UA-45667900-1
Showing posts with label BCAA. Show all posts
Showing posts with label BCAA. Show all posts

Wednesday 13 September 2017

Verapamil still working after 3+ years, for SIB in Autism


There are numerous ideas about how to treat self injurious behavior (SIB) associated with autism. ARI (the former home of Defeat Autism Now) have just had their take on the subject published.
In this blog we have seen that Tyler has developed a BCAA (branch chained amino acid) therapy, based on the idea of Acute Tryptophan Depletion, to control his son’s type of self injury.
The silver bullet for my son’s summer time raging and self-injury continues to be the L-type calcium channel blocker Verapamil.
I think many people will be skeptical of both BCAAs and Verapamil, which is entirely understandable. Unlike other aspects of autism, which are hard to measure, self-injury is really easy to measure and so you know when you have cracked the problem; what other people think tends not to matter.  
Now that Monty, aged 14 with ASD, has moved to secondary/high school the routine has changed a little and his assistant forgets to give him his midday dose of verapamil.
On the days she forgets, between 4.00pm and 4.30pm Monty starts to punch himself. On all other days and during the entire summer there has been no sign of self injury.
So when asked is it really necessary Monty keeps taking his pills, my answer remains yes.  In the case of verapamil I now have further evidence that after more than three years of use, his pollen allergy driven self injury continues to be entirely controllable using this therapy.
I do not know what ARI have put forward in their book. If your child has SIB that does not respond to whatever therapies you have tried, it might well be a helpful read. 

Other readers have noted GI and behavioral improvement from Verapamil and our doctor reader Agnieszka did try and collect case reports, but it seems parents are more interested in reading reports than writing them.
           







Thursday 18 May 2017

Amino Acids in Autism


Amino Acids (AAs) are very important to health and it is important that all 20 are within the reference ranges, or there can be serious consequences.  Inborn errors of amino acid metabolism do exist and there are metabolic disorders which impair either the synthesis and/or degradation of amino acids.
It has been suggested that a lack of certain amino acids might underlie some people’s autism. This seems to be the basis of one new autism drug, CM-AT, being developed in the US, but this idea remains somewhat controversial.

In those people who have normal levels of amino acids, potential does exist to modify their level for some therapeutic effect. 

Examples include:-

·        Using histidine to inhibit mast cells de-granulating and so reducing symptoms of allergy

·       Using the 3 branch chained AAs to reduce the level of the AA, phenylanine, which can drive movement disorders/tics

·       Methionine seems to promote speech in regressive autism, but for no known reason.

·        Some AAs, such as leucine, activate mTOR. It is suggested that others (histidine, lysine and threonine) can inhibit it, which might have a therapeutic benefit in those with too much mTOR signaling.

·        D-Serine, synthesized in the brain by from L-serine, serves as a neuromodulator by co-activating NMDA receptors.  D-serine has been suggested for the treatment of negative symptoms of schizophrenia

·        Aspartic acid is an NMDA agonist

·       Threonine is being studied as a possible therapy for Inflammatory Bowel Disease (IBD), because it may increase intestinal mucin synthesis.


Amino acids, the building blocks for proteins

To make a protein, a cell must put a chain of amino acids together in the right order. It makes a copy of the relevant DNA instruction in the cell nucleus, and takes it into the cytoplasm, where the cell decodes the instruction and makes many copies of the protein, which fold into shape as they are produced.

There are 20 standard or “canonical” amino acids, which can be thought of as protein building blocks.
Humans can produce 10 of the 20 amino acids; the others must be supplied in the food and are called “essential”. The human body does not store excess amino acids for later use, so these amino acids must be in your food every day.

The 10 amino acids that we can produce are alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, serine and tyrosine. Tyrosine is produced from phenylalanine, so if the diet is deficient in phenylalanine, tyrosine will be required as well.

The essential amino acids (marked * below) are arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine.

The three so-called branched-chain amino acids (BCAAs) are leucine, isoleucine and valine

The so-called aromatic amino acids (AAAs) are histidine, phenylanine, tryptophan and tyrosine

When plasma levels of BCAAs increase, this reduces the absorption of aromatic AAs; so the level of tryptophan, tyrosine, and phenylalanine will fall and this directly affects the synthesis and release of serotonin and catecholamines.
Many sportsmen, and indeed soldiers, take BCAA supplements in an attempt to build stronger muscles, but within the brain this will cause a cascade of other effects.
In people with tardive dyskinesia, which is a quite common tic disorder found in schizophrenia and autism, taking phenylalanine may make their tics worse.  It seems that taking BCAA supplements may make their tics reduce, because reducing the level of phenylalanine will impact dopamine (a catecholamine). Most movement disorders ultimately relate to dopamine.



In effect, BCAA supplements affect the synthesis and release of serotonin and catecholamines.  This might be good for you, or might be bad for you; it all depends where you started from.

   Alanine
   Arginine *
   Asparagine
   Aspartic acid
   Cysteine
   Glutamic acid
   Glutamine
   Glycine
   Histidine * Aromatic
   Isoleucine * BCAA
   Leucine * BCAA
   Lysine *
   Methionine *
   Phenylalanine *  Aromatic
   Proline
   Serine
   Threonine *
   Tryptophan * Aromatic
   Tyrosine  Aromatic
   Valine
*  BCAA


Blood levels of the BCAAs are elevated in people with obesity and those with insulin resistance, suggesting the possibility that BCAAs contribute to the pathogenesis of obesity and diabetes.  BCAA-restricted diets improve glucose tolerance and promote leanness in mice.


In the brain, BCAAs have two important influences on the production of neurotransmitters. As nitrogen donors, they contribute to the synthesis of excitatory glutamate and inhibitory gamma-aminobutyric acid (GABA) They also compete for transport across the blood-brain barrier (BBB) with tryptophan (the precursor to serotonin), as well as tyrosine and phenylalanine (precursors for catecholamines)Ingestion of BCAAs therefore causes rapid elevation of the plasma concentrations and increases uptake of BCAAs to the brain, but diminishes tryptophan, tyrosine, and phenylalanine uptake. The decrease in these aromatic amino acids directly affects the synthesis and release of serotonin and catecholamines. The reader is referred to Fernstrom (2005) for a review of the biochemistry of BCAA transportation to the brain. Oral BCAAs have been examined as treatment for neurological diseases such as mania, motor malfunction, amyotrophic lateral sclerosis, and spinocerebral degeneration. Excitotoxicity as a result of excessive stimulation by neurotransmitters such as glutamate results in cellular damage after traumatic brain injury (TBI). However, because BCAAs also contribute to the synthesis of inhibitory neurotransmitters, it is unclear to what extent the role of BCAAs in synthesis of both excitatory and inhibitory neurotransmitters might contribute to their potential effects in outcomes of TBI.

A list of human studies (years 1990 and beyond) evaluating the effectiveness of BCAAs in providing resilience or treating TBI or related diseases or conditions (i.e., subarachnoid hemorrhage, intracranial aneurysm, stroke, anoxic or hypoxic ischemia, epilepsy) in the acute phase is presented in Table 8-1; this also includes supporting evidence from animal models of TBI. The occurrence or absence of adverse effects in humans is included if reported by the authors.

Cell Signaling

Leucine indirectly activates p70 S6 kinase as well as stimulates assembly of the eIF4F complex, which are essential for mRNA binding in translational initiation. P70 S6 kinase is part of the mammalian target of rapamycin complex (mTOR) signaling pathway.



The present study provides the first evidence that mTOR signalling is enhanced in response to an acute stimulation with the proteinogenic amino acid, leucine, within cultured human myotubes. While these actions appear transient at the leucine dose utilised, activation of mTOR and p70S6K occurred at physiologically relevant concentrations independently of insulin stimulation. Interestingly, activation of mTOR signalling by leucine occurred in the absence of changes in the expression of genes encoding both the system A and system L carriers, which are responsible for amino acid transport. Thus, additional analyses are required to investigate the molecular mechanisms controlling amino acid transporter expression within skeletal muscle. Of note was the increased protein expression of hVps34, a putative leucine-sensitive kinase which intersects with mTOR. These results demonstrate the need for further clinical analysis to be performed specifically investigating the role of hVps34 as a nutrient sensing protein for mTOR signalling.

