UA-45667900-1

Friday 3 June 2016

Mefenamic acid (Ponstan) for some Autism


Caution:-

Ponstan (Mefenamic Acid) contains a warning:-
Caution should be exercised when treating patients suffering from epilepsy.

At lower doses Ponstan is antiepileptic, but at high doses it can have the opposite effect.  This effect depends on the biological origin of the seizures.
In an earlier post I wrote about a paper by Knut Wittkowski who applied statistics to interpret the existing genetic data on autism. 


“Autism treatments proposed by clinical studies and human genetics are complementary” & the NSAID Ponstan as a Novel AutismTherapy




His analysis suggested the early use of Fenamate drugs could potentially reduce the neurological anomalies that develop in autism as the brain develops.  The natural question arose in the comments was to whether it is too late to use Fenamates in later life.

Knut was particularly looking at a handful of commonly affected genes (ANO 2/4/7 & KCNMA1) where defects should partially be remedied by use of fenamates.

I recently received a comment from a South African reader who finds that his children’s autism improves when he gives them Ponstan and he wondered why.  Ponstan (Mefenamic Acid) is a fenamate drug often used in many countries as a pain killer, particularly in young children.

Ponstan is a cheap NSAID-type drug very widely used in some countries and very rarely used in other countries like the US.  It is available without prescription in some English-speaking countries (try a pharmacy in New Zealand, who sell online) and, as Petra has pointed out, it is widely available in Greece.

I did some more digging and was surprised what other potentially very relevant effects Ponstan has.  Ponstan affects GABAA receptors, where it is a positive allosteric modulator (PAM).  This may be very relevant to many people with autism because we have seen that fine-tuning the response of the sub-units that comprise GABAA receptors you can potentially improve cognition and also modulate anxiety. 

Anxiety seems to be a core issue in Asperger’s, whereas in Classic Autism, or Strict Definition Autism (SDA) the core issue is often actually cognitive function rather than “autism” as such.

In this post I will bring together the science showing why Ponstan should indeed be helpful in some types of autism.

Professor Ritvo from UCLA read Knut’s paper and also the bumetanide research and suggested that babies could be treated with Ponstan and then, later on, with  Bumetanide.

Autism treatments proposed by clinical studies and human genetics are complementary



I do not think the professor or Knut are aware of Ponstan’s effect on GABA.

The benefits from Ponstan may very well be greater if given to babies at risk of autism, but there does seem to be potential benefit for older children and adults, depending on their type of autism.

Professor Ritvo points out that that Ponstan is safely used in 6 month old babies, so trialing it in children and adults with autism should not be troubling.

Being an NSAID, long term use at high doses may well cause GI side effects.  An open question is the dosage at which Ponstan modulates the calcium activated ion channels that are implicated in some autism and also what dosage affects GABAA receptors.  It might well be lower than that required for Ponstan’s known ant-inflammatory effects.


Ponstan vs Ibuprofen

Ibuprofen is quite widely used in autism.  Ibuprofen is an NSAID but also a PPAR gamma agonist.  Ponstan is an NSAID but has no effect on PPAR gamma.

Research shows that some types of autism respond to PPAR gamma agonists.

So it is worth trying both Ponstan and Ibuprofen, but for somewhat different reasons.

They are both interesting to deal with autism flare-ups, which seem common.

Other drugs that people use short term, but are used long term in asthma therapy,  are Singulair (Montelukast) and an interesting Japanese drug called Ibudilast.  Singulair is a Western drug for maintenance therapy in asthma.  Ibudilast is widely used in Japan as maintenance therapy in Asthma, but works in a different way.  Ibudilast is being used in clinical trials in the US to treat Multiple Sclerosis.  Singulair is cheap and widely available, Ibudilast is more expensive and available mainly in Japan.


Pre-vaccination Immunomodulation

In spite of there being no publicly acknowledged link between vaccinations and autism secondary to mitochondrial disease (AMD), I read that short term immunomodulation is used prior to vaccination at Johns Hopkins, for some babies.

Singulair is used, as is apparently ibuprofen.  Ponstan and Ibudilast would also likely be protective.   Ponstan might well be the best choice; it lowers fevers better than ibuprofen.

For those open minded people, here is what a former head of the US National Institutes of Health, Bernadine Healy, had to say about the safe vaccination.  Not surprisingly she was another Johns Hopkins trained doctor, as is Hannah Poling’s Neurologist father.

