UA-45667900-1
Showing posts with label Pitt Hopkins. Show all posts
Showing posts with label Pitt Hopkins. Show all posts

Friday 24 May 2024

Cilantro (Coriander leaves) for sound sensitivity? cGPMax for some Pitt Hopkins and Rett syndrome. Plus, microdeletion of 2P16.3 NRXN1 and mutations in GPC5

 


Today’s post combines a very simple therapy for sound sensitivity that landed in my inbox from New Zealand with two more genes that I was recently asked about.

Before I get started I would like to thank our reader Daniel who is trying to spread that word that the IGF-1 targeting therapy cGPMax works in some Rett syndrome (half a capsule daily). I did go into the science of IGF-1 related therapies at the recent conference in Abu Dhabi. In that presentation I pointed out that the cGPMax therapy might well be helpful in Pitt Hopkins syndrome. I saw today that Soko, an 8 year old girl with Pitt Hopkins, had already made a trial and her parents are impressed:-

“Equally significant has been the positive shift in Soko's emotional well-being. Her struggles with irritability and mood fluctuations feel like are not as frequent and we feel like there is more often a sense of calm and emotional regulation. This has had a profound ripple effect on our little family and our stress levels.

Perhaps most striking has been the accelerated rate at which Soko acquires new skills. CGP Max has seemingly unlocked hidden potentials within her. This rapid skill acquisition has been very exciting for us. In the last year she has gone from being unable to walk to walking unassisted and even tackling steps no handed!”

I did some checking and some other parents have tried cGPMax for Pitt Hopkins. For Rett syndrome Daniel found that a lower dose was more beneficial than a higher dose. It is always best to start with low doses and gradually increase them.

This does link to today’s post because a  microdeletion of NRXN1 can cause Pitt Hopkins Like Syndrome 2 (PHLS2). In theory all these syndromes are untreatable, but try telling that to Soko’s parents.

 

Back to sound sensitivity

Today’s sound sensitivity is the type that is moderated by Ponstan (mefenamic acid) and indeed Diclofenac. It might well include those whose sound sensitivity responds to a simple potassium supplement.

If you want to look into the details, you can see from previous posts how potassium and potassium ion channels play a fundamental role in both hearing and its sensory processing. They also play a key role in excitability of neurons and so can play a key role in some epilepsy and some intellectual disability.

It turns out that Cilantro/Coriander leaves contains a chemical that activates the ion channels  KCNQ2 (Kv7.2) and KCNQ3 (Kv7.3). This effect is shared by Ponstan and Diclofenac.

In the case of Andy from New Zealand the effect of a 425mg Cilantro supplement lasts very much longer than taking a low dose of Ponstan or Diclofenac.

So, if your child responds well to Ponstan and can then happily take off his/her ear defenders, but you do not want to medicate every day, then a trial of Cilantro could be interesting.

I was curious as to why the effect would last so much longer than Ponstan/Diclofenac.  All of these drugs lower potassium levels within neurons.  Is the beneficial effect coming from lowering potassium levels and so reducing neuronal excitability?  Or, is the effect coming directly from a specific ion channel?

Nobody can tell you the half-life of the active component of cilantro,  (E)-2-dodecenal, in humans.  Andy thinks it must have a long half-life.

 

Cilantro (Coriander leaves)

If you live in North America you will know what cilantro is, for everyone else it means coriander leaves. Coriander seeds are the dried fruit of the coriander plant and, confusingly, in American English coriander means coriander seeds.

The medicinal properties of the leaves and seeds are not the same.

Cilantro leaves contain a compound called (E)-2-dodecenal, which has been shown to activate a specific family of potassium ion channel called KCNQ, otherwise known as Kv7 . These channels are found in neurons, and they play an important role in regulating the electrical activity of the brain.

When (E)-2-dodecenal binds to KCNQ/Kv7 channels, it causes them to open, which allows potassium ions to flow out of the neuron. This outflow of potassium ions helps to stabilize the neuron's membrane potential and makes it less likely to fire an action potential.

The level of potassium inside neurons is much higher than the level outside. Having it too high, or indeed too low, would affect the excitability of the neuron.

I am wondering if the problem with potassium is mirroring the problem we have with chloride; perhaps both are elevated inside neurons. That would be nice and simple.

The discovery that cilantro can activate KCNQ channels helps to explain its potential anticonvulsant properties.  KCNQ channel dysfunction has been linked to certain types of epilepsy, and drugs that activate these channels are being investigated as potential treatments for these conditions.

Research suggests cilantro's active compound, (E)-2-dodecenal, targets multiple KCNQ channels, particularly:

  • KCNQ2/KCNQ3: This is the most common type of KCNQ channel found in neurons.
  • KCNQ1 in complex with KCNE1: This form is mainly present in the heart. KCNE1 acts as a regulatory subunit that influences KCNQ1 channel function.

