Jump to content

Onemorestep

Members
  • Posts

    521
  • Joined

  • Last visited

  • Days Won

    61

Everything posted by Onemorestep

  1. GABAergic signaling linked to autophagy enhances host protection against intracellular bacterial infections well this is kinda cooool
  2. I read about a woman, I think on here, who had her hppd go away after giving birth. I think that becoming pregnant signals many things to the body— one of which is it’s time to clean things up. Become as strong as possible. If you are cleaning up bad pathogens and cells, it’s possible that is effecting your hppd because doing requires the immune system. Hppd may not be immune related, but having inflammatory reactions (not uncommon in pregnancy) can cause all sorts of emotional symptoms. im sorry to hear you are not feeling well best of luck and much love.
  3. This is a symptom of mold and candida overgrowth. I’m only sharing because I’m going through it now. If you have hppd it may make you sensitive to these two things. Or vice versa. Idk. All I know is my mold tox and candida are making my hppd worse or they helped create an environmental condition around me and in my body that helped cause the hppd. we are full of toxic chemicals. It sucks. But that’s capitalism.
  4. Hppd or not getting healthy is always the way to go!
  5. The last time I accrued hppd my biome test mentioned the word “war zone” about my gut. Viral loads. Fungi. Mold. Inability to process proteins correctly. They suggested 200 different lifestyle and supplements. I am going to do it again and expect it to be different. I think it is easy to do a drug, while something simultaneously happens that causes a neurological event, perhaps helped to be spurned on by the drug, but also perhaps not at all. Then, the brain, damaged and disordered from the event, cannot tolerate any drug now. You try a drug again, and it makes you worse, you assume the drugs are the root cause. It seems logical, but maybe you didn’t notice the glass you shared that night with a friend who had visited a foreign country, or the Amazon product you took you thought had proper COAs but… like most… are not good. I can link products with lead in them on Amazon rn with 4.5 star reviews. many patients with brain disorders cannot use drugs. This doesn’t mean drugs caused them. It also doesn Before anyone pops off this doesn’t mean you can do drugs, obviously, if you read that paragraph. our brain were simply not made for drugs. But they also are not made for the 80,000 new chemicals introduced into our environment by the fda since 1990. Anyone else notice everyone young getting sick? I do. Half my friends have autoimmune diseases and they are in their 20s. One of my families employees just died of cancer at 26. 10% of us homes have very toxic levels of mold in them. antibiotic use is rampant. we eat a credit card of plastic every year. our brains can’t handle a lot rn. A lot is being thrown at all of us. It’s a very sick planet rn. It’s a sad state. if there is a root etiology to hppd, all I can say is it’s going to become more prevalent because every individual at risk will be way more at risk. you don’t have to do drugs to get hppd. Just throwing that out there too.
  6. Tried it. Crappy med. makes it easier to have seizures
  7. C60 fullerene permanently made me negative reactions to drugs that increase inflammation lessened. Was a very interesting experience. Cleaned me out for sure haha. it may have also deactivated EBV in me. At least in the lab it has been shown to allow it to go lytic from latent. but then, since almost everyone has EBV, you’d think those taking c60 would always get it? I was also being exposed to mold at the time. And a high level of strenuous exercise. /shrug. very cool read man
  8. Hppd made me gain weight. This doesn’t seem to happen to everyone but some people gain weight. It could be stress eating for some but wasn’t for me. when I first got hppd I lost a lot of vocabulary and I would stop talking mid sentence. It was like I couldn’t find the words. I knew what I wanted to say meaning wise, but it was as if I had lost language. I had a huge amount of anxiety about people thinking I was weird or whatever. Most people didn’t even notice. and lastly—- these things went away. I don’t have pausing issues with speech and the weight went away. Give it some time focus on helping yourself feel more safe around people. Their just people—most are obsessed with what’s going on with them and pay you very little attention unless you’re hot. In which case you can walk funny and talk weird. They’ll notice then but more because they’re watching how hot you are. No one cares when you’re hot. So just focus on getting hot and then nothing matters. You could get a muscle suit like in arrested development but that’s lame. Fly to Korea and plasticize yourself. Take Chinese steroids. DO WHATEVER IT TAKES TO MAKE YOURSELF THE ADONIS THAT DISTRACTS FROM THE GOOP THAT IS OUR BRAINS. Make a show about it. Become the YouTube. You have some speech issues but You’ll get follows because guess what—I’m not listening anyway I’m just staring at you and imaging the ocean because you’re giving Jason Momoa a run for it. ignore all of this.
