OH NO! Comorbidity!

OH NO!- Comorbidity!

Autism Spectrum Disorder (ASD) is a neurodevelopmental condition that is known for its challenges in social situations such as social communication, repetitive behaviors, and problems with social cues. Those with ASD have difficulty expressing and understanding emotions, however this expands beyond the normal symptomology

Comorbidity is an additional disease on top of a primary diagnosis. They are more common in those with ASD than those without. Research shows a high rate of comorbidities- about 70% of those with ASD have co-occuring conditions that blur diagnosis and treatment for patients with ASD. Symptoms that develop with comorbidities add to the original symptoms of ASD making a healthy life difficult to have.

Genetic complexity

Autism does not have a single genetic cause. The developmental disorder is influenced by a multitude of factors. Primarily, multiple genetic factors like genetic mutations or copy number variations (CNVs). CNVs are a type of structural variation in the genome where DNA is deleted or duplicated. This changes normal function of genes and changes the number of gene copies present therefore, leading to developmental issues.

Figure 1: Normal X chromosome compared to fragile X chromosome

 

Fragile X Chromosome (FXS)

FXS is one condition that is associated with autism that is linked with DA dysfunction. It is the number 1 inherited cause of a wide range of intellectual disabilities. 1 in 3 individuals with ASD have FXS. This condition results from the repetition and mutation of the gene FMR1 which null mutants have significant increase in the synthesis of DA and 5-HT which is another name for serotonin- a neurotransmitter in charge of various functions including mood and helping out the nervous system.

In Figure 2, a typical chromosome the FMR1 gene will repeat just enough to maintain regulation for CGG repeat sequence, which will not interfere with functioning of the gene, however, a fragile X chromosome will exceed the number of regulated CGG repeat sequence and the FMR1 gene will slice, leaving an absence of the gene. Without the FMR1 gene neurons struggle to make synaptic connections in the brain.

Figure 2: Typical mutation of genes versus full mutation present in fragile X syndrome

 

When working with CNV’s you’re working with a lot of DNA! CNV’s make large changes to genetic material. This is one of the reasons why they are common in people who are neurodivergent. These genetic risk factors result in subtypes that  ultimately have a cascade of behavioral symptoms. For example, deletion of specific gene like 16p11.2 can result in delayed speech and struggling with social intelligence. So rather than focus on one gene, like finding a needle in a haystack, its better to try and adapt and understand the subtypes and comorbities of ASD.

The Role of Dopamine and Dopamine Transporter DAT

Dopamine (DA) is a widely known neurotransmitter that influences an array of functions within the brain. As one of the top three numerous neurotransmitters found in the brain, dysfunction of this molecule can lead to neurodevelopemental and psychiatric disorders including Autism. Too much or too little dopamine in those with ASD cause symptoms such as issues with sensory processing, repetitive behaviors, motor control and social reward processing.

There are two primary dopamine receptors, D1 and D2. These receptors are critical for managing the effects that dopamine has on the brain. Dysfunction of these receptors has been proven to be linked in ASD. These two receptors contribute mainly to behavioral disruptions that are present in ASD such as, social deficits, behavioral flexibility, and cognitive attention.

D1 vs D2

D1 Receptors: During activation D1 receptors  are primarily known for their synaptic plasticity, the strength in connection between neurons, and facilitating excitatory signaling, which is increasing neural activity. Synaptic plasticity and facilitating excitatory signaling regulates executive function and cognitive flexibility. They’re crucial for learning and memory. Dysfunction in D1 leads to deficits in communication and attention.

D2 Receptors: Unlike D1 receptors, D2 is mainly involved  inhibitory signaling-which is the decrease of neural activity- and involvement with motor control and coordination, in ASD, an overactive reward system leads to repetitive behaviors. In this situation the brains reward pathways are eing hyperstimulated and overly sensitive. When D2 receptors are not functioning there is no ‘manager’ to modulate motor control and coordination within movements.

DAT is the dopamine transporter in charge of regulating dopamine. The process in which dopamine levels are regulated from the synapse back into the neuron This means a dysfunction in DAT is a dysfunction in dopamine signaling. DA signaling drives behavioral activation which increases with reward rate.

What does This Have to do With Comorbidities? 

ASD and its comorbid conditions can help research within dopamine receptors to understand where dysregulation occurs. ASD contributes to neurological development of existing disorders as behavior, cognition, and emotion are all affected. Comorbidities are harmful to an individuals quality of life as with autism, it can be difficult to express how you are feeling. If we evaluate dopaminergic dysfunction we can understand core symptomology and be closer to the answer.

Al-Beltagi M. (2021). Autism medical comorbidities. World journal of clinical pediatrics10(3), 15–28. https://doi.org/10.5409/wjcp.v10.i3.15

DiCarlo, G. E., & Wallace, M. T. (2022, February). Modeling dopamine dysfunction in autism spectrum disorder: From invertebrates to vertebrates. Neuroscience and biobehavioral reviews. https://pmc.ncbi.nlm.nih.gov/articles/PMC8792250/

Hunter, J. E. (2024, May 16). FMR1 disorders. GeneReviews® [Internet]. https://www.ncbi.nlm.nih.gov/books/NBK1384/

Autism and the Gut-Brain connection

What is Autism?

Autism Spectrum Disorder is a developmental condition that affects an individual’s cognitive, social, and emotional functioning[1]. It is a spectrum, meaning that its symptoms and severity vary widely among individuals. While some individuals may have significant impairments in social interactions, others may demonstrate only mild deficits. The spectrum nature of autism means that no two people with autism experience the same set of challenges or strengths. Autism is not a disease but rather a neurodevelopmental condition that typically appears in early childhood. It is diagnosed more often in boys than in girls, with current estimates suggesting that 1 in 36 children and 1 in 45 adults are affected by the disorder[2].

Autism can be diagnosed at various ages, but most individuals are diagnosed around the age of 5, even though signs may be present as early as age 2[2]. Diagnosing autism early can help in providing the necessary support and interventions that can assist in improving long-term outcomes. Furthermore, autism often co-occurs with other psychiatric and medical conditions, a phenomenon known as comorbidity[2]. Common comorbidities include Attention-Deficit/Hyperactivity Disorder, anxiety disorders, depression, gastrointestinal issues, seizures, and sleep disorders[2]. Understanding these co-occurring conditions is crucial for providing comprehensive care for individuals with autism. Other sources on Autism symptoms and care can be found here.

Symptoms

Symptoms of autism involve deficits in social communication and interactions and restrictive or repetitive behaviors or interests[1].