 

Cilantro leaf harbors a potent potassium channel-activating anticonvulsant

Herbs have a long history of use as folk medicine anticonvulsants, yet the underlying mechanisms often remain unknown. Neuronal voltage-gated potassium channel subfamily Q (KCNQ) dysfunction can cause severe epileptic encephalopathies that are resistant to modern anticonvulsants. Here we report that cilantro (Coriandrum sativum), a widely used culinary herb that also exhibits antiepileptic and other therapeutic activities, is a highly potent KCNQ channel activator. Screening of cilantro leaf metabolites revealed that one, the long-chain fatty aldehyde (E)-2-dodecenal, activates multiple KCNQs, including the predominant neuronal isoform, KCNQ2/KCNQ3 [half maximal effective concentration (EC50), 60 ± 20 nM], and the predominant cardiac isoform, KCNQ1 in complexes with the type I transmembrane ancillary subunit (KCNE1) (EC50, 260 ± 100 nM). (E)-2-dodecenal also recapitulated the anticonvulsant action of cilantro, delaying pentylene tetrazole-induced seizures. In silico docking and mutagenesis studies identified the (E)-2-dodecenal binding site, juxtaposed between residues on the KCNQ S5 transmembrane segment and S4-5 linker. The results provide a molecular basis for the therapeutic actions of cilantro and indicate that this ubiquitous culinary herb is surprisingly influential upon clinically important KCNQ channels

Activation of KCNQ5 by cilantro could also contribute to its gut stimulatory properties, as KCNQ5 is also expressed in gastrointestinal smooth muscle, and its activation might therefore relax muscle, potentially being therapeutic in gastric motility disorders such as diabetic gastroparesis.

The KCNQ activation profile of (E)-2-dodecenal bears both similarities and differences to that of other KCNQ openers. We recently found that mallotoxin, from the shrub Mallotus oppositifolius that is used in African folk medicine, also activates KCNQ1-5 homomers, prefers KCNQ2 over KCNQ3, and in docking simulations binds in a pose reminiscent to that predicted for (E)-2-dodecenal, between (KCNQ2 numbering) R213 and W236 In addition to the widespread use of cilantro in cooking and as an herbal medicine, (E)-2-dodecenal itself is in broad use as a food flavoring and to provide citrus notes to cosmetics, perfumes, soaps, detergents, shampoos, and candles (59).

Our mouse seizure studies suggest it readily accesses the brain, and it is likely that its consumption as a food or herbal medicine (in cilantro) or as an added food flavoring would result in KCNQ-active levels in the human body; we found the 1% cilantro extract an efficacious KCNQ activator, and (E)-2-dodecenal itself showed greater than half-maximal opening effects on KCNQ2/3 at 100 nM (.10 mV shift at this concentration) (EC50, 60 6 20 nM). We anticipate that its activity on KCNQ channels contributes significantly to the broad therapeutic spectrum attributed to cilantro, which has persisted as a folk medicine for thousands of years throughout and perhaps predating human recorded history.

 

From the University of California: 


How cilantro works as a secret weapon against seizures

In a new study, researchers uncovered the molecular action that enables cilantro to effectively delay certain seizures common in epilepsy and other diseases.

The study, published in FASEB Journal, explains the molecular action of cilantro (Coriandrum sativum) as a highly potent KCNQ channel activator. This new understanding may lead to improvements in therapeutics and the development of more efficacious drugs.

“We discovered that cilantro, which has been used as a traditional anticonvulsant medicine, activates a class of potassium channels in the brain to reduce seizure activity,” said Geoff Abbott, Ph.D., professor of physiology and biophysics at the UC Irvine School of Medicine and principal investigator on the study.

“Specifically, we found one component of cilantro, called dodecenal, binds to a specific part of the potassium channels to open them, reducing cellular excitability.”

 

KCNQ channels and autism

There is a growing body of research suggesting a connection between KCNQ channels and autism.

·        KCNQ channel mutations: Genetic studies have identified mutations in several KCNQ channel genes (including KCNQ2, KCNQ3) in individuals with ASD. These mutations might disrupt the normal function of KCNQ channels, leading to abnormal brain activity.

  • Neuronal excitability: KCNQ channels help regulate the electrical activity of neurons by controlling the flow of potassium ions. Mutations or dysfunction in KCNQ channels could lead to increased neuronal excitability, which has been implicated in ASD. 
  • Shared features: Epilepsy is a common comorbidity with autism. Interestingly, KCNQ channel dysfunction is also linked to certain types of epilepsy. This suggests some shared mechanisms between these conditions.

 

KCNQ Dysfunction and Intellectual Disability

Mutations in certain KCNQ genes can lead to malfunctions in the corresponding potassium channels. These malfunctions can disrupt normal neuronal activity and contribute to intellectual disability.

  • KCNQ2/3 Mutations: Research suggests increased activity in KCNQ2 and KCNQ3 channels, due to mutations in their genes, might be associated with a subset of patients with intellectual disability alongside autism spectrum disorder. 
  • KCNQ5 Mutations: Studies have identified mutations in the KCNQ5 gene, leading to both loss-of-function and gain-of-function effects on the channel. These changes in KCNQ5 channel activity can contribute to intellectual disability, sometimes accompanied by epilepsy.

 

The other naming system

KCNQ channels belong to a larger potassium channel family called Kv7. So, you might see them referred to as Kv7.1 (KCNQ1), Kv7.2 (KCNQ2), and so on, based on their specific gene and protein sequence.

 

Mefenamic acid and Kir channels (inwards rectifying potassium ion channels)

Ponstan (mefenamic acid) affects Kir channels and KCNQ channels.

