/avoidance might be more closely associated with pain medication use than are other symptom clusters. In fact, we find that all PTSD symptom clusters are highly associated with increased pain medication use. Interestingly, the cluster of hyperarousal appears to be the most highly associated with analgesic prescription rates.

 


#132 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 05 February 2020 - 11:56 PM

Now this isn't fresh in my memory, but I better note it down before I completely forget.

 

So last Thursday I had an emotional flashback, after which I've felt very calm since then, makes me think I'll never experience another one, but I've thought that before.

 

Anyway I think it was Saturday night, for the first time in a long time I smoked some weed. I got the usual feelings of shame and seeing my actions and thoughts from a different perspective. But here's the big thing that happened to me. After a long while I finally decide to go to bed. Just as I'm alseep, I sit up in my bed because it feels like my brain is about to explode, pop so to speak. This lasted maybe 3-5 minutes, and then I went to sleep. But it was a big enough thing to wake me up and make me sit up.

 

The next day I had a little bit of the joint left, so I tried to experiment, but nothing happened.

 

But I've noted my weird experiences on weed previously when prior I've done a lot of tickling. The main instances was when I was in my garden chair, and I was just quiet for 15 minutes as I could feel my amygdalas swirling. The other time was painful electric shocks going from my leg to my brain and my whole body convulsing weirdly. And also that same time my thoughts would be sucked into a train of thoughts, seeing images one after another fast, and then coming back to reality. And the common seeing things from different perspective (outside your own ego construction I think).

 

So smoking weed, after a period of time of not smoking it and having done tickling many days prior, gives me effects of amygdala tickling.

 

So I'm thinking there is a connection to serotonin (because weed releases that) or the cannabidiol system in the brain and amygdala tickling.

 

 

 

https://www.ncbi.nlm...les/PMC5928403/

Amygdala–medial prefrontal cortex connectivity relates to stress and mental health in early childhood Abstract

Early life stress has been associated with disrupted functional connectivity between the amygdala and medial prefrontal cortex (mPFC), but it is unknown how early in development stress-related differences in amygdala–mPFC connectivity emerge. In a resting-state functional connectivity (rs-FC) analysis with 79 four- to seven-year-old children, we found a significant correlation between more adverse experiences and weaker amygdala–mPFC rs-FC. We also found that weaker amygdala–mPFC rs-FC was associated with higher levels of aggressive behavior and attention problems. These findings suggest that the impact of stress on emotional circuitry is detectable in early childhood and that this impact is associated with mental health difficulties. Connectivity in this circuit may be useful as a marker for mental health risk and for tracking the efficacy of early interventions.

 

 

https://www.nature.c...1386-018-0055-3

Deficient endocannabinoid signaling in the central amygdala contributes to alcohol dependence-related anxiety-like behavior and excessive alcohol intake
Abstract

Negative emotional states that are associated with excessive alcohol intake, particularly anxiety-like states, have been linked to opponent processes in the central nucleus of the amygdala (CeA), affecting stress-related transmitters and monoamines. This study extends these observations to include endocannabinoid signaling in alcohol-dependent animals. Rats and mice were exposed to chronic intermittent alcohol with vapor inhalation or liquid diet to induce dependence. In vivo microdialysis was used to estimate interstitial concentrations of endocannabinoids [N-arachidonoylethanolamine (anandamide; AEA) and 2-arachidonoylglycerol (2-AG)] and amino acids (glutamate and GABA) in rat CeA. Additionally, we evaluated the inhibition of endocannabinoids clearance enzymes [monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase] on anxiety-like behavior and alcohol consumption in alcohol-dependent rats and mice. Results revealed that alcohol dependence produced decreases in baseline 2-AG dialysate levels and increases in baseline levels of glutamate and GABA. Acute alcohol abstinence induced an enhancement of these dependence-induced effects and the levels of 2-AG and GABA were restored upon alcohol re-exposure. Additional studies showed that the increased CeA 2-AG levels induced by restraint stress and alcohol self-administration were blunted in alcohol-dependent rats. Pharmacological studies in rats and mice showed that anxiety-like behavior and alcohol consumption were increased in alcohol-dependent animals, and these behavioral effects were attenuated mainly by MAGL inhibitors [MJN110 (10 and 20 mg/kg) in rats and JZL184 (1 and 3 mg/kg) in mice]. The present results suggest a key role for endocannabinoid signaling in motivational neuroadaptations during alcohol dependence, in which a deficiency in CeA 2-AG signaling in alcohol-dependent animals is linked to stress and excessive alcohol consumption.

 

 

https://www.ncbi.nlm...les/PMC6482919/

Cannabidiol in the Treatment of Post-Traumatic Stress Disorder: A Case Series
Abstract

Objectives: Cannabidiol (CBD) is a non-psychotomimetic cannabinoid compound that is found in plants of the genus Cannabis. Preclinical research has suggested that CBD may have a beneficial effect in rodent models of post-traumatic stress disorder (PTSD). This effect is believed to be due to the action of CBD on the endocannabinoid system. CBD has seen a recent surge in research regarding its potential value in a number of neuro-psychiatric conditions. This is the first study to date examining the clinical benefit of CBD for patients with PTSD.

Methods: This retrospective case series examines the effect of oral CBD administration on symptoms of PTSD in a series of 11 adult patients at an outpatient psychiatry clinic. CBD was given on an open-label, flexible dosing regimen to patients diagnosed with PTSD by a mental health professional. Patients also received routine psychiatric care, including concurrent treatment with psychiatric medications and psychotherapy. The length of the study was 8 weeks. PTSD symptom severity was assessed every 4 weeks by patient-completed PTSD Checklist for the DSM-5 (PCL-5) questionnaires.

Results: From the total sample of 11 patients, 91% (n = 10) experienced a decrease in PTSD symptom severity, as evidenced by a lower PCL-5 score at 8 weeks than at their initial baseline. The mean total PCL-5 score decreased 28%, from a mean baseline score of 51.82 down to 37.14, after eight consecutive weeks of treatment with CBD. CBD was generally well tolerated, and no patients discontinued treatment due to side effects.

Conclusions: Administration of oral CBD in addition to routine psychiatric care was associated with PTSD symptom reduction in adults with PTSD. CBD also appeared to offer relief in a subset of patients who reported frequent nightmares as a symptom of their PTSD. Additional clinical investigation, including double-blind, placebo-controlled trials, would be necessary to further substantiate the response to CBD that was observed in this study.

 

 

 

https://www.frontier...2019.00971/full

Structural and Functional Connectivity of the Anterior Cingulate Cortex in Patients With Borderline Personality Disorder

Background: Emerging evidences supported the hypothesis that emotional dysregulation results from aberrant connectivity within the fronto-limbic neural networks in patients with borderline personality disorder (BPD). Considering its important role in emotional regulation, the anterior cingulate cortex (ACC) has not yet been fully explored in BPD patients. Therefore, using the seed-based resting state functional connectivity (rsFC) and probabilistic fiber tracking, we aimed to explore the alterations of functional and structural connectivity (SC) of the ACC in patients with BPD.

Methods: A cohort of 50 unmedicated, young BPD patients and 54 sex-, age-, and education-matched healthy controls (HCs) completed psychological tests and underwent rs-fMRI and diffuse tensor imaging (DTI) scanning. Rs-FC analysis and probabilistic fiber tracking were used to plot SC and FC of the ACC.

Results: With the left ACC selected as a seed, BPD patients exhibited increased rsFC and abnormal SC with the right middle frontal gyrus (MFG), and decreased rsFC with the left middle temporal gyrus (MTG), compared with HCs. Additionally, negative cognitive emotion regulation and depressive symptoms both correlated negatively with the rsFC of the left ACC in BPD patients.

Conclusion: Abnormal SC and FC of the ACC underlie the deficient emotional regulation circuitry in BPD patients. Such alterations may be important biomarkers of BPD and thus could point to potential BPD treatment targets.

 

 

https://medicalxpres...line-brain.html

After the 18-month pre-study phase, the monkeys were divided into two groups balanced for body weight, body mass index and depressive behavior. For the next 18 months, 21 monkeys received sertraline in daily doses comparable to those taken by humans while a group of 20 received a placebo. This treatment regimen is analogous to a human taking an antidepressant for approximately five years.

MRI images taken at the end of the treatment phase revealed that in depressed subjects the drug significantly increased the volume of one region of the brain, the anterior cingulate cortex, while decreasing the volume of this same region and the hippocampus in non-depressed subjects. Both of these areas are highly interconnected with other areas of the brain; are critical in a wide array of functions including memory, learning, spatial navigation, will, motivation and emotion; and are implicated in major depressive disorder.

In humans, Shively said, volume differences in neural structures have been noted in depressed and non-depressed individuals, with the most commonly reported differences being smaller volumes of the cingulate cortex and hippocampus in depressed people. One potential mechanism through which drugs such as Zoloft can be effective as antidepressants is by promoting neuron growth and connectivity in these brain regions.

 

 

Zoloft seems to increase the anterior cingulate cortex (ACC). It's also interesting to me the connection between BPD and a smaller ACC, as I think BPD is similar to C-PTSD and emotional flashbacks, as both are thought to occur from childhood trauma. But what's more interesting is Zoloft is prescribed for OCD and is meant to be quite effective. So this is sort of the serotonin connection and this part of the brain, i.e. an SSRI increases the volume of the anterior cingulate cortex.

