Review Article

Modulation of Excitatory and Inhibitory Systems in Autism Spectrum Disorder: The Role of Cannabinoids

Stefano Marini1,*, Lucia D’Agostino1, Marika Mentana2, Carla Ciamarra1 and Alessandro Gentile1

1National Health Service, Department of Mental Health, Termoli, Italy
2Optimus Social Cooperative, Milano, Italy

Received Date: 24/05/2023; Published Date: 20/09/2023

*Corresponding author: Stefano Marini, National Health Service, Department of Mental Health, Termoli, Italy

DOI: 10.46998/IJCMCR.2023.30.000730

Abstract

Autism Spectrum Disorder includes a group of developmental disabilities characterized by patterns of delay and deviance in the development of social, communicative, cognitive skills and the presence of repetitive and stereotyped behaviors as well as restricted interests. Although the etiopathogenesis of autism has not yet been elucidated, past literature has highlighted an imbalance between glutamatergic and Gamma-Aminobutyric Acid (GABA)-ergic neurotransmission. A cortical deficiency of GABA in young people with ASD has been reported. Endocannabinoids act in numerous synapses of the central nervous system, maintaining adequate synaptic homeostasis, preventing excess stimulation at the level of excitatory or inhibitory synapses. The endocannabinoid system appears to play an important role in some clinical presentations of autism, such as socialization. Indeed, Autism Spectrum Disorder seems to be characterized by a hypo-functionality of the endocannabinoid system.

The present work aims to describe the current state of the art regarding the possible role of cannabinoids in the modulation of the excitatory and inhibitory systems in individuals with ASD.

A literature search was conducted for relevant studies on PubMed database, concerning the randomized clinical trials on the modulating effect of excitatory and inhibitory cannabinoid systems in autism.

Three eligible articles were found according to the purpose of the present study. The results of the three articles considered highlighted a cannabinoid-related increase in glutamate in subcortical regions and a decrease in cortical regions, both in subjects with and without Autism. CBD increased gamma-aminobutyric acid transmission in the subcortical regions of neurotypical subjects, while it decreased it in the same areas of the ASD group. Furthermore, Cannabinoid modulated low-frequency activity used as a measure of brain activity and functional connectivity in the brains of adults with autism spectrum disorder.

Data from the three fMRI studies demonstrated that CBD influences cortical and subcortical connectivity in an adult sample. This effect was notable only in the ASD group but not in the controls. However, further studies are needed to confirm the results obtained so far.

Keywords: Autism Spectrum Disorder; Endocannabinoid System; Cannabinoids

Introduction

Autism Spectrum Disorder (ASD) includes a group of developmental disabilities characterized by patterns of delay and deviance in the development of social, communicative, cognitive skills and the presence of repetitive and stereotyped behaviors as well as restricted interests [1]. In addition to core symptoms, people with ASD often have numerous medical and psychiatric comorbidities that worsen the quality of life of patients and their caregivers [2]. Clinical features are accompanied by an atypical sensory experience present in up to approximately 95% of people with ASD [3].

The clinical presentation can vary considerably over the years, depending on the characteristics of the person, the surrounding environment and above all the habilitation/rehabilitation interventions undertaken [4].

Although the etiopathogenesis of autism has not yet been elucidated, the data in the literature agree that the causes of autism are multifactorial, including genetic, epigenetic, inflammatory, immunological and environmental factors [5-10].

More than one study has reported dysfunctional alterations in E/I neurotransmission in cortical neurons, using glutamatergic and dysfunctional GABAergic neurotransmission, and abnormal levels of GABA concentrations in brain tissue or plasma (for extensive reviews see [11-15]). Different mechanisms could be responsible for E/I imbalance, such as alterations in genes encoding glutamatergic receptors or synaptic proteins [16,17]. Particularly, the role of neurexins, neuroligins and SHANK proteins, implicated in the formation and maintenance of excitatory and inhibitory synapses, has been widely established in the pathogenesis of neurodevelopmental disorders [18-22].

Some recent studies have shown abnormalities of the GABAergic and glutamatergic pathways in the prefrontal cortex and Basal Ganglia (BG) regions, now proven to be crucial regions of the core signs and symptoms of ASD [23-26].