Different Kir channel subtypes contribute to various brain functions, including:

  • Neuronal excitability: Kir channels help regulate the resting membrane potential of neurons, influencing their firing activity.
  • Potassium homeostasis: They play a role in maintaining the proper balance of potassium ions within and outside neurons, crucial for normal electrical signaling.
  • Synaptic inhibition: Some Kir channels contribute to inhibitory neurotransmission, which helps balance excitatory signals in the brain.

Kir Channels are primarily inward rectifiers, meaning they allow potassium ions to flow more easily into the cell than out. They play a role in setting the resting membrane potential of cells, influencing their excitability.

KCNQ Channels can be voltage-gated or regulated by other factors. They contribute to various functions like regulating neuronal firing in the brain,

 

Other effects of Cilantro

It is certainly could be just a coincidence that Cilantro and Ponstan affect KCNQ channels. Cilantro has many other effects.

Coriandrum sativum and Its Utility in Psychiatric Disorders

Recent research has shown that Coriandrum sativum offers a rich source of metabolites, mainly terpenes and flavonoids, as useful agents against central nervous system disorders, with remarkable in vitro and in vivo activities on models related to these pathologies. Furthermore, studies have revealed that some compounds exhibit a chemical interaction with γ-aminobutyric acid, 5-hydroxytryptamine, and N-methyl-D-aspartate receptors, which are key components in the pathophysiology associated with psychiatric and neurological diseases. 

 

Bioactivities of isolated compounds from Coriandrum sativum by interaction with some neurotransmission systems involved in psychiatric and neurological disorders.

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10385770/table/molecules-28-05314-t002/?report=objectonly

 

 

Understanding 2p16.3 (NRXN1) deletions



One parent contacted me to ask about the genetic test results they had received for their child.

To understand what happens when parts of the NRXN1 gene are missing you need to read up on neurexins and neuroligins.

 

Neurexins and Neuroligins

Neurexins ensure the formation of proper synaptic connections, fine-tune their strength, and contribute to the brain's adaptability. Understanding their role is crucial for understanding brain development, function, and various neurological disorders.

Neurexins and neuroligins are cell adhesion molecules that work together to ensure proper synapse formation, function, and ultimately, a healthy and functioning brain.

Neuroligins are located on the postsynaptic membrane (receiving neuron) of a synapse.

Neurexins are located on the presynaptic membrane (sending neuron) of a synapse.

Mutations in either neurexin or neuroligin genes have been linked to various neurodevelopmental disorders, including autism.

A comprehensive presentation for families is below:

 

Understanding 2p16.3 (NRXN1) deletions

https://www.rarechromo.org/media/information/Chromosome%20%202/2p16.3%20(NRXN1)%20deletions%20FTNW.pdf

 

A microdeletion in the NRXN1 gene on chromosome 2p16.3 can cause a condition similar to Pitt-Hopkins syndrome, but referred to as Pitt-Hopkins like syndrome 2 (PHLS2).

 

NRXN1 Gene:

  • NRXN1 codes for a protein called neurexin 1 alpha, which plays a critical role in the development and function of synapses, the junctions between neurons in the brain.
  • Neurexin 1 alpha helps neurons connect with each other and transmit signals.

Microdeletion:

  • A microdeletion is a small deletion of genetic material from a chromosome.
  • In PHLS2, a microdeletion occurs in the NRXN1 gene, removing some of the genetic instructions needed to produce functional neurexin 1 alpha protein.

Pitt-Hopkins Like Syndrome 2 (PHLS2):

  • PHLS2 is a genetic disorder characterized by intellectual disability, developmental delays, and various neurodevelopmental features.
  • Symptoms can vary depending on the size and specific location of the NRXN1 microdeletion.
  • Common features include:
    • Intellectual disability (ranging from mild to severe)
    • Speech and language impairments
    • Developmental delays in motor skills
    • Stereotypies (repetitive movements)
    • Seizures
    • Behavioral problems (e.g., hyperactivity, anxiety)
    • Distinctive facial features (not always present)

 

What has this got to do with Pitt Hopkins syndrome (loss of TCF4)?

“TCF4 may be transcribed into at least 18 different isoforms with varying N-termini, which impact subcellular localization and function. Functional analyses and mapping of missense variants reveal that different functional domains exist within the TCF4 gene and can alter transcriptional activation of downstream genes, including NRXN1 and CNTNAP2, which cause Pitt-Hopkins-like syndromes 1 and 2.”

 

NRXN1 interactions with other genes/proteins

Given the function of neurexins and neuroligins, you would expect that the common interactions of NRXN1 are with neuroligins. We see below the NLGNs (neuroligin genes/proteins)

Our more avid readers may recall that neuroligins are one mechanism for regulating the GABA switch. This is the developmental switch that should occur in all humans about two weeks after birth.  If it does not occur, the brain cannot develop and function normally. Autism and intellectual disability are the visible symptoms.

 

An unexpected role of neuroligin-2 in regulating KCC2 and GABA functional switch

https://molecularbrain.biomedcentral.com/articles/10.1186/1756-6606-6-23#:~:text=Novel%20function%20of%20neuroligin%2D2,expression%20level%20was%20significantly%20decreased.