Skeletal muscle mass is determined by the balance between the synthesis and degradation of muscle proteins. Several hormones and nutrients, such as branched-chain amino acids (BCAAs), stimulate protein synthesis via the activation of the mammalian target of rapamycin (mTOR).
BCAAs (i.e., leucine, isoleucine, and valine) also exert a protective effect against muscle atrophy. We have previously reported that orally administered BCAA increases the muscle weight and cross-sectional area (CSA) of the muscle in rats



3.4. BCAAs in Brain Functions
BCAAs may also play important roles in brain function. BCAAs may influence brain protein synthesis and production of energy and may influence synthesis of different neurotransmitters, that is, serotonin, dopamine, norepinephrine, and so forth, directly or indirectly. Major portion of dietary BCAAs is not metabolized by liver and comes into systemic circulation after a meal. BCAAs and aromatic AA, such as tryptophan (Trp), tyrosine (Tyr), and phenylalanine (Phe), share the same transporter protein to transport into brain. Trp is the precursor of neurotransmitter serotonin; Tyr and Phe are precursors of catecholamines (dopamine, norepinephrine, and epinephrine). When plasma concentration of BCAAs increases, the brain absorption of BCAAs also increases with subsequent reduction of aromatic AA absorption. That may lead to decrease in synthesis of these related neurotransmitters [3]. Catecholamines are important in lowering blood pressure. When hypertensive rats were injected with Tyr, their blood pressure dropped markedly and injection with equimolar amount of valine blocks that action [49]. In vigorous working persons, such as in athletes, depletion of muscle and plasma BCAAs is normal. And that depletion of muscle and plasma BCAAs may lead to increase in Trp uptake by brain and release of serotonin. Serotonin on the other hand leads to central fatigue. So, supplementation of BCAAs to vigorously working person may be beneficial for their performance and body maintenance


Example of a treatable Amino Acid variant of Autism


Autism Spectrum Disorders (ASD) are a genetically heterogeneous constellation of syndromes characterized by impairments in reciprocal social interaction. Available somatic treatments have limited efficacy. We have identified inactivating mutations in the gene BCKDK (Branched Chain Ketoacid Dehydrogenase Kinase) in consanguineous families with autism, epilepsy and intellectual disability (ID). The encoded protein is responsible for phosphorylation-mediated inactivation of the E1-alpha subunit of branched chain ketoacid dehydrogenase (BCKDH). Patients with homozygous BCKDK mutations display reductions in BCKDK mRNA and protein, E1-alpha phosphorylation and plasma branched chain amino acids (BCAAs). Bckdk knockout mice show abnormal brain amino acid profiles and neurobehavioral deficits that respond to dietary supplementation. Thus, autism presenting with intellectual disability and epilepsy caused by BCKDK mutations represents a potentially treatable syndrome.

The data suggest that the neurological phenotype may be treated by dietary supplementation with BCAAs. To test this hypothesis, we studied the effect of a chow diet containing 2% BCAAs or a BCAA-enriched diet, consisting of 7% BCAAs, on the neurological phenotypes of the Bckdk−/− mice. Mice raised on the BCAA-enriched diet were phenotypically normal. On the 2% BCAA diet, however, Bckdk−/− mice had clear neurological abnormalities not seen in wild-type mice, such as seizures and hindlimb clasping, that appeared within 4 days of instituting the 2% BCAA diet (Fig. 3B). These neurological deficits were completely abolished within a week of the Bckdk−/− mice starting the BCAA-enriched diet, which suggests that they have an inducible yet reversible phenotype (Fig. 3C).

Our experiments have identified a Mendelian form of autism with comorbid ID and epilepsy that is associated with low plasma BCAAs. Although the incidence of this disease among patients with autism and epilepsy remains to be determined, it is probably quite a rare cause of this condition. We have shown that murine Bckdk−/− brain has a disrupted amino acid profile, suggesting a role for the BBB in the pathophysiology of this disorder. The mechanism by which abnormal brain amino acid levels lead to autism, ID, and epilepsy remains to be investigated. We have shown that dietary supplementation with BCAAs reverses some of the neurological phenotypes in mice. Finally, by supplementing the diet of human cases with BCAAs, we have been able to normalize their plasma BCAA levels (table S10), which suggests that it may be possible to treat patients with mutations in BCKDK with BCAA supplementation.


(Look at the three red rows, the BCAAs, all lower than the reference range, before supplementation)


Threonine, Mucin and Akkermansia muciniphila in Autism
Mucins are secreted as principal components of mucus by mucous membranes, like the lining of the intestines.  People with Inflammatory Bowel Disease (IBD) have mucus barrier changes.

The low levels of the mucolytic bacterium Akkermansia muciniphila found in children with autism, apparently suggests mucus barrier changes.

The amino acid Threonine is a component of mucin and Nestle have been researching for some time the idea of a threonine supplement to treat Inflammatory Bowel Disease (IBD), being a serious Swiss company they publish their research.      

Threonine Requirement in Healthy Adult Subjects and in Patients With Crohn's Disease and With Ulcerative Colitis Using the Indicator Amino Acid Oxidation (IAAO) Methodology

Threonine is an essential amino acid which must be obtained from the diet. It is a component of mucin. Mucin, in turn, is a key protein in the mucous membrane that protects the lining of the intestine.

Inflammatory bowel disease (IBD) is a group of inflammatory conditions that affect the colon and small intestine. IBD primarily includes ulcerative colitis (UC) and Crohn's disease (CD). In UC, the inflammation is usually in the colon whereas in CD inflammation may occur anywhere along the digestive tract. Studies in animals have shown that more threonine is used when there is inflammation in the intestine.

The threonine requirement in healthy participants and in IBD patients will be determined using the indicator amino acid oxidation method. The requirement derived in healthy participants will be compared to that derived in patients with IBD.

Each participant will take part in two x 3 day study periods. The first two days are called adaptation days where the subjects will consume a liquid diet specially designed for him. The diet will be consumed at home. It contains all vitamins, minerals, protein and all other nutrients required. On the third day, the participant will come to the Hospital for Sick Children in Toronto. Subjects will consume hourly meals for a total of 8 meals and a stable isotope 13C-phenylalanine. Breath and urine samples will be collected to measure the oxidation of phenylalanine from which the threonine requirement will be determined. 



We determined whether the steady-state levels of intestinal mucins are more sensitive than total proteins to dietary threonine intake. For 14 d, male Sprague-Dawley rats (158 ± 1 g, n = 32) were fed isonitrogenous diets (12.5% protein) containing 30% (group 30), 60% (group 60), 100% (control group), or 150% (group 150) of the theoretical threonine requirement for growth. All groups were pair-fed to the mean intake of group 30. The mucin and mucosal protein fractional synthesis rates (FSR) did not differ from controls in group 60. By contrast, the mucin FSR was significantly lower in the duodenum, ileum, and colon of group 30 compared with group 100, whereas the corresponding mucosal protein FSR did not differ. Because mucin mRNA levels did not differ between these 2 groups, mucin production in group 30 likely was impaired at the translational level. Our results clearly indicate that restriction of dietary threonine significantly and specifically impairs intestinal mucin synthesis. In clinical situations associated with increased threonine utilization, threonine availability may limit intestinal mucin synthesis and consequently reduce gut barrier function.
  


It has been proposed that excessive mucin degradation by intestinal bacteria may contribute to intestinal disorders, as access of luminal antigens to the intestinal immune system is facilitated. However, it is not known whether all mucin-degraders have the same effect. For example A. muciniphila may possess anti-inflammatory properties, as a high proportion of the bacteria has been correlated to protection against inflammation in diseases such as type 1 diabetes mellitus, IBD, atopic dermatitis, autism , type 2 diabetes mellitus, and.



Gastrointestinal disturbance is frequently reported for individuals with autism. We used quantitative real-time PCR analysis to quantify fecal bacteria that could influence gastrointestinal health in children with and without autism. Lower relative abundances of Bifidobacteria species and the mucolytic bacterium Akkermansia muciniphila were found in children with autism, the latter suggesting mucus barrier changes. 