  9. virus’s are super interesting to me. They’re these things—dead? Alive?— that can enter a host and tell their cells to do… well anything. Even making zombies. If you took all the virus’s on the planet, and put them in a block, it would completely encompass the Eiffel Tower. There are so many virus’s on this planet we’ve never identified. But what happens when a virus inters your brain? Could hppd be caused by our bodies pruning infected cells? 1) hallucinogens are powerful immune suppressing substances 2) all of us have dormant virus’s 3) some virus’s aren’t noticeable in the body but cause extreme havoc in the brain 4) the immune system functions differently in the brain than the body. It does mechanical things related to our functioning. So when it has to then also fight an infection—it’s bad 5) virus’s rarely enter the brain. But they CAN. This possibly helps explain why some people get hppd after so many trips. Perhaps it took 50 trips before the person was subjected to the virus in real time AND it got into their nose (or they have high BBB permeability). (more reasons to tell your kids to stop picking lol) 6) once the virus is in your brain, it rapidly starts infecting cells. But cannabis and hallucinogens suppress the initial immune response one should start right away. This, causing the virus to be able to enter a large amount of cells, is followed by a large cytokines response when the brain finally registers the threat. That makes you feel really awful—anxious and depressed. but what happens next is what makes the virus hypotheses so interesting to me—it’s the culling of infected cells all at once—- —essentially, that would cause… the equivalent of a very unique brain injury. Lose a few cells? Not a problem. Lose a ton of cell at once? The brain can’t rewire well when the loss is that quickly. You have to rebuild instead and that takes a lot of time…. personally, I always felt I became a different person after hppd. So much of the skills I had worked hard to make physical in my mind—poofed. it’s not a super long video and very interesting! I hope you all find something cool about it. on a personal note— I recently had a very bad spike of something. I had a lot of tests etc and it’s becoming clear that I had a virus enter my brain. It was by far the worst feeling I have ever had. My hppd’s visual symptoms almost returned—I had a full blown return for about 40 min but got super lucky and found some meds to help stop it from coming back. I can’t say from any of this that hppd can be caused by a virus but hot damn if it’s not that it can be make your hppd worse. Because cytokines make hppd worse. so what happens if you get a virus and you never completely fight it off (as almost always with some virus’s)??? It might just be a tug of war back and forth for years before you get it under control. Maybe 10 or 20 years later you go through a stressful time and it returns because stress causes your immune system to weaken and viruses NOTICE that and make their move. Or you decide to trip again. Or you come into contact with a random chemical in your environment that causes the virus to go lytic again. You’d be surprised. This also helps explain why some people’s symptoms “remit” one day in the same way they came. The brain got the virus under control. You probably won’t be the same because of the clipping but without the cytokines you feel stable again. has anyone noticed that some people don’t have visual symptoms? For those who made the correlation between hallucinogens and hppd and didn’t have visual symptoms just a few Google searches might lead them here. I’m certainly seeing them around here and Reddit. There is also an enormous array of different visuals. It could be that whatever causes hppd only causes visual symptoms if the virus gets to that part of the brain. lastly, I did a fair amount of posting about MTOR recently. Many virus’s hijack this system for their own use. They have to, since it’s essentially the body’s steering wheel. or it’s aliens idk.
  10. unfortunately i dont think you will be able to know whether this was covid or the psuedo. I mean people get covid and complain of a lot of the same things ppl with hppd do.
  11. I found freddds protocol to work for me for quite some time. That is until I got the covid shot. Now my nutritional needs have shifted quite a large bit. Very strange.
  12. It made me sleepy at first but now it causes quite severe insomnia with very small doses. Dont know why haha
  13. I would like that too. I’m very distrustful of these scientific studies sometimes in that they are trying to prove something. So yea they may show x y or a but did they decide to include the debilitating withdrawal the person went through? Or that “hey visuals permanently reduced—but they now have worse anxiety. Let’s not include that part”. 2 is a lot better though gotta say. Wonder what the improvement timeline is. Thing is too—there are 100 percent people who have gotten hppd and have been put on 2mg of klonopin off the bat. Hell my doctor gave me one mg no questions asked back in 2012 and I didn’t even seem that anxious. You’d think we’d see more anecdotal reports. but then again, playing devils advocate, if a person got hppd and then got klonopin for a few months and went off with Improvements— would they know to post that? I think 2mg is worth a shot if someone wants to do it. I wouldn’t cold Turkey it though. Even a few days is enough for some people to feel tolerance and withdrawal especially if they’re brains are already weighted towards glutamate vs gaba.
  14. “ In these five patients with positive respond, four of them were given therapy based on LEV, which may indicate LEV as a preferential choice for patients with DEPDC5 variants. Considering the fact that the loss-of-function variants in DEPDC5 will lead to over-activation of the mTOR pathway, the mTOR inhibitor, such as sirolimus or everolimus, may be a complementary treatment for DEDPC5related epilepsy.” https://aepi.biomedcentral.com/articles/10.1186/s42494-020-0011-9
  15. That’s wonderful!! bso makes me worse but for other it very well might make them better. It didn’t hurt me permanently. it’s a pretty complicated mechanism since it has many different chemicals in it. One of which is alterations in apoptosis and autophagy. it interacts with ampk/mtor, p13k/akt.
  16. Absolutely. As someone who has been through benzo withdrawal and never felt quite the same I would say caution is important haha. I also worry that the withdrawn brain is more primed for withdrawals in the future via kindling. This has been my experience at least. Still, it’s a tempting offer isn’t it. Take 6mg of klonopin a day for 2 months and reduce your hppd?... ...can I do it on a beach too somewhere ?
  17. So I have a theory on hppd that involves hyperactivity (of dysregulation) of mtorc1 and mtorc2. One of the things that happens in this scenario, is gaba a receptors... well the easiest way to describe it is they go bad hide. This is part of a cyclic loop that causes this brain state that is excitatory and cannot heal itself. I think there is some merit to the idea that a high dose of benzodiazepine for a short period may help. But it seems that it’s important it be clonazepam. Clonazepam is a pretty unique benzo. It also acts on serotonin systems seemingly without causing panic in people with hppd which is unusual. And I do think if you were to take 6mg daily for 2 months straight and cold Turkey you may experience withdrawals. And not only that, cold Turkeying benzos is bad for the shape of the receptor. I do not see a lot of harm in trying this. A short taper would be all that is needed after use. that being said, if the experiment fails, it is harder for our brains, imo, to get off benzos as we are in such a hyper excitable state. still, other experiments with hppd are much riskier. Like the microdose fire with fire method... yikes. see this is interesting: https://www.frontiersin.org/articles/10.3389/fpsyt.2020.00844/full ketamine induced rapid antidepressant effects by up regulating mtorc1 (as does lsd). When I see things that dampen that effect, I see potential. Naltrexone is another drug I find effective (if you can get over the short dysphoria event) and also inhibits mtorc1. Anecdotally, I heard a guy doing this and it working. But it’s a vague memory and I’m not sure where to find the post. I have a rem sleep disorder that the only medication to treat it is clonazepam. I may give this a shot haha.
  18. https://link.springer.com/article/10.1007/s12031-020-01611-x
  19. https://www.google.com/search?q=amiltryptyline+mtor&rlz=1CDGOYI_enUS931US931&oq=amiltryptyline+mtor&aqs=chrome..69i57.7449j0j4&hl=en-US&sourceid=chrome-mobile&ie=UTF-8 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4317004/
  20. speaking of which: https://www.pnas.org/content/118/5/e2020705118 this really falls in line with your thoughts as a possible etiology. Having hyper activated mTorc1 basically can cause everything you’re saying and more.