 

We report here that KCC2 is unexpectedly regulated by neuroligin-2 (NL2), a cell adhesion molecule specifically localized at GABAergic synapses. The expression of NL2 precedes that of KCC2 in early postnatal development. Upon knockdown of NL2, the expression level of KCC2 is significantly decreased, and GABA functional switch is significantly delayed during early development. Overexpression of shRNA-proof NL2 rescues both KCC2 reduction and delayed GABA functional switch induced by NL2 shRNAs. Moreover, NL2 appears to be required to maintain GABA inhibitory function even in mature neurons, because knockdown NL2 reverses GABA action to excitatory. 

Our data suggest that in addition to its conventional role as a cell adhesion molecule to regulate GABAergic synaptogenesis, NL2 also regulates KCC2 to modulate GABA functional switch and even glutamatergic synapses. Therefore, NL2 may serve as a master regulator in balancing excitation and inhibition in the brain.

 

It would seem plausible that in the case of microdeletions of the NRXN1 gene there will be a direct impact on the expression of NLGN2 gene that encodes neuroligin 2.

So plausible therapies to trial for microdeletions of the NRXN1 gene would include bumetanide, as well as cGPMax, due to the link with Pitt Hopkins.

 

GPC5 gene 

Finally, we move on to our last gene which is GPC5.

The protein Glpycan 5/GPC5 plays a role in the control of cell division and growth regulation.

Not surprising, GPC5 acts a tumor suppressor, making it a cancer gene. Because of this it is also an autism gene. It also plays a role in Alzheimer’s disease.

I was not sure I would be able to say anything about how you might treat autism caused by a mutation in GPC5.

 

Glycan susceptibility factors in autism spectrum disorders

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5556687/

 

I am assuming the mutation causes a loss of function, meaning there is a reduced level of the protein Glpycan 5.

Since one role of this gene is to suppress Wnt/beta-catenin signaling, you might want to replace this action.

This is actually covered in my blog in various places. One way is via a GSK-3β inhibitor.

GSK-3β inhibitor include drugs designed to block GSK-3β activity, examples include lithium (used for bipolar disorder), kenpaullone, and tideglusib. Certain natural compounds like curcumin and quercetin have been shown to possess GSK-3β inhibitory effects.

Atorvastatin, which my son has taken for 10 years, is indirectly a GSK-3β inhibitor

Some natural compounds like fisetin (found in fruits and vegetables) have been shown to promote beta-catenin phosphorylation, leading to its degradation.

In previous posts I pointed out that the cheap kids’ anthelmintic medication Mebendazole is indirectly another Wnt inhibitor. This is because it reduces TNIK. TNIK promotes Wnt signaling by stabilizing beta-catenin, a key player in the pathway. By reducing TNIK levels, mebendazole indirectly disrupts Wnt signaling. Mebendazole is therefore a novel cancer therapy and is being investigated to treat brain cancers, colon cancer, breast cancers etc.

Unlike what is says in the literature about GPC5, there actually are many options that can be safely trialed.

Note that you may not know for sure that any mutation is actually causal/pathogenic. Some people have several “likely pathogenic” mutations, some likely are not.

 

Conclusion

We have covered the potassium ion channel Kv7.1 previously. In Pitt Hopkins syndrome this ion channel is over expressed and so you would want to inhibit it. Do not take Cilantro, it would have the opposite effect to what you want.

It looks like cGPMAX is one thing you need to trial for Pitt Hopkins syndrome and Rett syndrome. For idiopathic autism it may, or may not help. Try a low dose first, observe the effect, then try a higher dose.

In Rett syndrome we know that people with have as much NKCC1 RNA — a molecule that carries the instructions to make the protein — as healthy individuals. However, their levels of KCC2 RNA are much lower, potentially disrupting the excitation/inhibition balance of nerve cell signaling. This will result in elevated chloride in neurons. This is correctable today using bumetanide.

People with NRXN1 microdeletions do seem to have treatment options, as do people with GPC5 mutations.

Note that out reader Janu, treating a mutation in GABRB2, reports success with a combination of the SSRI drug Lexapro and sodium valproate.

I am a fan of low dose Ponstan for sound sensitivity, it has numerous potentially beneficial mechanisms. It has been even shown to protect against Alzheimer’s disease.  There is no reason not to give cilantro a try as an alternative or complement to improve sound sensitivity.

Dried coriander is normally made from the seeds and is not what you need. In your supermarket you can buy fresh coriander leaves (Cilantro). The fresh herb is about 90% water, but when you dry the herb you will lose at lot of the active substance because it is volatile and will evaporate. My guess is that you will need 2-3 g of the fresh herb to equal Andy’s 425mg supplement.  You can eat the stalks as well as the leaves, it all has the same pungent taste.




Wednesday 10 May 2023

Low dose Clonazepam for MIA Autism, Ponstan and TRPM3 in Intellectual Disability, Clemastine to restore myelination in Pitt Hopkins, Improving Oxytocin therapy with Maca, Lamotrigine for some autism

 

Monty in Ginza, Tokyo

Today’s post comes from Tokyo and looks at 5 therapies already discussed in previous posts and follows up on recent coverage in the research. They all came up in recent conversations I have been having.