The Vaccines-Autism War: Détente Needed

“Finally, are certain groups of people especially susceptible to side effects from vaccines, and can we identify them? Youngsters like Hannah Poling, for example, who has an underlying mitochondrial disorder and developed a sudden and dramatic case of regressive autism after receiving nine immunizations, later determined to be the precipitating factor. Other children may have a genetic predisposition to autism, a pre-existing neurological condition worsened by vaccines, or an immune system that is sent into overdrive by too many vaccines, and thus they might deserve special care. This approach challenges the notion that every child must be vaccinated for every pathogen on the government's schedule with almost no exception, a policy that means some will be sacrificed so the vast majority benefit.”


So if I was an American running the FDA/CDC I would suggest giving parents the option of paying a couple of dollars for 10 days of Ponstan prior to these megadose vaccinations and a few days afterwards.  No harm or good done in 99.9% of cases, but maybe some good done for the remainder.

The fact the fact that nobody paid any attention to the late Dr Healy on this subject tells you a lot.



Fenamates (ANO 2/4/7 & KCNMA1)

Here Knut is trying to target the ion channels expressed by the genes ANO 2/4/7 & KCNMA1. 

·        ANO 2/4/7 are calcium activated chloride channels. (CACCs)


·        KCNMA1 is a calcium activated potassium channel.  KCNMA1encodes the ion channel KCa1.1, otherwise known as BK (big potassium).  This was the subject of post that I never got round to publishing.
  
Fenamates are an important group of clinically used non-steroidal anti-inflammatory drugs (NSAIDs), but they have other effects beyond being anti-inflammatory.  They act as CaCC inhibitors and also stimulate BKCa channel activity.


But fenamates also have a potent effect on what seems to be the most dysfunctional receptor in classic autism, the GABAA receptor.




The fenamate NSAID, mefenamic acid (MFA) prevents convulsions and protects rats from seizure-induced forebrain damage evoked by pilocarpine (Ikonomidou-Turski et al., 1988) and is anti-epileptogenic against pentylenetetrazol (PTZ)-induced seizure activity, but at high doses induces seizures (Wallenstein, 1991). In humans, MFA overdose can lead to convulsions and coma (Balali-Mood et al, 1981; Young et al., 1979; Smolinske et al., 1990). More recent data by Chen and colleagues (1998) have shown that the fenamates, flufenamic, meclofenamic and mefenamic acid, protect chick embryo retinal neurons against ischaemic and excitotoxic (kainate and NMDA) induced neuronal cell death in vitro (Chen et al., 1998a; 1998b). MFA has also been reported to reduce neuronal damage induced by intraventricular amyloid beta peptide (Aβ1-42) and improve learning in rats treated with Aβ1-42 (Joo et al., 2006). The mechanisms underlying these anti-epileptic and neuroprotective effects are not well understood but together suggest that fenamates may influence neuronal excitability through modulation of ligand and/or voltage-gated ion channels. In the present study, therefore, we have investigated this hypothesis by determining the actions of five representative fenamate NSAIDs at the major excitatory and inhibitory ligand-gated ion channels in cultured hippocampal neurons


This study demonstrates for the first time that mefenamic acid and 4 other representatives of the fenamate NSAIDs are highly effective and potent modulators of native hippocampal neuron GABAA receptors. MFA was the most potent and at concentrations equal to or greater than 10 μM was also able to directly activate the GABAA gated chloride channel. A previous study from this laboratory reported that mefenamic acid potentiated recombinant GABAA receptors expressed in HEK-293 cells and in Xenopus laevis oocytes (Halliwell et al., 1999). Together these studies lead to the conclusion that fenamate NSAIDs should now also be considered a robust class of GABAA receptor modulators.


Also demonstrated for the first time here is the direct activation of neuronal GABAA receptors by mefenamic acid. Other allosteric potentiators, including the neuroactive steroids and the depressant barbiturates share this property, with MFA at least equipotent to neurosteroids and significantly more potent than the barbiturates. The mechanism(s) of the direct gating of GABAA receptor chloride channels by MFA requires further investigation using ultra-fast perfusion techniques but may be distinct from that reported for neurosteroids (see, Hosie et al., 2006). Mefenamic acid induced a leftward shift in the GABA dose-response curve consistent with an increase in receptor affinity for the agonist. This is an action observed with other positive allosteric GABAA receptor modulators, including the benzodiazepine agonist, diazepam, the neuroactive steroid, allopregnanolone, and the intravenous anesthetics, pentobarbitone and propofol (e.g. Johnston, 2005). To our knowledge, a unique property of MFA was that it was significantly (F = 10.35; p≤ 0.001) more effective potentiating GABA currents at hyperpolarized holding potentials (especially greater than −60mV). Further experiments are required however to determine the underlying mechanism(s).