 

 



#133 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 06 February 2020 - 12:13 AM

https://www.ncbi.nlm...les/PMC3629308/

 

 

Modulation of cognitive and emotional processing by cannabidiol: the role of the anterior cingulate cortex
Effect of CBD on the ACC
 
CBD is associated with increased resting cerebral regional blood flow (rCBF) in the left parahippocampal gyrus and decreased rCBF in the amygdala-hippocampus complex, including the posterior cingulate cortex (Crippa et al., 2004). A functional neuroimaging (fMRI) study found evidence for attenuation of the blood-oxygen level dependent (BOLD) signal in the amygdala and the posterior and ACC in response to the presentation of fearful faces, combined with a reduction in subjective anxiety (Fusar-Poli et al., 2009). CBD also disrupts the functional connectivity between the ACC and amygdala (Fusar-Poli et al., 2010). Taken together, these results point to both an anxiolytic effect of CBD and a critical modulatory role of the ACC. However, Bhattacharyya et al. (2010) found no effect of CBD on ACC activity in a task identical to the one used by Fusar-Poli et al. (2009)—a discrepancy we will be getting back to. To summarize, apart from the emotion-regulating properties of CBD, the CBD–ACC relationship has not been systematically investigated.

 

The interest of CBD came when I read an anecdotal report on reddit that it was able to lessen their emotional flashback significantly. And with my own experiences with weed and amygdala tickling, weed I can confirm on many occasions now has given me amplified amygdala tickling experiences as opposed to subtle ones when being in a sober state.

 

So I'm trying to find the connection why weed can amplify my amygdala tickling experiences a lot, and I think it has something to do with either serotonin, the cannabidol receptors, and the anterior cingulate cortex.

 

It would be interesting to find if there are any natural supplements that can grow the ACC rather than zoloft.



#134 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 06 February 2020 - 12:15 AM

lol googling increase anterior cingulate cortex, this is the first result, how spooky.

 

https://www.ncbi.nlm...pubmed/22156722

 

Increased anterior cingulate cortex and hippocampus activation in Complex PTSD during encoding of negative words.
Abstract

Post-traumatic stress disorder (PTSD) is associated with impaired memory performance coupled with functional changes in brain areas involved in declarative memory and emotion regulation. It is not yet clear how symptom severity and comorbidity affect neurocognitive functioning in PTSD. We performed a functional magnetic resonance imaging (fMRI) study with an emotional declarative memory task in 28 Complex PTSD patients with comorbid depressive and personality disorders, and 21 healthy non-trauma-exposed controls. In Complex PTSD patients--compared to controls--encoding of later remembered negative words vs baseline was associated with increased blood oxygenation level dependent (BOLD) response in the left ventral anterior cingulate cortex (ACC) and dorsal ACC extending to the dorsomedial prefrontal cortex (dmPFC) together with a trend for increased left hippocampus activation. Patients tended to commit more False Alarms to negative words compared to controls, which was associated with enhanced left ventrolateral prefrontal and orbitofrontal cortex (vlPFC/OFC) responses. Severity of child abuse was positively correlated with left ventral ACC activity and severity of depression with (para) hippocampal and ventral ACC activity. Presented results demonstrate functional abnormalities in Complex PTSD in the frontolimbic brain circuit also implicated in fear conditioning models, but generally in the opposite direction, which may be explained by severity of the trauma and severity of comorbid depression in Complex PTSD.

 



#135 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 06 February 2020 - 12:26 AM

Ahh even more interesting things appear.

 

https://www.longecit...ase-motivation/

 

 

 

Evidence for a modulatory effect of sulbutiamine on glutamatergic and dopaminergic cortical transmissions in the rat brain.

Trovero FGobbi MWeil-Fuggaza JBesson MJBrochet DPirot S.

Source

Key-Obs S.A., Centre d'Innovation, 16, rue Leonard de Vinci, 45074 Orleans, 2, Cedex, France. fabricetrovero@aol.com

Abstract

Chronic treatment of rats by sulbutiamine induced no change in density of N-methyl-D-aspartate (NMDA) and (+/-)-alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors in the cingular cortex, but a significant decrease of the kainate binding sites, as measured by quantitative autoradiography. In the same treated animals, an increase of D1 dopaminergic (DA) binding sites was measured both in the prefrontal and the cingular cortex, while no modification of the D2 binding sites was detected. Furthermore, an acute sulbutiamine administration induced a decrease of kainate binding sites but no change of the density of D1 and D2 DA receptors. Acute sulbutiamine injection led to a decrease of the DA levels in the prefrontal cortex and 3,4-dihydroxyphenylacetic acid levels in both the cingular and the prefrontal cortex. These observations are discussed in terms of a modulatory effect of sulbutiamine on both dopaminergic and glutamatergic cortical transmissions.
PMID:10996447

 

 

 

Learn Mem. 2006 Nov-Dec; 13(6): 777–782.
doi: 10.1101/lm.409306

PMCID: PMC1783632

Dopamine D1 receptors in the anterior cingulate cortex regulate effort-based decision making

Abstract

The anterior cingulate cortex (ACC) has been implicated in encoding whether or not an action is worth performing in view of the expected benefit and the cost of performing the action. Dopamine input to the ACC may be critical for this form of effort-based decision making; however, the role of distinct ACC dopamine receptors is yet unknown. Therefore, we examined in rats the effects of an intra-ACC D1 and D2 receptor blockade on effort-based decision making tested in a T-maze cost-benefit task. In this task, subjects could either choose to climb a barrier to obtain a high reward in one arm or a low reward in the other arm without a barrier. Unlike vehicle-treated rats, rats with intra-ACC infusion of the D1 receptor antagonist SCH23390 exhibited a reduced preference for the high-cost– high-reward response option when having the choice to obtain a low reward with little effort. In contrast, in rats with intra-ACC infusion of the D2 receptor antagonist eticlopride, the preference for the high-cost–high-reward response option was not altered relative to vehicle-treated rats. These data provide the first evidence that D1 receptors in the ACC regulate effort-based decision making.

 

I posted in post 107, my previously written down experience of sulbutiamine giving me amygdala tickling type effects when I wasn't tickling years back.

 



#136 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 06 February 2020 - 12:29 AM

https://www.longecit...us/#entry617427

 

 

 

For example, while the anterior cingulate cortex is implicated in executive function and most well known for being a major part of OCD

 

I did not know that. Shows how dumb I am in all this, I should have been googling OCD causes years ago, as I've suffered from it forever, except it's gone into remission since taking NAC (and maybe emotional flashbacks releasing trauma?).

 

Anyway this is probably why Zoloft works so well for OCD (from what I've read), as we know Zoloft increases the volume of the anterior cingulate cortex.



#137 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 06 February 2020 - 12:32 AM

https://www.longecit...n-the-long-run/

 

Int J Neuropsychopharmacol. 2010 May;13(4):421-32. doi: 10.1017/S1461145709990617. Epub 2009 Sep 24.

Modulation of effective connectivity during emotional processing by Delta 9-tetrahydrocannabinol and cannabidiol.

Cannabis sativa, the most widely used illicit drug, has profound effects on levels of anxiety in animals and humans. Although recent studies have helped provide a better understanding of the neurofunctional correlates of these effects, indicating the involvement of the amygdala and cingulate cortex, their reciprocal influence is still mostly unknown. In this study dynamic causal modelling (DCM) and Bayesian model selection (BMS) were used to explore the effects of pure compounds of C. sativa [600 mg of cannabidiol (CBD) and 10 mg Delta 9-tetrahydrocannabinol (Delta 9-THC)] on prefrontal-subcortical effective connectivity in 15 healthy subjects who underwent a double-blind randomized, placebo-controlled fMRI paradigm while viewing faces which elicited different levels of anxiety. In the placebo condition, BMS identified a model with driving inputs entering via the anterior cingulate and forward intrinsic connectivity between the amygdala and the anterior cingulate as the best fit. CBD but not Delta 9-THC disrupted forward connectivity between these regions during the neural response to fearful faces. This is the first study to show that the disruption of prefrontal-subocritical connectivity by CBD may represent neurophysiological correlates of its anxiolytic properties.

 

Neuropsychopharmacology. 2004 Feb;29(2):417-26.

Effects of cannabidiol (CBD) on regional cerebral blood flow.

Animal and human studies have suggested that cannabidiol (CBD) may possess anxiolytic properties, but how these effects are mediated centrally is unknown. The aim of the present study was to investigate this using functional neuroimaging. Regional cerebral blood flow (rCBF) was measured at rest using (99m)Tc-ECD SPECT in 10 healthy male volunteers, randomly divided into two groups of five subjects. Each subject was studied on two occasions, 1 week apart. In the first session, subjects were given an oral dose of CBD (400 mg) or placebo, in a double-blind procedure. SPECT images were acquired 90 min after drug ingestion. The Visual Analogue Mood Scale was applied to assess subjective states. In the second session, the same procedure was performed using the drug that had not been administered in the previous session. Within-subject between-condition rCBF comparisons were performed using statistical parametric mapping (SPM). CBD significantly decreased subjective anxiety and increased mental sedation, while placebo did not induce significant changes. Assessment of brain regions where anxiolytic effects of CBD were predicted a priori revealed two voxel clusters of significantly decreased ECD uptake in the CBD relative to the placebo condition (p<0.001, uncorrected for multiple comparisons). These included a medial temporal cluster encompassing the left amygdala-hippocampal complex, extending into the hypothalamus, and a second cluster in the left posterior cingulate gyrus. There was also a cluster of greater activity with CBD than placebo in the left parahippocampal gyrus (p<0.001). These results suggest that CBD has anxiolytic properties, and that these effects are mediated by an action on limbic and paralimbic brain areas.