·      Low dose Clonazepam  – Maternal Immune Activation model of autism

·      Ponstan – TRPM3 causing intellectual disability  (ID/MR)

·      Clemastine – improving myelination in Pitt Hopkins syndrome model

·      Oxytocin – Maca supplement to boost effect

·      Lamotrigine (an anti-epilepsy drug) to moderate autism

The good news is that many of same therapies keep coming up.


Ponstan and TRPM3 caused ID/MR

There is a lot in this blog about improving cognition, which is how I called treating ID/MR.  There are very many causes of ID and some of them are treatable.

ID/MR was always a part of classic autism and in the new jargon is part of what they want to call profound autism.

I was recently sent a paper showing how the cheap pain reliever Ponstan blocks the TRMP3 channel and that this channel when mutated can lead to intellectual disability and epilepsy.

Mefenamic acid selectively inhibits TRPM3-mediated calcium entry.

My own research has established that mefenamic acid seems to improve speech and cognition, as well as sound sensitivity.  The latter effect I am putting down to its effect on potassium channels. 

De novo substitutions of TRPM3 cause intellectual disability and epilepsy

The developmental and epileptic encephalopathies (DEE) are a heterogeneous group of chronic encephalopathies frequently associated with rare de novo nonsynonymous coding variants in neuronally expressed genes. Here, we describe eight probands with a DEE phenotype comprising intellectual disability, epilepsy, and hypotonia. Exome trio analysis showed de novo variants in TRPM3, encoding a brain-expressed transient receptor potential channel, in each. Seven probands were identically heterozygous for a recurrent substitution, p.(Val837Met), in TRPM3’s S4–S5 linker region, a conserved domain proposed to undergo conformational change during gated channel opening. The eighth individual was heterozygous for a proline substitution, p.(Pro937Gln), at the boundary between TRPM3’s flexible pore-forming loop and an adjacent alpha-helix. General-population truncating variants and microdeletions occur throughout TRPM3, suggesting a pathomechanism other than simple haploinsufficiency. We conclude that de novo variants in TRPM3 are a cause of intellectual disability and epilepsy.

 

Fenamates as TRP channel blockers: mefenamic acid selectively blocks TRPM3

This study reveals that mefenamic acid selectively inhibits TRPM3-mediated calcium entry. This selectivity was further confirmed using insulin-secreting cells. KATP channel-dependent increases in cytosolic Ca2+ and insulin secretion were not blocked by mefenamic acid, but the selective stimulation of TRPM3-dependent Ca2+ entry and insulin secretion induced by pregnenolone sulphate were inhibited. However, the physiological regulator of TRPM3 in insulin-secreting cells remains to be elucidated, as well as the conditions under which the inhibition of TRPM3 can impair pancreatic β-cell function. Our results strongly suggest mefenamic acid is the most selective fenamate to interfere with TRPM3 function. 

Here, we examined the inhibitory effect of several available fenamates (DCDPC, flufenamic acid, mefenamic acid, meclofenamic acid, niflumic acid, S645648, tolfenamic acid) on the TRPM3 and TRPV4 channels using fluorescence-based FLIPR Ca2+ measurements. To further substantiate the selectivity, we tested the potencies of these fenamates on two other TRP channels from different subfamilies, TRPC6 and TRPM2. In addition, single-cell Ca2+ imaging, whole-cell voltage clamp and insulin secretion experiments revealed mefenamic acid as a selective blocker of TRPM3.

  

Oxytocin

 Oxytocin does increase how emotional you feel; the difficulty is how to administer it in a way that provides a long lasting effect.  The half-life of oxytocin is a just minutes. The traditional method uses a nose spray.

I favour the use of a gut bacteria that stimulates the release of oxytocin in the brain.  The effect should be much longer lasting. Even then the effect is more cute than dramatic.

The supplement Maca does not itself produce oxytocin, but “it restores social recognition impairments by augmenting the oxytocinergic neuronal pathways”.

So Maca looks like an interesting potential add-on therapy to boost the effect of oxytocin.

One reader wrote to me with a positive report on using Maca by itself, without any oxytocin.

 

Oral Supplementation with Maca Improves Social Recognition Deficits in the Valproic Acid Animal Model of Autism Spectrum Disorder

Autism spectrum disorder (ASD) is a congenital, lifelong neurodevelopmental disorder whose main symptom is impaired social communication and interaction. However, no drug can treat social deficits in patients with ASD, and treatments to alleviate social behavioral deficits are sorely needed. Here, we examined the effect of oral supplementation of maca (Lepidium meyenii) on social deficits of in utero-exposed valproic acid (VPA) mice, widely used as an ASD model. Although maca is widely consumed as a fertility enhancer and aphrodisiac, it possesses multiple beneficial activities. Additionally, it benefits learning and memory in experimental animal models. Therefore, the effect of maca supplementation on the social behavioral deficit of VPA mice was assessed using a social interaction test, a three-stage open field test, and a five-trial social memory test. The oral supplementation of maca attenuated social interaction behavior deficit and social memory impairment. The number of c-Fos-positive cells and the percentage of c-Fos-positive oxytocin neurons increased in supraoptic and paraventricular neurons of maca-treated VPA mice. These results reveal for the first time that maca is beneficial to social memory and that it restores social recognition impairments by augmenting the oxytocinergic neuronal pathways, which play an essential role in diverse social behaviors.