The highly effective modulation of GABAA receptors in cultured hippocampal neurons suggests the fenamates may have central actions. Consistent with this hypothesis, mefenamic acid concentrations are 40–80μM in plasma with therapeutic doses (Cryer & Feldman, 1998); fenamates can also cross the blood brain barrier (Houin et al., 1983; Bannwarth et al., 1989) Coyne et al. Page 5 Neurochem Int. Author manuscript; available in PMC 2008 November 1. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript and in overdose in humans are associated with coma and convulsions (Smolinske et al., 1990). In animal studies, mefenamic acid is anticonvulsant and neuroprotective against seizureinduced forebrain damage in rodents (Ikonomidou-Turski et al., 1988). The present study would suggest that the anticonvulsant effects of fenamates may be related, in part, to their efficacy to potentiate native GABAA receptors in the brain, although a recent study has suggested that activation of M-type K+ channels may contribute to this action (Peretz et al., 2005) Finally, Joo and co-workers (2006) have recently reported that mefenamic acid provided neuroprotection against β-amyloid (Aβ1-42) induced neurodegeneration and attenuated cognitive impairments in this animal model of Alzheimer’s disease. The authors proposed that neuroprotection may have resulted from inhibition of cytochrome c release from mitochondria and reduced caspase-3 activation by mefenamic acid. Clearly it would also be of interest to evaluate the role of GABA receptor modulation in this in vivo model of Alzheimer’s disease. Moreover, considerable evidence has emerged in the last few years indicating that GABA receptor subtypes are involved in distinct neuronal functions and subtype modulators may provide novel pharmacological therapies (Rudolf & Mohler, 2006). Our present data showing that fenamates are highly effective modulators of native GABAA receptors and that mefenamic acid is highly subtype-selective (Halliwell et al., 1999) suggests that further studies of its cognitive and behavioral effects would be of value.

  

Note in the above paper that NSAIDs other than mefenamic acid also modulate GABAA receptors.

Just a couple of months ago a rather complicated paper was published, again showing that NSAIDs modulate GABAA receptors and showing that this is achieved via the same calcium activated chloride channels (CaCC) referred to by Knut.

NSAIDs modulate GABA-activated currents via Ca2+-activated Cl channels in rat dorsal root ganglion neurons






"Schematic displaying the effects of CaCCs on GABA-activated inward currents and depolarization. GABA activates the GABAA receptor to open the Cl  channel and the Cl efflux induces the depolarization response (inward current) of the membrane of dorsal root ganglion (DRG) neurons. Then, voltage dependent L-type Ca2+ channels are activated by the depolarization, and give rise to an increase in intracellular Ca2+. CaCCs are activated by an increase in intracellular Ca2+ concentration which, in turn, increases the driving force for Cl efflux. Finally, the synergistic action of the chloride ion efflux through GABAA receptors and NFA-sensitive CaCCs causes GABA-activated currents or depolarization response in rat DRG neurons."


Note in the complex explanation above the L-type calcium channels, which are already being targeted by Verapamil, in the PolyPill.



Mefenamic Acid and Potassium Channels

We know that Mefenamic acid also affects Kv7.1 (KvLQT1).

A closely related substance called meclofenamic acid is known to act as novel KCNQ2/Q3 channel openers and is seen as having potential for the treatment of neuronal hyper-excitability including epilepsy, migraine, or neuropathic pain.