Previous studies in rats by MacFabe et al. have shown that intraventricular administration of propionate induces behaviors resembling autism (e.g., repetitive dystonic behaviors, retropulsion, seizures, and social avoidance) (12, 13). We have also reported increased fecal propionate concentrations in ASD children compared with that in controls in the same fecal samples (25). However, the abundance of a key propionate-producing bacterium, Prevotella sp., was not significantly different between the study groups. This suggests that other untargeted bacteria, such as those from Clostridium cluster IX, which also includes major propionate producers (24), may be responsible for the observed differences in fecal propionate concentrations. Moreover, it is possible that the activities of the bacteria responsible for producing propionate, rather than bacterial numbers, have been altered. Other factors, such as differences in GI function that change GI transit time in ASD children, should also be considered.
In summary, the current findings of depleted populations of A. muciniphila and Bifidobacterium spp. add to our knowledge of the changes in the GI tracts of ASD children. These findings could potentially guide implementation of dietary/probiotic interventions that impact the gut microbiota and improve GI health in individuals with ASD.


Conclusion
I think that modifying levels of amino acids can have merit for some people, but it looks like another case for personalized medicine, rather than the same mix of powders given to everyone.
Threonine is interesting given the incidence of Inflammatory Bowel Disease (IBD) in autism.  IBD mainly describes ulcerative colitis and Crohn's disease.
The research into Threonine, is being funded by Nestle, the giant Swiss food company, who fortunately do publish their research.
The trial in the US of CM-AT is unusual because no results have ever been published in the literature, so we just have press releases. It likely that CM-AT is a mixture of pancreatic enzymes from pigs and perhaps some added amino acids.



This 14-week, double-blind, randomized, placebo-controlled Phase 3 study is being conducted to determine if CM-AT may help improve core and non-core symptoms of Autism. CM-AT, which has been granted Fast Track designation by FDA, is designed to enhance protein digestion thereby potentially restoring the pool of essential amino acids. Essential amino acids play a critical role in the expression of several genes important to neurological function and serve as precursors to key neurotransmitters such as serotonin and dopamine.


Based on the study I referred to early this year:-


·        Amino acids, his, lys and thr, inhibited mTOR pathway in antigen-activated mast cells

·     Amino acids, his, lys and thr inhibited degranulation and cytokine production of mast cells

·     Amino acid diet reversed mTOR activity in the brain and behavioral deficits in allergic and BTBR mice.

in my post:



I for one will be evaluating both lysine and threonine, having already found a modest dose histidine very beneficial in allergy (stabilizing mast cells).




Thursday 11 May 2017

Tardive Dyskinesia (TD)  - Amino Acids, VMAT2, Diamox, B6 etc


Today’s post is about Tardive Dyskinesia which is a side effect eventually experienced by about 30% of people taking antipsychotic drugs, like risperidone, widely prescribed in both autism and schizophrenia.




Enough money for your lifetime supply of a VMAT2 inhibitor?


Tardive Dyskinesia (TD) is a disorder resulting in involuntary, repetitive body movements, which might think of as tics or grimaces.

It appears that the longer the drug is taken the greater the chance of developing Tardive Dyskinesia.


Tics are quite common in autism and not just in those taking psychiatric drugs.

Tourette’s syndrome is a well-known tic disorder that does overlap with autism, it used to be considered rare, but now 1% of children are thought to be affected.  Some common Tourette’s tics are eye blinking, coughing, throat clearing, sniffing, and facial movements.

People diagnosed with Tourette’s might also be diagnosed with ADHD, OCD or indeed autism.  

We saw in some Italian research that young children with Tourette’s type autism have a fair chance of seeing their symptoms of autism substantially fade away. It was called Dysmaturational Syndrome.


The part of the brain that is thought to be affected in  Tardive Dyskinesia is that same part suspected in Tourette’s and indeed PANDAS/PANS; it is the basal ganglia.  


Avoiding Tardive Dyskinesia

The best way to avoid Tardive Dyskinesia is not to use antipsychotic/ neuroleptic drugs.

It appears that high doses of melatonin and other antioxidants may give a protective effect from developing Tardive Dyskinesia. 


Treating Tardive Dyskinesia

Much is written about treating Tardive Dyskinesia (TD), because nobody yet has found a cure for it, nonetheless there is a long list of partially effective therapies.

Given that the underlying basis of TD may very likely to overlap to some extent with Tourette’s and PANDAS/PANS it is over broader interest.  


A Review of off-label Treatments  for Tardive Dyskinesia 

The Spanish study below gives a good overview of most therapies, but exclude the very recent therapies based on VMAT2. 






  
All of the studies in the review were small, but you can see that some therapies did seem to help, including:-

·        Vitamin E

·        Vitamin B6

·        Acetazolamide/Diamox

·        Amino Acids

·        Piracetam


I proposed Acetazolamide/Diamox to potentially treat some autism a while back and some readers of this blog do find it effective.

Piracetam is the world’s first cognitive enhancing (nootropic) drug.  It was discovered by mistake when trying to make a cure for motion sickness.

Amino acids may look an odd choice, but in males, and only males, branched chain  amino acids (BCAAs) valine, isoleucine, and leucine in a 3:3:4 ratio appears to be beneficial.  Another amino acid called Phenylalanine is associated with tardive dyskinesia in men but not in women. It is an established fact that an increase in BCAAs will cause a reduction in phenylalanine in the brain, among a range of other effects.

Phenylalanine is a precursor for dopamine (as well as  tyrosine, norepinephrine, and epinephrine). 

One theory is that tardive dyskinesia results primarily from neuroleptic-induced dopamine supersensitivity. So if the BCAAs reduce the amount of dopamine produced, this might explain their effect.



VMAT2

VMAT2 transports monoamines - particularly neurotransmitters such as dopamine, norepinephrine, serotonin, and histamine - from cellular cytosol into synaptic vesicles. Inhibiting VMAT2 will reduce the release of dopamine. 

In some circumstances VMAT2 is necessary for the release of the neurotransmitter GABA. 

Drugs that inhibit VMAT2 appear to help treat Tardive Dyskinesia and one drug Valbenazine/Ingrezza was FDA approved for this purpose in April 2017. Not surprisingly, it is now being investigated to treat Tourette’s syndrome. 

Valbenazine is known to cause a reversible reduction of dopamine release by selectively inhibiting VMAT2. 



Conclusion

Since our regular reader Valentina is dealing with Tardive Dyskinesia, she will probably be very interested in Valbenazine.

The problem is the price. The drug will apparently cost $60,000 a year in the US.

So for the time being it is best to work through the list of very cheap interventions that do seem to be partially effective, at least in some people.













Thursday 11 June 2015

mTOR – Indirect inhibition, the Holy Grail for Life Extension and Perhaps Some Autism




 Not cheap at about $1,000 for just 140mg


Life extension may come as a surprise, but it is interesting because it is well studied and, in mice at least, easy to measure.  Most research into mTOR relates to cancer, but this is a very complex condition. With various feedback loops it means that sometimes the actual effect is the opposite of what was predicted.  For example, a substance that can help prevent cancer can actually become harmful later and promote its growth.

Direct inhibition of mTOR with Everolimus and similar drugs (variants/analogs of Rapamycin, all called Rapalogs) has not been as successful as hoped in cancer research.  Trials of direct inhibition of mTOR will shortly start in one rare single gene type of autism (TSC).  The drugs are so expensive that many providers do not want to pay for them.

As you will see mTOR is just one process in a cloud of interrelated processes.  Almost everything has a role/effect:- growth factors, cytokines, amino acids, mitochondria, dendritic spines, PPAR gamma, hormones, oxidative stress, autophagy ….

While it would be nice to think that a single protein complex like mTORC1 or mTORC2 is the root of all evil in autism, I rather doubt it can be so simple.

The knowledge that one factor controlling mTORC1 and mTORC2 is oxidative stress, does raise the possibility that, yet again, the root problem could be oxidative stress.  
Nonetheless, we will see in today’s post that too much mTOR activity is clearly not good and that it is associated with lots of bad things:-

·        Epilepsy
·        Autistic behaviours
·        Food allergies
·        Mitochondrial dysfunction
·        Cognitive impairment

as well as aging, cancer ….