#138 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 06 February 2020 - 01:21 AM

So I found an article on another longecity thread about inositol and ocd. I've heard about it, but never tried it. Lots of interesting anecdotal reports, but I'm quite sensitive to supplements these days, so will try to take lower doses rather than 18g.

 

Here is some more googling:

 

Lower In vivo Myo-Inositol in the Anterior Cingulate Cortex Correlates with Delayed Melatonin Rhythms in Young Persons with Depression
Myo-inositol, a second messenger glucose isomer and glial marker, is potentiated by melatonin. In addition to common abnormalities in melatonin regulation, depressive disorders have been associated with reduced myo-inositol in frontal structures. This study examined associations between myo-inositol in the anterior cingulate cortex and the timing of evening melatonin release. Forty young persons with unipolar depression were recruited from specialized mental health services (20.3 ± 3.8 years old). Healthy controls were recruited from the community (21.7 ± 2.6 years old). The timing of dim light melatonin onset (DLMO) was estimated using salivary melatonin sampling. Myo-inositol concentrations (MI/CrPCr ratio) in the anterior cingulate cortex were obtained using proton magnetic resonance spectroscopy. After controlling for age, sex, and CrPCr concentration the depression group had significantly lower MI/CrPCr ratios than healthy controls [F(4, 75) = 11.4, p = 0.001]. In the depression group, later DLMO correlated with lower MI/CrPCr ratio (r = −0.48, p = 0.014). These findings suggest that neurochemical changes in the frontal cortex are associated with circadian disruptions in young persons with depression.

 

 

 



#139 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 06 February 2020 - 01:24 AM

https://www.biologic...0155-0/fulltext

 

Myo-inositol, Glutamate, and Glutamine in the Prefrontal Cortex, Hippocampus, and Amygdala in Major Depression
Abstract
Background

The brains of patients with depression exhibit many changes in various regions. Recently, proton magnetic resonance spectroscopy has been used to measure brain metabolites, using saturation bands to shape the volume of interest. Our a priori hypothesis was that myo-inositol and glutamate were downregulated in the hippocampus and amygdala in depression.

Methods

We measured brain metabolites from the medial prefrontal cortex, hippocampus, and amygdala of 22 drug-naïve, first-episode patients with major depressive disorder and 27 healthy control subjects using 3T proton magnetic resonance spectroscopy.

Results

Compared with healthy control subjects, patients showed statistically significant reductions in myo-inositol levels in all three regions and reductions in glutamate levels in the medial prefrontal cortex. Furthermore, we found significant decreases in the ratios of glutamate to creatine plus phosphocreatine in the medial prefrontal cortex and amygdala. Additionally, the ratios of glutamine to creatine plus phosphocreatine were also decreased in all three regions examined, although not all the participants presented reliable data. Finally, glutamate levels in the medial prefrontal cortex and amygdala have significant correlations with executive function and those in the hippocampus with memory function. Hippocampal myo-inositol was significantly related to blood cortisol.

Conclusions

Our findings indicated abnormal myo-inositol, glutamate, and glutamine levels in the brains of major depressive disorder patients.

 



#140 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 06 February 2020 - 01:33 AM

https://www.ncbi.nlm...pubmed/27053114 The role of inositol 1,4,5-trisphosphate 3-kinase A in regulating emotional behavior and amygdala function.
Abstract

Inositol 1,4,5-trisphosphate 3-kinase A (IP3K-A) is a molecule enriched in the brain and neurons that regulates intracellular calcium levels via signaling through the inositol trisphosphate receptor. In the present study, we found that IP3K-A expression is highly enriched in the central nucleus of the amygdala (CeA), which plays a pivotal role in the processing and expression of emotional phenotypes in mammals. Genetic abrogation of IP3K-A altered amygdala gene expression, particularly in genes involved in key intracellular signaling pathways and genes mediating fear- and anxiety-related behaviors. In agreement with the changes in amygdala gene expression profiles, IP3K-A knockout (KO) mice displayed more robust responses to aversive stimuli and spent less time in the open arms of the elevated plus maze, indicating high levels of innate fear and anxiety. In addition to behavioral phenotypes, decreased excitatory and inhibitory postsynaptic current and reduced c-Fos immunoreactivity in the CeA of IP3K-A KO mice suggest that IP3K-A has a profound influence on the basal activities of fear- and anxiety-mediating amygdala circuitry. In conclusion, our findings collectively demonstrate that IP3K-A plays an important role in regulating affective states by modulating metabotropic receptor signaling pathways and neural activity in the amygdala.

 



#141 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 06 February 2020 - 02:28 AM

https://www.reddit.c...ol_mood_swings/

 

 

This is going to be a long story; but since you asked:

It is apparent that inositol has to be cycled. It's hard to explain the effects, but I would definitely describe my initial inositol experience has very pleasurable. At a 6 gram dose, taken 1 hour before falling asleep at 2 am, I noticed sights, sounds, emotions and tactile sensations had become subjectively more enjoyable. I mixed in another 6 gram dose the following morning in my coffee and I actually ended up taking a 30 minute shower; whereby I was relishing in the wonderful feeling of the warm water hitting my neck and body. I spent much of the next few hours rediscovering some music in my iTunes library; finding songs I had associated with fond memories as a teenager, reconnecting with them, feeling a positive emotional impact. I felt optimistic, not only about my own life situation, but about the state of affairs of people around me. Things got even better when I took my next heaping teaspoon mid afternoon before going to my girlfriend's family's house for dinner. Usually, I dread doing this task every other weekend, but I found myself to be extremely extroverted and genuinely enjoying connecting with my girlfriend's family members. I felt like I was absolutely high on life. I returned home, blended up an inositol smoothie with about 7-8 grams of the powder mixed with some orange juice and frozen berries. I felt wonderfully drowsy, and zonked off to sleep for a solid 13 hours. Day 3 was when the shit turned on me. My tablespoon dose in my coffee made me off the rails hyper. Instead of the usual anxiolytic effects, drowsiness, and hedonistic experiences, I had felt like I had downed a couple 5 hour energy shots. I decided to take a break from the vitamin B8. The next evening, I gave the inositol another shot, albiet finding a very different experience (at around 5 grams). To me, this dosing round felt much like the lexapro trial I had been on a few years back. I had felt sleep deprived despite having slept well the night before, intense brain fog, and hyperawareness to physical sensations such as the soles of my feet toughing the floor. I thought I may have a tolerance issue, so I bombed another 6 grams and felt nice and drowsy again. I had went out later that night, had a few drinks and noticed that I was beginning to feel a general sense of malaise and hopelessness. I felt extremely worn down, irritable, agitated and lethargic whilst paradoxically not feeling the usual calmness inositol was giving me.

It was clear inositol megadosing wasn't a long term solution for anxiety, depression, and well-being I was looking for. Things got very scary 36 hours after my last dose. I felt the worst I had since tapering from benzos the year before. I was experiencing widespread, diffuse pain, depersonalization, powerful and unpleasant hypersensitivity to light and sound. Subjectively I felt a sense of despair, fear and anxiety. The worst was the akathisia; I could not sit still and chill out for any amount of time. Any menial task I would undertake would become overwhelming and I would have to distract myself with something else until that other thing became too unbearable.

I had to go to my parents that night for thanksgiving dinner and it was imperative that I get my shit together before going completely psychotic. I picked up some enteric coated 5HTP, swallowed 100 mg of the natural factors brand pills, and downed 3.5 gs of phenibut with a nice big glass of whiskey. A few beers into dinner and I was feeling no more insanity, save for an odd sensation of electric current running through my body.

Fast forward 4 months later, I give inositol another shot during my girlfriend's birthday part. At this point I had been on several other serotonergic supplements/nootropics/amino acids/herbs to try to replicate the profound experience I had with inositol with no success (actually many of these worsened my mood). I dropped a heaping teaspoon into some juice and chugged it down. I had another wonderful experience, but I again noticed I was more energetic, pleasant, extroverted, and chatty. I genuinely enjoyed connecting with new people I would have otherwise no interest in (waiters, friends of friends, bartenders). About 7 hours after dosing, I felt like I had hit a an emotional wall. I didn't feel awful, but definitely burnt out, angry, irritable, agitated and small things like sound of a wrapper opening were very bothersome and annoying. The feeling subsided the next morning. $70 more of inositol went into the toilet the following day.

I'm having great difficulty with supplements as of late. I want to self medicate with them. I hate most anxiolytic pharms (except for the fun ones that I can't get anymore because I was improperly taking [abusing] them), and I need to find a supplement that gives me the euphoria, mood lift, and anxiety drown-out that I get initially with inositol, phenibut and rhodiola without the unpredictable effects after repeated dosages.

 

While not a long term positive experience with high doses of inositol. The thing that stood out for me was the "I was feeling no more insanity, save for an odd sensation of electric current running through my body.". This is very interesting because of the electric shocks throughout my body when I practice amygdala tickling a lot. Also his first initial dose was very pleasurable, and made music and tactile sensations more enjoyable.

 

It will be interesting if low doses of 2-3g of inositol a day might help accelerate my amygdala tickling work. Also I'm pretty much cycling my supplements these days naturally, as I don't seem to be in an everyday routine with them at the moment, so this along with lower doses should help prevent tolerance. 

 

The last point I wanted to say was, I haven't be explaining some of the nice things I've been experiencing lately. I've been going through periods where I'm really enjoying music, and when driving, I sometimes look at the beautiful country side the world around me, as a beautiful painting. These aren't all the time and consistent but glimpses of joy, compared to where I was a year back where I was very OCD and anhedonic and everything was boring. Hell I spent a big chunk of my time one day watching sonic 3 speedruns and retro gaming history for no reason other than enjoyment. Oh and I forgot one other important thing, I seem to be remembering things from my past, memories I'd forgotten. My long term memory has been pretty blank for awhile now, and my short term memory is pretty crap also. But I'm now remembering more memories from my childhood and growing up more now. So I definitely view all of this as progress, hopefully I'll start unlocking more fun things as I continue tickling and test other things into my stack.

