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

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).




Wednesday 4 January 2017

Histidine for Allergy, but as an effective MTOR inhibitor?



Today’s post is likely to be of interest to those dealing with allergy and mast cell activation, but it may have broader implications for those with excess brain mTOR activity.
In the jargon, we are told that:
enhanced mammalian target of rapamycin (mTOR) signaling in the brain has been implicated in the pathogenesis of autism spectrum disorder”.
I have discussed mTOR and mTOR inhibitors previously on this blog.



Amino acids, not just for body builders?


mTOR plays a key role in aging and many human diseases ranging from cancer, diabetes and obesity to autism and Alzheimer’s.

The greatest interest in mTOR seems to be in cancer care.  Many cancer genes and pathways are also involved in autism, so we can benefit from the cancer research.  Another autism gene that is also a cancer gene is PTEN.  PTEN is a tumor suppressor and in the most common male cancer, prostate cancer (PCa), what happens is that PTEN gets turned off and so the cancer continues to grow.  If you upregulate PTEN you slow the cancer growth and if you upregulated this gene in those people at risk of Pca perhaps they would never develop this cancer in the first place?  PTEN is upregulated by statin-type drugs and people already on this type of drug have better PCa prognoses.   The beneficial of effect of statins on PCa is known, but the mechanism being PTEN upregulation does not seem to have been noticed. No surprise there.

Inhibiting mTOR using cancer drugs is very expensive.

Other substances affecting mTOR include amino acids, growth factors, insulin, and oxidative stress.

The amino acid Leucine is an mTOR activator, we don’t need that.  We actually want the opposite effect and, at least in mice, we can get it from some of the other amino acids. 


          Highlights 

·        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.



Neuroprotective and anti-inflammatory diet reduced behavioral deficits only in allergic mice.

              Abstract

Enhanced mammalian target of rapamycin (mTOR) signaling in the brain has been implicated in the pathogenesis of autism spectrum disorder (ASD). Inhibition of the mTOR pathway improves behavior and neuropathology in mouse models of ASD containing mTOR-associated single gene mutations. The current study demonstrated that the amino acids histidine, lysine, threonine inhibited mTOR signaling and IgE-mediated mast cell activation, while the amino acids leucine, isoleucine, valine had no effect on mTOR signaling in BMMCs. Based on these results, we designed an mTOR-targeting amino acid diet (Active 1 diet) and assessed the effects of dietary interventions with the amino acid diet or a multi-nutrient supplementation diet (Active 2 diet) on autistic-like behavior and mTOR signaling in food allergic mice and in inbred BTBR T + Itpr3tf/J mice. Cow’s milk allergic (CMA) or BTBR male mice were fed a Control, Active 1, or Active 2 diet for 7 consecutive weeks. CMA mice showed reduced social interaction and increased self-grooming behavior. Both diets reversed behavioral impairments and inhibited the mTOR activity in the prefrontal cortex and amygdala of CMA mice. In BTBR mice, only Active 1 diet reduced repetitive self-grooming behavior and attenuated the mTOR activity in the prefrontal and somatosensory cortices. The current results suggest that activated mTOR signaling pathway in the brain may be a convergent pathway in the pathogenesis of ASD bridging genetic background and environmental triggers (food allergy) and that mTOR over-activation could serve as a potential therapeutic target for the treatment of ASD.

  

So in mice a combination of the three amino acids Histidine, Lysine and Threonine reduced brain mTOR activity and improved autism.

I did look at all three of these amino acids and their other effects and I choose Histidine. 
Histidine can be produced in adult humans in very small amounts, but in young children they need to obtain some from other sources, usually dietary.

Histidine is the precursor of histamine.  Histamine has both good and bad effects.

Histidine decarboxylase (HDC) is the enzyme that catalyzes the reaction that produces histamine from histidine with the help of vitamin B6 as follows:



You can treat allergy by inhibiting HDC.