Past literature has proven that Endocannabinoid System (or Endogenous Cannabinoid System, ECS) functions as retrograde signal molecules in synapses, especially in glutamatergic and GABAergic synapses, preventing excess excitation or inhibition, respectively [27,28]. Moreover, the ECS can enhance GABAergic transmission and reduce glutamate transmission in different brain regions [29,30].

The ECS is a complex biological system implicated within the pharmacological effects of cannabis [31], neuronal plasticity [32,33], postnatal development [34], pain sensation [35], emotionality [36], appetite [37], learning and memory [38]. It is also concerned with the homeostasis of the organism [39] through the modulation of more than one system such as the cardiovascular (CVS), central nervous (CNS), peripheral nervous (SNP), endocrine, reproductive, immune and digestive systems [40,41].

The ECS includes three major components: endogenous cannabinoids (or endocannabinoids, eCBs), metabotropic receptors and enzymes answerable for the synthesis and degradation of eCBs [42].

The most studied eCBs are N-arachidonylethanolamine (or anandamide, AEA) [43] and 2-arachidonylglycerol (2-AG) [44]. Cannabis contains more than 500 compounds and the most abundant are represented by ∆-9-tetrahydrocannabinol (THC), cannabidiol (CBD), flavonoids and terpenes [45]. Cannabidiol (CBD) and delta-9-tetrahydrocannabinol (Δ9-THC) are the most intensively studied phytocannabinoids. In addition to eCBs, phytocannabinoids and synthetic derivative compounds represent other substances that act as ligands for ECS receptors.

THC has a similar affinity for both CB1 and CB2 receptors, but most of the psychoactive effects of THC are related to the activation of CB1 receptors [46]. Cannabidiol (CBD) does not have psychotropic properties, as recently confirmed [47]. CBD has a very low affinity for CB1 and CB2 receptors, and several results have suggested that CBD operates as a negative allosteric modulator/inverse agonist in both CB1 and CB2 receptors [48-50].

There are other potentially therapeutic phytocannabinoids, which have been tested in pre-clinical studies, but not yet extensively in vivo and are represented by: ∆8-Tetrahydrocannabinol [51], Cannabinol [52], Cannabigerol [53], Cannabichromene [54], ∆9-Tetrahydrocannabivarin (∆ 9-THCV) [55] and Cannabidivarin (CBDV) [56].

The two main receptors of the ECS are the cannabinoid receptor type 1 (CB1) and the cannabinoid receptor type 2 (CB2). The CB1 is expressed in both the peripheral and the central nervous systems such as in the hippocampus, cortical regions, basal ganglia (BG) and cerebellum. On the other hand, the CB2 receptor is mostly expressed in peripheral cells and tissues of the immune system. Although the presence of CB2 in the brain is very low compared to CB1, CB2 appears to play a crucial role in macrophage/microglia functions [57]. Indeed, CB2 expression drastically increases in activated microglia and CB2 activation decreases the production of pro-inflammatory molecules [58]. Both of these receptors are coupled to the G protein signaling pathway [59].

The ECS appears to play an important role in some clinical presentations of autism as well as in the regulation of emotional responses, behavioral reactivity, reciprocity, social play and social interaction [60]. Indeed, numerous murine studies have shown that the ECS plays a crucial role in the pathophysiological mechanisms underlying ASD [61-63]. Human studies support growing evidence of ECS abnormalities in people with ASD [64-67].

Currently, only risperidone and aripiprazole are approved drugs for the treatment of irritability associated with ASD [68]. Due to the high incidence of adverse effects of conventional psychotropic therapies [69], the use of phytocannabinoids could become an alternative therapeutic strategy in these situations [70].

In the literature, there is preclinical evidence to support the positive effects of the use of cannabinoids in balancing the pathological mechanisms of ASD [71-74] and ASD-like symptoms [75-78].

Cannabinoids are already being used for a wide range of conditions such as multiple sclerosis, Tourette's syndrome, Parkinson's disease, epilepsy, glaucoma, nausea and pain [79], but clinical data on the use of cannabinoids in people with autism are still very limited.

Materials and Methods

The present work aims to describe the current state of the art regarding the possible role of cannabinoids in the modulation of the excitatory and inhibitory systems in individuals with ASD.