 

Oh I even forget, I got a very subtle epiphany the other day. It was the bigger world view, all my problems seems to trivial. It was subtle, not like my mini pop. But to me, it makes me think trauma, neurosis, whatever negativity gets you stuck in your subconscious for years, constricts your senses, keeps to seeing the world through a dirty glass. But like my mini pop I experienced back in late 2010, I remember your brain really wakes up, like really wakes up. And so this subtle epiphany felt like the brain woke up slightly for a small period of time, and my problems felt so trivial, and it was a good feeling.

 

Anyway this is going to be a very interesting year.



#142 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 07 February 2020 - 07:29 PM

Core I love this forum, but rarely do I just browse it without search terms.

 

Anyway after browsing brain health just now I came across this thread: https://www.longecit...rders/?p=885315

 

https://www.mdedge.c...erasure-anxiety

 

Single-dose propranolol tied to ‘selective erasure’ of anxiety disorders
 

 

BARCELONA – A single 40-mg dose of oral propranolol, judiciously timed, constitutes an outside-the-box yet highly promising treatment for anxiety disorders, and perhaps for posttraumatic stress disorder as well, Marieke Soeter, PhD, said at the annual congress of the European College of Neuropsychopharmacology.

Soeter_Marieke_NETH_web.jpgBruce Jancin/MDedge News

Dr. Marieke Soeter

The concept here is that the beta-blocker, when given with a brief therapist-led reactivation of a fear memory, blocks beta-adrenergic receptors in the brain so as to interfere with the specific proteins required for reconsolidation of that memory, thereby disrupting the reconsolidation process and neutralizing subsequent expression of that memory in its toxic form. In effect, timely administration of one dose of propranolol, a drug that readily crosses the blood/brain barrier, achieves pharmacologically induced amnesia regarding the learned fear, explained Dr. Soeter, a clinical psychologist at TNO, the Netherlands Organization for Scientific Research, an independent nonprofit translational research organization.

“It looks like permanent fear erasure. You can never say that something is erased, but we have not been able to get it back,” she said. “Propranolol achieves selective erasure: It targets the emotional component, but knowledge is intact. They know what happened, but they aren’t scared anymore. The fear association is affected, but not the innate fear response to a threat stimulus, so it doesn’t alter reactions to potentially dangerous situations, which is important. If there is a bomb, they still know to run away from it.”

 

This single-session therapy addressing what psychologists call fear memory reconsolidation is totally outside the box relative to contemporary psychotherapy for anxiety disorders, which typically entails gradual fear extinction learning requiring multiple treatment sessions. But contemporary psychotherapy for anxiety disorders leaves much room for improvement, given that up to 60% of patients experience relapse. That’s probably because the original fear memory remains intact and resurfaces at some point despite initial treatment success, according to Dr. Soeter.

Nearly 2 decades ago, other investigators showed in animal studies that fear memories are not necessarily permanent. Rather, they are modifiable, and even erasable, during the vulnerable period that occurs when the memories are reactivated and become labile.

 

Later, Dr. Soeter – then at the University of Amsterdam – and her colleagues demonstrated the same phenomenon using Pavlovian fear-conditioning techniques involving pictures and electric shocks in healthy human volunteers. They showed that a dose of propranolol given before memory reactivation blocked the fear response, while nadolol, a beta-blocker that does not cross the blood/brain barrier, did not.

However, since the fear memories they could ethically induce in the psychology laboratory are far less intense than those experienced by patients with anxiety disorders, the researchers next conducted a randomized, double-blind clinical trial in 45 individuals with arachnophobia. Fifteen received 40 mg of propranolol after spending 2 minutes in proximity to a large tarantula, 15 got placebo, and another 15 received propranolol without exposure to a tarantula. One week later, all patients who received propranolol with spider exposure were able to approach and actually pet the tarantula. Pharmacologic disruption of reconsolidation and storage of their fear memory had turned avoidance behavior into approach behavior. This benefit was maintained for at least a year after the brief treatment session (Biol Psychiatry. 2015 Dec 15;78[12]:880-6).

“Interestingly, there was no direct effect of propranolol on spider beliefs. Therefore, do we need treatment that targets the cognitive level? These findings challenge one of the fundamental tenets of cognitive-behavioral therapy that emphasizes changes in cognition as central to behavioral modification,” Dr. Soeter said.

 
 
 

Most recently, she and a coinvestigator have been working to pin down the precise conditions under which memory reconsolidation can be targeted to extinguish fear memories. They have shown in a 30-subject study that the process is both time- and sleep-dependent. The propranolol must be given within roughly an hour before to 1 hour after therapeutic reactivation of the fear memory to be effective. And sleep is an absolute necessity: When subjects were rechallenged 12 hours after memory reactivation and administration of propranolol earlier on the same day, with no opportunity for sleep, there was no therapeutic effect: The disturbing fear memory was elicited. However, when subjects were rechallenged 12 hours after taking propranolol the previous day – that is, after a night’s sleep – the fear memory was gone (Nat Commun. 2018 Apr 3;9[1]:1316. doi: 10.1038/s41467-018-03659-1).

“Postretrieval amnesia requires sleep to happen. Sleep may be the final and necessary link to prevent the process of reconsolidation,” Dr. Soeter said. It’s still unclear, however, how much sleep is required. Perhaps a nap will turn out to be sufficient, she said.

Colleagues at the University of Amsterdam are now using single-dose propranolol-based therapy in patients with a wide range of phobias.

“The effects are pretty amazing,” Dr. Soeter said. “Everything is treatable. It’s almost too good to be true, but these are our findings.”

Based upon her favorable anecdotal experience in treating a Dutch military veteran with severe combat-related PTSD of 10 years’ duration which had proved resistant to multiple conventional and unconventional interventions, a pilot study of single-dose propranolol with traumatic memory reactivation is now being planned in patients with war-related PTSD.

“After one pill and a 20-minute session, this veteran with severe chronic PTSD has no more nightmares, insomnia, or alcohol problems, and he now travels the world,” she said.

Her research met with an enthusiastic reception from other speakers at the ECNP session on PTSD. Eric Vermetten, MD, PhD, welcomed the concept that pharmacologic therapy upon reexposure to fearful cues can impede the molecular and cellular cascade required to reestablish fearful memories. This also is the basis for the extremely encouraging, albeit preliminary, clinical data on ketamine, an N-methyl-D-aspartate receptor antagonist, as well as 3,4-Methylenedioxymethamphetamine (MDMA) for therapeutic manipulation of trauma memories.

Vermetten_Eric_2_of_NETH_web.jpgBruce Jancin/MDedge News

Dr. Eric Vermetten

“Targeting reconsolidation of existing fear memories is worthy of looking into further,” declared Dr. Vermetten, professor of psychiatry at Leiden (the Netherlands) University and a military mental health researcher for the Dutch Ministry of Defense.

New thinking regarding pharmacotherapy for PTSD is sorely needed, he added. He endorsed a consensus statement by the PTSD Psychopharmacology Working Group that decried what was termed a crisis in pharmacotherapy of PTSD (Biol Psychiatry. 2017 Oct 1;82[7]:e51-e59. doi: 10.1016/j.biopsych.2017.03.007. Epub 2017 Mar 14).

“We only have two [Food and Drug Administration]-approved medications for PTSD – sertraline and paroxetine – and they were approved back in 2001,” Dr. Vermetten noted. “Research has stalled, and there is a void in new drug development.”

Dr. Soeter’s study of the time- and sleep-dependent nature of propranolol-induced amnesia was supported by the Netherlands Organization for Scientific Research, where she is employed.

 

I've made the text of the juicy bits RED. Sounds like a wonder drug for PTSD. I wonder if I can pop one single 40mg dose of propranalol during or building up to an emotional flashbacks and erase the fear/negative emotions?

 

Only problem is I would never really be able to test them, as I don't know where they originate from, as they're emotional flashbacks, not flashbacks.

 


Edited by Jesus is King, 07 February 2020 - 07:30 PM.


#143 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 07 February 2020 - 07:36 PM

https://www.ncbi.nlm...pubmed/11822998

 

Pilot study of secondary prevention of posttraumatic stress disorder with propranolol.

 

 

Abstract
BACKGROUND:

Preclinical considerations suggest that treatment with a beta-adrenergic blocker following an acute psychologically traumatic event may reduce subsequent posttraumatic stress disorder (PTSD) symptoms. This pilot study addressed this hypothesis.

METHODS:

Patients were randomized to begin, within 6 hours of the event, a 10-day course of double-blind propranolol (n = 18) versus placebo (n = 23) 40 mg four times daily.

RESULTS:

The mean (SD) 1-month Clinician-Administered PTSD Scale (CAPS) score of 11 propranolol completers was 27.6 (15.7), with one outlier 5.2 SDs above the others' mean, and of 20 placebo completers, 35.5 (21.5), t = 1.1, df = 29, p =.15. Two propranolol patients' scores fell above, and nine below, the placebo group's median, p =.03 (sign test). Zero of eight propranolol, but six of 14 placebo, patients were physiologic responders during script-driven imagery of the traumatic event when tested 3 months afterward, p =.04 (all p values one-tailed).

CONCLUSIONS:

These pilot results suggest that acute, posttrauma propranolol may have a preventive effect on subsequent PTSD.