Tritoqualine, is an inhibitor of the enzyme histidine decarboxylase and therefore an atypical antihistamine,

You might think that having extra histidine would result in extra histamine, but this appears not to be the case.  There is a paradoxical reaction where increasing histadine actually seems to reduce the release of histamine from the mast cells that store it.  This may indeed be a case of feedback loops working in our favour.

So it seems that histidine may give two different benefits, it reduces IgE-mediated mast cell activation and it reduces mTOR signalling in the brain.

If the effect on mTOR is sufficient we would then benefit from an increase in autophagy, the cellular garbage disposal service that does not work well in autism.  We might eventually see a benefit from increased synaptic pruning which might be seen in improved cognition.  



Recap on mTOR and Synaptic Pruning

This has been covered in earlier posts.

In autism loss of mTOR-dependent macro-autophagy causes synaptic pruning deficits; this results in too many dendritic spines.









A dendritic spine (or spine) is a small membranous protrusion from a neuron's dendrite that typically receives input from a single axon at the synapse. Dendritic spines serve as a storage site for synaptic strength and help transmit electrical signals to the neuron's cell body. The dendrites of a single neuron can contain hundreds to thousands of spines. In addition to spines providing an anatomical substrate for memory storage and synaptic transmission, they may also serve to increase the number of possible contacts between neurons.

A feature of autism is usually too many, but can be too few, dendritic spines.  In an earlier post we saw how the shape of individual spines affects their function.  The shape is constantly changing and can be influenced by external therapy. Wnt signaling affects dendritic spine morphology and so using this pathway you could fine-tune dendritic spine shape.  We did look at PAK1 inhibitors in connection with this.

Synaptic pruning is an ongoing process well into adolescence.

So it may be possible to improve synapse density and structure well after the onset of autism.

It should be noted that using Rapalogs, the usual mTOR inhibiting drugs, would have a negative effect in the minority of autism that feature hypo-active growth signalling.  That would be people born with small heads and small bodies.  So a child affected by the zika virus, might very likely exhibit autism and ID, but likely has too few dendritic spines and would then need more mTOR, rather than less.

Rapalog drugs like Everolimus are very expensive, but as in this recent paper do show effect in some autism. 



The mTOR pathway is a central regulator of mammalian metabolism and physiology, with important roles in the function of tissues including liver, muscle, white and brown adipose tissue, and the brain, and is dysregulated in human diseases, such as diabetes, obesity, depression, and certain cancers.

mTOR Complex 1 (mTORC1) is composed of MTOR, regulatory-associated protein of MTOR (Raptor), mammalian lethal with SEC13 protein 8 (MLST8) and the non-core components PRAS40 and DEPTOR. This complex functions as a nutrient/energy/redox sensor and controls protein synthesis. The activity of mTORC1 is regulated by rapamycin, insulin, growth factors, phosphatidic acid, certain amino acids and their derivatives (e.g., L-leucine and β-hydroxy β-methylbutyric acid), mechanical stimuli, and oxidative stress

Rapamycin inhibits mTORC1, and this appears to provide most of the beneficial effects of the drug (including life-span extension in animal studies). Rapamycin has a more complex effect on mTORC2.



How do amino acids affect mTOR?

This is not fully understood by anyone, but here is a relevant paper, for those interested.




Mammalian target of rapamycin (mTOR) controls cell growth and metabolism in response to nutrients, energy, and growth factors. Recent findings have placed the lysosome at the core of mTOR complex 1 (mTORC1) regulation by amino acids. Two parallel pathways, Rag GTPase-Ragulator and Vps34-phospholipase D1 (PLD1), regulate mTOR activation on the lysosome. This review describes the recent advances in understanding amino acid-induced mTOR signaling with a particular focus on the role of mTOR in insulin resistance.

We then discuss how mTORC1 activation by amino acids controls insulin signaling, a key aspect of body metabolism, and how deregulation of mTOR signaling can promote metabolic disease. 