Indirect reduction in mTOR activity

Rather than the very expensive first and second generation mTOR inhibiting drugs developed for cancer,  I think the safe way forward for autism (and aging) may be indirect reduction in mTOR activity, and there is already a wide choice of methods.

Ketogenic Diet, (or just reduction in carbohydrate intake)

This diet has been used for a hundred years to control epilepsy, which it now seems can be triggered by elevated mTOR.  Research has shown that the ketogenic diet reduces mTOR. 

Low glycemic index diet

This is a low carbohydrate, no sugar diet, typical of someone with diabetes.  It avoids rapid change in blood sugar.  This will lower mTOR and has recently been shown in a mouse model to improve autistic behaviors.

Growth factors

The blood levels of growth factors such as insulin and IGF-1 reflect the fed status of the organism. When food is plentiful, levels of these growth factors are sustained and promote anabolic cell processes such as translation, lipid biosynthesis, and nutrient storage via mTORC1.  So, dietary restriction, which lowers IGF-1, will reduce mTOR; but it will also reduce growth.
Note that one autism therapy under trial does just the opposite, it is to increase IGF-1 levels via injections of IGF-1.

Increase amino acids, particularly leucine

Ask any body builder about BCAA (Branch Chained Amino Acids)

  
Reduce oxidative stress

We know how to do that

NMDA agonists

NMDA receptor activation decreases mTOR signaling activity. 


Note that D-Cycloserine is used in autism and D-Serine is used in schizophrenia


Increase PTEN, for example with a Statin drug


Reduce RAS signaling, for example with a Statin drug


I am not the first person to realize this.  Here is a very highly cited paper:-


  
Since the body is controlled via feedback loops, there might exist a clever way to “trick” the body into lowing mTOR.  For example PPAR gamma, which we have come across in earlier posts, is controlled via mTOR.  If you stimulate PPAR gamma externally this might well have an effect back stream on mTOR activity, via these feedback loops.  Just like if you supplement Melatonin, you will likely affect the behaviour back stream of the pineal gland.


mTOR and Aging

A surprising number of emerging autism therapies are actually also put forward by the life extension people.  In case you did not know, there is a small industry of pills and potions dedicated to making you live longer.  Some serious institutions like MIT and Harvard are involved, as in the paper below.



We earlier saw that PAK-1 is probably there to make sure you do eventually die, reducing mTOR signaling can probably extend your lifetime and, more importantly, your healthy lifetime.


Ketogenic Diet

We did see a case report a while back from Martha Herbert, from Harvard, who has a good result with the ketogenic diet



  
  
The Science of mTOR

In the following section there are numerous scientific papers explaining mTOR, so you can choose just how deep you want to go into the details.

You may notice on the complex diagram below various substances that we have already encountered in this blog as relevant to autism.

·        PTEN ( increased by Statins) reduced in some autism
·        Growth factors (disturbed in autism and therapeutic to some)
·        Ras / Rasopathy (increased by statins, linked to some autism and MR/ID )
·        Wnt (affects morphology of those dendritic spines, malformed in autism)
·        Lipid metabolism/synthesis (disturbed in autism)
·        TSC1  (tuberous sclerosis autism variant)
·        PPAR alpha and gamma affecting inflammation
·        Mitochondrial metabolism, dysfunctional in autism
·        Autophagy was explained in recent post and, if impaired, will degrade cellular health and function, particularly in mitochondria
·        Note Stress/Hypoxia, we have mentioned Hypoxia before.  REDD1 inhibits mTOR.  REDD1 was first identified as a gene induced by hypoxia and DNA damage, other environmental stresses such as energy stress, glucocorticoid treatment and reactive oxygen species have also been reported to induce REDD1 transcription  





Pathway Description: The mechanistic target of Rapamycin (mTOR) is an atypical serine/threonine kinase that is present in two distinct complexes.
The first, mTOR complex 1 (mTORC1), is composed of mTOR, Raptor, GβL, and DEPTOR and is inhibited by Rapamycin. It is a master growth regulator that senses and integrates diverse nutritional and environmental cues, including growth factors, energy levels, cellular stress, and amino acids. It couples these signals to the promotion of cellular growth by phosphorylating substrates that potentiate anabolic processes such as mRNA translation and lipid synthesis, or limit catabolic processes such as autophagy. The small GTPase Rheb, in its GTP-bound state, is a necessary and potent stimulator of mTORC1 kinase activity, which is negatively regulated by its GAP, the tuberous sclerosis heterodimer TSC1/2. Most upstream inputs are funneled through Akt and TSC1/2 to regulate the nucleotide-loading state of Rheb. In contrast, amino acids signal to mTORC1 independently of the PI3K/Akt axis to promote the translocation of mTORC1 to the lysosomal surface where it can become activated upon contact with Rheb. This process is mediated by the coordinated actions of multiple complexes, notably the v-ATPase, Ragulator, the Rag GTPases, and GATOR1/2.

The second complex, mTOR complex 2 (mTORC2), is composed of mTOR, Rictor, GβL, Sin1, PRR5/Protor-1, and DEPTOR. mTORC2 promotes cellular survival by activating Akt, regulates cytoskeletal dynamics by activating PKCα, and controls ion transport and growth via SGK1 phosphorylation.
Aberrant mTOR signaling is involved in many disease states including cancer, cardiovascular disease, and diabetes.






Growth factors regulate mTORC1
Energy and stress regulate mTORC1
mTOR regulates metabolism in mammals
mTOR in fasting and starvation
mTOR, over-feeding, and insulin sensitivity
One of the most efficient forms of energy storage are triglycerides, because they provide a high energetic yield per unit of mass. mTORC1 mediates lipid accumulation in fat cells
mTORC1 may impact on PPAR-γ activity by increasing its translation118 and by activating the transcription factor SREBP-1c . Active SREBP-1c enhances PPAR-γ activity and transactivates a set of genes directly involved in lipid synthesis. At present, the molecular links between mTORC1, SREBP-1c and PPAR-γ activity remain to be clarified.

Thus, mTORC1 coordinates food intake with energy storage at multiple levels, from central control of food seeking to energy storage and expenditure in peripheral tissues. This multi-level regulation explains the profound consequences that dysregulated mTOR signaling exerts on human metabolism.

Aging

Due to its role at the interface of growth and starvation, mTOR is a prime target in the genetic control of ageing, and evidence from genetic studies supports the view that mTOR may be a master determinant of lifespan and ageing in yeast, C. elegans, flies and mice.
The only ‘natural’ method available to counter ageing is dietary restriction (DR), where the caloric intake is decreased anywhere from 10% to 50%. DR appears to act mainly through the inhibition of mTORC1, and genetic inactivation of mTORC1 pathway components provides no additional benefit over DR. In mice, DR causes lifespan extension and changes in gene expression profile similar to those resulting from loss of S6K1 further supporting the view that DR acts through inhibition of mTORC1
Finally, it remains to be seen whether limiting mTOR activity in adult humans would really enable a longer lifespan, or it would only bring an increase in the quality of life and the way we age, without necessarily affecting how long we live.


mTOR in food allergy


Highlights
mTOR pathway is implicated in gut–brain axis of food allergy-induced ASD-like behavior.
Food allergy is associated with enhanced mTOR signaling in the brain and gut.
mTORC1 inhibitor Rapamycin improved the behavioral deficits of allergic mice.
Rapamycin reduced mTORC1 activity in the brain and gut of allergic mice.
Rapamycin inhibited food allergy and increased the number of Treg cells in the ileum.