  21. Hello everyone! The following is a bunch of research on how, through the modulating of mTOR, we could arrive at our present situation. my apologies for the lackluster organization. its a lot. First off LSD has JUST been shown to increase mTORC1: "Social behavior (SB) is a fundamental hallmark of human interaction. Repeated administration of low doses of the 5-HT2A agonist lysergic acid diethylamide (LSD) in mice enhances SB by potentiating 5-HT2A and AMPA receptor neurotransmission in the mPFC via an increasing phosphorylation of the mTORC1, a protein involved in the modulation of SB. Moreover, the inactivation of mPFC glutamate neurotransmission impairs SB and nullifies the prosocial effects of LSD. Finally, LSD requires the integrity of mTORC1 in excitatory glutamatergic, but not in inhibitory neurons, to produce prosocial effects. This study unveils a mechanism contributing to the role of 5-HT2A agonism in the modulation of SB." Why is this important? Well mTor is how our body regulates EVERYTHING. The mammalian target of rapamycin (mTOR) is an evolutionary conserved serine/threonine kinase that is present in two complexes, mTORC1 and mTORC2. mTORC1 is the main energy and nutrient sensor of the cell: it senses the presence of amino acids, glucose, lipids and ATP to allow efficient activation of the network in response to growth factors, Toll-like receptor ligands and cytokines. Activation of the mTOR pathway usually promotes an anabolic response that induces the synthesis of nucleic acids, proteins and lipids. In addition, it stimulates glycolysis as well as mitochondrial respiration. Emerging data suggest that this metabolic reconfiguration is required for specific effector functions in myeloid cells. Translational control of gene expression in myeloid immune cells emerges as one way in which mTORC1 controls cellular processes such as migration, interferon and pro- or anti-inflammatory cytokine expression as well as metabolic reprogramming. The mammalian target of rapamycin (mTOR) integrates the intracellular signals to control cell growth, nutrient metabolism, and protein translation. mTOR regulates many functions in the development of the brain, such as proliferation, differentiation, migration, and dendrite formation. In addition, mTOR is important in synaptic formation and plasticity. Abnormalities in mTOR activity is linked with severe deficits in nervous system development, including tumors, autism, and seizures. Dissecting the wide-ranging roles of mTOR activity during critical periods in development will greatly expand our understanding of neurogenesis. Inhibition of mTORC1 in macrophages promotes autophagy, which is important for intracellular pathogen killing and clearance of ingested complex lipids such as LDL cholesterol. lets start with autophagy Soo... whats autophagy? and how does it relate to mTOR? Autophagy and mTOR As a key regulator of autophagy, the mTOR plays an important role in autophagy, translation, cell growth and survival (Hwang et al., 2017). Mammalian target of rapamycin and autophagy are tightly bound within cells, and defects of mTOR and autophagy process might lead to a variety of human diseases (Hoeffer and Klann, 2010). Studies have shown that mTOR is widely involved in autophagy activation and synaptic plasticity (Ryskalin et al., 2018). The mTOR modulates long-lasting synaptic plasticity, memory and learning via regulating the synthesis of dendritic proteins (Liu et al., 2018a). Macroautophagy can degrade organelles and long-lived proteins in case of mTOR inactivation. Synaptic plasticity is further modulated by mTOR and neurodegeneration occurs when macroautophagy is absent (Hernandez et al., 2012). Therefore, macroautophagy following mTOR inactivation at the presynaptic terminal rapidly changes the neural transmission and presynaptic structure (Hernandez et al., 2012). The mechanisms for the target of rapamycin have been involved in modulating neurodegeneration and synaptic plasticity, but the role of mTOR in regulating presynaptic function via autophagy has not been clarified clearly (Torres and Sulzer, 2012). In summary, there is a close relationship among mTOR, brain plasticity and autophagy. The mTOR related pathways play important role in regulating the process of autophagy and brain plasticity. Autophagy is a lysosome-reliant degradation mechanism that regulate many biological courses, such as neuroprotection and cellular stress reactions (Shen and Ganetzky, 2009). There are different kinds of autophagy in most mammalian cells, and each type of autophagy performs very specific tasks in the course of intracellular degradation (Tasset and Cuervo, 2016). The autophagy-lysosomal pathway is a main proteolytic pathway, which mainly embraces chaperone-mediated autophagy and macroautophagy in mammalian systems (Xilouri and Stefanis, 2010). Macroautophagy, as a lysosomal pathway in charge of the circulation of long-lived proteins and organelles, is mainly considered as the inducible course in neurons, which is activated in conditions of injury and stress (Boland and Nixon, 2006). Coupled with macro-autophagy, chaperone-mediated autophagy (CMA) is crucial for maintaining intracellular survival and homeostasis via selectively reducing oxidized, misfolded, or degraded cytoplasmic proteins (Cai et al., 2015). The plasticity of the central nervous system(CNS) can be regarded as changes of functional interaction between different types of cells, astrocytes, neurons, and oligodendrocytes (Aberg et al., 2006). The mature brain, as a highly dynamic organ, constantly alters its structure via eliminating and forming new connections. In general, these changes are known as brain plasticity and are related to functional changes (Viscomi and D’Amelio, 2012). Brain plasticity can be divided into structure plasticity and function plasticity. The structural plasticity of the brain refers to the fact that the connections between synapses and neurons in the brain can be established due to the influence of learning and experience. It includes the plasticity of synapses and neurons. Synaptic plasticity refers to the changes of pre-existing relationship between two neurons including structure and function alteration (De Pitta et al., 2016). Synaptic plasticity is considered as the representative of cellular mechanisms of memory and learning. Mitochondria are related to the modulation of complicated course of synaptic plasticity (Todorova and Blokland, 2017). For a long period, synaptic plasticity has been considered as a neuronal mechanism under the regulation of neural network action (Ronzano, 2017). Recent data indicate that autophagy is a homeostatic