Concluding remarks


Recent findings of new mediators and their regulatory mechanisms have broadened our understanding of amino acid-induced mTOR signaling. In addition to the role of the TSC1-TSC2-Rheb hub in transducing upstream signals from growth factors, stressors and energy to mTOR, the lysosomal regulation of mTOR functions as a platform to connect nutrient signals to the Rheb axis. Furthermore, two parallel pathways of amino acid signaling explain the diverse regulation of mTOR signaling. It is yet to be determined which regulators sense amino acids directly and whether the two pathways require separate amino acid sensing mechanisms. The identification of a direct amino acid sensor will shed light on these uncertainties.

A more integrated understanding of mTOR regulation in amino acid signaling will open the door for new therapeutic approaches for metabolic diseases, especially type 2 diabetes. Already, metformin, an antidiabetic drug, inhibits mTOR in an AMP-activated kinase (AMPK)-independent and Rag-dependent manner,64 providing further support for the idea that the regulation of amino acid sensing could be a therapeutic target for diabetes.



How typical is the level of amino acids in autism?



As regards essential amino acid levels, autistic children had significant lower plasma levels of leucine, isoleucine, phenylalanine, methionine and cystine than controls (P < 0.05),while there was no statistical difference in the level of tryptophan, valine, threonine, arginine, lysine and histidine (P > 0.05). In non-essential amino acid levels, phosphoserine was significantly raised in autistic children than in controls (P < 0.05). Autistic children had lower level of hydroxyproline, serine and tyrosine than controls (P < 0.05). On the other hand there was no significant difference in levels of taurin, asparagine, alanine, citrulline, GABA, glycine, glutamic acid, and ornithine (P > 0.05).

There was no significant difference between cases and controls as regards the levels of urea, ammonia, total proteins, albumin and globulins (alpha 1, alpha 2, beta and gamma) (P > 0.05).



  

Conclusion 

For the more common hyperactive pro growth signaling pathway types of autism, histidine should be a good amino acid, whereas for the hypoactive type, that might feature microcephaly, leucine should be a good choice.

Histidine is already used by some people to treat allergy.

Histidine does have numerous other functions and one relates to zinc, so it is suggested that people who supplement histidine add a little zinc. For this reason German histidine supplements thoughtfully all seem to include zinc.

Histidine also has some direct antioxidant effects and has an effect on Superoxide dismutase (SOD).

It is not clear how much histidine would be needed in humans to achieve the mTOR inhibiting effect found in mice.

The RDA for younger teenagers is histidine  850 mg and leucine 2450 mg.  What the therapeutic dose to affect mTOR in humans remains to be seen.

Histidine is also claimed to help ulcers, which is plausible.

For allergy some people are taking 1,500mg of histidine a day.






Monday 30 May 2016

Sense, Missense or Nonsense - Interpreting Genetic Research in Autism (TCF4, TSC2 , Shank3 and Wnt)




Some clever autism researchers pin their hopes on genetics, while some equally clever ones are not convinced.

One big problem is that genetic testing is still not very rigorous, it is fine if you know what you are looking for, like a specific single gene defect, but if it is a case of find any possible defect in any of the 700+ autism genes it can be hopeless.

Most of the single gene types of autism can be diagnosed based on known physical differences and then that specific gene can be analyzed to confirm the diagnosis.

Today’s post includes some recent examples from the research, and they highlight what is often lacking - some common sense.

There are numerous known single gene conditions that lead to a cascade of dysfunctions that can result in behaviors people associate with autism.  However in most of these single gene conditions, like Fragile X or Pitt-Hopkins, there is a wide spectrum, from mildly affected to severely affected.

There are various different ways in which a gene can be disturbed and so within a single gene condition there can be a variety of sub-dysfunctions.  A perfect example was recently forwarded to me, a study showing how a partial deletion of the Pitt Hopkins gene (TCF4) produced no physical features of the syndrome, but did unfortunately produce intellectual disability.