The voltage-dependent M-type potassium current (M-current) plays a major role in controlling brain excitability by stabilizing the membrane potential and acting as a brake for neuronal firing. The KCNQ2/Q3 heteromeric channel complex was identified as the molecular correlate of the M-current. Furthermore, the KCNQ2 and KCNQ3 channel  subunits are mutated in families with benign familial neonatal convulsions, a neonatal form of epilepsy. Enhancement of KCNQ2/Q3 potassium currents may provide an important target for antiepileptic drug development. Here, we show that meclofenamic acid (meclofenamate) and diclofenac, two related molecules previously used as anti-inflammatory drugs, act as novel KCNQ2/Q3 channel openers. Extracellular application of meclofenamate (EC50  25 M) and diclofenac (EC50  2.6 M) resulted in the activation of KCNQ2/Q3 K currents, heterologously expressed in Chinese hamster ovary cells. Both openers activated KCNQ2/Q3 channels by causing a hyperpolarizing shift of the voltage activation curve (23 and 15 mV, respectively) and by markedly slowing the deactivation kinetics. The effects of the drugs were stronger on KCNQ2 than on KCNQ3 channel  subunits. In contrast, they did not enhance KCNQ1 K currents. Both openers increased KCNQ2/Q3 current amplitude at physiologically relevant potentials and led to hyperpolarization of the resting membrane potential. In cultured cortical neurons, meclofenamate and diclofenac enhanced the M-current and reduced evoked and spontaneous action potentials, whereas in vivo diclofenac exhibited an anticonvulsant activity (ED50  43 mg/kg). These compounds potentially constitute novel drug templates for the treatment of neuronal hyperexcitability including epilepsy, migraine, or neuropathic pain. Volt




BK channel

KCNMA1encodes the ion channel KCa1.1, otherwise known as BK (big potassium). BK channels are implicated not only by Knut’s statistics, but numerous studies ranging from schizophrenia to Fragile X. 

Usually it is a case of too little BK channel activity.

The BK channel is implicated in some epilepsy.

  

Pharmacology

BK channels are pharmacological targets for the treatment of several medical disorders including stroke and overactive bladder. Although pharmaceutical companies have attempted to develop synthetic molecules targeting BK channels, their efforts have proved largely ineffective. For instance, BMS-204352, a molecule developed by Bristol-Myers Squibb, failed to improve clinical outcome in stroke patients compared to placebo. However, BKCa channels are reduced in patients suffering from the Fragile X syndrome and the agonist, BMS-204352, corrects some of the deficits observed in Fmr1 knockout mice, a model of Fragile X syndrome.
BK channels have also been found to be activated by exogenous pollutants and endogenous gasotransmitters carbon monoxide and hydrogen sulphide.
BK channels can be readily inhibited by a range of compounds including tetraethylammonium (TEA), paxilline and iberiotoxin.



Achieving a better understanding of BK channel function is important not only for furthering our knowledge of the involvement of these channels in physiological processes, but also for pathophysiological conditions, as has been demonstrated by recent discoveries implicating these channels in neurological disorders. One such disorder is schizophrenia where BK channels are hypothesized to play a role in the etiology of the disease due to the effects of commonly used antipsychotic drugs on enhancing K+ conductance [101]. Furthermore, this same study found that the mRNA expression levels of the BK channel were significantly lower in the prefrontal cortex of the schizophrenic group than in the control group [101]. Similarly, autism and mental retardation have been linked to haploinsufficiency of the Slo1 gene and decreased BK channel expression [102].
Two mutations in BK channel genes have been associated with epilepsy. One mutation has been identified on the accessory β3 subunit, which results in an early truncation of the protein and has been significantly correlated in patients with idiopathic generalized epilepsy [103]. The other mutation is located on the Slo1gene, and was identified through genetic screening of a family with generalized epilepsy and paroxysmal dyskinesia [104]. The biophysical properties of this Slo1 mutation indicates enhanced sensitivity to Ca2+ and an increased average time that the channel remains open [104107]. This increased Ca2+ sensitivity is dependent on the specific type of β subunit associating with the BK channel [106, 107]. In association with the β3 subunit, the mutation does not alter the Ca2+-dependent properties of the channel, but with the β4 subunit the mutation increases the Ca2+ sensitivity [105107]. This is significant considering the relatively high abundance of the β4 subunit compared to the weak distribution of the β3 subunit in the brain [12, 13,15, 106, 107]. It has been proposed that a gain of BK channel function may result in increases in the firing frequency due to rapid repolarization of APs, which allows a quick recovery of Na+ channels from inactivation, thereby facilitating the firing of subsequent APs [104]. Supporting this hypothesis, mice null for the β4 subunit showed enhanced Ca2+ sensitivity of BK channels, resulting in temporal lobe epilepsy, which was likely due to a shortened duration and increased frequency of APs [108]. An interesting relevance to the mechanisms of BK channel activation as discussed above, the Slo1 mutation associated with epilepsy only alters Ca2+ dependent activation originated from the Ca2+ binding site in RCK1, but not from the Ca2+bowl, by altering the coupling mechanism between Ca2+ binding and gate opening [100]. Since Ca2+dependent activation originated from the Ca2+ binding site in RCK1 is enhanced by membrane depolarization, at the peak of an action potential the binding of Ca2+ to the site in RCK1 contributes much more than binding to the Ca2+ bowl to activating the channel [84, 109].
Although these associations between specific mutations in BK channel subunits and various neurological disorders have been demonstrated by numerous studies, it is also important to point out certain caveats with these studies, such as genetic linkage between BK channels and different diseases do not necessary show causation as these studies were performed based on correlation between changes in the protein/genetic marker and overall phenotype. Furthermore, studies performed using a mouse model also can fail to indicate what may happen in higher-order species, and this is especially true for BK channels, where certain β subunits are only primate specific [110].