mechanism which is compatible with the microenvironment of the synapse, with the purpose of serving local functions linked with synaptic transmission (Todorova and Blokland, 2017). Neuronal plasticity is maintained by the fine modulation of organelle biogenesis and degradation and protein synthesis and degradation to assure high-efficiency turnover (Viscomi and D’Amelio, 2012). Protein degradation plays an important role in the course of synaptic plasticity, but the involved molecular mechanisms are unclear (Haynes et al., 2015). Therefore, Autophagy is a quality control mechanism of organelles and proteins in neurons, which plays a crucial role in their physiology and pathology (Viscomi and D’Amelio, 2012). In a word, there is a close relationship between autophagy and brain plasticity, and the related mechanisms are summarized in this review paper (as Table 1 and Figure 1 demonstrate). https://www.frontiersin.org/articles/10.3389/fncel.2019.00228/full#:~:text=Autophagy%20is%20the%20core%20regulator,et%20al.%2C%202017). mTOR complex 1 (mTORC1) was unveiled as a master regulator of autophagy since inhibition of mTORC1 was required to initiate the autophagy process. _______________________________________________ So... weve hyperactived our mTORC1.... we cannot initiate the autophagy process... what does this mean??? 1) Hyperactivation of mTORC1 by TSC1/2 deletion induces aberrant growth, proliferation, and differentiation of neurons and astrocytes, resulting in neuronal dysplasia, abnormal neuronal architecture, reactive astrogliosis, and seizures (27, 40,–42). Hyperactivation of mTORC1 disrupts cellular homeostasis in cerebellar Purkinje cells Mammalian target of rapamycin (mTOR) is a central regulator of cellular metabolism. The importance of mTORC1 signaling in neuronal development and functions has been highlighted by its strong relationship with many neurological and neuropsychiatric diseases. Previous studies demonstrated that hyperactivation of mTORC1 in forebrain recapitulates tuberous sclerosis and neurodegeneration. In the mouse cerebellum, Purkinje cell-specific knockout of Tsc1/2 has been implicated in autistic-like behaviors. However, since TSC1/2 activity does not always correlate with clinical manifestations as evident in some cases of tuberous sclerosis, the intriguing possibility is raised that phenotypes observed in Tsc1/2 knockout mice cannot be attributable solely to mTORC1 hyperactivation. Here we generated transgenic mice in which mTORC1 signaling is directly hyperactivated in Purkinje cells. The transgenic mice exhibited impaired synapse elimination of climbing fibers and motor discoordination without affecting social behaviors. Furthermore, mTORC1 hyperactivation induced prominent apoptosis of Purkinje cells, accompanied with dysregulated cellular homeostasis including cell enlargement, increased mitochondrial respiratory activity, and activation of pseudohypoxic response. These findings suggest the different contributions between hyperactivated mTORC1 and Tsc1/2 knockout in social behaviors, and reveal the perturbations of cellular homeostasis by hyperactivated mTORC1 as possible underlying mechanisms of neuronal dysfunctions and death in tuberous sclerosis and neurodegenerative diseases. neurogenesis, dendrite formation, and synaptic integration: Effects of mTOR activation during neurogenesis. Neural stem cells (blue) undergo proliferation and either give rise to more stem cells (self-renewal) or daughter cells (green, differentiation). Activation of mTORC2 promotes neural stem cells (NSC) cell cycle entry through Akt. Hyperactivation of mTORC1 results in diminished self-renewal, favoring differentiation and lineage expansion. Daughter cells then migrate (red) from proliferation zones to their terminal positions. Activation of mTORC1 results in aberrant migration of daughter cells. Upon reaching their terminal positions, newly born neurons (gray) extend neurites and properly form dendritic arbors. Cells with high levels of mTORC1 activity can severely alter dendrite formation and synaptic integration. Upward pointing arrows indicate increased activity of designated genes or proteins. Downward pointing arrows indicate decreased activity or knockdown of designated genes or proteins. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5983636/ Mitochondria: Induction of cellular stresses by mTORC1 hyperactivation We explored the molecular mechanisms underlying Purkinje cell death by mTORC1 hyperactivation in PC-mTOR Tg mice. Activation of mTORC1 enhances the mitochondrial biogenesis by forming a complex with PGC1α and YY124. We observed mitochondrial morphology in Purkinje cells by using the electron microscopy (Fig. 6a–f). As expected, the remarkably enlarged mitochondria were often found in PC-mTOR Tg mice in both cell bodies (Fig. 6a and b) and dendrites (Fig. 6e and f) compared to control mice. Despite their abnormal morphology, the internal lamellar structure of cristae was almost preserved even in enlarged mitochondria in PC-mTOR Tg mice (Fig. 6c and d). To test the mitochondrial respiratory activity, the cytochrome c oxidase activity was visualized in the cerebellar slices. Mitochondrial activity was detected in both molecular and Purkinje cell layers of control mice (Fig. 6g and h). Although similar staining patterns were also observed in PC-mTOR Tg mice, the cell bodies of Purkinje cells were stained more densely than control mice. Thus, despite their abnormal morphology, the mitochondrial respiratory function was not impaired but rather enhanced in Purkinje cells of PC-mTOR Tg mice. Glutamate To assess the pre- and postsynaptic function of each mTOR complex in glutamatergic synaptic transmission, we inactivated mTORC1 signaling by conditionally deleting Raptor, or mTORC2 signaling by conditionally deleting Rictor, postmitotically in primary neuron cultures from mouse hippocampus. We then performed morphological and whole-cell patch-clamp analysis of synaptic and membrane properties of glutamatergic neurons. Our results showed that both mTOR complexes were necessary to support normal neuron growth and evoked excitatory synaptic transmission. Despite these similarities, the effects of mTORC1 on evoked EPSCs (eEPSCs) were postsynaptic, via reductions in synapse number, whereas mTORC2 regulated the presynaptic Ca2+ dependence of evoked SV release. Furthermore, although the mechanism through which mTORC1 inactivation decreased eEPSCs was postsynaptic, it also increased spontaneous SV release and SV pool replenishment, which are thought to be presynaptic processes. Overall, each mTOR complex affected distinct modes of SV release: mTORC1 inactivation