The study goes on to suggest that “screening for mutations in TCF4 could be considered in the investigation of NSID (non-syndromic intellectual disability)”

Partial deletion of TCF4 in three generation family with non-syndromic intellectual disability, without features of Pitt-Hopkins syndrome



This all matters because one day when therapies for Pitt Hopkins are available, they would very likely be effective on the cognitive impairment of those with undiagnosed partial-Pitt Hopkins.



Another reader sent me links to the studies showing:-


Rapamycin reverses impaired social interaction in mouse models of tuberous sclerosis complex.

Reversal of learning deficits in a Tsc2+/- mouse model of tuberous sclerosis.


But isn’t that Tuberous sclerosis (TSC) extremely rare? like Pitt Hopkins.  Is it really relevant?

Tuberous sclerosis (TSC)  is indeed a rare multisystem genetic disease that causes benign tumors to grow in the brain and on other vital organs such as the kidneys, heart, eyes, lungs, and skin. A combination of symptoms may include seizures, intellectual disability, developmental delay, behavioral problems, skin abnormalities, and lung and kidney disease. TSC is caused by a mutation of either of two genes, TSC1 and TSC2, 

About 60% of people with TSC have autism (biased to TSC2 mutations) and many have epilepsy.

How rare is TSC?  According to research between seven and 12 cases per 100,000, with more than half of these cases undetected.  

Call it 0.01%, rare indeed.

How rare is partial TSC?  What is partial TSC?  That is just my name for what happens when you have just a minor missense mutation, you have a mutation in TSC2 but have none of the characteristic traits of tuberous sclerosis, except autism.
In a recent study of children with autism 20% has a missense mutation of TSC2. 

Not so rare after all.


Mutations in tuberous sclerosis gene may be rife in autism


Mutations in TSC2, a gene typically associated with a syndrome called tuberous sclerosis, are found in many children with autism, suggests a genetic analysis presented yesterday at the 2016 International Meeting for Autism Research in Baltimore.
The findings support the theory that autism results from multiple ‘hits’ to the genome.
Tuberous sclerosis is characterized by benign tumors and skin growths called macules. Autism symptoms show up in about half of all people with tuberous sclerosis, perhaps due to abnormal wiring of neurons in the brain. Tuberous sclerosis is thought to result from mutations in either of two genes: TSC1 or TSC2.
The new analysis finds that mutations in TSC2 can also be silent, as far as symptoms of the syndrome go: Researchers found the missense mutations in 18 of 87 people with autism, none of whom have any of the characteristic traits of tuberous sclerosis.
“They had no macules, no seizure history,” says senior researcher Louisa Kalsner, assistant professor of pediatrics and neurology at the University of Connecticut School of Medicine in Farmington, who presented the results. “We were surprised.”
The researchers stumbled across the finding while searching for genetic variants that could account for signs of autism in children with no known cause of the condition. They performed genetic testing on blood samples from 87 children with autism.

Combined risk:

To see whether silent TSC2 mutations are equally prevalent in the general population, the researchers scanned data from 53,599 people in the Exome Aggregation Consortium database. They found the mutation in 10 percent of the individuals.
The researchers looked more closely at the children with autism, comparing the 18 children who have the mutation with the 69 who do not.
Children with TSC2 mutations were diagnosed about 10 months earlier than those without a mutation, suggesting the TSC2 mutations increase the severity of autism features. But in her small sample, Kalsner says, the groups show no differences in autism severity or cognitive skills. The researchers also found that 6 of the 18 children with TSC2 mutations are girls, compared with 12 of 69 children who don’t have the mutation.
TSC2 variants may combine with other genetic variants to increase the risk of autism. “We don’t think TSC is the sole cause of autism in these kids, but there’s a significant chance that it increases their risk,” Kalsner says.


"hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1) is a consequence of tuberous sclerosis complex (TSC) 1/2 inactivation."

"the combination of rapamycin and resveratrol may be an effective clinical strategy for treatment of diseases with mTORC1 hyperactivation."


So for the 20% of autism with partial TSC, so-called Rapalogs and other mTOR inhibitors could be helpful, but Rapalogs all have side effects.