  

Possible role of potassium channel, big K in etiology of schizophrenia.

Schizophrenia (SZ), a common severe mental disorder, affecting about 1% of the world population. However, the etiology of SZ is still largely unknown. It is believed that molecules that are in an association with the etiology and pathology of SZ are neurotransmitters including dopamine, 5-HT and gamma-aminobutyric acid (GABA). But several lines of evidences indicate that potassium large conductance calcium-activated channel, known as BK channel, is likely to be included. BK channel belongs to a group of ion channels that plays an important role in regulating neuronal excitability and transmitter releasing. Its involvement in SZ emerges as a great interest. For example, commonly used neuroleptics, in clinical therapeutic concentrations, alter calcium-activated potassium conductance in central neurons. Diazoxide, a potassium channel opener/activator, showed a significant superiority over haloperidol alone in the treatment of positive and general psychopathology symptoms in SZ. Additionally, estrogen, which regulates the activity of BK channel, modulates dopaminergic D2 receptor and has an antipsychotic-like effect. Therefore, we hypothesize that BK channel may play a role in SZ and those agents, which can target either BK channel functions or its expression may contribute to the therapeutic actions of SZ treatment.




Conclusion

It appears that Ponstan and related substances have some interesting effects that are only now emerging in the research.

People with autism, and indeed schizophrenia, may potentially benefit from Ponstan and for a variety of different reasons.

I think it will take many decades for any conclusive research to be published on this subject, because this is an off-patent generic drug.

As with most NSAIDS, it is simple to trial Ponstan.

Thanks to Knut for the idea, Professor Ritvo for his endorsement of the idea and our reader from South Africa for sharing his positive experience with Ponstan. 







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.







Monday 23 May 2016

More Melatonin!




  Older people, those with autism, those with reflux, IBS/IBD and other GI problems generally have low levels of melatonin.  Poor sleep is but one consequence.



I have previously written about the potential for melatonin in autism and I do not just mean to improve sleeping disorders.  Melatonin does a great deal more than that.

Melatonin for Kids with Autism, and indeed their Parents


MitoE, MitoQ and Melatonin as possible therapies for Mitochondrial Dysfunction in Autism. Or Dimebon (Latrepirdine) from Russia?




Most substances I write about in this blog are either prescription drugs or quite expensive supplements.

Other than in a small number of countries like the United Kingdom, melatonin is widely available as a cheap supplement, but that does not mean it is not a drug.

In humans melatonin is produced in two different places and it appears in two orders of magnitude.  Traditionally melatonin is considered to be a hormone produced by the pineal gland in the brain, but far more melatonin is actually produced in your intestines, where it has completely different functions.

Many people have low levels of melatonin, for example people with autism/schizophrenia/bipolar, older people and people with intestinal problems ranging from reflux/GERD/GORD to ulcerative colitis.

We know that melatonin is a potent antioxidant, but there are numerous other antioxidants.  Damaging oxidants vary both by type, but also by their location and so if you are clever you would match your antioxidant(s) very specifically to the oxidant(s).  

So if you have elevated risk of prostate cancer, take lycopene, it accumulates in fatty tissue and the prostate is surrounded by a fatty deposit called periprostatic adipose tissue (PPAT).  It is not agreed whether lycopene can cross the blood brain barrier in humans; it does for sure in rats.  

It seems that in people with type 2 diabetes there is oxidative stress in the mitochondria of the beta cells in their pancreas.  Beta cells make insulin and in type 2 diabetes there is often a gradual loss in beta cells resulting in type 1 diabetes.  Numerous cancer studies have shown the potential of different antioxidants in different cancers, NAC in breast cancer, Sulforaphane is esophageal cancer etc.  It seems to be agreed that antioxidants are most helpful in disease prevention, rather than cure.  
  