 



#144 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 09 February 2020 - 07:47 PM

https://europepmc.or...le/med/29543135

 

One-week cocoa flavanol intake increases prefrontal cortex oxygenation at rest and during moderate-intensity exercise in normoxia and hypoxia.
 
Abstract 
During exercise in hypoxia, O2 delivery to brain and muscle is compromised, and oxidative stress is elicited. Cocoa flavanols (CF) have antioxidant capacities and can increase blood flow by stimulating endothelial function. We aimed to examine the effects of 7-day CF intake on oxidative stress, nitric oxide production, and tissue oxygenation in response to exercise in normobaric hypoxia (14.3% O2). In a randomized, double-blind, cross-over study, 14 well-trained male cyclists completed four trials: exercise in normoxia or hypoxia, after 7-day CF or placebo intake. Flow-mediated dilation (FMD) was measured before intake of the last dose CF or placebo. One hundred minutes later, 20-min steady-state (SS; 45% V̇o2max) and 20-min time trial (TT) (cycling) were performed. Blood samples were taken. Prefrontal and muscular oxygenation was assessed by near-infrared spectroscopy. At baseline, FMD was increased by CF. Hypoxia increased exercise-induced elevations in lipid peroxidation and antioxidant capacity. CF suppressed exercise-induced lipid peroxidation but did not influence antioxidant capacity. At rest and during SS, prefrontal and muscular oxygenation was decreased by hypoxia. CF elevated prefrontal oxygenation but did not impact muscular oxygenation. During TT, hypoxia accelerated the exercise-induced decrease in prefrontal oxygenation, but not in muscular oxygenation. During TT, CF did not alter prefrontal and muscular oxygenation. CF did not change plasma nitrite, nitrate, and arginine:citrulline. During high-intensity exercise, CF improved neither tissue oxygenation nor performance in well-trained athletes. At rest and during moderate-intensity exercise, CF reduced exercise-induced lipid peroxidation and partially restored the hypoxia-induced decline in prefrontal oxygenation. NEW & NOTEWORTHY For the first time, we showed that CF had beneficial effects on endothelial function at rest, as well as on prefrontal oxygenation at rest and during moderate-intensity exercise, both in normoxia and hypoxia. Moreover, we showed that CF intake inhibited oxidative stress during exhaustive exercise in hypoxia.

 



#145 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 09 February 2020 - 07:57 PM

https://link.springe...rticle-renderer

 

Acute cocoa flavanols intake improves cerebral hemodynamics while maintaining brain activity and cognitive performance in moderate hypoxia
Abstract
Introduction

Acute cocoa flavanols (CF) intake has been suggested to modulate cognitive function and neurovascular coupling (NVC). Whether increased NVC is solely driven by improved vascular responsiveness or also by neuronal activity remains unknown. This study investigated the effects of acute CF intake on cognitive performance, NVC, and neuronal activity in healthy subjects in normoxia and hypoxia (4000 m simulated altitude; 12.7% O2).

Methods

Twenty healthy subjects (age 23.2 ± 4.3 years) performed four trials. Participants performed a Stroop task and “cognition” battery 2 h after acute CF (530 mg CF, 100 mg epicatechin) or placebo intake, and 30 min after initial exposure to hypoxia or normoxia. Electroencephalogram and functional near-infrared spectroscopy were used to analyze hemodynamic changes and neuronal activity.

Results

CF enhanced NVC in the right prefrontal cortex during several tasks (risk decision making, visual tracking, complex scanning, spatial orientation), while neuronal activity was not affected. CF improved abstract thinking in normoxia, but not in hypoxia and did not improve other cognitive performances. Hypoxia decreased accuracy on the Stroop task, but performance on other cognitive tasks was preserved. NVC and neuronal activity during cognitive tasks were similar in hypoxia vs. normoxia, with the exception of increased β activity in the primary motor cortex during abstract thinking.

Conclusions

Acute CF intake improved NVC, but did not affect neuronal activity and cognitive performance in both normoxia and hypoxia. Most cognitive functions, as well as NVC and neuronal activity, did not decline by acute exposure to moderate hypoxia in healthy subjects.

 



#146 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 10 February 2020 - 05:29 PM

Now I've had some interesting experiences with cocoa in the past. I can't remember why I stopped, possibly insomnia. But I do remember being filled with hope and motivation and that I could do anything, as I use to describe it, delusions of grandeur.

 

I then came across a reddit post that mentioned anandamide is also present in cocoa. This is the natural neurotransmitter that works on humans cannabinoid receptors in the brain. So I'm hoping drinking more cocoa will increase anandamide, and I won't have to continue taking expensive cbd oil.

 

Interesting enough reading anandamide wiki page just now: https://en.wikipedia...wiki/Anandamide

 

I found this: 

 

 

A Scottish woman with a rare genetic mutation in her FAAH gene with resultant elevated anandamide levels was reported to be immune to anxiety, unable to experience fear and insensitive to pain. The frequent burns and cuts she suffered due to her hypoalgesia healed quicker than average.[31][32][33]

 

Immune to anxiety, unable to experience fear, things usually associated with the amygdala. So it's interesting this natural cannabinoid elevated in someone can do this.

 

I will say, I haven't experienced an emotional flashback since I started taking cbd oil occasionally. It makes me wonder if I was low in the natural anandamide neurotransmitter. But also it's too soon to tell to conclude anything.

 

What's more interesting according to here: https://www.dummies....e-affects-mood/

 

 

 

Anandamide is a cannabinoid, a chemical that hooks up to the same brain receptors that catch similar ingredients in marijuana smoke. Your brain produces some anandamide naturally, but you also get very small amounts of the chemical from cocoa bean products — chocolate. In addition, chocolate contains two chemicals similar to anandamide that slow the breakdown of the anandamide produced in your brain, thus intensifying its effects.

 

So not only is there small amounts of anandamide in cocoa, but two chemicals similar to it which slow the breakdown of it (not sure what these chemicals are).

 

Plus when experimenting with cocoa before, I was interested in theobromine, because I thought that was the component that use to give me that delusions of granduer, motivation and hope, maybe a little euphoric. But cocoa also has caffeine, PEA, flavonols, and probably other stuff I haven't looked into. A whole bunch of good crap.

 

So my experiment with cocoa has already begun. The good thing this time round is while yesterday I did the usual make it with milk in a pan thing. I actually never knew cocoa can taste good in boiling water, hence why I always made it with milk previously. But the problem with milk and a pan, is I'd always have to clean the pan afterward, because it stuck to the bottom. And I didn't want to microwave my milk + cocoa before because I was worried it would destroy the nutrients.

 

Anyway now I can make it just like people make tea. Boil water, use a big mug and add 3 teaspoons of cocoa, and add some honey. BOOM

 

So the experiment sort of started yesterday, but regardless it has started. I should be consuming around 3 big mugs of cocoa a day, so 9 teaspoons a day, i.e. roughly 50-60g of cocoa.

 

The thing is I pretty much cut all stimulants except cigarettes, because caffeine stresses me the hell out. But cocoa doesn't seem to do that, even on the come down (I wrote about it previously here). But even when I live my life without stimulants, there's no umph, no high so to speak, everything is blah, boring, even with cutting stimulants for a long time. While consuming cocoa today and yesterday, there's a feeling of enthusiasm and non anxiety, even if I'm not overly productive or doing anything interesting. So it will be interesting to see was effects I experience consuming 50g+ of cocoa a day for a couple of weeks.

 

Also you can see here I posted a study showing there are flavanols in processed cocoa: https://www.longecit...ndpost&p=870643

 

 

In this study, the degree of cocoa alkalization caused a progressive, but not complete loss, of flavanol antioxidants, with about 40% retained in lightly dutched cocoas, 25% retained in medium dutched cocoas, and 10% retained in heavily dutched cocoas.

 

And I've had bad side effects with raw cocoa in the past that I chucked it away (can't remember what they were). But store bought cadbury's bornville cocoa works fine for me.

 

Let the cocoa and amygdala tickling experiment begin! And that Scottish woman immune to anxiety and unable to experience fear due to elevated anandamide levels is very very very very interesting!



#147 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 10 February 2020 - 08:24 PM

BOOM BOOM BOOM BOOM!

 

The nutritional connection to amygdala tickling has been found!

 

I am going to post a few studies, and then end on the most important one yet!



#148 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 10 February 2020 - 08:29 PM

https://criobru.com/...onquer-your-day

 

 

Cacao and Cannabinoids: Working To Empower You To Conquer Your Day
 
 

For centuries, people have enjoyed dark chocolate for both recreational and medicinal reasons. The ancient Americans had over 1000 medicinal uses of cacao and its derivatives. It has long been known to have restorative properties as well as elevating mood. We are going to discuss some of the reasons for these mood-enhancing benefits and the similarities with cannabinoids.

Cannabinoids are fat-soluble (lipophilic), often-medicinal, chemical compounds found in plants and mammals and some have medicinal properties as well as causing feelings of euphoria. Of the 85, or so, known cannabinoids, the most well-known natural cannabinoid chemical is Delta-9-tetrahydrocannabinol (THC). THC is the primary psychoactive compound found in the cannabis genus of plants.

In 1996, Daniele Piomelli and fellow researchers isolated a cannabinoid neurotransmitter called anandamide (n-arachidonoylethanolamine or AEA) in chocolate. The interesting thing about anandamide is that it is also a naturally endogenous (internally occurring) cannabinoid found in the human brain and nervous system that plays a role in feelings of overall well-being.      The word anandamide is derived from the Sanskrit word, ananda, meaning, bliss.