One interesting option that arose in my earlier post on Type 3 diabetes and intranasal insulin is Metformin. The common drug used for type 2 diabetes.

 








Metformin regulates mTORC1 signaling (but so does insulin).

'Metformin activates AMPK by inhibiting oxidative phosphorylation, which in turn negatively regulates mTORC1 signaling via activation of TSC2 and inhibitory phosphorylation of raptor. In parallel, metformin inhibits mTORC1 signaling by suppressing the activity of the Rag GTPases and upregulating REDD1."

Source:  Rapalogs and mTOR inhibitors as anti-aging therapeutics



Clearly you could also just use intranasal insulin.  It might be less potent but should have less side effects because it acting only within the CNS (Metfornin would be given orally).



The Shank protein and the Wnt protein family

Mutations in a gene called Shank3 occur in about 0.5 percent of people with autism.  
But what about partial Shank3 dysfunction?

Shank proteins also play a role in synapse formation and dendritic spine maturation.

Mutations in this gene are associated with autism spectrum disorder. This gene is often missing in patients with 22q13.3 deletion syndrome

Researchers at MIT have just shown, for the first time, that loss of Shank3 affects a well-known set of proteins that comprise the Wnt signaling pathway.  Without Shank3, Wnt signaling is impaired and the synapses do not fully mature.


“The finding raises the possibility of treating autism with drugs that promote Wnt signaling, if the same connection is found in humans”

I have news for MIT, people already do use drugs that promote Wnt signaling, FRAX486 and Ivermectin for example.  All without any genetic testing, most likely.


Reactivating Shank3, or just promote Wnt signaling

The study below showed that in mice, aspects of autism were reversible by reactivating the Shank3 gene.  You might expect that in humans with a partial Shank3 dysfunction you might jump forward to the Wnt signaling pathway and intervene there.

Mouse study offers promise of reversing autism symptoms


One reader of this blog finds FRAX486 very helpful and to be without harmful side effects.  FRAX 486 was recently acquired by Roche and is sitting over there on a shelf gathering dust.



Where from here?

I think we should continue to look at the single gene syndromes but realize that very many more people may be partially affected by them.

Today’s genetic testing gives many false negatives, unless people know what they are looking for; so many dysfunctions go unnoticed.

This area of science is far from mature and there may be many things undetected in the 97% of the genome that is usually ignored that affect expression of the 3% that is the exome.

So best not to expect all the answers, just yet, from genetic testing; maybe in another 50 years.

Understanding and treating multiple-hit-autism, which is the majority of all autism, will require more detailed consideration of which signaling pathways have been disturbed by these hits.  There are 700 autism genes but there a far fewer signaling pathways, so it is not a gargantuan task.  For now a few people are figuring this out at home.   Good for them.

I hope someone does trials of metformin and intranasal insulin in autism.  Intranasal insulin looks very interesting and I was surprised to see in those earlier posts is apparently without side effects.

The odd thing is that metformin is indeed being trialed in autism, but not for its effect on autism, but its possible effect in countering the obesity caused by the usual psychiatric drugs widely prescribed in the US to people with autism.

My suggestion would be to ban the use of drugs like Risperdal, Abilify, Seroquel, Zyprexa etc.

Vanderbilt enrolling children with autism in medication-related weight gain study



Here are details of the trial.


Metformin will be dispensed in a liquid suspension of 100 mg/mL. For children 6-9 years of age, metformin will be started at 250 mg at their evening meal for 1 week, followed by the addition of a 250 mg dose at breakfast for 1 week. At the Week 2 visit, if metformin is well-tolerated, the dose will be increased to 500 mg twice daily. For children from 10-17 years of age, metformin will be started at 250 mg at their evening meal for 1 week, followed by the addition of a 250 mg dose at breakfast for 1 week. At the Week 2 visit, if metformin is well-tolerated, the dose will be increased to 500 mg twice daily. At the Week 4 visit, if metformin is well-tolerated, the dose will be increased to 850 mg twice daily.