Maca (Lepidium meyenii) belongs to the cruciferous family and grows at high altitudes in Peru. In 2002, it was transplanted from Peru to the Yunnan Province of China. It is rich in dietary fiber; has many essential amino acids and nutrients including vitamin C, copper, and iron; and its root contains bioactive compounds. It is globally consumed and is popularly used as a fertility enhancer and aphrodisiac. On the other hand, with its potential to possess multi-nutritious components, it is reported to have diverse functions, including immunomodulation, antioxidant, antidepressant, antirheumatic, UV radiation protection, hepatoprotective, anti-fatigue, and neuroprotective effects. Interestingly, although the mechanism of the neuronal effect of maca is unclear, the uptake of maca extract improves learning and memory in memory-impaired model mice induced by either ethanol, ovariectomy, or scopolamine. However, the effects of maca on social memory impairment in neurodevelopmental disorders, including ASD, have not yet been tested.

In this study, the effects of maca on ASD animal models, in utero VPA-exposed mice, were investigated. The effect on social recognition by maca uptake with gavage was assessed using the social interaction test, a three-stage open field test, and the five-trail social recognition test. We also explored whether maca intake affects oxytocinergic signaling pathways, which play an important role in various social behaviors.

In this study, we showed that maca uptake rescues the deficits of social behavior and social recognition memory in VPA mice, a mouse model of autism. The c-Fos immunoreactivity of oxytocinergic neurons in SON and PVN increased significantly after maca treatment in VPA mice. Following previous studies indicating that OT administration ameliorates the impairment of social behavior in VPA mice, maca may also have improving effects on the deficit of social behavior and social recognition memory of VPA mice, probably by activating the OT neuronal pathway. Previous studies showed that maca could improve cognitive function in the mice model of impaired cognitive memory induced by either ovariectomy, ethanol, or scopolamine. Further studies are necessary to elucidate the potential link between maca and OT and to determine which components are involved in improving social recognition memory.

We have shown that maca improves the impairment of social memory and social behavioral deficits through oxytocinergic system modulation in this study. Although maca may not have an immediate effect on social behavioral deficits and takes days or weeks to demonstrate the effects, behavioral improvements, were visible regardless of the time of oral intake. The time between the very last oral intake of maca and the start of the social behavioral experiments in this study was more than 16 h. The duration of the maca’s effect on social behavioral deficits after the supplementation period is being investigated in our follow-up experiments. The possibility of the persistent effect of maca is very appealing, given that OT does not have a sustained effect due to its rapid metabolism, despite its immediate effects. Therefore, taking maca as a supplement while also receiving repeated OT treatment may have a synergistic, sustainable effect on improving social impairment in patients with ASD. Maca is already being used as a dietary supplement worldwide and has a high potential for practical applications.

 

This study showed for the first time that maca supplementation improves the impairment of social recognition memory in ASD model mice. We added the mechanism that social memory improvement may occur through the upregulation of oxytocinergic pathways. Maca highlights the possibility of treating social deficits sustainably in individuals with ASDs.

 

Low dose clonazepam

Professor Catterall was the brains behind low dose clonazepam for mice, I just translated it across to humans. It is one way to modify the E/I (excitatory/inhibitory) imbalance in autism.

I found that it gave a boost to cognition. Not as big as bumetanide, but worth having nonetheless.

I do not believe you have to be a bumetanide responder to respond well to low dose clonazepam.

Several people have written to me recently to say it works for their child.

Our reader Tanya is interested in the Maternal Immune Activation (MIA) trigger to autism. She highlighted a recent study showing how and why clonazepam can reverse autism in the MIA mouse model of autism. 

Clonazepam attenuates neurobehavioral abnormalities in offspring exposed to maternal immune activation by enhancing GABAergic neurotransmission

Ample evidence indicates that maternal immune activation (MIA) during gestation is linked to an increased risk for neurodevelopmental and psychiatric disorders, such as autism spectrum disorder (ASD), anxiety and depression, in offspring. However, the underlying mechanism for such a link remains largely elusive. Here, we performed RNA sequencing (RNA-seq) to examine the transcriptional profiles changes in mice in response to MIA and identified that the expression of Scn1a gene, encoding the pore-forming α-subunit of the brain voltage-gated sodium channel type-1 (NaV1.1) primarily in fast-spiking inhibitory interneurons, was significantly decreased in the medial prefrontal cortex (mPFC) of juvenile offspring after MIA. Moreover, diminished excitatory drive onto interneurons causes reduction of spontaneous gamma-aminobutyric acid (GABA)ergic neurotransmission in the mPFC of MIA offspring, leading to hyperactivity in this brain region. Remarkably, treatment with low-dose benzodiazepines clonazepam, an agonist of GABAA receptors, completely prevented the behavioral abnormalities, including stereotypies, social deficits, anxiety- and depression-like behavior, via increasing inhibitory neurotransmission as well as decreasing neural activity in the mPFC of MIA offspring. Our results demonstrate that decreased expression of NaV1.1 in the mPFC leads to abnormalities in maternal inflammation-related behaviors and provides a potential therapeutic strategy for the abnormal behavioral phenotypes observed in the offspring exposed to MIA.