A literature search was conducted for relevant studies using PubMed database concerning the randomized clinical trials using functional Magnetic Resonance Imaging (fMRI) or Magnetic Resonance Spectroscopy (MRS) on the modulating effect of cannabinoids on the excitatory and inhibitory systems in people with autism.

Results and Discussion

Three eligible papers [80-82] were found according to the purpose of the present study.

Over the years, technological progress has made it possible to measure the cerebral effects of psychotropic substances. fMRI has already been used in people with ASD, to study the brain effects of riluzole, propranolol and oxytocin during cognitive tasks [23,83,84]. MRS has been already used to determine alteration in E/I dynamics in BG and dorsomedial prefrontal cortex (DMPFC) [24].

All three studies used a randomized double-blind, cross-over design. Two studies [80,81] acquired data following a single oral dose of 600 mg CBD or a matched placebo while one study utilized CBDV [82] (see Table 1).

Two studies compared MRS measures of glutamate and GABA [81] and Glx (glutamate + glutamine) and GABA+ (GABA+ macromolecules) [82] levels in the BG and DMPFC in men with and without ASD.

Considering that differentiating the cognitive response to a task from normal brain activation can be challenging in people with ASD [85,86], one study rightly used a resting state design (task-free) to examine the fractional amplitude of low-frequency fluctuations (fALFF) as a measure of spontaneous regional brain activity [87]. Indeed, low-frequency oscillations appear to be important in synchronizing activity between brain regions [88].

All three studies taken into consideration show how cannabinoids shift both fALFF and FC [80] as well as the levels of excitatory and inhibitory neurotransmitters [81,82] in the living adult human brain both in the ASD group and in the control group.

Two studies [81,82] showed no differences in baseline glutamate and GABA levels in BG and DMPFC as evidenced by some MRS studies [23,24], but not all [89]. However, it should be noted that individual responses in autistic brains varied according to baseline Glx levels.

Changes in fALFF were more prominent in the ASD group and not significant in controls [80]. Furthermore, in ASD the shift in fALFF in the cerebellum was accompanied by diffuse changes in vermal FC with many of its subcortical and cortical targets. As suggested by the authors, CBD appears to ‘tune’ FC in a region or connection-specific manner. These data are supported by previous studies correlating levels of E/I cerebellar and cerebro-cerebellar FC in adolescents and adults with ASD [90].

The most important FC alterations were attributable to the vermis but not to the fusiform probably, as explained by the authors, due to reduced expression of GABA receptors [91], decreased levels of enzymes responsible for converting glutamate to GABA, alterations of inhibitory Purkinje cells [92] and structural alterations in ASD [93,94], although data in the literature are conflicting [95,96].

Given the impairments [85,97] of functional spindle-shaped abnormalities in face processing [98] and the study evidence, one could hypothesize the use of CBD to improve the recognition of faces in ASD.

Regarding Glx, both people with ASD and neurotypicals responded to CBD in an overlapping manner [81]. On the other hand, CBD decreased GABA+ levels in the BG and (markedly) in the DMPFC voxel of autistic adults, probably due to metabolic or genetic alterations of GABA receptors [92,99-101]. Probably these results are explained by the ability of CBD to suppress the activity of prefrontal glutamatergic neurons through the 5-HT1A receptors [102,103]. Furthermore, 5-HT1a receptor dysfunction in ASD is now known [104]. These data support on the one hand the theory of E/I alteration in ASD and on the other the possible use of CBD as a therapeutic approach.

The cortical, sub-cortical, and thalamic connections of the BG appear to be important for the clinical presentation of the features of the autism diagnosis [105-107].

CBDV induced an increase in BG Glx levels (left BG) in both groups. In contrast, CBDV had no impact on Glx in DMPFC, nor on GABA+ levels in either voxel [82]. Only in the ASD group, the drug-induced shift in Glx levels in BG was significantly negatively correlated with baseline Glx value. Moreover, based on the basal levels of Glx, a different response was highlighted in subjects with ASD. Indeed, in autistic individuals with lower baseline Glx levels, CBDV increased Glx levels, while individuals with higher baseline Glx values experienced a decrease in Glx. From the data collected, CBDV seems to normalize the altered levels of Glx.

These effects could be explained by the binding of CBDV to TRP receptors coupled to pyramidal excitatory neurons highly represented in BG as demonstrated by preclinical studies [108,109]. Furthermore, CBDV could modulate excitatory neurotransmission through the activation of microglial TRP receptors, a mechanism already known to increase extracellular vesicular shedding and subsequent glutamate release [110].