We know that melatonin is potent at combatting oxidants in the mitochondria, so logically people with mitochondrial dysfunction might well benefit from melatonin.  It is vastly cheaper than the antioxidant drugs that target the mitochondria (MitoE, MitoQ etc).

An interesting recent study has linked low levels of melatonin in the parents of those with autism.


  
Background: Low melatonin levels are a frequent finding in autism spectrum disorder (ASD) patients. Melatonin is also important for normal neurodevelopment and embryonic growth. As a free radical scavenger and antioxidant melatonin is highly effective in protecting DNA from oxidative damage. Melatonin deficiency, possibly due to low CYP1A2 activity, could be a major factor, and well a common heritable variation. ASD is already present at birth. As the fetus does not produce melatonin, low maternal melatonin levels should be involved. Methods: We measured 6-sulfatoxymelatonin in urine of mothers of a child with ASD that attended our sleep clinic for people with an intellectual disability (ID), and asked for parental coffee consumption habits, as these are known to be related to CYP1A2 activity. Results: 6-Sulfatoxymelatonin levels were significantly lower in mothers than in controls (p = 0.005), as well as evening coffee consumption (p = 0.034). In mothers with a second child with ASD and/or ID, 6-sulfatoxymelatonin levels were lower compared to mothers with one child with ASD (p = 0.084), 

Conclusions: Low parental melatonin levels, likely caused by low CYP1A2 activity, seem to be a major contributor to ASD and possibly ID etiology.


I think you would also find, more generally, high levels of oxidative stress in parents of those with autism, and more importantly oxidative stress during pregnancy would have negative effects.  I think autism produces stress and stress helps produce autism.

  

Potency of pre–post treatment of coenzyme Q10 and melatoninsupplement in ameliorating the impaired fatty acid profile in rodent model ofautism


  

  
"It is now almost 60 years since the discovery of melatonin and new physiological functions of the indole continuously appear in the most recent studies worldwide. Besides the pineal gland, the existence and value of other sources of synthesis force us to rethink the established premises about the biological role of this molecule, such as the well-known regulation of circadian and reproductive cycles (Hardeland et al., 2008). In the last few years, other properties of melatonin such as antioxidant power, immunoregulatory capacity, and oncostatic action have enriched our knowledge about the pleiotropic nature of the hormone.

The role of melatonin in mitochondrial homeostasis has gained strength in the scientific community. Experimental evidence emphasizes its importance as a stabilizer of organular bioenergetics, which could be related to the             prevention of development of aging and several diseases.

  
Role of melatonin on mitochondrial dysfunction and diseases

The idea that mitochondrial dysfunction is implicated in the etiology of various diseases has been strengthened after several years of research. Initially, studies of mitochondrial diseases have focused on mitochondrial respiratory-chain diseases associated with mutations of mtDNA. However, more recent evidence shows that oxidative damage is responsible for the impairment of mitochondrial function, leading to a self-induced vicious cycle that finally culminates in necrosis and apoptosis of cells and organ failure. We are now starting to understand the mechanisms of a large list of mitochondrial-related diseases (cancer, diabetes, obesity, cardiovascular and neurodegenerative diseases, and aging); all of them seem to share the common features of disturbances of mitochondrial Ca2+, ATP, or ROS metabolism (Sheu et al., 2006). Therefore, selective prevention of such phenomena should be an effective therapy in a wide range of human diseases (Smith et al., 1999; Sheu et al., 2006). Melatonin, as was described in the previous section, has many of the characteristics of a perfect candidate for the treatment of these kinds of illnesses.

  
Conclusion

Mitochondrial dyshomeostasis and related events have begun to reveal themselves as possible etiologies of several diseases of unknown origin. In the next years, conscientious investigation about this topic should be undertaken by scientists of different research areas to achieve a better understanding of the molecular mechanisms implied, which will ultimately allow the development and clinical application of efficacious treatments."


Recent posts looked at disturbed calcium homeostasis in autism, particularly low bone density.  Melatonin may play a role here as well.



Melatonin osteoporosis prevention study (MOPS): a randomized, double-blind, placebo-controlled study examining the effects of melatonin on bone health and quality of life in perimenopausal women.