enhanced modes with low rates of SV fusion, such as spontaneous release, and mTORC2 inactivation impaired modes with high rates of SV fusion, such as action potential-evoked release. Thus, via differential activation of these two complexes, the mTOR pathway is ideally poised to respond to external cues and make fine adjustments to glutamatergic synaptic transmission to maintain normal neural network function. Previous studies showed that mTOR inhibition by rapamycin treatment reduces the number of AMPA receptors at the synapse (Wang et al., 2006), the number of synapses (Weston et al., 2012), and the number of SVs per synapse (Hernandez et al., 2012). Accordingly, mTOR hyperactivation increases mEPSC amplitude (Xiong et al., 2012), AMPA receptor number, and spine density (Tang et al., 2014; Williams et al., 2015), and these effects are blocked by rapamycin. Thus, integrating our findings on specific mTORC1 inactivation with these previous findings, several lines of evidence now indicate that mTORC1 acts via a postsynaptic mechanism to bidirectionally regulate evoked glutamatergic synaptic strength. https://elifesciences.org/articles/51440 GABA: We have FUCKED gaba (but you knew that didn't you) The following is evidence of the reduced gaba hypothesis that is thrown around a lot. basically, disrupting the autophagy from mTOR (such as that which would occure through hyperactive MTORC1) causes a reduction in the surface expression of GABA A receptors ( thats the ones benzos increase activity on). The following is two studies, one is a good tldr and the other is much mor indepth. Autophagy links MTOR and GABA signaling in the brain The disruption of MTOR-regulated macroautophagy/autophagy was previously shown to cause autistic-like abnormalities; however, the underlying molecular defects remained largely unresolved. In a recent study, we demonstrated that autophagy deficiency induced by conditional Atg7 deletion in either forebrain GABAergic inhibitory or excitatory neurons leads to a similar set of autistic-like behavioral abnormalities even when induced following the peak period of synaptic pruning during postnatal neurodevelopment. Our proteomic analysis and molecular dissection further revealed a mechanism in which the GABAA receptor trafficking function of GABARAP (gamma-aminobutyric acid receptor associated protein) family proteins was compromised as they became sequestered by SQSTM1/p62-positive aggregates formed due to autophagy deficiency. Our discovery of autophagy as a link between MTOR and GABA signaling may have implications not limited to neurodevelopmental and neuropsychiatric disorders, but could potentially be involved in other human pathologies such as cancer and diabetes in which both pathways are implicated. GABARAPs dysfunction by autophagy deficiency in adolescent brain impairs GABAA receptor trafficking and social behavior https://advances.sciencemag.org/content/5/4/eaau8237 Excessive p62 accumulation in autophagy-deficient and mTOR-hyperactivated neurons results in reduced GABAA receptor surface expression due to mislocalized GABARAPs We next asked whether the observed disruption of GABAA receptor trafficking is specific to autophagy-deficient conditions or is due to a general increase in p62 levels. First, by immunofluorescence, we found that p62 overexpression in WT neurons led to the formation of p62+ aggregates that also sequestered GABARAPs (Fig. 5A and fig. S7, A to C). Second, we observed a significant reduction in surface GABAA receptors in WT neurons with p62 overexpression (Fig. 5B). Third, if the reduced surface expression of GABAA receptors was caused by sequestration of GABARAPs by p62, a reduction of p62 in Atg7 cKO neurons would be expected to restore such deficits. To test this hypothesis, we performed surface receptor biotinylation experiments on Atg7 cKO and control neurons with or without Sqstm1 (p62) knockdown. Consistent with our prediction, Sqstm1 knockdown in Atg7 cKO neurons reversed the reduction of surface GABAA receptors to control levels (Fig. 5C). Together, this series of experiments suggests that the pathologic accumulation of p62 in Atg7 cKO neurons sequesters GABARAPs and disrupts the normal functions of GABARAPs, resulting in a reduction of surface GABAA receptor levels. Astrocytes: here we go Astrocyte activation has been implicated in the pathogenesis of several neurological conditions, such as neurodegenerative diseases, infections, trauma, and ischemia. Reactive astrocytes are capable of producing a variety of pro-inflammatory mediators, including interleukin-6 (IL-6), IL-1β, tumor necrosis factor-α (TNF-α), neurotrophic factors [1], as well as potentially neurotoxic compounds, like nitric oxide (NO). NO, one of the smallest known bioactive products of mammalian cells, is biosynthesized by three distinct isoforms of NO synthase (NOS): the constitutively expressed neuronal (n)NOS and endothelial (e)NOS, and the inducible (i)NOS [2]. The expression of iNOS can be induced in different cell types and tissues by exposure to immunological and inflammatory stimuli [3]. In vitro, primary astrocyte cultures express iNOS in response to cytokines such as IL-1β [4], interferon γ (IFNγ), TNFα and/or the bacterial endotoxin, lipopolysaccharide (LPS) [5, 6]. Once induced, iNOS leads to continuous NO production, which is terminated by enzyme degradation, depletion of substrates, or cell death [7]. iNOS activity generates large amounts of NO (within the μM range) that can have antimicrobial, anti-atherogenic, or apoptotic actions [8]. However, aberrant iNOS induction exerts detrimental effects and seems to be involved in the pathophysiology of several human diseases [9, 10]. soo... this is where it gets bad. Neuronal mTORC1 Is Required for Maintaining the Nonreactive State of Astrocytes This study has revealed that the inactivation of mTORC1 in postmitotic neurons causes moderate reactive astrogliosis. The loss of neural mTORC1 activity may induce astrogliosis by reducing the neuronal secretion of FGF-2, thereby inhibiting FGF receptor signaling in astrocytes, which is required to maintain their nonreactive state (36) (Fig. 7). Although our present data could not identify the exact role of FGF-2 in this process, and the underlying mechanism needs to be further investigated, our findings have uncovered a novel mechanism for the regulation of astrocytes by dysfunctional neurons and have established a potential important link between mTORC1 signaling and CNS pathologies. The function of mTORC1 in neurons and astrocytes has been extensively studied in conditional knock-out mice (26, 37,–39). Hyperactivation of mTORC1 by TSC1/2 deletion induces