5. Conclusions

In conclusion, the current studies provide strong and first evidence
that the enhanced mTOR signaling pathway in the brain as well as in the intestines plays a pivotal role in the behavioral and immunological changes in CMA mice. mTOR might be the linking pin involved in gut-immune-brain axis in ASD and the intestinal tract could be a potential target in the treatment of patients with ASD and comorbid intestinal symptoms. It is a compelling hypothesis that an enhanced mTOR activity throughout the body may account for both the behavioral as well as the gastrointestinal dysfunctions in patients with ASD. Whether inhibition of mTOR is able to treat both allergic and behavioral deficits of ASD patients remains to be further investigated. Importantly, increased gastrointestinal deficits and in particular behavioral abnormalities are commonly reported in other neurodevelopmental diseases such as attention deficit hyperactivity disorder (ADHD), multiple sclerosis , schizophrenia, Parkinson's disease , however the role of mTOR needs to be investigated. Our findings on the gut-immune-brain connection in a murine model of CMA indicate that targeting mTOR signaling pathway might be applicable to various neurological disorders. Future studies focusing on the mTOR signaling pathway should shed more light on the effective treatment of ASD and other neurodevelopmental disorders.


  
mTOR and Autism



Hyperconnectivity of neuronal circuits due to increased synaptic protein synthesis is thought to cause autism spectrum disorders (ASDs). The mammalian target of Rapamycin (mTOR) is strongly implicated in ASDs by means of upstream signaling; however, downstream regulatory mechanisms are ill-defined. Here we show that knockout of the eukaryotic translation initiation factor 4E-binding protein 2 (4E-BP2)—an eIF4E repressor downstream of mTOR—or eIF4E overexpression leads to increased translation of neuroligins, which are postsynaptic proteins that are causally linked to ASDs. Mice that have the gene encoding 4E-BP2 (Eif4ebp2) knocked out exhibit an increased ratio of excitatory to inhibitory synaptic inputs and autistic-like behaviours (that is, social interaction deficits, altered communication and repetitive/stereotyped behaviours). Pharmacological inhibition of eIF4E activity or normalization of neuroligin 1, but not neuroligin 2, protein levels restores the normal excitation/inhibition ratio and rectifies the social behaviour deficits. Thus, translational control by eIF4E regulates the synthesis of neuroligins, maintaining the excitation-to-inhibition balance, and its dysregulation engenders ASD-like phenotypes.



 Reversing autism by targeting downstream mTOR signaling
 Autism spectrum disorders (ASDs) are a group of clinically and genetically heterogeneous neurodevelopmental disorders characterized by impaired social interactions, repetitive behaviors and restricted interests. The genetic defects in ASDs may interfere with synaptic protein synthesis. Synaptic dysfunction caused by aberrant protein synthesis is a key pathogenic mechanism for ASDs Understanding the details about aberrant synaptic protein synthesis is important to formulate potential treatment for ASDs. The mammalian target of the Rapamycin (mTOR) pathway plays central roles in synaptic protein. Recently, Gkogkas and colleagues published exciting data on the role of downstream mTOR pathway in autism





Previous studies have indicated that upstream mTOR signaling is linked to ASDs. Mutations in tuberous sclerosis complex (TSC) 1/TSC2, neurofibromatosis 1 (NF1), and Phosphatase and tensin homolog (PTEN) lead to syndromic ASD with tuberous sclerosis, neurofibromatosis, or macrocephaly, respectively. TSC1/TSC2, NF1, and PTEN act as negative regulators of mTOR complex 1 (mTORC1), which is activated by phosphoinositide-3 kinase (PI3K) pathway. Activation of cap-dependent translation is a principal downstream mechanism of mTORC1. The eIF4E recognizes the 5′ mRNA cap, recruits eIF4G and the small ribosomal subunit. The eIF4E-binding proteins (4E-BPs) bind to eIF4E and inhibit translation initiation. Phosphorylation of 4E-BPs by mTORC1 promotes eIF4E release and initiates cap-dependent translation. A hyperactivated mTORC1–eIF4E pathway is linked to impaired synaptic plasticity in fragile X syndrome, an autistic disorder caused by lack of fragile X mental retardation protein (FMRP) due to mutation of the FMR1 gene, suggesting that downstream mTOR signaling might be causally linked to ASDs. Notably, one pioneering study has identified a mutation in the EIF4E promoter in autism families, implying that deregulation of downstream mTOR signaling (eIF4E) could be a novel mechanism for ASDs.As an eIF4E repressor downstream of mTOR, 4E-BP2 has important roles in synaptic plasticity, learning and memory. Writing in their Nature article, Gkogkas and colleagues reported that deletion of the gene encoding 4E-BP2 (Eif4ebp2) leads to autistic-like behaviors in mice. Pharmacological inhibition of eIF4E rectifies social behavior deficits in Eif4ebp2 knockout mice. Their study in mouse models has provided direct evidence for the causal link between dysregulated eIF4E and the development of ASDs.Are these ASD-like phenotypes of the Eif4ebp2 knockout mice caused by altered translation of a subset mRNAs due to the release of eIF4E? To test this, Gkogkas et al. measured translation initiation rates and protein levels of candidate genes known to be associated with ASDs in hippocampi from Eif4ebp2 knockout and eIF4E-overexpressing mice. They found that the translation of neuroligin (NLGN) mRNAs is enhanced in both lines of transgenic mice. Removal of 4E-BP2 or overexpression of eIF4E increases protein amounts of NLGNs in the hippocampus, whereas mRNA levels are not affected, thus excluding transcriptional effect. In contrast, the authors did not observe any changes in the translation of mRNAs coding for other synaptic scaffolding proteins. Interestingly, treatment of Eif4ebp2 knockout mice with selective eIF4E inhibitor reduces NLGN protein levels to wild-type levels. These data thus indicate that relief of translational suppression by loss of 4E-BP2 or by the overexpression of eIF4E selectively enhances the NLGN synthesis. However, it cannot be ruled out that other proteins (synaptic or non-synaptic) may be affected and contribute to animal autistic phenotypes.Aberrant information processing due to altered ratio of synaptic excitation to inhibition (E/I) may contribute to ASDs. The increased or decreased E/I ratio has been observed in ASD animal models  In relation to these E/I shifts, Gkogkas et al then examined the synaptic transmission in hippocampal slices of Eif4ebp2 knockout mice. They found that 4E-BP2 de-repression results in an increased E/I ratio, which can be explained by the increase of vesicular glutamate transporter and spine density in hippocampal pyramidal neurons. As expected, application of eIF4E inhibitor restores the E/I balanceFinally, in view of the facts that genetic manipulation of NLGNs results in ASD-like phenotypes with altered E/I balance in mouse models  and NLGN mRNA translation is enhanced concomitant with increased E/I ratio in Eif4ebp2 knockout mice, Gkogkas et al. tested the effect of NLGN knockdown on synaptic plasticity and behaviour in these mice . NLGN1 is predominantly postsynaptic at excitatory synapses and promotes excitatory synaptic transmission. The authors found that NLGN1 knockdown reverses changes at excitatory synapses and partially rescues the social interaction deficits in Eif4ebp2 knockout mice. These findings thus established a strong link between eIF4E-dependent translational control of NLGNs, E/I balance and the development of ASD-like animal behaviors (Figure 1).
In summary, Gkogkas et al. have provided a model for mTORC1/eIF4E-dependent autism-like phenotypes due to dysregulated translational control (Gkogkas et al., 2013). This novel regulatory mechanism will prompt investigation of downstream mTOR signaling in ASDs, as well as expand our knowledge of how mTOR functions in human learning and cognition. It may narrow down therapeutic targets for autism since targeting downstream mTOR signaling reverses autism. Pharmacological manipulation of downstream effectors of mTOR (eIF4E, 4E-BP2, and NLGNs) may eventually provide therapeutic benefits for patients with ASDs.

  



3.3. Autism
As with epilepsy, the link between aberrant mTOR activation and autism is strongest in tuberous sclerosis complex; between 20 and 60% of tuberous sclerosis patients are diagnosed with autism [219, 237], which may account for 1–4% of all autism cases [238]. In addition to tuberous sclerosis, however, there is growing evidence that dysregulated mTOR activity may contribute to a wider variety of autism spectrum disorders. As with epilepsy, mutations in PTEN that lead to aberrant activation of mTOR are associated with autism [239]. In addition, mutations in the downstream mTOR target eukaryotic translation initiation factor 4E (eIF4E) have also been associated with autism [240]. There is also evidence for a strong association between macrocephaly (large head size) early in life and autism spectrum disorders, as well as genetic diseases linked to autism and mTOR hyperactivation, including tuberous sclerosis complex, neurofibromatosis type I, Lhermitte-Duclos syndrome, and Fragile X syndrome [241]. Taken together these data suggest that disinhibited mTOR may cause, or at least contribute to, many cases of autism spectrum disorder. Clinical trials are ongoing to assess whether Everolimus can reduce autistic symptoms in tuberous sclerosis patients.