  • Lack of facial expressions (by 9 months )
  • Delayed language skills
  • Delayed movement skills
  • Avoiding eye contact
  • Lack of social gestures ( shaking hands or waving goodbye by 12 months old)
  • Disinterest in social interaction (by 24 months old)
  • Difficulties identifying others’ emotions
  • Obsessive interests
  • Strong negative emotional response in response to minor changes
  • Excessive repeating of words or phrases
  • Strong reaction to certain sensory stimuli
  • Dependance on strong routines

The Gut- Brain Connection

Fig 1
Figure 1 [4] 
Research on mice that lack gut microbiota showed that these animals display altered social behaviors and increased anxiety-like traits[3]. This suggests that gut bacteria contribute to brain function and behavior, and these interactions can be seen in Figure 1. Additionally, gut microbiota influence neurotransmitter systems, including dopamine and serotonin, both of which play critical roles in ASD-related symptoms[3].

The Enteric nervous system regulates gut function and shares neurotransmitters and signaling pathways with the central nervous system. Dysregulation of these pathways has been observed in Autism, supporting the idea that gut dysfunction may be linked to behavioral and neurological symptoms[3]. Studies have also found differences in the expression of synaptic proteins in Autism models, reinforcing the potential impact of gut microbiota on brain development and function[3].

Conclusion

In conclusion, autism is a complex and multifaceted disorder that requires a personalized and holistic approach to diagnosis and treatment. Ongoing research into the gut-brain connection may eventually uncover new strategies to improve the quality of life for individuals on the autism spectrum.

 

 

 

 

[1] Centers for Disease Control and Prevention. (2024, November 25). About autism spectrum disorder. Centers for Disease Control and Prevention. https://www.cdc.gov/autism/about/index.html

[2] Autism spectrum disorder (ASD). Autism Speaks. (2025). https://www.autismspeaks.org/what-autism

[3]DiCarlo, G. E., & Wallace, M. T. (2022, February). Modeling dopamine dysfunction in autism spectrum disorder: From invertebrates to vertebrates. Neuroscience and biobehavioral reviews. https://pmc.ncbi.nlm.nih.gov/articles/PMC8792250/

[4]Liu, L., Huh, J., & Shah, K. (2022, March). Microbiota and the gut-brain-axis: Implications for new therapeutic design in the CNS. EBioMedicine. https://pubmed.ncbi.nlm.nih.gov/35255456/

Fragile X Syndrome and its ties to Autism

Artstract created by Ren Lind

There are many different theories as to the causes of Autism, however, Fragile X Syndrome is a direct cause for an estimated 2-6% of Autism cases. [1] Theories for other causes include genetics, environmental factors, the communication between neurons being disrupted, and others, but today we’ll be focusing on Fragile X.

Fragile X Background

Fragile X Syndrome (FXS) is an inherited genetic disorder. It gets its name from the X chromosome appearing “fragile” compared to a typical X chromosome, as figure 1 depicts. 

Figure 1: A mutation in the FMR1 gene leaves a gap in the X chromosome [2]
Since it’s on the X chromosome, males with XY chromosomes will always have FXS if inherited, while females with XX chromosomes can be carriers for FXS if it’s only on one X chromosome. Typically, males will have more severe symptoms compared to females. [3]

FXS individuals have a mutation on the FMR1 gene. This gene is responsible for making a protein that manages and develops synapses between neurons. Synapses are where communication between neurons gets passed along and important messages can be spread to the brain, or the body so bodily processes can happen. 

This gene mutation can lead to behavioral and social challenges, intellectual deficits, alterations in physical features, anxiety, and delayed speech and learning in childhood, among other symptoms. Typically, FXS is diagnosed between 12 months and 3.5 years old, but it can be diagnosed later in life. Symptoms of FXS greatly overlap with symptoms presented in Autism Spectrum Disorder. 

Autism Background

Autism Spectrum Disorder (ASD) is characterized by social deficits and repetitive motions or interests throughout life. [4] Symptoms such as social anxiety, avoiding eye contact, social impairment, and other diagnostic symptoms overlap between FXS and ASD. 

FXS causes Autism because the symptoms presented can be the same, depending on which symptoms the individual with FXS displays. It’s important to note that not all people with FXS will be diagnosed with ASD because they may have different symptoms than those of Autism. However, about 60% of males with FXS have Autism, and 20% of females with FXS have Autism. [5]

ADHD and Seizures Co-occurring 

Interestingly, there are also comorbidities of Attention-Deficit/Hyperactivity Disorder (ADHD) and seizure for people with Fragile X and Autism. Around 50% of people with Fragile X and ASD also have ADHD. [6]

Figure 2: Overlap between ADHD, Autism, and Fragile X Syndrome [7]
Seizures are a known comorbidity of ASD, [8] but around 15-20% of people with Fragile X also have seizures. A literature article written by Dicarlo and Wallace hypothesizes that ADHD and seizures may have similar biological pathologies to Autism, however, more research is needed to understand why these conditions seem to travel together. 

Summary

Autism has many theorized causes and risk factors, but Fragile X Syndrome is a confirmed cause for a small percentage of Autism cases. This occurs because Fragile X can present the same way as Autism, creating an overlap between the conditions, however, not every person with Fragile X will have the same symptoms as Autism. 

Resources

[1] Rajaratnam, A., Shergill, J., Salcedo-Arellano, M., Saldarriaga, W., Duan, X., & Hagerman, R. (2017). Fragile X syndrome and fragile X-associated disorders. F1000Research6, 2112. https://doi.org/10.12688/f1000research.11885.1

[2] Image from https://healthjade.net/fragile-x-syndrome/

[3] Rajaratnam, A., Shergill, J., Salcedo-Arellano, M., Saldarriaga, W., Duan, X., & Hagerman, R. (2017). Fragile X syndrome and fragile X-associated disorders. F1000Research6, 2112. https://doi.org/10.12688/f1000research.11885.1

[4] Dicarlo, G., Wallace, M. (2022). Modeling dopamine disfunction in autism spectrum disorder: from invertebrates and vertebrates. Neuroscience and Biobehavioral Reviews, 133. https://doi.org/10.1016/j.neubiorev.2021.12.017

[5, 6, 7] Rajaratnam, A., Shergill, J., Salcedo-Arellano, M., Saldarriaga, W., Duan, X., & Hagerman, R. (2017). Fragile X syndrome and fragile X-associated disorders. F1000Research6, 2112. https://doi.org/10.12688/f1000research.11885.1

[8] Dicarlo, G., Wallace, M. (2022). Modeling dopamine disfunction in autism spectrum disorder: from invertebrates and vertebrates. Neuroscience and Biobehavioral Reviews, 133. https://doi.org/10.1016/j.neubiorev.2021.12.017

How Genetic Epilepsies Relate to Autism Spectrum Disorder Symptoms

Introduction

Could epilepsy and autism be caused by your genes? Epilepsy is a common disorder that accompanies autism spectrum disorder (ASD), and ASD has been linked with multiple random genetic mutations in the DNA that comprises various genes.1 While genetics is not the only cause of ASD, the question of what mutated genes cause the epileptic seizures in many people with ASD can be asked.