Abstract


The purpose of this double-blind study was to assess the effects of nightly melatonin supplementation on bone health and quality of life in perimenopausal women. A total of 18 women (ages 45-54) were randomized to receive melatonin (3mg, p.o., n=13) or placebo (n=5) nightly for 6months. Bone density was measured by calcaneal ultrasound. Bone turnover marker (osteocalcin, OC for bone formation and NTX for bone resorption) levels were measured bimonthly in serum. Participants completed Menopause-Specific Quality of Life-Intervention (MENQOL) and Pittsburgh Sleep Quality Index (PSQI) questionnaires before and after treatment. Subjects also kept daily diaries recording menstrual cycling, well-being, and sleep patterns. The results from this study showed no significant change (6-month-baseline) in bone density, NTX, or OC between groups; however, the ratio of NTX:OC trended downward over time toward a ratio of 1:1 in the melatonin group. Melatonin had no effect on vasomotor, psychosocial, or sexual MENQOL domain scores; however, it did improve physical domain scores compared to placebo (mean change melatonin: -0.6 versus placebo: 0.1, P<0.05). Menstrual cycling was reduced in women taking melatonin (mean cycles melatonin: 4.3 versus placebo: 6.5, P<0.05), and days between cycles were longer (mean days melatonin: 51.2 versus placebo: 24.1, P<0.05). No differences in duration of menses occurred between groups. The overall PSQI score and average number of hours slept were similar between groups. These findings show that melatonin supplementation was well tolerated, improved physical symptoms associated with perimenopause, and may restore imbalances in bone remodeling to prevent bone loss. Further investigation is warranted.

           Melatonin Effects on Hard Tissues: Bone and Tooth


Melatonin, as an endogenous hormone, participates in many physiological and pharmacological processes. The above analyzed data indicate that melatonin may be involved in the development of the hard tissues bone and teeth. Decreased melatonin levels may be related to bone disease and abnormality. Due to its ability of regulating bone metabolism, enhancing bone formation, promoting osseointegration of dental plant and cell and tissue protection, melatonin may used as a novel mode of therapy for augmenting bone mass in bone diseases characterized by low bone mass and increased fragility, bone defect/fracture repair and dental implant surgery. The investigation of melatonin on tooth still insufficient and requires further research.

The following very interesting study, looking at the broader effects of high dose melatonin in autism, has been completed, but the results have yet to be published

Melatonin Dose-effect Relation in Childhood Autism (MELADOSE)

the objective of this clinical trial is to study the relation between the melatonin dose administered and its effect on severity of autistic impairments especially in verbal communication and play.


Experimental: 2 mg melatonin
1 tablet of 2mg melatonin and 4 tablets of its placebo once a day, an hour before falling asleep, for 6 weeks.
Experimental: 4 mg melatonin
2 tablets of 2mg melatonin and 3 tablets of its placebo once a day, an hour before falling asleep, for 6 weeks.
Experimental: 10 mg melatonin
5 tablets of 2mg melatonin once a day, an hour before falling asleep, for 6 weeks.




The science part

The following is an extract from an excellent paper about the use of melatonin to treat ulcerative colitis:-







Melatonin was first described as a secretion from the pineal gland with multiple neurohormonal functions, including regulation of the circadian rhythm, reproductive physiology, and body temperature, but has since also been found to inhibit the Cox-2 and NF-_B pathways and several aging processes. The multifactorial role of this hormone, however, has only relatively recently been appreciated (Fig. 1) as it circulates unimpeded across anatomical barriers, the blood– brain barrier included, and exhibits both receptor-dependent and receptor-independent effects.

Furthermore, melatonin exhibits a high degree of conservation across the evolutionary ladder, pointing to a critical function in various forms of life, even in organisms devoid of a pineal gland. In fact, the analysis of extrapineal sources of melatonin have highlighted the GI tract as a major source of this factor, with concentrations of melatonin as much as 100 times that found in blood and 400 times that found in the pineal gland.40 GI melatonin comes from both pineal melatonin and de novo synthesis in the GI tract and may have a direct effect on many GI tissues, serving as an endocrine, paracrine, or autocrine hormone, influencing the regeneration and function of epithelium, modulating the immune milieu in the gut, and reducing the tone of GI muscles by targeting smooth muscle cells.40 Melatonin may also influence the GI tract indirectly, through the central nervous system and the mucosa, by a receptor-independent scavenging of free radicals leading to reduction of inflammation, reduction of secretion
of hydrochloric acid, stimulation of the immune system, COX-2 fostering tissue repair and epithelial regeneration, and increasing microcirculation. Human intestinal motility follows a circadian rhythm with reduced nocturnal activity. Abnormalities in colonic motor function in patients with UC have been well documented.