5. Conclusion
Given the breadth of pathological conditions where mTOR has already been implicated, it seems likely that additional therapeutic uses for mTOR inhibitors will be discovered in the near future. While potential negative effects of mTOR inhibition need to be addressed, they appear generally manageable and, as new mTOR inhibitors continue to be developed, it may be possible to maximize the beneficial effects of targeted mTOR inhibition while reducing adverse effects.






This paper is very comprehensive and this graphic has everything you could ever need to know.  You can use it to figure out your own therapy.












mTOR and seizures




Epilepsy, a common neurological disorder and cause of significant morbidity and mortality, places an enormous burden on the individual and society. Presently, most drugs for epilepsy primarily suppress seizures as symptomatic therapies but do not possess actual antiepileptogenic or disease-modifying properties. The mTOR (mammalian target of Rapamycin) signaling pathway is involved in major multiple cellular functions, including protein synthesis, cell growth and proliferation and synaptic plasticity, which may influence neuronal excitability and be responsible for epileptogenesis. Intriguing findings of the frequent hyperactivation of mTOR signaling in epilepsy make it a potential mechanism in the pathogenesis as well as an attractive target for the therapeutic intervention, and have driven the significant ongoing efforts to pharmacologically target this pathway. This review explores the relevance of the mTOR pathway to epileptogenesis and its potential as a therapeutic target in epilepsy treatment by presenting the current results on mTOR inhibitors, in particular, Rapamycin, in animal models of diverse types of epilepsy. Limited clinical studies in human epilepsy, some paradoxical experimental data and outstanding questions have also been discussed.



  
The ketogenic diet (KD) is an effective treatment for epilepsy, but its mechanisms of action are poorly understood. We investigated the hypothesis that KD inhibits mammalian target of Rapamycin (mTOR) pathway signaling. The expression of pS6 and pAkt, markers of mTOR pathway activation, was reduced in hippocampus and liver of rats fed KD. In the kainate model of epilepsy, KD blocked the hippocampal pS6 elevation that occurs after status epilepticus. As mTOR signaling has been implicated in epileptogenesis, these results suggest that the KD may have anticonvulsant or antiepileptogenic actions via mTOR pathway inhibition.







Highlights

Tsc1 deletion in neurons causes epilepsy and autism-like behaviors in mice.
Epileptiform activity spreads to the brainstem.
mTOR becomes hyperactivated in 5-HT neurons following seizure onset.
mTOR hyperactivity in 5-HT neurons causes autism behaviors.
Autism-like behaviors can be reversed following treatment with Rapamycin.

Abstract
Epilepsy and autism spectrum disorder (ASD) are common comorbidities of one another. Despite the prevalent correlation between the two disorders, few studies have been able to elucidate a mechanistic link. We demonstrate that forebrain specific Tsc1 deletion in mice causes epilepsy and autism-like behaviors, concomitant with disruption of 5-HT neurotransmission. We find that epileptiform activity propagates to the raphe nuclei, resulting in seizure-dependent hyperactivation of mTOR in 5-HT neurons. To dissect whether mTOR hyperactivity in 5-HT neurons alone was sufficient to recapitulate an autism-like phenotype we utilized Tsc1flox/flox;Slc6a4-cre mice, in which mTOR is restrictively hyperactivated in 5-HT neurons. Tsc1flox/flox;Slc6a4-cre mice displayed alterations of the 5-HT system and autism-like behaviors, without causing epilepsy. Rapamycin treatment in these mice was sufficient to rescue the phenotype. We conclude that the spread of seizure activity to the brainstem is capable of promoting hyperactivation of mTOR in the raphe nuclei, which in turn promotes autism-like behaviors. Thus our study provides a novel mechanism describing how epilepsy can contribute to the development of autism-like behaviors, suggesting new therapeutic strategies for autism.




mTOR inhibition via carbohydrate restriction







  


  



Amino acids and mTOR




The activity of mammalian target of Rapamycin (mTOR) complexes regulates essential cellular processes, such as growth, proliferation or survival. Nutrients such as amino acids are important regulators of mTOR Complex 1 (mTORC1) activation, thus affecting cell growth, protein synthesis and autophagy.
Here, we show that amino acids may also activate mTOR Complex 2 (mTORC2). This activation is mediated by the activity of class I PI3K and of Akt. Amino acids induced a rapid phosphorylation of Akt at Thr308 and Ser473. Whereas both phosphorylations were dependent on the presence of mTOR, only Akt phosphorylation at Ser473 was dependent on the presence of rictor, a specific component of mTORC2. Kinase assays confirmed mTORC2 activation by amino acids. This signaling was functional, as demonstrated by the phosphorylation of Akt substrate FOXO3a. Interestingly, using different starvation conditions, amino acids can selectively activate mTORC1 or mTORC2. These findings identify a new signaling pathway used by amino acids underscoring the crucial importance of these nutrients in cell metabolism and offering new mechanistic insights.

Finally, this report shows the crucial importance of dietary restriction/starvation conditions for understanding the amino acid signaling. Several studies show the effects of amino acid intake in obesity [23,27,28], and of dietary restriction in human cancers [79,80]. Although more physiological studies are needed to link these effects to mTOR complex regulation, it is noteworthy that a study in human muscle shows activation of both mTORC1 and mTORC2 by ingestion of
a leucine-enriched amino acid-carbohydrate mixture [86]. It has been recently described that branched-chain amino acid dietary supplementation increased the average life span in mice and cardiac and skeletal muscle improvement [87] validating the physiological relevance of amino acid supplementation. In this context, we now report that cell supplementation with amino acids can activate both mTOR complexes (Figures 10 and 11). In summary, this manuscript shows for the first time that amino acids can activate mTORC1 and mTORC2 complexes, thus underscoring the crucial importance of these nutrients in cell metabolism and offering new mechanistic insights with potential therapeutic applications in cancer, obesity and aging.

  

Recent evidence points to a strong relationship between increased mitochondrial biogenesis and increased survival in eukaryotes. Branched-chain amino acids (BCAAs) have been shown to extend chronological life span in yeast. However, the role of these amino acids in mitochondrial biogenesis and longevity in mammals is unknown. Here, we show that a BCAA-enriched mixture (BCAAem) increased the average life span of mice. BCAAem supplementation increased mitochondrial biogenesis and sirtuin 1 expression in primary cardiac and skeletal myocytes and in cardiac and skeletal muscle, but not in adipose tissue and liver of middle-aged mice, and this was accompanied by enhanced physical endurance. Moreover, the reactive oxygen species (ROS) defense system genes were upregulated, and ROS production was reduced by BCAAem supplementation. All of the BCAAem-mediated effects were strongly attenuated in endothelial nitric oxide synthase null mutant mice. These data reveal an important antiaging role of BCAAs mediated by mitochondrial biogenesis in mammals.

  

Amino acid deficiency causing Autism



A rare, hereditary form of autism has been found — and it may be treatable with protein supplements.

Genome sequencing of six children with autism has revealed mutations in a gene that stops several essential amino acids being depleted. Mice lacking this gene developed neurological problems related to autism that were reversed by dietary changes, a paper published today in Science shows1.
Some children with autism have low blood levels of amino acids that can't be made in the body.
“This might represent the first treatable form of autism,” says Joseph Gleeson, a child neurologist at the University of California, San Diego, who led the study. “That is both heartening to families with autism, and also I think revealing of the underlying mechanisms of autism.”

He emphasizes, however, that the mutations are likely to account for only a very small proportion of autism cases. “We don’t anticipate this is going to have implications for patients in general with autism,” says Gleeson. And there is as yet no proof that dietary supplements will help the six children, whose mutations the researchers identified by sequencing the exome — the part of the genome that codes for proteins.

In mice, at least, the chemical imbalance can be treated. The mutant mice had neurological problems typical of mouse versions of autism, including tremors and epileptic seizures. But those symptoms disappeared in less than a week after the mice were put on diets enriched in branched-chain amino acids.