To start off, most epilepsies are a combination of environmental and genetic factors, with genetic epilepsies only making up a small portion of epilepsies. With those that are genetic though, called idiopathic epilepsies, there are various mechanisms that initiate the process by which someone with a normal brain becomes susceptible to developing epilepsy (epileptogenesis). Those mechanisms include ion-channel disorders, the mechanisms underlying progressive myoclonus epilepsies (“myoclonus” meaning causing involuntary muscle spasms), developmental abnormalities, energy metabolism defects, and neuronal migration disorders.2

Table 1. This table displays idiopathic epileptic syndromes, their associated genes, which chromsomes these genes are on, and the mode of inheritance by which these genes are passed on. Oligogen refers to how the trait is influenced by a few genes.2

As you can see in Table 1, there are many epileptic syndromes linked to various genes on different chromosomes, and the way these syndromes are inherited varies. Clearly, genetics plays a role in epilepsy, but how does that correlate to ASD?

 

ASD Theories

To determine this, we must look at what the impacted genes do within the body and determine parallels between these results and the symptomatic characteristics of ASD. One theory regarding the cause of ASD is the excitatory-inhibitory (E/I) balance disruption theory, which proposes that ASD is due to alterations in the ratio of excitatory to inhibitory neurotransmission. This theory would explain the hyperactivity/hyperexcitability and reduced ability to maintain attention found in ASD patients. Another theory is the altered network connectivity theory, which hypothesizes that changes in neuronal connections within the brain are the main drivers of behaviors observed in ASD.1

Genes Related to Epilepsy & How They Correlate to ASD

Looking at Table 1 again, we see the epileptic syndrome abbreviated as ADNFLE. This is caused by mutations in the CHRNA4 or CHRNB2 genes located on chromosome 20. These genes typically encode for the alpha and beta subunits of nicotinic acetylcholine receptors, which are ion channels. When ADNFLE occurs, the wall of this ion channel is disrupted, disrupting cholinergic system function.2 The system plays a large role in memory, attention, and neuronal connectivity, so this epileptic syndrome, if co-occurring with ASD, could perhaps explain the working memory deficits seen in ASD and ties in well with the altered network connectivity theory of ASD.

Below ADNFLE on Table 1 we see BFNC, which is an ASD-inherited seizure disorder that causes mutations in voltage-gated K+ channel genes KCNQ2 and KCNQ3. In this disorder, the structure of a K+ channel pore is disrupted, reducing potassium current, which as can be seen in Figure 1, is a powerful controller of neuronal firing by controlling repolarization of the neuron.3 If K+ current is reduced, a neuron cannot return to its resting membrane potential, and this causes increased excitability of neurons since there are more easily brought to the threshold potential that causes an action potential to be sent.2 Thus, BFNC ties in nicely with the E/I balance disruption theory of autism and shows how epileptic disorders commonly cause the epileptic symptoms characteristic to ASD.

Figure 1. This diagram shows the flow of ions as an action potential is initiated during depolarization and is subsided during repolarization.3

There are other epileptic syndromes associated with genetic mutations that have similar impacts on ion channels, disrupting neuronal excitability, as well as other mutations related to disruptions of neuronal connectivity. For more examples of these genetic mutations and those that cause the earlier mechanisms characteristic to idiopathic epilepsies, click here.

In conclusion, idiopathic epilepsies are caused by various mechanisms, but the ones that correlate most with ASD are those that disrupt neuronal excitability and/or neuronal connectivity. Therefore, further research should be done to determine if medications that target these processes can improve the symptoms of epileptic phenotypes of ASD.

 

Footnotes

1Maximilians, Ludwig. “Genetics and epilepsy.” Dialogues Clin Neurosci, vol. 10, no. 1, 2008, pp. 29-36, doi:10.31887/DCNS.2008.10.1/oksteinlein

2DiCarlo, Gabriella E., Wallace, Mark T. “Modeling dopamine dysfunction in autism spectrum disorder: From invertebrates to vertebrates.” Neurosci and Biobehavioral Reviews, vol. 133, 2022, pp. 1-11, https://doi.org/10.1016/j.neubiorev.2021.12.017

3Changes in Sodium and Potassium Conductances. Cellular Physiology. https://neurotext.library.stonybrook.edu/C4/C4_5/C4_5.html

The Future of Autism Diagnosis: Integrating Subtyping into Diagnosis

Autism spectrum disorder (ASD) is recognized as a highly heritable neurodevelopmental condition. It impacts a significant percentage of children, with estimates suggesting that approximately 1 in 59 children are affected.1 ASD is typically characterized by challenges in social interactions, as well as patterns of restricted interests and repetitive behaviors.

There is no singular cause of this disorder. ASD is often diagnosed based on observable behavioral symptoms, reflecting a massive array of genetic and mechanistic differences among individuals.1

Understanding the Science

Discussing ASD often presents challenges when it comes to diagnosing individuals. The symptoms of ASD manifest across a spectrum of severity and can vary significantly from person to person.2 In males, symptoms are typically more noticeable than in females. For instance, females may often go undiagnosed because their symptoms can be less severe, and they may be more adept at masking them.

There are several theories regarding the potential causes of ASD and why it may be more prevalent in certain individuals.1 Some common theories include:

  • Excitatory/Inhibitory Imbalance: This theory suggests that there may be excessive excitation of neurons or a reduced level of inhibition, which can help explain the occurrence of co-occurring conditions such as epilepsy and ADHD.
  • Altered Network Connectivity: Research has shown that brains of individuals with ASD may exhibit both overconnectivity and underconnectivity among neurons.
  • Predictive Coding: This theory suggests that the brain struggles to update its internal model of the world, potentially leading to hypersensitivity and repetitive behaviors.

Here is a link to an article discussing the role that toxins might play in ASD development.

Numerous other hypotheses have been proposed regarding the development of ASD, including the role of dopamine dysregulation in contributing to ASD-like traits. Dopamine is involved in four major pathways1:

  • Nigrostriatal: Related to movement
  • Mesolimbic: Involved in reward and motivation
  • Mesocortical: Associated with cognition and decision-making
  • Tuberoinfundibular: Regulates hormone levels

These pathways highlight the complexity of dopamine’s role in the brain and its potential implications for individuals with autism. Here is another link to an article that further discussed the idea of dopamine playing a role in ASD.

Subtyping ASD

Currently, there are no specific treatments designed exclusively for individuals with ASD that can fully address the complexities of the condition. Instead, we primarily have options that focus on alleviating co-occurring symptoms, such as those associated with ADHD.1

Dividing ASD into distinct subcategories, such as “ASD with epilepsy” or “ASD with ADHD,” could significantly improve treatment outcomes for individuals. By recognizing these subtypes, healthcare providers could develop more targeted and effective intervention strategies tailored to each individual’s unique profile of symptoms and challenges.