This discovery came about through research into cannabinoid receptor sites. A receptor site is a structure on the surface of a cell that can lock onto certain molecules, making it possible to carry a signal through the cell wall — a “lock-and-key” system. It had long been known that the human brain contains receptor sites that interact with cannabis’ THC molecule. It was inevitable that an endogenous, naturally-occurring, chemical — namely: anandamide — would be found to explain the presence of these receptors. Anandamide is very different from THC and, as previously mentioned, is naturally occurring in the brain.  Anandamide gives a general sense of bliss and happiness. 

High levels of anandamide were found in young men who ran or cycled at a moderate rate for about an hour, according to a study published in the journal NeuroReport by the Georgia Institute of Technology and the University of California, Irvine. Anandamide may be responsible for the euphoric feeling some people experience when they exercise that we sometimes call “runner’s high.” Arne Dietrich, the study’s principal investigator believes the body releases cannabinoids to help cope with the prolonged stress and pain of moderate or intense exercise. Studies have linked anandamide to analgesic reactions produced by the body during exercise, especially by running.

people-2592247_1920_medium.jpg?v=1557254

While there is very little anandamide in cacao, there are 2 important chemicals found in much higher concentrations in cacao that are more than likely the reason why we notice the mood elevating effects from cacao: N-oleolethanolamine (OEA), a known inhibitor of weight gain in mice, and N-linoleoylethanolamine (18:3 NAE), a known anti-inflammatory molecule — like anandamide — via the TRPV1 receptor). These two structural cousins of anandamide both inhibit the metabolic breakdown of anandamide and could potentially inhibit the breakdown of other cannabinoids including THC and CBD causing these compounds to stick around longer, providing even more benefits

All of these mood-enhancing benefits are compounded by theobromine, the primary stimulant in chocolate. Theobromine is a known vasodilator that acts as a “driver” of nutrients and chemicals into the tissues. It could also be responsible for amplifying the effects of the anandamide found in chocolate as it is also a mood enhancer.

In addition to anandamide and theobromine, cacao has serotonin, Phenylethylamine, and dopamine, all of which help us to feel more optimistic and peaceful. All of these combined provide us with an overall sense of well being while providing gentle, natural, long-lasting energy.

The reasons to love chocolate are many and if you consume high cacao content dark chocolate, or better yet, Crio Bru, you will not have to worry about the fat and calories. So Please...Drink in peace!

 



#149 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 10 February 2020 - 08:32 PM

https://hellomd.com/...-bliss-molecule

 

The word anandamide may sound like chemistry book jargon, but its name was actually derived from the Sanskrit word for “bliss” or “joy.” Anandamide is often dubbed the “bliss molecule,” because it helps drive feelings of pleasure. As a neurotransmitter, it’s also involved in modulating memory, motivation and appetite, among other functions.

 

THE CONNECTION BETWEEN ANANDAMIDE & CANNABIS

Anandamide was one of the first endocannabinoids discovered by scientists. What makes anandamide especially unique is that it interacts with the same endocannabinoid receptors that the tetrahydrocannabinol (THC) in cannabis does. This is why some folks are able to experience feelings of euphoria and contentment when consuming THC.

RELATED: WHEN MARIJUANA MAKES YOU FEEL ANXIOUS

Even though THC and anandamide bind to the same receptors, anandamide is a much more fragile molecule. After it’s produced in the brain, anandamide is quickly broken down into other compounds by an enzyme called fatty acid amide hydrolase (FAAH). The slower FAAH works, the longer anandamide stays in the body—and the longer it promotes the blissful feeling it’s named for. This chemical pathway is of major interest to scientists, who think that it may lead to treatments for disorders that involve the nervous system, like anxiety.

What’s more, anandamide has been discovered in two foods: chocolate and black truffles.

ANANDAMIDE MAY BE ONE REASON CHOCOLATE IS SO CRAVEABLE

Chocolate contains more than 300 compounds, including anandamide. While sugar is mostly responsible for making chocolate delicious, scientists think these other compounds, such as caffeine and the stimulant theobromine, also enhance the satisfaction we get when we eat it.

An often-cited study found that chocolate not only contains small amounts of anandamide, but two other compounds that slow down its breakdown, which in theory would allow anandamide to circulate longer in the system.

Researchers found anandamide in cocoa solids, which give dark chocolate its intense flavor. This suggests that the darker the chocolate, the higher the likelihood that it contains anandamide. Milk chocolate, on the other hand, is usually made with a much lower percentage of cocoa solids.

If you’ve ever wondered how to choose dark chocolate with the right percentage of cacao, check out this handy buying guide. And consider applying what you’ve learned the next time you shop for a chocolate-covered cannabis edible.

BLACK TRUFFLES ALSO CONTAIN ANANDAMIDE

The winter black truffle also contains anandamide. Initially, this discovery puzzled researchers. Truffles don’t contain endocannabinoid receptors, so why would they need to produce anandamide?

They theorized that it might be because of evolution. Certain animals, like pigs and dogs, have endocannabinoid receptors. The anandamide could provide a positive reward to the animals that eat the truffles, encouraging them to seek out more and spread the spores.

It’s no secret that truffles carry a hefty price tag. (An ounce of black truffles will set you back at least $100.) So unless you have access to a truffle discount, there may be more cost-efficient ways to feel a little bliss.

HEADS UP: TOO MUCH ANANDAMIDE MAY HAVE A DOWNSIDE

While anandamide is best known for promoting bliss, research done on mice suggests that it’s also involved in stimulating the appetite. In fact, some researchers think that the entire endocannabinoid system, including anandamide, may play an important role in the development of obesity.

As more research is done on endocannabinoids, it’ll be interesting to find out how people can harness the power of anandamide, and if you can really feel its blissful effects from eating certain foods. In the meantime, you can always count on a little chocolate to boost your mood.



sponsored ad

  • Advert
Advertisements help to support the work of this non-profit organisation. To go ad-free join as a Member.

#150 Mr Serendipity

  • Topic Starter
  • Guest
  • 982 posts
  • 19
  • Location:UK
  • NO

Posted 10 February 2020 - 08:37 PM

https://www.frontier...2017.00166/full

 

 

Metabolism of the Endocannabinoid Anandamide: Open Questions after 25 Years
 

Cannabis extracts have been used for centuries, but its main active principle ∆9-tetrahydrocannabinol (THC) was identified about 50 years ago. Yet, it is only 25 years ago that the first endogenous ligand of the same receptors engaged by the cannabis agents was discovered. This “endocannabinoid (eCB)” was identified as N-arachidonoylethanolamine (or anandamide (AEA)), and was shown to have several receptors, metabolic enzymes and transporters that altogether drive its biological activity. Here I report on the latest advances about AEA metabolism, with the aim of focusing open questions still awaiting an answer for a deeper understanding of AEA activity, and for translating AEA-based drugs into novel therapeutics for human diseases.

 
From Phytocannabinoids to Endocannabinoids

Cannabis (Cannabis sativa or Cannabis indica) extracts have been used in folklore medicine for centuries, and at present the potential benefits and harms to human health of legalizing the therapeutic and/or recreational cannabis use are a major issue for public authorities and opinion leaders worldwide. It took almost 30 years after the isolation of the psychoactive ingredient of cannabis, ∆9-tetrahydrocannabinol (THC) in 1964 (Gaoni and Mechoulam, 1964) to discover the first endogenous lipophilic molecule able to activate the same G protein-coupled type-1 (CB1) and type-2 (CB2) cannabinoid receptors activated by THC (Pertwee et al., 2010). In 1992 this “endocannabinoid (eCB)” was found to be N-arachidonoylethanolamine, also known as anandamide (AEA), the “amide of the inner bliss = ananda” (Devane et al., 1992). Shortly after the identification of AEA, another major eCB was discovered, 2-arachidonoylglycerol (Mechoulam et al., 1995Sugiura et al., 1995), and both compounds are still recognized as the two main members of an ever-growing family of bioactive lipids (Mechoulam et al., 2014). Indeed, other important ω-6 (n-6) fatty acid compounds with cannabimimetic properties, such as N-arachidonoyldopamine, 2-arachidonoylglycerylether (noladin ether) and O-arachidonoylethanolamine (virodhamine), have been listed among eCBs (Fezza et al., 2014), along with ω-3 (n-3) fatty acid derivatives like N-docosahexaenoylethanolamine (Brown et al., 2010). Additionally, “eCB-like” compounds such as N-palmitoylethanolamineN-oleoylethanolamine, and N-stearoylethanolamine have been shown to exert a CB1CB2-independent “entourage effect” that potentiates the activity of eCBs at their receptor targets (Ben-Shabat et al., 1998). More recently, two novel “eCB-like” compounds derived from juniperonic acid, the ω-3 structural isomer of arachidonic acid (AA), were identified in the plant kingdom, suggesting that distinct N-acylethanolamines may occur in different monophyletic taxa (Gachet et al., 2017).

In the last 25 years, AEA and congeners have been shown to play key biological activities, in both the central nervous system (Maccarrone et al., 2014Di Marzo et al., 2015Soltesz et al., 2015Curran et al., 2016), and the periphery (Maccarrone et al., 2015Benyó et al., 2016Jourdan et al., 2016Sharkey and Wiley, 2016Wang et al., 2016). Such a multifaceted ability of AEA to impact on virtually every system of human body (and well beyond humans along the phylogenetic tree) depends on a multiplicity of receptor targets that include, besides CB1 and CB2, transient receptor potential vanilloid-1 (TRPV1) channels, G-protein coupled receptors 55 (GPR55) and 119 (GPR119), and peroxisome proliferator activated receptors (PPARs; reviewed in Maccarrone et al., 20142015Di Marzo et al., 2015Soltesz et al., 2015Benyó et al., 2016Curran et al., 2016Jourdan et al., 2016Sharkey and Wiley, 2016Wang et al., 2016). These receptor-mediated activities of AEA, the underlying signal transduction pathways and the related target diseases, will not be covered in the present review, which focuses on metabolism, storage and trafficking that control endogenous content, and thence biological activity, of AEA. The aim is to put in a better perspective open questions that remain to be answered for a deeper understanding of AEA activity, and for the possible translation of AEA-based drugs into novel therapeutics for human diseases.