 

Pitt Hopkins – Clemastine and Sobetirome

Poor myelination is a feature of much autism and is a known problem in Pitt Hopkins syndrome.

I did cover a paper a while back where the Pitt Hopkins researchers showed that genes involved in myelination are down-regulated not only in Pitt Hopkins, but in several other popular models of autism.

From the multiple sclerosis (MS) research we have assembled a long list of therapies to improve different processes involved in myelination. Today we can add to that list sobetirome (and the related Sob-AM2). Sobetirome shares some of its effects with thyroid hormone (TH), it is a thyroid hormone receptor isoform beta-1 (THRβ-1) liver-selective analog.

Some people do use thyroid hormones to treat autism, and indeed US psychiatrists have long used T3 to treat depression.

The problem with giving T3 or T4 hormones is that it has body-wide effects and if you give too much the thyroid gland will just produce less.

One proposed mechanism I wrote about long ago is central hypothyroidism, that is a lack of the active T3 hormone just within the brain. One possible cause proposed was that oxidative stress reduces the enzyme D2 that is used to convert circulating prohormone T4 to T3. The result is that your blood test says your thyoid function is great, but in your brain you lack T3.

It looks like using sobetirome you can spice up myelination in the brain, without causing any negative effects to your thyroid gland.

Rather surprisingly, sobetirome is already sold as a supplement, but it is not cheap like Clemastine, the other drug used in the successful study below.

 

Promyelinating drugs promote functional recovery in an autism spectrum disorder mouse model of Pitt–Hopkins syndrome

Pitt–Hopkins syndrome is an autism spectrum disorder caused by autosomal dominant mutations in the human transcription factor 4 gene (TCF4). One pathobiological process caused by murine Tcf4 mutation is a cell autonomous reduction in oligodendrocytes and myelination. In this study, we show that the promyelinating compounds, clemastine, sobetirome and Sob-AM2 are effective at restoring myelination defects in a Pitt–Hopkins syndrome mouse model. In vitro, clemastine treatment reduced excess oligodendrocyte precursor cells and normalized oligodendrocyte density. In vivo, 2-week intraperitoneal administration of clemastine also normalized oligodendrocyte precursor cell and oligodendrocyte density in the cortex of Tcf4 mutant mice and appeared to increase the number of axons undergoing myelination, as EM imaging of the corpus callosum showed a significant increase in the proportion of uncompacted myelin and an overall reduction in the g-ratio. Importantly, this treatment paradigm resulted in functional rescue by improving electrophysiology and behaviour. To confirm behavioural rescue was achieved via enhancing myelination, we show that treatment with the thyroid hormone receptor agonist sobetirome or its brain penetrating prodrug Sob-AM2, was also effective at normalizing oligodendrocyte precursor cell and oligodendrocyte densities and behaviour in the Pitt–Hopkins syndrome mouse model. Together, these results provide preclinical evidence that promyelinating therapies may be beneficial in Pitt–Hopkins syndrome and potentially other neurodevelopmental disorders characterized by dysmyelination.

 

Sobetirome  (also called GC-1)

Sobetirome is a thyroid hormone receptor isoform beta-1 (THRβ-1) liver-selective analog.

In humans, sobetirome lowers plasma LDL cholesterol and reduced plasma triglycerides, while its liver-selective activity helped avoid the side effects seen with many other thyromimetic agents.

 

Myelin repair stimulated by CNS-selective thyroid hormone action

Oligodendrocyte processes wrap axons to form neuroprotective myelin sheaths, and damage to myelin in disorders, such as multiple sclerosis (MS), leads to neurodegeneration and disability. There are currently no approved treatments for MS that stimulate myelin repair. During development, thyroid hormone (TH) promotes myelination through enhancing oligodendrocyte differentiation; however, TH itself is unsuitable as a remyelination therapy due to adverse systemic effects. This problem is overcome with selective TH agonists, sobetirome and a CNS-selective prodrug of sobetirome called Sob-AM2. We show here that TH and sobetirome stimulated remyelination in standard gliotoxin models of demyelination. We then utilized a genetic mouse model of demyelination and remyelination, in which we employed motor function tests, histology, and MRI to demonstrate that chronic treatment with sobetirome or Sob-AM2 leads to significant improvement in both clinical signs and remyelination. In contrast, chronic treatment with TH in this model inhibited the endogenous myelin repair and exacerbated disease. These results support the clinical investigation of selective CNS-penetrating TH agonists, but not TH, for myelin repair.

 

Compound protects myelin, nerve fibers

 

Research could be important in treating, preventing progression of multiple sclerosis, other neurodegenerative diseases

A compound appears to protect nerve fibers and the fatty sheath, called myelin, that covers nerve cells in the brain and spinal cord. The new research in a mouse model advances earlier work to develop the compound - known as sobetirome - that has already showed promise in stimulating the repair of myelin.

Lead author Priya Chaudhary, M.D., assistant professor of neurology in the OHSU School of Medicine who is focused on developing therapies for neurodegenerative diseases, said that the technique is a common step in drug discovery.

"It is important to show the effectiveness of potential drugs in a model that is most commonly used for developing new therapies," Chaudhary said.