Categorizing ASD in this way would allow for a more personalized approach to treatment, enabling clinicians to select therapies that specifically address each individual’s conditions. For some individuals, this might involve traditional medical interventions, while others may benefit more from behavioral therapies or other therapeutic options tailored to their specific needs. This approach could lead to better management of symptoms, improved quality of life, and more effective support for individuals with ASD and their families.

In summary, refining our understanding of ASD through subcategorization has the potential to enhance treatment options, allowing for a more comprehensive and tailored approach to care that meets the diverse needs of those on the autism spectrum.

Final Thoughts

While medications and therapies can be effective options for supporting individuals with ASD, they may not always be the most suitable solution. It is crucial for the public to develop a deeper understanding of ASD and to avoid isolating those who are affected by it.

Instead of viewing individuals with ASD as needing to be “fixed,” we should focus on understanding their unique experiences and challenges so we can provide the appropriate support when they seek it. It is important to empower individuals with ASD to maintain their independence and to make their own choices regarding their future and quality of life.

References

(1)      DiCarlo, G. E.; Wallace, M. T. Modeling Dopamine Dysfunction in Autism Spectrum Disorder: From Invertebrates to Vertebrates. Neuroscience and Biobehavioral Reviews. Elsevier Ltd February 1, 2022. https://doi.org/10.1016/j.neubiorev.2021.12.017.

(2)      Rossignol, D. A.; Genuis, S. J.; Frye, R. E. Environmental Toxicants and Autism Spectrum Disorders: A Systematic Review. Translational Psychiatry. Nature Publishing Group January 1, 2014. https://doi.org/10.1038/tp.2014.4.

 

The ADNP Gene and Its Role in Autism Spectrum Disorder

Autism Spectrum Disorder (ASD) is a highly heterogeneous neurodevelopmental condition affecting communication, social behavior, and cognitive functions. Recent advances in genetics have revealed a complex interplay between multiple genes and neurobiological pathways contributing to ASD. Among these, the Activity-Dependent Neuroprotective Protein (ADNP) gene has emerged as a crucial player in neurodevelopment, with mutations leading to severe cognitive and behavioral impairments. This paper explores the findings from the provided article, discussing the ADNP gene’s role in ASD and the implications for future research and therapy.

The ADNP gene encodes a protein essential for brain development and synaptic plasticity. It is one of the most frequently mutated genes associated with ASD, particularly in syndromic cases like Helsmoortel-Van der Aa syndrome (HVDAS). The article outlines how mutations in ADNP result in disrupted synaptic formation, leading to altered dopamine (DA) signaling, a neurotransmitter crucial for cognitive function, reward processing, and motor control (DiCarlo & Wallace, 2022).

One key finding is the link between dopamine dysfunction and ASD. Dopaminergic pathways are known to regulate attention, learning, and social behavior, all of which are impaired in individuals with ASD. Studies in animal models with ADNP mutations show altered DA transmission, providing a possible explanation for the repetitive behaviors and cognitive deficits seen in ASD (DiCarlo & Wallace, 2022).

Additionally, ADNP is implicated in regulating chromatin remodeling and gene expression during neural development. Mutations in this gene lead to widespread transcriptional dysregulation, affecting multiple pathways involved in neurogenesis, synaptic connectivity, and neuronal survival (DiCarlo & Wallace, 2022). Given these roles, ADNP has been proposed as a biomarker for early ASD diagnosis and a potential therapeutic target (DiCarlo & Wallace, 2022)

 Implications and Future Directions

The discovery of ADNP’s role in ASD represents a significant leap forward in understanding the genetic basis of the disorder. However, several challenges remain in translating this knowledge into effective treatments. Below are some key considerations:

1. Personalized Medicine and Targeted Therapies

Given the impact of ADNP mutations on dopamine signaling, pharmacological interventions targeting dopaminergic pathways may hold promise. Drugs such as dopamine agonists or modulators of synaptic plasticity could potentially mitigate cognitive and behavioral symptoms. However, the variability in ASD presentation necessitates a personalized approach to treatment (DiCarlo & Wallace, 2022).

2. Gene Therapy Prospects

Recent advances in CRISPR-Cas9 technology open new possibilities for correcting mutations in ADNP at the genetic level. Although gene-editing therapies for neurodevelopmental disorders are still in their infancy, research in this direction could pave the way for long-term solutions to ADNP-related ASD (DiCarlo & Wallace, 2022).

3. ADNP as a Diagnostic Biomarker

Current ASD diagnosis relies on behavioral assessments, which can be subjective. The identification of ADNP mutations as a genetic marker could lead to early and more precise diagnostic methods. This would enable early intervention, which is known to improve outcomes in children with ASD (DiCarlo & Wallace, 2022).

4. Environmental and Epigenetic Influences

While genetic mutations play a significant role, environmental factors and epigenetic modifications also contribute to ASD severity. Future research should explore how lifestyle, diet, and external stressors interact with ADNP mutations to influence ASD progression and symptomatology (DiCarlo & Wallace, 2022).

Conclusion

The ADNP gene provides a crucial link between genetic mutations and the neurobiological mechanisms underlying ASD. Its role in dopamine regulation, synaptic plasticity, and neural development makes it a prime target for future research. While challenges remain, ongoing advances in genetics and neuroscience bring hope for novel therapeutic interventions, offering new possibilities for individuals affected by ASD. Understanding ADNP’s function not only enhances our comprehension of ASD but also lays the groundwork for developing innovative strategies for diagnosis and treatment.

References

Aalto, S., Brück, A., Laine, M., Någren, K., & Rinne, J. O. (2005). Frontal dopamine release during a working memory task in healthy humans: A positron emission tomography study. Neuroscience Letters, 379(3), 207–212. https://doi.org/10.1016/j.neulet.2004.12.073

DiCarlo, G. E., & Wallace, M. T. (2022). Modeling dopamine dysfunction in autism spectrum disorder: From invertebrates to vertebrates. Neuroscience & Biobehavioral Reviews, 133, 104494. https://doi.org/10.1016/j.neubiorev.2021.12.017

Fisher, H. E., Aron, A., & Brown, L. L. (2005). Romantic love: An fMRI study of a neural mechanism for mate choice. The Journal of Comparative Neurology, 493(1), 58–62. https://doi.org/10.1002/cne.20772

Gaugler, T., Klei, L., Sanders, S. J., Bodea, C. A., Goldberg, A. P., Lee, A. B., Mahajan, M., Manaa, D., Pawitan, Y., Reichert, J., Ripke, S., Sandin, S., Sklar, P., Sullivan, P. F., Hultman, C. M., Devlin, B., Roeder, K., & Buxbaum, J. D. (2014). Most genetic risk for autism resides with common variation. Nature Genetics, 46(8), 881–885. https://doi.org/10.1038/ng.3039