aberrant growth, proliferation, and differentiation of neurons and astrocytes, resulting in neuronal dysplasia, abnormal neuronal architecture, reactive astrogliosis, and seizures (27, 40,–42). Inactivation of mTORC1 in neuronal progenitors impairs the growth and proliferation of neurons and astrocytes, resulting in a smaller brain and in death shortly after birth (25, 43). Lets talk about INFLAMMATION: OMG THE SYMPTOMS So we see that mTORC1 Hyperactivation results in an increase in cytokines. One of those is il-1b.... and its nefarious in the brain. IL-1b Interleukin-1β Causes Anxiety by Interacting with the Endocannabinoid System Steps: 1. Increase in mTORC1 2. increase in IL-1b proinflammatory cytokine 3. decrease in cb1 recebtor binding 4. decreases CB1R's (gabaA) synapse binding in the striatum 5. causes behavioral manifestations closely resembling anxious-depressive symptoms in humans, including anhedonia, reduced exploratory behaviors, social withdrawal, fatigue, and sleep disturbances 6. This entire process requires "intact function of the transient receptor potential vanilloid 1 (TRPV1)" to work. 2. Interleukin-1 beta modulates AMPA receptor expression and phosphorylation in hippocampal neurons IL-1beta, but not IL-10 or tumour necrosis factor (TNF)-alpha, down-regulated the surface expression and Ser831 phosphorylation of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluR1. Agents that block IL-1beta receptor activity abolished these effects. In contrast, no change in the surface expression of the N-methyl-d-aspartate (NMDA) receptor subunit NR1 was observed. The inhibition of NMDA receptor activity or depletion of extracellular calcium blocked IL-1beta effects on GluR1 phosphorylation and surface expression. NMDA-mediated calcium influx was also regulated by IL-1beta. These findings suggest that IL-1beta selectively regulates AMPA receptor phosphorylation and surface expression through extracellular calcium and an unknown mechanism involving NMDA receptor activity. (PS Phosphorylation of the AMPA receptor GluR1 subunit is required for synaptic plasticity and retention of spatial memory) Blood brain barrier: On the other hand, interleukin (IL)-1β significantly induces the production of MMP 1, 3, 10, and 13 via a mechanism that is independent of Ca2+ In summary, the results presented here are the first to reveal the function of MMP3 in the BBB and suggest that it has an essential role in the brain microvasculature that differs from its function in other vessels. We have shown that MMP3 increases BBB permeability by upregulating the ERK signaling pathway, which subsequently reduces TJ and AJ protein abundance in BMVECs. Oxidative stress often leads to impairment of BBB. Since the BBB is the primary regulator of exchange between the peripheral blood and the brain, our observations likely have important implications for treating neuroinflammatory conditions and other CNS disorders involving the endothelial MMP3 pathway. https://www.hindawi.com/journals/omcl/2021/6655122/ TNF-a: this is a bit scary TNF released by microglia has an important role in regulating synaptic plasticity [110]. Specifically, it controls a process called synaptic scaling, i.e., the adjustment of synaptic strength in response to prolonged changes in the electrical activity of neurons [110,111]. Indeed, a reduction of glutamate transmission increases microglial TNF release, which promotes the expression of AMPA glutamate receptors in neurons. Conversely, increased extracellular glutamate concentration inhibits TNF release from microglia, additional glutamate receptor expression, and declines neuronal activity [111–113]. The increase of AMPA receptor GluR1 subunit expression does not occur at mRNA level, but this is controlled by TNF at post-transcriptional level [114]. Subsequent studies revealed that TNF facilitates the trafficking and membrane insertion of AMPA receptors at the neuron surface, which are crucial for the homeostatic synaptic plasticity. Specifically, hippocampal neurons exposed to TNF increase surface expression of GluR1 subunit through modulation of NF-κB and acid sphingomyelinase pathways [115]. TNF not only controls homeostatic synaptic activity, but also induces neurotoxicity via autocrine/paracrine loops involving other endogenous mediators. First, TNF activates TNFR1 on microglia, amplifying its production and release [95]. Second, microglia-derived TNF activates TNFR1 expressed on astrocytes, allowing glutamate release from the glial cells. This, in turn, activates its specific receptors, including the metabotropic mGluR2 receptor on microglia, potentiating microglial TNF production and affecting synaptic transmission [110]. ATP, released by microglia concurrently with TNF, contributes to TNF-mediated neuronal damage by inducing a prolonged activation of microglial P2X7 receptor and release of both IL-1β and TNF inflammatory cytokines. In addition, both microglial TNF and ATP trigger adjacent astrocytes to release additional ATP, that amplifies microglia response and promotes astroglial release of glutamate, aggravating neuronal dysfunction [110]. Moreover, TNF mediates neuronal death by increasing extracellular levels of the excitotoxic transmitter glutamate and excessive AMPA receptor activation via downregulation of the astrocytic glutamate transporter EAAT2/GLT1 [116]. The effects of TNF on N-methyl-D-aspartate receptors (NMDARs) trafficking are less characterized. However, it has been demonstrated that, in hippocampal neurons, TNF increases the expression of the NR1 subunit of NMDAR and its specific clustering into lipid rafts [117]. Accordingly, treatment of human neuronal cultures with competitive (2-APV) and noncompetitive (MK-801) NMDA receptor antagonists reduced the glutamate neurotoxicity induced by TNF [118]. https://www.mdpi.com/2073-4409/9/10/2145/pdf " In MS, glutamate-related excitotoxicity, caused by excessive activation of these receptors (leading to a Ca2+ overload), is responsible for neuronal and oligodendrocyte death [2, 16, 17]. In addition, microglia, the resident macrophages of the CNS, become activated by increased glutamate concentration. Activated microglia proliferate, secrete cytokines, chemokines, nitric oxide and ROS, and may become phagocytic; outcomes all of which cause further injury to the ailing CNS [9]. Oligodendrocytes have been found to be particularly susceptible to glutamate excitotoxicity, via the AMPA/kainate receptors. AMPA/kainate antagonists have been shown to increase oligodendrocyte survival as well as reducing axonal damage [16, 17]. " --this is important to note because if we have abysmal gaba and high glutamate from the