Melatonin appears to be involved in the regulation of GI motility, exerting both excitatory and inhibitory effects on the smooth musculature of the gut.  The precise mechanism through which melatonin regulates GI motility is not clear, although some studies suggest that this may be related to blockade of nicotonic channels by melatonin and/or the interaction between melatonin and Ca2+ activated K channels.

Melatonin may also function as a physiological antagonist of serotonin. In a recent rodent model, melatonin administration was shown to reverse lipopolysaccharide-induced GI motility disturbances through the inhibition of oxidative stress. The net motor regulation by melatonin is, therefore, likely multifactorial.

In addition, several lines of in vitro studies as well as animal studies, have reported that melatonin regulates the extensive gut immune system and has important general antiinflammatory and immunomodulatory effects. Given its
presence in GI tissue and its suggested importance in GI tract physiology, it is reasonable to hypothesize that melatonin could influence inflammation-related GI disorders, including UC. In various animal experiments, melatonin administration was (among other immunomodulatory effects) shown to increase
IL-10 production and inhibit production of IFN-_, TNF-_, IL-6, and NO, suggesting that melatonin may exert benefits in UC by reducing or controlling inflammation.

Melatonin administration has also inhibited the TNF-_-induced mucosal addressin cell adhesion molecule (MAd-CAM)-1 in vitro, and intercellular adhesion molecule (ICAM)-1 in vivo, limiting the influx of activated _4_7_ and LFA-1_ leukocytes to the mucosal environment. During inflammation, the mucosal microvasculature controls the selection and magnitude of influx of T-cell subsets into the gut through cell adhesion molecules expression and chemokine secretion, which further amplify the communication with other leukocytes and cells. In animal experiments neutralization of MAdCAM-1 and ICAM-1 led to attenuation of mucosal damage in colitis.


If you made your way through the above section, and regularly read this blog you will appreciate the multiple possible beneficial actions for many types of autism.

I was going to have a post about GI issues, but I will put some of the melatonin part in this post.  In summary, very many GI problems are associated with low levels and melatonin and numerous studies have shown that giving oral melatonin is an effective treatment to varying degrees. Melatonin is a useful adjunct (add-on) therapy in these conditions. 

Not only does melatonin appears to promote healing of the esophagus but also the tightening of the LES (The lower esophagealsphincter)

Failure of this sphincter to close is why people get reflux/GERD/GORD.

One possibility is that the night time spike in melatonin signals your brain that it is time to sleep and also signals your LES to shut tightly, so that during the night acid does not rise up your esophagus while you are horizontal.



The potential therapeutic effect of melatonin in gastro-esophageal reflux disease


Regression of gastroesophageal reflux disease symptoms using dietary supplementation with melatonin, vitamins and aminoacids: comparison with omeprazole.   




Oxidative Stress: An Essential Factor in the Pathogenesis of Gastrointestinal Mucosal Diseases





Conclusion

Melatonin may already be the most widely used drug to treat autism, but generally at the lower sleep-inducing doses.

It would seem that those with GI problems, mitochondrial problems or more general oxidative stress may very well benefit from the higher doses of melatonin already used by some.

Older people, people with esophagitis/duodenitis or IBS/IBD, people with type 1 or 2 diabetes and even people with osteoporosis may also want to look into melatonin supplementation.

Given the supplement is ending up in your intestines, where much melatonin should already be being produced, the impact on pineal melatonin production becomes less of an issue.  People giving thyroid hormones T3 and T4 to children who are euthyroid (ie normal thyroid function) should be aware of the consequences (thyroid shutdown).

For various reasons, production of ROS (reactive oxygen species) that are the oxidants varies throughout the day, the morning is the worst time supposedly.  Ideally you would match this with your antioxidant intake.  One combination would be melatonin before bed, a larger dose of NAC at breakfast and then NAC throughout the day.  As highlighted in an earlier post, sustained release NAC is also interesting, but it would help if there was a more potent version. 

Hopefully Dr Tordjman will publish the results of her high dose melatonin in autism study soon.
  
Most people struggle to access the really effective autism drugs, but antioxidants are available in abundance.

Oxidative stress is not a cause of autism, but it is a common side effect.  Treating oxidative stress does indeed seem to help many people with autism, but since the source of those oxidants may vary so should the most effective therapy.  Melatonin may be a useful part of that antioxidant mix, particularly if there are GI, mitochondrial or sleep issues.

Melatonin has a half-life of less than an hour, people who respond well might consider sustained release versions, which are available quite cheaply (5 and 10 mg sustained release forms look interesting).  There are even some clinical trials measuring the resulting plasma levels.