Gleeson’s team has tried supplementing the diets of the children with this form autism, using muscle-building supplements that contain branched-chain amino acids. The researchers found that the supplements restore the children's blood levels of amino acids to normal. As for their autism symptoms, Gleeson says, the “patients did not get any worse and their parents say they got better, but it’s anecdotal”.

  

  
This paper is very recent and suggests, at least in one mouse model, that oxygen consumption in the brain is dysfunction and that this was rescued using the mTOR inhibitor Rapamycin.

  
Tuberous sclerosis (TSC) is associated with autism spectrum disorders and has been linked to metabolic dysfunction and unrestrained signaling of the mammalian target of Rapamycin (mTOR). Inhibition of mTOR by Rapamycin can mitigate some of the phenotypic abnormalities associated with TSC and autism, but whether this is due to the mTOR-related function in energy metabolism remains to be elucidated. In young Eker rats, an animal model of TSC and autism, which harbors a germ line heterozygous Tsc2 mutation, we previously reported that cerebral oxygen consumption was pronouncedly elevated. Young (4 weeks) male control Long–Evans and Eker rats were divided into control and Rapamycin-treated (20 mg/kg once daily for 2 days) animals. Cerebral regional blood flow (14C-iodoantipyrine) and O2 consumption (cryomicrospectrophotometry) were determined in isoflurane-anesthetized rats. We found significantly increased basal O2 consumption in the cortex (8.7 ± 1.5 ml O2/min/100 g Eker vs. 2.7 ± 0.2 control), hippocampus, pons and cerebellum. Regional cerebral blood flow and cerebral O2 extractions were also elevated in all brain regions. Rapamycin had no significant effect on O2 consumption in any brain region of the control rats, but significantly reduced consumption in the cortex (4.1 ± 0.3) and all other examined regions of the Eker rats. Phosphorylation of mTOR and S6K1 was similar in the two groups and equally reduced by Rapamycin. Thus, a Rapamycin-sensitive, mTOR-dependent but S6K1-independent, signal led to enhanced oxidative metabolism in the Eker brain. We found decreased Akt phosphorylation in Eker but not Long–Evans rat brains, suggesting that this may be related to the increased cerebral O2 consumption in the Eker rat. Our findings suggest that Rapamycin targeting of Akt to restore normal cerebral metabolism could have therapeutic potential in tuberous sclerosis and autism.



Mitochondrial Dysfunction  and mTOR
  
  
Mitochondria are organelles that play a central role in processes related to cellular viability, such as energy production, cell growth, cell death via apoptosis, and metabolism of reactive oxygen species (ROS). We can observe behavioral abnormalities relevant to autism spectrum disorders (ASDs) and their recovery mediated by the mTOR inhibitor Rapamycin in mouse models. In Tsc2+/- mice, the transcription of multiple genes involved in mTOR signaling is enhanced, suggesting a crucial role of dysregulated mTOR signaling in the ASD model. This review proposes that the mTOR inhibitor may be useful for the pharmacological treatment of ASD. This review offers novel insights into mitochondrial dysfunction and the related impaired glutathione synthesis and lower detoxification capacity. Firstly, children with ASD and concomitant mitochondrial dysfunction have been reported to manifest clinical symptoms similar to those of mitochondrial disorders, and it therefore shows that the clinical manifestations of ASD with a concomitant diagnosis of mitochondrial dysfunction are likely due to these mitochondrial disorders. Secondly, the adenosine triphosphate (ATP) production/oxygen consumption pathway may be a potential candidate for preventing mitochondrial dysfunction due to oxidative stress, and disruption of ATP synthesis alone may be related to impaired glutathione synthesis. Finally, a decrease in total antioxidant capacity may account for ASD children who show core social and behavioral impairments without neurological and somatic symptoms.



PTEN-type Autism and mTOR



Germline mutations in PTEN, which encodes a widely expressed phosphatase, was mapped to 10q23 and identified as the susceptibility gene for Cowden syndrome, characterized by macrocephaly and high risks of breast, thyroid, and other cancers. The phenotypic spectrum of PTEN mutations expanded to include autism with macrocephaly only 10 years ago. Neurological studies of patients with PTEN-associated autism spectrum disorder (ASD) show increases in cortical white matter and a distinctive cognitive profile, including delayed language development with poor working memory and processing speed. Once a germline PTEN mutation is found, and a diagnosis of phosphatase and tensin homolog (PTEN) hamartoma tumor syndrome made, the clinical outlook broadens to include higher lifetime risks for multiple cancers, beginning in childhood with thyroid cancer. First described as a tumor suppressor, PTEN is a major negative regulator of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of Rapamycin (mTOR) signaling pathway—controlling growth, protein synthesis, and proliferation. This canonical function combines with less well-understood mechanisms to influence synaptic plasticity and neuronal cytoarchitecture. Several excellent mouse models of Pten loss or dysfunction link these neural functions to autism-like behavioral abnormalities, such as altered sociability, repetitive behaviors, and phenotypes like anxiety that are often associated with ASD in humans. These models also show the promise of mTOR inhibitors as therapeutic agents capable of reversing phenotypes ranging from overgrowth to low social behavior. Based on these findings, therapeutic options for patients with PTEN hamartoma tumor syndrome and ASD are coming into view, even as new discoveries in PTEN biology add complexity to our understanding of this master regulator


Intellectual Disability (MR) and mTOR




Protein synthesis regulation via mammalian target of Rapamycin complex 1 (mTORC1) signaling pathway has key roles in neural development and function, and its dysregulation is involved in neurodevelopmental disorders associated with autism and intellectual disability. mTOR regulates assembly of the translation initiation machinery by interacting with the eukaryotic initiation factor eIF3 complex and by controlling phosphorylation of key translational regulators. Collybistin (CB), a neuron-specific Rho-GEF responsible for X-linked intellectual disability with epilepsy, also interacts with eIF3, and its binding partner gephyrin associates with mTOR. Therefore, we hypothesized that CB also binds mTOR and affects mTORC1 signaling activity in neuronal cells. Here, by using induced pluripotent stem cell-derived neural progenitor cells from a male patient with a deletion of entire CB gene and from control individuals, as well as a heterologous expression system, we describe that CB physically interacts with mTOR and inhibits mTORC1 signaling pathway and protein synthesis. These findings suggest that disinhibited mTORC1 signaling may also contribute to the pathological process in patients with loss-of-function variants in CB.



mTORC2 as opposed to mTORC1 as a target in Autism Research



The goal of my DOD-supported research is determine the role of the new mTOR complex (mTORC2) in Autism Spectrum Disorder (ASD). ASD individuals exhibit impaired social interactions, seizures and abnormal repetitive behavior. In addition, 70-80% of autistic individuals suffer from mental retardation. Autism is a heritable genetically heterogeneous disorder and mutations in negative regulators of the mammalian target of Rapamycin complex 1 (mTORC1) signaling pathway, such as PTEN were associated with ASD. Here, we show that in the hippocampus of Pten fb-KO mice – where Pten is conditionally deleted in the murine forebrain – the activity of both mTORC1 and mTORC2 is increased. In addition, Pten fb-KO mice exhibit seizures, learning and memory and social deficits. Our remarkable preliminary data show that genetic inhibition of mTORC2 activity in Pten-deficient mice significantly promotes survival. In addition, Pten-rictor fb- double KO (DKO) mice, in which mTORC2 activity is restored to normal levels, EEG seizures, learning and memory as well as social phenotypes, are all rescued. In the second year, we will study the molecular mechanism underlying this process. These insights hold the promise for new treatment of ASD.



1. Introduction:

Autism represents a heterogeneous group of disorders, which are defined as “autism spectrum disorders” (ASDs). ASD individuals exhibit common features such as impaired social interactions, language and communication, and abnormal repetitive behavior. In addition, 70-80% of autistic individuals suffer from mental retardation1-3. The major goal of this award is to determine the role of mTORC2 in two mouse models of ASD.