mTORC1 and from astrocyte issues then excitotoxic events are easily understandable. Here you can see how these events could lead to an out-of-control inflammatory response. What do we do???? BEATS ME REALLY but heres some options reduce mtor? Alleviation of neuronal energy deficiency by mTOR inhibition as a treatment for mitochondria-related neurodegeneration mTOR inhibition is beneficial in neurodegenerative disease models and its effects are often attributable to the modulation of autophagy and anti-apoptosis. Here, we report a neglected but important bioenergetic effect of mTOR inhibition in neurons. mTOR inhibition by rapamycin significantly preserves neuronal ATP levels, particularly when oxidative phosphorylation is impaired, such as in neurons treated with mitochondrial inhibitors, or in neurons derived from maternally inherited Leigh syndrome (MILS) patient iPS cells with ATP synthase deficiency. Rapamycin treatment significantly improves the resistance of MILS neurons to glutamate toxicity. Surprisingly, in mitochondrially defective neurons, but not neuroprogenitor cells, ribosomal S6 and S6 kinase phosphorylation increased over time, despite activation of AMPK, which is often linked to mTOR inhibition. A rapamycin-induced decrease in protein synthesis, a major energy-consuming process, may account for its ATP-saving effect. We propose that a mild reduction in protein synthesis may have the potential to treat mitochondria-related neurodegeneration. Autophagy Dysfunction and mTOR Hyperactivation Is Involved in Surgery: Induced Behavioral Deficits in Aged C57BL/6J Mice Autophagy is crucial for cell survival, development, division, and homeostasis. The mammalian target of rapamycin (mTOR), which is the foremost negative controller of autophagy, plays a key role in many endogenous processes. The present study investigated whether rapamycin can ameliorate surgery—induced cognitive deficits by inhibiting mTOR and activating autophagy in the hippocampus. Both adult and aged C57BL/6J mice received an intraperitoneal injection of rapamycin (10 mg/kg/day) for 5 days per week for one and a half months. Mice were then subjected to partial hepatectomy under general anesthesia. Behavioral performance was assessed on postoperative days 3, 7, and 14. Hippocampal autophagy-related (Atg)-5, phosphorylated mTOR, and phosphorylated p70S6K were examined at each time point. Brain derived neurotrophic factor (BDNF), synaptophysin, and tau hyperphosphorylation (T396) in the hippocampus were also examined. Surgical trauma and anesthesia exacerbated spatial learning and memory impairment in aged mice on postoperative days 3 and 7. Following partial hepatectomy, the levels of phosphorylated mTOR, phosphorylated 70S6K, and phosphorylated tau were all increased in the hippocampus. A corresponding decline in BDNF and synaptophysin were observed. Rapamycin treatment restored autophagy function, attenuated phosphorylation of tau protein, and increased BDNF and synaptophysin expression in the hippocampus of surgical mice. Furthermore, surgery and anesthesia induced spatial learning and memory impairments were also reversed by rapamycin treatment. Autophagy impairments and mTOR hyperactivation were detected along with surgery—induced behavioral deficits. Inhibiting the mTOR signaling pathway with rapamycin successfully ameliorated surgery-related cognitive impairments by sustaining autophagic degradation, inhibiting tau hyperphosphorylation, and increasing synaptophysin and BDNF expression. https://link.springer.com/article/10.1007/s11064-019-02918-x Neuronal mTORC1 Is Required for Maintaining the Nonreactive State of Astrocytes In summary, this study has demonstrated that the inactivation of mTORC1 in postmitotic neurons induces reactive astrogliosis, possibly by inhibiting FGF-2 secretion. mTORC1 activity in postmitotic neurons is required for maintaining astrocytes in a nonreactive state. Astrogliosis is likely to be regulated by various signaling pathways and various cell types in different nuclei on the CNS. In our studies, neuronal mTORC1 activity regulates astrocyte activation, possibly via multiple potential signals directly or indirectly. Further investigation is required to define the pathological consequences of astrogliosis induced by the loss of neuronal mTORC1 and its association with CNS disease. Manipulating mTORC1 in neurons or FGF-2 signaling in astrocytes may represent a novel therapeutic mechanism for treating CNS disorders and improving functional recovery in neuropathological conditions. Medications: IM NOT A DOCTOR and honestly some of these are pretty extreme. Don't take these without research and consulting a medical professional. The point of these medications: reduce damage caused by reaction. this includes microglial and astrocyte dysfunction and resulting inflammatory markers treat reaction at source Repair damage from the reaction Pramipexole: curative action: DRD3 (dopamine receptor D3) but not DRD2 activates autophagy through MTORC1 inhibition preserving protein synthesis The results revealed that pramipexole induces autophagy through MTOR inhibition and a DRD3-dependent but DRD2-independent mechanism. DRD3 activated AMPK followed by inhibitory phosphorylation of RPTOR, MTORC1 and RPS6KB1 inhibition and ULK1 activation. Interestingly, despite RPS6KB1 inhibition, the activity of RPS6 was maintained through activation of the MAPK1/3-RPS6KA pathway, and the activity of MTORC1 kinase target EIF4EBP1 along with protein synthesis and cell viability, were also preserved. This pattern of autophagy through MTORC1 inhibition without suppression of protein synthesis, contrasts with that of direct allosteric and catalytic MTOR inhibitors and opens up new opportunities for G protein-coupled receptor ligands as autophagy inducers in the treatment of neurodegenerative and psychiatric diseases. https://pubmed.ncbi.nlm.nih.gov/31538542/ Palliative: this will help with the astrocyte insanity/out of control inflammatory cycle Experimental autoimmune encephalomyelitis (EAE) is the most used animal model of multiple sclerosis (MS) for the development of new therapies. Dopamine receptors can modulate EAE and MS development, thus highlighting the potential use of dopaminergic agonists in the treatment of MS, which has been poorly explored. Herein, we hypothesized that pramipexole (PPX), a dopamine D2/D3 receptor-preferring agonist commonly used to treat Parkinson's disease (PD), would be a suitable therapeutic drug for EAE. Thus, we report the effects and the underlying mechanisms of action of PPX in the prevention of EAE. PPX (0.1 and 1 mg/kg) was administered intraperitoneally (i.p.) from day 