Sanders, S. J., He, X., Willsey, A. J., Ercan-Sencicek, A. G., Samocha, K. E., Cicek, A. E., Murtha, M. T., Bal, V. H., Bishop, S. L., Dong, S., Goldberg, A. P., Jinlu, C., Keaney, J. F., Klei, L., Mandell, J. D., Neale, B. M., De Rubeis, S., Smith, L., & Buxbaum, J. D. (2015). Insights into autism spectrum disorder genomic architecture and biology from 71 risk loci. Neuron, 87(6), 1215–1233. https://doi.org/10.1016/j.neuron.2015.09.016

Selective Signaling in the endocannabinoid System : The ECS’s Secret to Keeping You Balanced

The human body is a finely tuned machine, constantly regulating pain, mood, metabolism, and more to maintain balance. One important system responsible for this regulation is the endocannabinoid system (ECS), a network of cannabinoid receptors(CB1 and CB2) , endocannabinoids (Anandamide (AEA) and 2-Arachidonoylglycerol (2-AG) ) , and enzymes (FAAH and MAGL) that work together to keep everything running smoothly. This means that the ECS helps regulate things like pain perception, mood, immune function, and metabolism. It’s constantly monitoring the body’s needs and adjusting accordingly, ensuring that everything stays in balance. [1]

  • Endocannabinoids (AEA and 2-AG) are produced when needed and act locally to help regulate these functions.
  • Cannabinoid Receptors (CB1 and CB2) are activated by these endocannabinoids to trigger specific responses.
  • Enzymes (FAAH and MAGL) break down these molecules once they’ve done their job, ensuring the ECS doesn’t overdo it.

But maintaining balance isn’t as simple as turning functions on and off. If the ECS activated all its pathways randomly or continuously, it could lead to dysfunction instead of stability. That’s why it relies on selective signaling, only activating specific pathways when needed to ensure precise control.

Therefore, understanding how the ECS’s selective signaling works is essential for improving health. By learning how to support this system, we can regulate vital functions more effectively while avoiding unwanted side effects, helping the body stay in perfect balance.

So, how does the ECS know when and where to act? It carefully monitors the body’s needs and responds accordingly. Without selective signaling, ECS pathways would activate constantly and unpredictably, disrupting balance rather than maintaining it.

What Is Selective Signaling and Why Does It Matter?

Selective signaling is the ability of the ECS to target specific pathways or receptors at just the right time and place [2]. It’s like a light switch, when you need light in a room, you flip the switch, and it turns on just the right amount. If the light was on everywhere, all at once, it would be too bright and overwhelming. 

The ECS uses selective signaling to activate only the receptors that are needed in specific tissues, reducing unnecessary effects elsewhere. This process is achieved through factors such as GPCR signalling and β-Arrestin Pathway.

The GPCR Pathway in the Endocannabinoid System (ECS)

CB1 and CB2 receptors are G-protein coupled receptors (GPCRs) that help control pain, mood, and immune function [3]. When endocannabinoids like  AEA or 2-AG attach to these receptors, they start a Gi/o protein signaling process, which leads to:

  • Blocking adenylate cyclase (AC) → Lowers cAMP levels, slowing down cell activity.
  • Reducing protein kinase A (PKA) activation → Less activation of other proteins inside the cell.
  • Decreasing neurotransmitter release → Less glutamate, GABA, and dopamine are sent between brain cells.

But the effects of CB1 and CB2 activation depend on where they are located in the body. CB1 receptors in the brain and nervous system help regulate pain, mood, and neurotransmission by:

  • Reducing glutamate & GABA release, altering pain perception and mood (leading to relaxation or, in some cases, anxiety).[4]
  • Changing dopamine levels, which influences pleasure, motivation, and addiction.
  • Activating potassium (K+) channels and blocking calcium (Ca2+) channels, making neurons less excitable—reducing pain but sometimes causing drowsiness.

Meanwhile, CB2 receptors in the immune system and peripheral tissues focus on reducing inflammation and regulating immune responses by:

  • Lowering immune cell activity, which helps control inflammation.
  • Modulating chronic pain and autoimmune diseases, calming an overactive immune system.

Therefore, selective signaling is essential for ensuring these processes remain controlled and beneficial. Without it, the ECS could become overactive or unbalanced. 

β-Arrestin Pathway and CB1 Receptor Regulation

When CB1 receptors are overstimulated, such as with excessive THC use, the β-arrestin pathway helps regulate their activity to prevent overstimulation and build-up of tolerance. [5] This happens through three key processes:

  1. Desensitization: β-arrestin attaches to the CB1 receptor, blocking it from sending signals and reducing its activity.
  2. Internalization: The receptor is pulled inside the cell through clathrin-coated vesicles, making it temporarily inactive.
  3. Downregulation and Tolerance: If CB1 receptors stay internalized for too long, they may be broken down instead of recycled, leading to fewer receptors available for activation. This makes the body less responsive to THC over time, requiring higher doses to achieve the same effects.


Figure 1: Shows how GPCR signaling is regulated and how β-arrestin affects CB1 receptors. (a) When an agonist binds, GPCRs activate G proteins. (b) GRK phosphorylates the receptor, allowing β-arrestin to bind and stop signaling (desensitization). (c) β-arrestin helps remove the receptor through clathrin-coated vesicles for recycling or breakdown. With too much THC, CB1 receptors may be broken down instead of reused, reducing their numbers and leading to tolerance. [6]

But, if CB1 receptors are overstimulated too often, this desensitization can lead to tolerance, meaning the user will need more of the substance to experience the same effect. Over time, this could increase the risk of dependence. Since excessive β-arrestin activation plays an important role in this process, researchers are looking for ways to fine-tune CB1 receptor signaling.

This is where ligand bias comes in. Instead of activating all pathways equally, ligand bias allows for more accurate control, favoring beneficial signaling while minimizing unwanted effects.

Ligand Bias: Activating the Right Pathway

CB1 receptors don’t always respond the same way to different molecules. Some activate the G-protein pathway, which helps with pain relief, while others activate the β-arrestin pathway as shown in figure 2, which can lead to tolerance and side effects.

  • G-protein signaling → Helps with pain & inflammation
  • β-arrestin pathway → Leads to tolerance & side effects

This idea, called ligand bias (biased agonism), is helping scientists develop better treatments. By creating drugs that only activate the helpful G-protein pathway while avoiding too much β-arrestin activation, we can improve pain relief and neuroprotection without causing tolerance or unwanted effects.