Metabolism of AEA

Shortly after the discovery of AEA, it was found that its biosynthesis occurs by release from membrane phospholipid precursors (Di Marzo et al., 1994Cadas et al., 1997). A striking feature that emerged in the following years was that AEA biosynthetic pathways are apparently redundant (for reviews see Ueda et al., 2013Fezza et al., 2014Cascio and Marini, 2015Battista and Maccarrone, 2017). The best characterized enzymes that synthesize AEA are shown in Table 1, and their reactions are schematically depicted in Figure 1. Among these routes, the sequential action of a Ca2+-dependent (Cadas et al., 1997Ogura et al., 2016) or independent N-acyltransferase (NAT or iNAT, respectively; Jin et al., 20072009), and then of N-acyl-phosphatidylethanolamine (NAPE)-specific phospholipase D (NAPE-PLD; Okamoto et al., 2004) appears the most relevant biosynthetic pathway of AEA.

 
TABLE 1
fnmol-10-00166-t001.gif

Table 1. Main biosynthetic, hydrolytic and oxidative enzymes of N-arachidonoylethanolamine (or anandamide (AEA)).

 
 
FIGURE 1
fnmol-10-00166-g001.gif

Figure 1. Metabolic pathways of AEA synthesis, degradation and oxidation. See text for details. Abbreviations: AEA, N-arachidonoylethanolamine (anandamide); NAT, N-acyltransferase; NAPE, N-acyl-phosphatidylethanolamine; NAPE-PLD, NAPE-specific phospholipase D; NArPE, N-arachidonoyl-phosphatidylethanolamine; ABHD4, α/β-hydrolase domain 4; GDE, glycerophosphodiesterase; PLC, phospholipase C; PTPN22, non-receptor protein tyrosine phosphatase 22; sPLA2, soluble phospholipase A2; FAAH, fatty acid amide hydrolase; NAAA, N-acylethanolamine-hydrolyzing acid amidase; COX-2, cyclooxygenase-2; LOX, lipoxygenase; P450s, cytochrome P450 monooxygenases; PG-EA, prostaglandin-ethanolamide; HETE-EA, hydroxyeicosatetraenoyl-ethanolamide (hydroxy-AEA); EET-EA, epoxyeicosatrienoyl-ethanolamide.

 
 

Degradation of AEA into AA and ethanolamine also depends on multiple enzymes. Shortly after the discovery of the first AEA amidase activity in different cell types (Koutek et al., 1994Hillard et al., 1995), it was found that fatty acid amide hydrolase (FAAH) is the main responsible for AEA cleavage (Cravatt et al., 1996Giang and Cravatt, 1997). Cloning, crystal structure, kinetic properties and body distribution of FAAH have been extensively reviewed (Cravatt and Lichtman, 2003McKinney and Cravatt, 2005), and faah knockout mice have definitively confirmed its pivotal role in controlling AEA tone in vivo, because these animals have ~15-fold higher levels of AEA than wild-types (Cravatt et al., 20012004). The best characterized enzymes that degrade AEA are shown in Table 1, and their reactions are schematically depicted in Figure 1 (see Fezza et al., 2014; and Battista and Maccarrone, 2017; for extensive reviews).

In addition to hydrolytic pathways, AEA can be subjected to oxygenation by cyclooxygenase-2 (COX-2; Kozak et al., 20012002Rouzer and Marnett, 2011Hermanson et al., 2013), 5-,12- and 15-lipoxygenase (5-/12-/15-LOX; Hampson et al., 1995Edgemond et al., 1998Van der Stelt et al., 2000), as well as by several cytochrome P450 monooxygenases (P450s; Snider et al., 2010Urquhart et al., 2015), as shown in Table 1. COX-2 turns AEA into prostaglandin-ethanolamides (PGs-EA), while LOXs convert it into hydroxy-anandamides or hydroxyeicosatetraenoyl-ethanolamides (HETEs-EA), and P450s into epoxyeicosatrienoyl-ethanolamides (EETs-EA), as schematically depicted in Figure 1. Accumulated evidence suggests that oxygenated derivatives of AEA have a biological activity of their own, but their impact on human health and disease remains to be clarified. Incidentally, it should be noted that recent data suggest that in vivo COX-2 might even prefer eCBs over AA as natural substrates (Hermanson et al., 2013), widening the spectrum of lipid signals that can be affected by COX-2 inhibitors like nonsteroidal anti-inflammatory drugs (Hermanson et al., 2014). Therefore, it seems urgent to further interrogate the impact of these large and widely consumed therapeutic drugs on eCB metabolism in pathophysiological processes of our body.

Storage and Trafficking of AEA

The classical “dogma” that AEA is synthesized and released on demand via hydrolysis of cell membrane phospholipid precursors has been recently revisited (Maccarrone et al., 2010Min et al., 2010), also on the basis of unexpected evidence for intracellular reservoirs and transporters of eCBs. These new entities have been shown to drive intracellular trafficking of eCBs, thus adding a new dimension to the regulation of their biological activity (Maccarrone et al., 2010).

Storage of AEA has been documented in adiposomes (lipid droplets), that constitute a dynamic reservoir for the sequestration of this eCB (Oddi et al., 2008). Remarkably, confocal microscopy and biochemical studies revealed that also FAAH-1 (Oddi et al., 2008) and FAAH-2 (Kaczocha et al., 2010) are spatially associated with lipid droplets, and that cells with a larger adiposome compartment have enhanced AEA catabolism by both enzymes. FAAH-2 displays also a putative N-terminal hydrophobic region as a functional lipid droplet localization sequence (Kaczocha et al., 2010). Overall, these findings suggest that adiposomes may have a critical role in accumulating AEA, and possibly in connecting plasma membrane to internal organelles along the metabolic route of this eCB. In line with these data, depletion of a pre-existing pool of 2-arachidonoylglycerol has been recently shown as a key event in sperm activation (Miller et al., 2016), speaking against the on demand synthesis of this eCB much alike that of AEA.

Whatever the physiological relevance of AEA accumulation, the number of alternate targets that a single cell can have for this eCB poses the question of how AEA can reach the right target in a timely manner and at a suitable concentration for effective action. It should be recalled that the lipid nature of AEA hampers its free movement in aqueous mediums like cytosol. Thus, intracellular AEA transporters (AITs) should exist that ferry AEA to the correct final destination, like: (i) endoplasmic reticulum (ER) for degradation by FAAH-1/FAAH-2; (ii) adiposomes for accumulation, degradation by FAAH-1/-2 or oxidation by COX-2 or LOXs; (iii) mitochondria for oxidation by COX-2 or P450 s, and possible activation of CB1 (Bénard et al., 2012); (iv) lysosomes for degradation by P450s or NAAA; or (v) nucleus for activation of PPARs (Figure 2). Interestingly, AITs have been indeed found in different cell types, and include fatty acid binding proteins (FABPs; Kaczocha et al., 2009), heat shock protein 70 (HSP70) and albumin (Oddi et al., 2009), FAAH-1-like AEA transporter (FLAT-1; Fu et al., 2011), and potentially sterol carrier protein 2 (SCP-2; Liedhegner et al., 2014). Of note, the role of a specific AIT that may deliver AEA where and when needed has been recently demonstrated in a study showing the ability of FABP5 to drive AEA to nuclear PPARs (Kaczocha et al., 2012).

 
FIGURE 2
fnmol-10-00166-g002.gif

Figure 2. Intracellular and extracellular trafficking of AEA. See text for details. 3D structures were drawn by using the following source files: 5TGZ.pdb human (CB1); 5IRZ.pdb rattus norvegicus (TRPV1); 2I4J.pdb ppar-gamma human (PPARs); 4LKP.pdb apo human epidermal fatty acid binding protein (FABP5); 1MT5.pdb rattus norvegicus (FAAH-1); 4NRE.pdb human 15-lipoxygenase-2 (LOXs); 4RRW.pdb apo murine COX-2; 2JJN.pdb closed cytochrome P450 EriK (P450 s). Crystal structures are not yet available for CB2, GPR55 and GPR119, therefore to represent these G protein coupled receptors data of β1-adrenergic receptor (5A8E.pdb) were used. Finally, for FAAH-2 and NAAA the same 3D structure as FAAH-1 was used, in the absence of crystallographic data. All 3D structures were drawn by means of the Jsmol software, a JavaScript framework (open source) available at www.RCSB.org. Abbreviations: CB1, type-1 cannabinoid receptor; CB2, type-2 cannabinoid receptor; AEA, N-arachidonoylethanolamine (anandamide); TRPV1, transient receptor potential vanilloid-1; GPR55, G-protein coupled receptor 55; GPR119, G-protein coupled receptor 119; PPARs, peroxisome proliferator activated receptors; FAAH, fatty 523 acid amide hydrolase; NAAA, N-acylethanolamine-hydrolyzing acid amidase; COX-2, cyclooxygenase-2; LOXs, lipoxygenases; P450s, cytochrome P450 monooxygenases; AET, AEA extracellular transporter; AITs, intracellular AEA transporters; AMT, AEA membrane transporter; ER, endoplasmic reticulum; FABP5, fatty acid binding protein 5; LRs, lipid rafts.