In our view, these findings appear important to clinicians in further differentiating subgroups within the autistic disorder spectrum in such a way as to make treatments increasingly individualized.

The studies considered, have some limitations such as the low sample size, the acute use of cannabinoids only, having recruited only male subjects, the stringent inclusion criteria which excluded psychiatric comorbidities often frequent in people with ASD. Although the data are very encouraging, future research is needed to confirm the data exposed on a large scale and on more representative samples of the population of people with ASD.

Table 1: Summary of studies.

Conclusion

Data from the three studies considered in this review demonstrated that CBD and CBDV influence cortical and subcortical connectivity in an adult sample. This effect was notable only in the ASD group but not in the controls. Indeed, data showed how cannabinoids shift both fALFF and Functional Connectivity as well as the levels of excitatory and inhibitory neurotransmitters in the living adult human brain more consistently in the ASD group. Furthermore, these data, in addition to confirming the hypothesis of alteration between excitatory and inhibitory systems, lay the foundations for the use of drugs acting on the GABA system as a treatment target in ASD. However, further studies are needed to confirm the results obtained so far.

Acknowledgments: This paper was entirely funded by the authors, and no pharmaceutical companies were informed of or were involved in the paper. The authors have no potential conflicts of interest that are directly relevant to the contents of the paper.