Figure 2. Ligand-Biased Signaling in CB1 and CB2 Receptors. This figure shows ligand-biased signaling in CB1 and CB2 receptors, where ligands primarily activate either the G protein pathway or the β-arrestin pathway, leading to different cellular responses. Antagonists block both pathways by preventing ligand binding.[7]

Selective Signaling in Neurodegenerative Diseases 

Neurodegenerative diseases like multiple sclerosis (MS), Huntington’s disease (HD), and Alzheimer’s disease (AD)are linked to problems in the endocannabinoid system (ECS). The CB1 receptor plays an important role in protecting the brain, but if it is overused, the body can build tolerance, making treatments less effective.

Multiple Sclerosis (MS)

✔ CB1 activation helps reduce brain inflammation, muscle stiffness, and pain.
✔ Sativex (a THC-CBD spray) improves movement and reduces symptoms in MS patients.[8]
✔ According to the paper “Cannabinoid Receptors in the Central Nervous System: Their Signaling and Roles in Disease” studies on CB1-deficient mice show increased brain damage, proving that CB1 protects neurons. [9]

Huntington’s Disease (HD)

✔ CB1 receptors start disappearing early in the disease, even before symptoms appear.
✔ Losing CB1 receptors makes movement problems worse and leads to faster brain cell damage.
✔ CB1 activation increases BDNF (Brain-Derived Neurotrophic Factor), which helps protect brain cells and supports neuron survival. [9]

Alzheimer’s Disease (AD) 

✔ CB1 activation helps clear harmful β-amyloid buildup, which is linked to memory loss in AD.
✔ Mice without CB1 receptors have worse memory problems, showing that CB1 is needed for brain function.
✔ CBD helps protect the brain by reducing tau buildup, which contributes to neuron damage. [9]

In diseases like MS, HD, and AD, selective CB1 activation can help protect brain cells and improve symptoms. However, too much activation can lead to tolerance, making treatments less effective over time.Therefore, Selective signaling plays an important role in ensuring that CB1 receptors are activated only when and where needed, allowing scientists to develop more effective therapies with fewer side effects.

What Happens When Selective Signaling Fails?

Without selective signaling, the body could suffer from several issues:

  • Increased side effects: If CB1 receptors are overstimulated in the brain, it could lead to memory loss, confusion, and impaired motor function. Overstimulation of CB2 receptors might suppress the immune system too much, making us vulnerable to infections.
  • Uncontrolled pain and inflammation: Without selective signaling, the body could either feel too much pain in some areas or too little in others. Pain relief might not be targeted properly, leaving some areas of the body still suffering.
  • Tolerance and receptor burnout: With constant activation of CB1 receptors, they can become desensitized. This means a person may need higher doses to achieve the same effect, which increases the risk of addiction and worsens the overall response.
  • Mood disruptions: Overactivation of the ECS could lead to psychological side effects like anxiety, paranoia, or depression.

Therefore, without selective signaling, the ECS would lose its ability to finely tune processes, and the body would suffer from overactivation or underactivation of critical pathways.

The Bottom Line: Balance Is Key

And the endocannabinoid system (ECS) plays a vital role in regulating key functions like pain, mood, and immune response. It’s like the body’s internal balancing act, making sure everything runs smoothly. But here’s the catch: when the ECS doesn’t work properly, it can lead to serious issues, from chronic pain to mood swings and even disorders like anxiety or inflammation. Without proper regulation, things can get out of control fast!

Therefore, by understanding how selective signaling works in the ECS, we can create better treatments that target exactly what’s needed without all the side effects. So, why should you care? Because the ECS affects everyone! Whether you’re dealing with pain, stress, or just want to stay healthy as you age, learning more about how the ECS works could lead to safer, smarter treatments for a better life. And who wouldn’t want that?







 

 

 

 

 

 

Footnotes

[1] Lu, H. C., & Mackie, K. (2016). An Introduction to the Endogenous Cannabinoid System. Biological psychiatry, 79(7), 516–525. https://doi.org/10.1016/j.biopsych.2015.07.028

[2] Bosier, B., Muccioli, G. G., Hermans, E., & Lambert, D. M. (2010). Functionally selective cannabinoid receptor signalling: therapeutic implications and opportunities. Biochemical pharmacology, 80(1), 1–12. https://doi.org/10.1016/j.bcp.2010.02.013

[3] Howlett, A. C., & Abood, M. E. (2017). CB1 and CB2 Receptor Pharmacology. Advances in pharmacology (San Diego, Calif.), 80, 169–206. https://doi.org/10.1016/bs.apha.2017.03.007

[4] Patel, S., & Hillard, C. J. (2009). Role of endocannabinoid signaling in anxiety and depression. Current topics in behavioral neurosciences, 1, 347–371. https://doi.org/10.1007/978-3-540-88955-7_14

[5] Nguyen, P. T., Schmid, C. L., Raehal, K. M., Selley, D. E., Bohn, L. M., & Sim-Selley, L. J. (2012). β-arrestin2 regulates cannabinoid CB1 receptor signaling and adaptation in a central nervous system region-dependent manner. Biological psychiatry, 71(8), 714–724. https://doi.org/10.1016/j.biopsych.2011.11.027

[6] Whalen, E. J., Rajagopal, S., & Lefkowitz, R. J. (2011). Therapeutic potential of β-arrestin- and G protein-biased agonists. Trends in Molecular Medicine, 17(3), 126-139. https://doi.org/10.1016/j.molmed.2010.11.004

[7] Ye, L., Cao, Z., Wang, W., & Zhou, N. (2019). New Insights in Cannabinoid Receptor Structure and Signaling. Current Molecular Pharmacology, 12(3), 239–248. https://doi.org/10.2174/1874467212666190215112036

[8] Russo, M., Calabrò, R. S., Naro, A., Sessa, E., Rifici, C., D’Aleo, G., Leo, A., De Luca, R., Quartarone, A., & Bramanti, P. (2015). Sativex in the management of multiple sclerosis-related spasticity: role of the corticospinal modulation. Neural plasticity, 2015, 656582. https://doi.org/10.1155/2015/656582

[9] Kendall, D. A., & Yudowski, G. A. (2017). Cannabinoid Receptors in the Central Nervous System: Their Signaling and Roles in Disease. Frontiers in Cellular Neuroscience, 10(294). https://doi.org/10.3389/fncel.2016.00294



 

CB1 Receptor Trafficking in Cannabinoid Signaling in the CNS: Why It Matters

The human brain is a sophisticated network of impulses that continually adapts to its surroundings.  Among its various functions, the endocannabinoid system (ECS) regulates mood, memory, and neurological health.  But what happens if the system is disrupted?  More significantly, how does the trafficking of CB1 receptors within our cells affect neurological disorders?  Kendall and Yudowski (2017)[1] investigate the complicated mechanics of CB1 receptor signaling, offer insight on how these receptors travel throughout neurons, and discuss the potential for illness treatment.