0 to 40 post-immunization (p.i.). Our results showed that PPX 1 mg/kg prevented EAE development, abolishing EAE signs by blocking neuroinflammatory response, demyelination, and astroglial activation in spinal cord. Moreover, PPX inhibited the production of inflammatory cytokines, such as IL-17, IL-1β, and TNF-α in peripheral lymphoid tissue. PPX was also able to restore basal levels of a number of EAE-induced effects in spinal cord and striatum, such as reactive oxygen species, glutathione peroxidase, parkin, and α-synuclein (α-syn). Thus, our findings highlight the usefulness of PPX in preventing EAE-induced motor symptoms, possibly by modulating immune cell responses, such as those found in MS and other T helper cell-mediated inflammatory diseases. https://pubmed.ncbi.nlm.nih.gov/26801190/ Levetiracetam: The results indicated that TPM and LEV alleviated behavioral deficits and reduced amyloid plaques in APPswe/PS1dE9 transgenic mice. TPM and LEV increased Aβ clearance and up‐regulated Aβ transport and autophagic degradation. TPM and LEV inhibited Aβ generation and suppressed γ‐secretase activity. TPM and LEV inhibited GSK‐3β activation and increased the activation of AMPK/Akt activation. Further, TPM and LEV inhibited histone deacetylase activity in vivo. activation of the ampk pathway reduces mtorc1 Baclofen: possible brain healing GABA receptors play an important role in ischemic brain injury. Studies have indicated that autophagy is closely related to neurodegenerative diseases. However, during chronic cerebral hypoperfusion, the changes of autophagy in the hippocampal CA1 area, the correlation between GABA receptors and autophagy and their influences on hippocampal neuronal apoptosis have not been well established. Here, we found that chronic cerebral hypoperfusion resulted in rat hippocampal atrophy, neuronal apoptosis, enhancement and redistribution of autophagy, down-regulation of Bcl-2/Bax ratio, elevation of cleaved caspase-3 levels, reduction of surface expression of GABAA receptor α1 subunit and an increase in surface and mitochondrial expression of connexin 43 (CX43) and CX36. Chronic administration of GABAB receptors agonist baclofen significantly alleviated neuronal damage. Meanwhile, baclofen could up-regulate the ratio of Bcl-2/Bax and increase the activation of Akt, GSK-3β and ERK which suppressed cytodestructive autophagy. The study also provided evidence that baclofen could attenuate the decrease in surface expression of GABAA receptor α1 subunit and down-regulate surface and mitochondrial expression of CX43 and CX36, which might enhance protective autophagy. The current findings suggested that, under chronic cerebral hypoperfusion, the effects of GABAB receptors activation on autophagy regulation could reverse neuronal damage. WARNING: baclofen interacts with ampa receptors causing an initial increase and then what seems like long term decrease. since there could be a loss of ampa function (whether through cell death or otherwise) this could worsen certain symptoms. Mainly low empathy/issues with anhedonia. Ampa receptors play a very complex role in synaptic plasticity, mood, and behavior. I took baclofen in very high doses (100mg per day) a few years ago. It restored my cognition, made me sociopathic (not permanent), hypomanic (god i wish permanent but no), and gave me anhedonia that took 2 years to cease. But hey it actually did work to heal my brain which is neato ** I think i just overdid it. PAWS set in. Homotaurine may help with this should anyone wish to try baclofen for restoring their cognition. Rapamycin: the ultimate mtor inhibitor, see basically all the studies. The mTOR kinase inhibitor rapamycin decreases iNOS mRNA stability in astrocytes In our previous studies, we observed that although rapamycin reduced iNOS expression mRNA and activity in microglial cells, and was without effect on astrocyte iNOS activity [21], it caused a rapid significant increase in iNOS mRNA levels in astrocytes induced by two different proinflammatory stimuli. Later time points were not examined; neither was the basis for this contrasting result examined. In the present paper we tested the hypothesis that while at early times rapamycin increases iNOS mRNA, at later times it modifies iNOS mRNA stability. Our results using primary rat astrocytes are consistent with this hypothesis, and suggest that inhibition of mTOR kinase activity in glial cells results in anti-inflammatory actions. Together with the marked anti-inflammatory effects observed in microglial cells [21], these data further provide pre-clinical evidence for a possible clinical use of mTOR inhibitors in the treatment of inflammatory-based CNS pathologies. https://jneuroinflammation.biomedcentral.com/articles/10.1186/1742-2094-8-1 but.... is it that simple?? Probably not. Inhibiting mtorc1 across the board surely has issues involved with it. These are very complex mechanisms at work... ive come across some stuff saying there are serious pros and cons for using drugs like rapamycin. Some good reads: https://europepmc.org/article/pmc/pmc6223325#S4 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5787061/#idm140239461448896title tldr: Our brains are going fucking nuts. We have out of control inflammation, autophagy problems, gaba is gonnneeeee, glutamate is out of control... the list is almost endless really. There is so much more not mentioned in this post. this is like a giant tree-- you can follow the branches out very far. But it makes sense-- especially with the weird onset times of hppd. Some of this needs time to get bad enough that you notice it. And if the mtor stays dysregulated you wont heal. For whatever reason, in some people it stays stuck. If we can reverse this issue it should provide at least a fair fight for healing. The sooner its dealt with the better as the more time spent in the "active" state of hppd the more damage is being done. So why cant you see this on an MRI? thats a whole nother post. Youd be surprised what you cant see on an mri though lol. One thing that is important to note--- if there is excitotoxicity of glutamate receptors in this process, reducing the thing that is boosting them, even if it is causing the pathology, may make a person feel WORSE initially. Or, likely, there are other things going on here too--- like how hallucinogens open up susceptibility to viral reactivation in the brain.... for another day.
  22. its very pro usage. Unfortunately a lot of what i come across in new psych research is all pro even when it has GLARING implications for this and other diseases
×
×
  • Create New...

Important Information

By using this site, you agree to our Terms of Use.