The researchers discovered that they were able to prevent damage to myelin and nerve fibers from occurring, by stimulating a protective response in the cells that make and maintain myelin. They also reduced the activity of migroglia, a type of inflammatory cell in the brain and spinal cord that's involved in causing damage in multiple sclerosis and other diseases.

"The effects are impressive and are at least in part consistent with a neuroprotective effect with particular inhibition of myelin and axon degeneration, and oligodendrocyte loss," the authors write.

The discovery, if proven in clinical trials involving people, could be especially useful for people who are diagnosed with multiple sclerosis early in the disease's progression.

"The drug could protect the nervous system from damage and reduce the severity of the disease," Bourdette said.

 

Does Lamotrigine have the potential to 'cure' Autism?

Recently headlines appeared like this one:-

Scientists 'CURE autism' in mice using $3 epilepsy drug

It referred to the use of the epilepsy drug Lamotrigine to treat a mouse model of autism, caused by reduced expression of the gene MYT1L.

What the tabloid journalists failed to notice was that there has already been a human trial of Lamotrigine in autism.  That trial was viewed as unsuccessful by the clinicians, although the parents did not agree.

There were many comments in the media from parents whose child already takes this drug for their epilepsy and they saw no reduction in autism. There were some who found it made autism worse.

 

MYT1L haploinsufficiency in human neurons and mice causes autism-associated phenotypes that can be reversed by genetic and pharmacologic intervention

 

Lamotrigine therapy for autistic disorder: a randomized, double-blind, placebo-controlled trial

In autism, glutamate may be increased or its receptors up-regulated as part of an excitotoxic process that damages neural networks and subsequently contributes to behavioral and cognitive deficits seen in the disorder. This was a double-blind, placebo-controlled, parallel group study of lamotrigine, an agent that modulates glutamate release. Twenty-eight children (27 boys) ages 3 to 11 years (M = 5.8) with a primary diagnosis of autistic disorder received either placebo or lamotrigine twice daily. In children on lamotrigine, the drug was titrated upward over 8 weeks to reach a mean maintenance dose of 5.0 mg/kg per day. This dose was then maintained for 4 weeks. Following maintenance evaluations, the drug was tapered down over 2 weeks. The trial ended with a 4-week drug-free period. Outcome measures included improvements in severity and behavioral features of autistic disorder (stereotypies, lethargy, irritability, hyperactivity, emotional reciprocity, sharing pleasures) and improvements in language and communication, socialization, and daily living skills noted after 12 weeks (the end of a 4-week maintenance phase). We did not find any significant differences in improvements between lamotrigine or placebo groups on the Autism Behavior Checklist, the Aberrant Behavior Checklist, the Vineland Adaptive Behavior scales, the PL-ADOS, or the CARS. Parent rating scales showed marked improvements, presumably due to expectations of benefits.


One reader of this blog who heard all about the news and was sceptical, since after all it is a mouse model. Her 8 year old non-verbal child was not happy taking the drug Keppra and was already scheduled to try Lamotrigine. 

Within a week his teacher called to say he was saying his ABCs, the next week he was counting out loud, the following month he’s attempting to repeat words of interest and this week he’s spelling animals by memory, dolphin, duck, wolf, chicken, pig, etc.

We are 2 months in and at 50mg, our target dose is 100mg bid. Obviously with our success, I’ve been working with his doctor and will continue to.”

 

Conclusion

Even though every day new autism research is published, there is so much already in this blog that not much appearing is totally new to regular readers.

We saw several years ago that low dose clonazepam should be beneficial to some people with autism, in particular Dravet syndrome. Today we learnt a little more about why Nav1.1 might be disturbed beyond those with Dravet syndrome. In the maternal immune activation model it seems to be a winner. It seems to benefit many of those who have trialed it.

Treating myelination deficits has been well covered in this blog. In previous posts we saw how Pitt Hopkins syndrome researchers showed how myelination gene expression was disturbed in a wide range of autisms. Today we saw evidence to support such therapy and we discovered a new drug.

Oxytocin does help some people with autism, but not as much as you might expect. Today we learnt of a potential add on therapy, a supplement called Maca.

The idea that anti-epilepsy drugs might help some autism has been well covered. From low dose valproate to low dose phenytoin from Dr Philip Bird in Australia.

Treatment of Autism with low-dose Phenytoin, yet another AED

Recent research suggested that Lamotrigine should help some with autism and today you learned that it really does help in one case. The fact that a tiny study a few years ago suggested no responders just tells us that only a small subgroup are likely to benefit.

We already know that some people's autism is made worse by their epilepsy therapy. This is just what you would expect. Time to find a different epilepsy therapy.

My favorite new therapy, low dose mefenemic acid / ponstan has numerous effects. One reader without autism, but with an unusual visual dysfunction (visual snow syndrome) and a sound sensitivity problem contacted me a while to see if NKCC1 might be the root of his problem. I suggested he try Ponstan, which did actually work for him and is easy to buy where he lives. Now he sends me research into all its possible modes of action. One mode of action relates to a cause of intellectual disability (ID/MR). Is this a factor in why Ponstan seems to improve speech and cognition in some autism? I really don't mind why it works - I just got lucky again, that is how I look at it. The more I read the luckier I seem to get.