Understanding the CB1 Receptor

CB1 receptors are a type of G protein-coupled receptor (GPCR) that is widely distributed throughout the central nervous system.  They are triggered by cannabinoids, which include both endogenous substances like anandamide and plant-derived molecules like tetrahydrocannabinol.  When activated, CB1 receptors alter neurotransmitter release, which influences mood, cognition, and pain perception.  However, these receptors do not remain stationary within cells; instead, they move about in a process known as receptor trafficking (Figure 1)

Figure adapted from Kendall and Yudowski, 2017[1]

www.frontiersin.org

Figure 1. Differential cannabinoid (CB) receptor signaling modalities can impact neuromodulation in health and disease in specific ways. (A)Endogenous ligands arachidonylethanolamine (AEA) and 2-arachidonylglycerol (2-AG) are produced by key enzymes, including diacylglycerol lipase(DGLα) and phospholipase D.  These activate the CB1 receptor in the central nervous system (CNS).  The outcome may include modification of adenylate cyclase activity to decrease cAMP buildup, voltage-gated calcium channels (VGCC), K+ channels, and neurotransmitter release in presynaptic excitatory and inhibitory synapses. After ligand binding activates the CB1 receptor, signaling through G protein and/or β-arrestin might occur at the plasma membrane, endocytic pits, or endosomes.  G proteins typically bind unphosphorylated receptors, whereas β-arrestin binds phosphorylated receptors via G protein receptor kinases[1].

CB1 Receptor Trafficking: A Key to Understanding Disease

When CB1 receptors are activated, they can undergo endocytosis, which is the process by which they move from the cell surface to intracellular compartments.  This movement is critical for controlling the strength and duration of cannabinoid signaling.  CB1 receptor trafficking is governed by interactions with proteins such β-arrestins, which can either encourage receptor recycling back to the surface or target receptors for destruction [2].

Why does this matter? Research suggests that dysregulation in CB1 receptor trafficking is linked to several neurological disorders, including:

  • Alzheimer’s Disease (AD): Impaired CB1 receptor signaling may contribute to memory deficits, as CB1 receptors are involved in synaptic plasticity[3].
  • Multiple Sclerosis (MS): Modulating CB1 receptor activity has been explored as a therapeutic approach for reducing neuroinflammation and spasticity[4].
  • Huntington’s Disease (HD): A decline in CB1 receptor expression correlates with disease progression, suggesting that preserving receptor function could be neuroprotective[5].

    Therapeutic Implications: The Future of CB1 Receptor Research

  • The ability to modulate CB1 receptor trafficking presents exciting therapeutic possibilities. By designing drugs that influence receptor movement and signaling bias, researchers aim to create targeted treatments with fewer side effects. For instance, ‘biased ligands’—molecules that preferentially activate either G-protein or β-arrestin pathways—could lead to more selective therapeutic outcomes (Figure 2).

    Figure 2. Adapted from Hua et al., “Crystal structure of the human cannabinoid

  • 1-s2.0-S009286741631385X-gr2.jpg

    Figure 2. Synthesis and Characterization of AM6538

    (A) Synthetic processes for AM6538[6].
    (B) Saturation [3H]-CP55,940 binding assays in the absence (control) or presence of rimonabant (100 nM) or AM6538 (50 nM) show that both antagonists cause displacement of the radioligand’s specific binding when present concurrently in the 1 hr binding assay[6].

    (C) Membranes were pretreated with buffer (none), rimonabant (100 nM), or AM6538 (50 nM) at 37°C for 6 hours. Membranes were rinsed with buffer 3× before [3H]-CP55,940 binding as described in (B)[6].
    (D) The percentage of residual binding (Bmax) was determined using the conditions specified in (B) (concurrent) and (C) (pretreat and wash).  When both antagonists were incubated together during the 1 hour binding assay, they reduced [3H]-CP55,940 binding by approximately 30%.  Rimonabant has no effect on future radioligand binding under pretreatment or washout conditions, but AM6538 competes even after membrane washing[6].

    Moreover, understanding the structure of CB1 receptors at the molecular level, as revealed in recent crystallography studies[6], opens doors to designing precise drugs that either enhance or suppress specific receptor functions

    Why Should the Public Care?

    The public’s interest in the ECS is expanding as cannabis-based medicines become more common.  However, without a more sophisticated knowledge of CB1 receptor dynamics, the effects of cannabinoids risk being oversimplified.  What’s the takeaway?  While cannabinoids show promise, their effects are heavily dependent on receptor signaling and trafficking.  Future treatments must account for these complications in order to realize the full potential of cannabinoid-based therapy.

     As research progresses, we get closer to understanding the entire therapeutic potential of CB1 receptor regulation.  This could open the path for new medicines that benefit millions of people suffering from neurological illnesses.

    References:

    1. Kendall DA, Yudowski GA. “Cannabinoid Receptors in the Central Nervous System: Their Signaling and Roles in Disease.” Front. Cell. Neurosci. 2017;10:294. DOI:10.3389/fncel.2016.00294
    2. Delgado-Peraza et al., “Mechanisms of biased β-arrestin-mediated signaling downstream from the cannabinoid 1 receptor.” Molecular Pharmacology. 2016;89(6):618-629. DOI: 10.1124/mol.115.103176.
    3. Mulder et al., “Molecular reorganization of endocannabinoid signalling in Alzheimer’s disease.” Brain. 2011;134(4):1041-1060. DOI:10.1093/brain/awr046.
    4. Pryce et al., “Cannabinoids inhibit neurodegeneration in models of multiple sclerosis.” Brain. 2003;126(10):2191-2202. DOI: 10.1093/brain/awg224.
    5. Mievis et al., “Worsening of Huntington disease phenotype in CB1 receptor knockout mice.” Neurobiology of Disease. 2011;42(3):524-529. DOI: 10.1016/j.nbd.2011.03.006.
    6. Hua et al., “Crystal structure of the human cannabinoid receptor CB1.” Cell. 2016;167(3):750-762.e14. DOI: 10.1016/j.cell.2016.10.004.

Endocannabinoid

The article we have covered in a previous week, “Cannabinoid Receptors in the Central
Nervous System: Their Signaling
and Roles in Disease” by Debra A. Kendall
and Guillermo A. Yudowski was an article about the purpose of Cannabinoid Receptors in the Central Nervous System. Basically, it can be summarized as the following; the endocannabinoid system has been connected to various essential roles in the synaptic ability to neurologically adapt and maintaining proper homeostasis in the brain. However, must those processes fail a myriad of things can go wrong to cause a tragedies in the subject.The topic today is why people should care about this topic and what the people must know, so without further ado let’s get to reading and clarifying!