Recently, we have shown that mTORC2 plays a crucial role in long-term memory formation. Briefly, mice lacking mTORC2 showed impaired long-lasting changes in synaptic strength (L-LTP) as well as impaired long-term memory (LTM). In addition, we have found that by promoting mTORC2 activity, with a new agent A-443654, it facilitates L-LTP and enhances long-term memory formation in WT mice. Interestingly, mTORC2 activity is altered in both ASD patients and ASD mouse models harboring mutation in Tsc and Pten5,6. Hence, in this proposal we will test the hypothesis that the neurological dysfunction in several ASD mouse models is caused by dysregulation of mTORC2 rather than mTORC1 activity.


4. Key Research Accomplishment

- We developed a way to specifically block mTORC2 activity in Pten-deficient mice.
- Genetic deletion of mTORC2 prolongs the survival of Pten-deficient mice.
- Genetic deletion of mTORC2 dramatically attenuates seizures in Pten-deficient mice.
- Genetic deletion of mTORC2 improves cognitive and social phenotypes in Pten-deficient mice.

5. Conclusion

It has been proposed that the increased mTORC1 in Pten-deficient or Tsc-deficient mice causes the cellular and behavioral phenotypes associated with ASD. Our new data challenge this view and posit that the neurological dysfunction in ASD, at least in the Pten-ASD mouse model, is caused by dysregulation of mTORC2. Hence, these preliminary data are very important since they identified a new signaling pathway involved in ASD and seizure disorders that could be targeted and lead to the development of new treatments for ASD and seizure disorders.


E/I Imbalance in Schizophrenia and Autism




This paper looks really useful and does refer to mTOR, but is not open access

Autism Spectrum Disorders (ASD) and Schizophrenia (SCZ) are cognitive disorders with complex genetic architectures but overlapping behavioral phenotypes, which suggests common pathway perturbations. Multiple lines of evidence implicate imbalances in excitatory and inhibitory activity (E/I imbalance) as a shared pathophysiological mechanism. Thus, understanding the molecular underpinnings of E/I imbalance may provide essential insight into the etiology of these disorders and may uncover novel targets for future drug discovery. Here, we review key genetic, physiological, neuropathological, functional, and pathway studies that suggest alterations to excitatory/inhibitory circuits are keys to ASD and SCZ pathogenesis.


NMDA activation, Sociability and mTOR



Highlights
Several syndromic forms of ASD are associated with disinhibited activity of mTORC1.
Rapamycin, an inhibitor of mTORC1, improved sociability in mouse models of TSC.
NMDA receptor-mediated neurotransmission regulates sociability in mice.
NMDA receptor activation decreases mTOR signaling activity.
D-Cycloserine improved sociability in the Balb/c and BTBR mouse models of ASD.
  
Abstract

Tuberous Sclerosis Complex is one example of a syndromic form of autism spectrum disorder associated with disinhibited activity of mTORC1 in neurons (e.g., cerebellar Purkinje cells). mTORC1 is a complex protein possessing serine/threonine kinase activity and a key downstream molecule in a signaling cascade beginning at the cell surface with the transduction of neurotransmitters (e.g., glutamate and acetylcholine) and nerve growth factors (e.g., Brain-Derived Neurotrophic Factor). Interestingly, the severity of the intellectual disability in Tuberous Sclerosis Complex may relate more to this metabolic disturbance (i.e., overactivity of mTOR signaling) than the density of cortical tubers. Several recent reports showed that Rapamycin, an inhibitor of mTORC1, improved sociability and other symptoms in mouse models of Tuberous Sclerosis Complex and autism spectrum disorder, consistent with mTORC1 overactivity playing an important pathogenic role. NMDA receptor activation may also dampen mTORC1 activity by at least two possible mechanisms: regulating intraneuronal accumulation of arginine and the phosphorylation status of a specific extracellular signal regulating kinase (i.e., ERK1/2), both of which are “drivers” of mTORC1 activity. Conceivably, the prosocial effects of targeting the NMDA receptor with agonists in mouse models of autism spectrum disorders result from their ability to dampen mTORC1 activity in neurons. Strategies for dampening mTORC1 overactivity by NMDA receptor activation may be preferred to its direct inhibition in chronic neurodevelopmental disorders, such as autism spectrum disorders.


Dendritic Spine Dysgenesis in Autism and mTOR




The activity-dependent structural and functional plasticity of dendritic spines has led to the long-standing belief that these neuronal compartments are the subcellular sites of learning and memory. Of relevance to human health, central neurons in several neuropsychiatric illnesses, including autism related disorders, have atypical numbers and morphologies of dendritic spines. These so-called dendritic spine dysgeneses found in individuals with autism related disorders are consistently replicated in experimental mouse models. Dendritic spine dysgenesis reflects the underlying synaptopathology that drives clinically relevant behavioral deficits in experimental mouse models, providing a platform for testing new therapeutic approaches. By examining molecular signaling pathways, synaptic deficits, and spine dysgenesis in experimental mouse models of autism related disorders we find strong evidence for mTOR to be a critical point of convergence and promising therapeutic target.






3. Spine dysgenesis in autism related disorders Spine dysgenesis has been described in autopsy brains of several ARDs, their genetic causes ranging from hundreds of affected genes to one, with their pervasiveness relating to both severity and number of clinical symptoms. By examining common clinical phenotypes correlated to spine and synaptic abnormalities between the disorders, we can work to recognize causalities in dysgenesis and identify potential targets for therapeutic intervention.

4. mTOR: a convergence point of spine dysgenesis and synaptopathologies in ASD Dysgenesis of dendritic spines occurs in the majority of individuals afflicted with ARDs, as well as in most experimental mouse models of these syndromes. It would, therefore, follow that there must be a converging deregulated molecular pathway downstream of the affected genes and upstream of dendritic spine formation and maturation. Identifying this pathway will not only define a causal common denominator in autism-spectrum disorders, but also open new therapeutic opportunities for these devastating conditions. The Ras/ERK and PI3K/mTOR pathways, which regulate protein translation in dendrites near excitatory synapses, have received the most attention as such candidate convergence points


5. Conclusion Cajal once postulated, “the future will prove the great physiological role played by the dendritic spines” [229]. And indeed, it is now widely accepted that dendritic spines are the site of neuronal plasticity of excitatory synapses and the focal point for synaptopathophysiologies of ARDs. Individuals and mouse models of ARDs all display spine dysgenesis, with mTOR-regulated protein translation being a critical point of convergence. Deviations from optimal levels of protein synthesis correlate with the magnitude of dendritic spine pruning and LTD in ARDs. Alleviation of heightened mTOR activity rescues both synaptic and behavioral phenotypes in FXS and TS animals. Correcting mTOR signaling levels also reversed ARD phenotypes in adult fully symptomatic mice, challenging the traditional view that genetic defects caused irreversible developmental defects [230]. More excitingly, these observations demonstrate the potential of pharmacological therapies for neurodevelopmental disorders. The list of ARDs that have been reversed in adult symptomatic mice continues to grow, and also includes RTT [231], DS [232,233], and AS [92]. Together, these findings demonstrate the remarkable plastic nature of the brain and imply that if the causal denominator of ARDs could be found and therapeutically targeted, we may be able to allow the ARD brain to rewire itself and relieve clinical symptoms once believed to be irreversible. The analysis of correlative physiological and behavioral phenotypes and identification of the common mTOR pathway will hopefully provide such potential targets.

  

Clinical Trials


It will be interesting to see the results of the current trials on children with Tuberous Sclerosis Complex, a rare type of autism, that is the most likely to respond to mTOR inhibition.


The purpose of this study is to assess the feasibility and safety of administering rapalogs sirolimus or everolimus, in participants with Tuberous Sclerosis Complex (TSC) and self-injury and to measure cognitive and behavioral changes, including reduction in autistic symptoms, self-injurious and aggressive behaviors, as well as improvements in cognition across multiple domains of cognitive function.



Tuberous sclerosis complex (TSC) is a genetic disease that leads to mental retardation in over 50% of patients, and to learning problems, behavioral problems, autism and epilepsy in up to 90% of patients. The underlying deficit of TSC, loss of inhibition of the mammalian target of Rapamycin (mTOR) protein due to dysfunction of the tuberin/hamartin protein complex, can be rescued by everolimus. Everolimus has been registered as treatment for renal cell carcinoma and giant cell astrocytoma (SEGA). Evidence in human and animal studies suggests that mTOR inhibitors improve learning and development in patients with TSC.