 
 

Much alike intracellular trafficking, cell-to-cell transport of AEA requires AEA extracellular transporters (AETs), as depicted in Figure 2. The identity of such AETs remains to be elucidated, but recent data point to microvesicles as good candidates (Gabrielli et al., 2015). Also FABP4 (Hotamisligil and Bernlohr, 2015), albumin and HSP70 (Shevtsov and Multhoff, 2016) are known to be secreted extracellularly, thus it is conceivable that they play a role as AETs. Yet, their actual contribution to AEA extracellular transport remains to be demonstrated. Noteworthy AETs might drive AEA to CB1 receptors localized within or outside cholesterol-enriched membrane microdomains like lipid rafts (LRs), thus modulating receptor activation. Indeed, it has been shown that cholesterol present in LRs reduces AEA binding to CB1, and hence signal transduction triggered thereof (Oddi et al., 2012).

Finally, transport of AEA across the plasma membrane has been highly debated over the last 20 years, with a number of studies pointing towards the existence of a true AEA membrane transporter (AMT), but as many pointing against it (see Fowler, 2013Nicolussi and Gertsch, 2015Deutsch, 2016). While the discussion of evidences in favor or against AMT goes beyond the scope of this review, is worthwhile to stress that a saturable uptake of AEA can certainly be due to a facilitated transport or to a passive diffusion driven by intracellular hydrolysis, most likely by FAAH-1. Yet, a saturable export of AEA, that has been reported shortly after the discovery of this eCB (Hillard et al., 1997), and then by several independent studies (see Chicca et al., 2012, and references therein), cannot depend on a concentration gradient driven by intracellular hydrolysis. Therefore, a clear evidence of a saturable bidirectional transport of AEA appears to strongly speak in favor of the existence of a true AMT. A reason for the still missing molecular identity of an AMT could be that it is a multimeric protein rather than a monomer, making it difficult to identify and isolate all subunits and reconstitute a functional AMT from them. Preliminary evidence for the assembly of an AEA uptake machinery within caveolae/lipid rafts has been indeed reported (McFarland et al., 2004), though its composition has not yet been determined.

Open Questions and Future Directions

The previous sections have shown a rather complex AEA metabolism, storage and trafficking, strongly suggesting that for every cell it is important to properly synthesize, degrade and transport this eCB. In fact the multiplicity of pathways that lead to AEA release from membrane phospholipid precursors, and then to its cleavage (Table 1; Figure 1), should have not passed natural selection unless they might have conferred an advantage for survival. It is apparent that FAAH-1 is a key controller of AEA tone in vivo, and therefore many inhibitors have been developed over the last decade to block its activity, thus enhancing content and biological activity of AEA. Despite the potential of these compounds as innovative therapeutics (reviewed by Bisogno and Maccarrone, 2013Fowler, 2015), better drugs are still needed for effective cure or slowing down of human pathologies. In this context, it seems noteworthy that subtle differences exist between rodent and human FAAH-1 that impact on the efficacy of inhibitors, thus leading to rodent/human ratios of IC50 values from ~0.3 to ~4.0 (Di Venere et al., 2012). The consequences on translation of preclinical studies to the patient’s bedside are apparent. Moreover, the selectivity of an inhibitor of FAAH-1 should be carefully checked towards effects on other components of the so-called “endocannabinoid system (ECS)” which includes different metabolic enzymes and carriers of AEA, in addition to the proteins that bind, metabolize and transport 2-arachidonoylglycerol (Maccarrone et al., 2014). Thus, a valuable FAAH-1 inhibitor should show at least very little (if any) effects on other ECS proteins, as well as potential non-ECS off-targets. Yet it is not always obvious that these tests are performed during new drug development programs (Di Venere et al., 2012). In this context, a dramatic outcome of a recent phase I clinical trial with the purported FAAH-1 inhibitor BIA 10-2474 led to an unanticipated severe toxic cerebral syndrome whose underlying mechanisms remain unknown (Kerbrat et al., 2016). This adverse event clearly demonstrates the importance of a thorough characterization of the specificity of any new FAAH-1-oriented drug. Indeed, conclusions of an ad hoc temporary specialized scientific committee and the fact that phenomena resembling those seen in humans with BIA 10-2474 have not been reported in the literature in any of the numerous animal studies and clinical trials with various inhibitors of FAAH (e.g., PF-04457845, JNJ-42165279, SSR-411298, V-158866 and URB597, just to list those with more advanced programs), strongly (yet not conclusively) suggest that off-target effects of BIA 10-2474 itself or a metabolite thereof, and not FAAH-1 inhibition was the cause of adverse reactions (Mallet et al., 2016Edan and Kerbrat, 2017). Moreover, it should be recalled that unexpected regulators of FAAH-1 activity are emerging, such as membrane cholesterol that favors the access of AEA to the enzyme active site (Dainese et al., 2014). Thus it remains to be clarified to what extent the lipid environment may tune FAAH-1 activity in vivo, and how it can be exploited to design more effective FAAH-1 inhibitors.

Unlike degradation, the key enzymes for AEA biosynthesis are still to be identified, though NAPE-PLD seems to play a pivotal role. Anyhow selective inhibitors of AEA biosynthetic enzymes are not yet available, making it difficult to dissect the contribution of each pathway to the overall synthesis of this eCB. In addition, as shown in knockout animals the lack of an enzyme can be compensated by an alternate route. Therefore, development of effective inhibitors of distinct AEA biosynthetic enzymes, and of conditional knockout mice where a specific enzyme can be switched off at will, is deemed necessary to boost our understanding of AEA biosynthesis. Better methods and more accurate measurements are required to this aim. This has been indeed the case for FAAH-1, whereby activity-based protein profiling has allowed to identify truly selective blockers with low or no activity towards off-targets (Simon and Cravatt, 2010). Additionally, the quest for 3D structures of AEA metabolic enzymes should be actively pursued, because this type of information can certainly favor drug development. To date, a 3D crystal structure has been obtained only for FAAH-1 (Bracey et al., 2002), for which unsurprisingly different classes of effective inhibitors have been developed and investigated in detail (Table 2).

 
TABLE 2
fnmol-10-00166-t002.gif

Table 2. Main classes of human fatty acid amide hydrolase-1 (FAAH-1) inhibitors.

 
 

Of basic importance appears to be the identification and characterization of in vivo activity of distinct AITs and AETs that may drive AEA to the right target within and outside the cell (Figure 2). Each AIT and/or AET could be a further regulatory player within the cell, adding a new dimension to the already complex AEA-dependent signaling thus representing a target for novel AEA-based drugs.

Finally, genetic manipulation of the faah-1 gene allowed to generate not only faah-1 knockout (Cravatt et al., 2001) but also faah-1knockdown animals, where this AEA hydrolase has been deleted from peripheral tissues only (Cravatt et al., 2004). Studies performed on these allelic variants highlighted a different susceptibility to drug/alcohol abuse (Sipe et al., 2002Chiang et al., 2004Zhou et al., 2016), supporting a potential link between functional abnormalities in eCB signaling and drug/alcohol dependance. This is just an example of the potential impact of genetic manipulation of eCB system on human health (Zimmer, 2015), for instance on the widespread cannabis use disorder (Boileau et al., 2016). In addition, accumulating evidence points to AEA as a unique natural repressor of gene transcription, via epigenetic mechanisms that include increased DNA methylation (Paradisi et al., 2008), reduced histone acetylation and microRNA (D’Addario et al., 2013). It should be recalled that DNA methylation is a fundamental epigenetic modification of the genome that is involved in a large number of cellular processes, like embryonic development, transcription, chromatin structure, X chromosome inactivation, and genomic imprinting and chromosome stability. Among many other diseases, a role for altered methylation has been established in cancer, for which DNA hypomethylation is a hallmark (Paradisi et al., 2008). Against this background, the potential of AEA as a natural anti-cancer agent appears very promising, and certainly worth of urgent investigations.

Conclusions

After 25 years much has been learned about AEA metabolism and transport, and this knowledge may serve as a paradigm to appreciate the complex machinery that can regulate many other lipid signals. Yet, much remains to be clarified in the next future, in order to better understand AEA signaling regulation and then translate basic AEA research into AEA-based therapeutics. In particular, it seems challenging that AEA and its metabolic enzymes hold potential also as much desired peripheral (blood) biomarkers of human diseases affecting tissues not easy to reach. For instance, FAAH-1 activity and expression are up-regulated in Alzheimer’s disease patients (D’Addario et al., 2012), unlike any other ECS element analyzed (including CB1, CB2, and NAPE-PLD). Moreover, AEA content, NAPE-PLD and FAAH-1 (reviewed by Rapino et al., 2014), and more recently also FAAH-2 (Tedeschi et al., 2017), have been shown to undergo distinct changes in distinct human reproductive disorders. In the new era of nanoscopy, it can be anticipated that also visualization of AEA and its metabolic machinery will greatly increase our understanding of signal transduction pathways triggered by this eCB. Availability of new tools like biotin-AEA (Fezza et al., 2008), positron-emission tomography (PET) probes (Boileau et al., 2016), and luciferin nanoparticles (Yuan et al., 2016) for FAAH-1 visualization, as well as of substrates for in vivo bioluminescence detection of FAAH-1 enzymatic activity (Mofford et al., 2015), indicates that the way to accurate location of distinct elements of AEA signaling within the cell has been already paved.

 







Also tagged with one or more of these keywords: amygdala, visualization, frontal lobes, brain exercise

0 user(s) are reading this topic

0 members, 0 guests, 0 anonymous users