The article informs us that certain kinds of unfortunate scenarios involving the endocannabinoid system are connected to various neurological diseases such as, Multiple Sclerosis, Huntington’s, Type Three Diabetes (Alzheimer’s Disease), and brain trauma.^1 Though, the article unfortunately leaves these tragic consequences at the article. As a result of these connection, we can now understand the essential nature of this information before us.

Figure one from the article mentioned is specifically excellent at explaining the essential nature of endocannabinoids. That piece is a diagram which explains the roles through the context of the membrane of neurons. Although, it could feel a tad bit overwhelming if you’re unfamiliar with the professional acronyms mentioned in figure one from the Neuroscience field. This was an excellent piece to me for it is maximized simplicity because, for clear reasons, that kind of thing strongly helps at a mere glance. The figure may also benefit people uninvolved in Neuroscience as well because figure one works similarly with the textbooks we grew up with in school with all the brief, yet descriptive, labeling and artistic style, and I find all of that effective myself in general because it’s easy on the eyes to track and logicate.

Now, at this point, one, such as yourself, may wonder why people really should care about all the above information. Well, let’s answer with essential basics to answer ourselves by quickly asking ourselves something simpler first; what really is so bad about what I considered consequences during a dysfunctional cannabinoid system? Well, Alzheimer’s disease for example is not very nice at all and looks even worse when examined scientifically. According to the NIH, Alzheimer’s disease has a wide spectrum of symptoms from memory loss and brain literal deterioration. Considering that we know the brain to be the most vital organ of all in the body, it’s no shock that such a scenario is serious and even severe. The severity of both mentioned examples of the consequences of Alzheimer’s disease in the last paragraph is clear and concerning when thought of this way in the clinical lens rather than “just another syndrome/disease” I personally notice in the general public awareness. I recommend reading the articles yourself to truly understand as I can only summarize so much of what we know!

Footnotes:
1: “Cannabinoid Receptors in the Central
Nervous System: Their Signaling
and Roles in Disease” by Debra A. Kendall
and Guillermo A. Yudowski

2: https://www.nia.nih.gov/health/alzheimers-causes-and-risk-factors/what-happens-brain-alzheimers-disease

From Treatment to Tolerance: Understanding Marijuana’s Long-Term Effects

Cannabis

Cannabis is a generic term used for the variety of products derived from the cannabis sativa plant. Cannabis goes by many names including marijuana which describes parts of the plant high in THC[1]. Delta 9 tetrahydrocannabinol (THC)  and Cannabidiol (CBD) are the most investigated[1]. Marijuana is one of the most used drugs in the United States, particularly among individuals aged 18-25.[1] In 2021, 35.4% of people aged 18 to 25 reported using marijuana in the past year[1]. The drug is surrounded by a significant amount of controversy and stigma as it is slowly being legalized for recreational use across the nation.

It is important to note that not all uses of marijuana are recreational. Before the drug was legalized for recreational use, it was legalized for medical use. Cannabis has been used in medicine to treat chronic pain, opioid withdrawals, seizures, multiple sclerosis, and decreased appetite[2].  The two main cannabis-derived medications available are Cannabidiol and Dronabinol[2].

Epidiolex

Epidolex (cannabiniol) is used in the treatment of seizures associated with Lennox-Gastaut syndrome or Dravet syndrome. The medication is approved for individuals 2 years and older. it contains a purified form of CBD derived from marijuana without any THC.  Epidiolex is administered orally with strawberry flavoring. Common side effects include drowsiness, diarrhea, decreased appetite, lack of energy, sleep problems, increased liver enzymes, and infections

Dronabinol

Dronabinol is commonly used to treat weight loss in individuals with AIDs. It is also used to treat nausea and vomiting associated with chemotherapy. Dronabinol is a synthetic form of THC. The medication can be taken as a pill ranging from 2.5-5mg of THC per dose. It can also be administered orally as a spray with a 2.7 mg/100ul dose. Common side effects include dizziness, euphoria, nausea, vomiting, stomach pain, paranoia, sleepiness, and abnormal thinking[4].

Cannabis use disorder

Cannabis use disorder is a mental health condition where cannabis use causes distress or impairments in day-to-day tasks and functioning. In more severe cases the diagnosis can be classified as cannabis addiction [5].

Symptoms

  • Strong urge to consume cannabis
  • Unsuccessful attempts to limit cannabis use
  • Disruptions in social, occupational, or recreational activities because of cannabis use
  • Developing a tolerance
  • Confusion
  • Delusions
  • Memory issues

Forceps minor | Radiology Reference Article | Radiopaedia.org
Figure 3 Forceps minor [6]
Figure 4 orbitofrontal cortex  [7]
 

 

 

 

 

 

 

Long term effects

In the study Long-term effects of marijuana use on the brain, researchers found that long-term marijuana use had significant impacts on cognition and brain anatomy. Cannabis users exhibited significantly lower gray matter volume in the right and left middle orbitofrontal cortex (Figure 3) than controls. The marijuana groups also displayed increased white matter growth in the Forceps minor (Figure 4). Marijuana users also displayed significantly lower IQ scores compared to the control group.

 

[1] WHO. (2024, October 24). Cannabis. World Health Organization. https://www.who.int/teams/mental-health-and-substance-use/alcohol-drugs-and-addictive-behaviours/drugs-psychoactive/cannabis

[2] Cleveland Clinic medical. (2025, February 21). Marijuana. Cleveland Clinic. https://my.clevelandclinic.org/health/articles/4392-marijuana-cannabis

[3] Abu-Sawwa, R., & Stehling, C. (2020). Epidiolex (Cannabidiol) Primer: Frequently Asked Questions for Patients and Caregivers. The Journal of Pediatric Pharmacology and Therapeutics : JPPT, 25(1), 75. https://doi.org/10.5863/1551-6776-25.1.75

[4] Drugbank. (2025, February 16). Dronabinol: Uses, interactions, mechanism of action | drugbank online. https://go.drugbank.com/drugs/DB00470

[5] Cleveland Clinic medical. (2025a, February 21). Cannabis use disorder. Cleveland Clinic. https://my.clevelandclinic.org/health/diseases/cannabis-use-disorder

[6] Filbey, F. M., Aslan, S., Calhoun, V. D., Spence, J. S., Damaraju, E., Caprihan, A., & Segall, J. (2014). Long-term effects of marijuana use on the brain. Proceedings of the National Academy of Sciences of the United States of America111(47), 16913–16918. https://doi.org/10.1073/pnas.1415297111

[7] https://en.wikipedia.org/wiki/Orbitofrontal_cortex

[8]Gaillard F, Murphy A, Hacking C, et al. Forceps minor. Reference article, Radiopaedia.org (Accessed on 25 Feb 2025) https://doi.org/10.53347/rID-4705

 

Spam prevention powered by Akismet