References

  1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5 Edition, VA, Washington. Arlington, 2013.
  2. Lord C, Elsabbagh M, Baird G, Veenstra-Vanderweele J. “Autism Spectrum Disorder,” Lancet, 2018; 392(10146): pp. 508-520.
  3. Chung S, Son JW. “Visual Perception in Autism Spectrum Disorder: A Review of Neuroimaging Studies,” Journal of the Korean Acadademy of Child and Adolescent Psychiatry, 2020; 31(3): pp. 105-120.
  4. Mukherjee SB. “Autism Spectrum Disorders—Diagnosis and Management,” Indian Journal of Pediatrics, 2017; 84(4): pp. 307-314.
  5. Tonhajzerova I, Ondrejka I, Mestanik M, Mikolka P, Hrtanek I, Mestanikova A, et al. “Inflammatory Activity in Autism Spectrum Disorder,” Advances in Experimental in Medicine and Biology, 2015; 861: pp. 93-98.
  6. Onore C, Careaga M, Ashwood P. “The role of immune dysfunction in the pathophysiology of autism,” Brain, Behavior, and Immunity, 2012; 26(3): pp. 383-392.
  7. Sanders SJ, He X, Willsey AJ, Ercan-Sencicek AG, Samocha KE, Cicek AE, et al. “Insights into Autism Spectrum Disorder Genomic Architecture and Biology from 71 Risk Loci,” Neuron, 2015; 87(6): pp. 1215-1233.
  8. Willsey AJ, State MW. “Autism spectrum disorders: from genes to neurobiology,” Current Opinion in Neurobiology, 2015; 30: pp. 92-99.
  9. Forsberg SL, Ilieva M, Maria Michel T. “Epigenetics and cerebral organoids: promising directions in autism spectrum disorders,” Translational Psychiatry, 2018; 8(1): p. 14.
  10. Bölte S, Girdler S, Marschik PB. “The contribution of environmental exposure to the etiology of autism spectrum disorder,” Cellular and Molecula Life Sciences, 2019; 76(7): p. 1275-12 97.
  11. Carlson GC. “Glutamate receptor dysfunction and drug targets across models of autism spectrum disorders,” Pharmacol, Biochemistry, and Behavior, 2012; 100(4): pp. 850-854.
  12. Spooren W, Lindemann L, Ghosh A, Santarelli L. “Synapse dysfunction in autism: a molecular medicine approach to drug discovery in neurodevelopmental disorders,” Trends in Pharmacological Sciences, 2012; 33(12): pp. 669-684.
  13. Gao R, Penzes P. “Common mechanisms of excitatory and inhibitory imbalance in schizophrenia and autism spectrum disorders,” Current Molecular Medicine, 2015; 15(2): pp. 146-167.
  14. Dickinson A, Jones M, Milne E. “Measuring neural excitation and inhibition in autism: different approaches, different findings and different interpretations,” Brain Research, 2016; 1648(Pt A): pp. 277-289.
  15. Goel A, Portera-Cailliau C. “Autism in the balance: elevated E-I ratio as a homeostatic stabilization of synaptic drive,” Neuron, 2019; 101(4): pp. 543-545.
  16. Lee E, Lee J, Kim E. “Excitation/inhibition imbalance in animal models of autism spectrum disorders,” Biological Psychiatry, 2017; 81(10): pp. 838-847.
  17. Culotta L, Penzes P. “Exploring the mechanisms underlying excitation/inhibition imbalance in human iPSC-derived models of ASD,” Molecular Autism, 2020; 11(1): p 32.
  18. Trobiani L, Meringolo M, Diamanti T, Bourne Y, Marchot P, Martella G, et al. “The neuroligins and the synaptic pathway in Autism Spectrum Disorder,” Neuroscience and Biobehavoral Reviews, 2020; 119: pp. 37-51.
  19. Kasem E, Kurihara T, Tabuchi K. “Neurexins and neuropsychiatric disorders,” Neuroscience Research, 2018; 127: pp. 53-60.
  20. Port RG, Oberman LM, Roberts TP. “Revisiting the excitation/inhibition imbalance hypothesis of ASD through a clinical lens,” Br J Radiol, 2019; 92(1101).
  21. Wang J, Gong J, Li L, Chen Y, Li L, Gu H, et al. “Neurexin gene family variants as risk factors for autism spectrum disorder,” Autism Research, 2018; 11(1): pp. 37-43.
  22. Howell BW, Smith KM. “Synaptic structural protein dysfunction leads to altered excitation inhibition ratios in models of autism spectrum disorder,” Pharmacologial Research, 2019; 139: 207-214.
  23. Ajram LA, Horder J, Mendez MA, Galanopoulos A, Brennan LP, Wichers RH, et al. “Shifting brain inhibitory balance and connectivity of the prefrontal cortex of adults with autism spectrum disorder,” Translational Psychiatry, 2017; 7(5): p. 1137.
  24. Horder J, Lavender T, Mendez MA, O’Gorman R, Daly E, Craig MC, et al. “Reduced subcortical glutamate/glutamine in adults with autism spectrum disorders: a [(1) HMRS study,” Translational Psychiatry, 2013; 3(7): e279.
  25. Cochran DM, Sikoglu EM, Hodge SM, Edden RA, Foley A, Kennedy DN, et al. “Relationship among glutamine, gamma-aminobutyric acid, and social cognition in autism spectrum disorders,” Journal of Child and Adolescent Psychopharmacology, 2015; 25(4): pp. 314-322.
  26. Roja DC, Singel D, Steinmetz S, Hepburn S, Brown MS. “Decreased left perisylvian GABA concentration in children with autism and unaffected siblings,” Neuroimage, 2014; 86: pp. 28-34.
  27. Kano M, Ohno-Shosaku T, Hashimotodani Y, Uchigashima M, Watanabe M. “Endocannabinoid-mediated control of synaptic transmission,” Physiological Reviews, 2009; 89(1): pp. 309-380.
  28. Lovinger DM. “Presynaptic modulation by endocannabinoids,” Handbook of Experimental Pharmacology, 2008; 184: pp. 435-477.
  29. Cohen K, Weizman A, Weinstein A. “Modulatory effects of cannabinoids on brain neurotransmission,” The European Journal of Neuroscience, 2019; 50(3): pp. 2322-2345.
  30. Bakas T, van Nieuwenhuijzen PS, Devenish SO, McGregor IS, Arnold JC, Chebib M. “The direct actions of cannabidiol and 2-arachidonoyl glycerol at GABAA receptors,” Pharmacological Research, 2017; 119: pp. 358-370.
  31. Wu J. “Cannabis, cannabinoid receptors, and endocannabinoid system: Yesterday, today, and tomorrow,” Acta Pharmacologica Sinica, 2019; 40(3): pp. 297-299.
  32. Augustin SM, Lovinger DM. “Functional relevance of endocannabinoid-dependent synaptic plasticity in the central nervous system,” ACS Chemical Neuroscience, 2018; 9(9): 2146-2161.
  33. Castillo PE, Younts TJ, Chávez AE, Hashimotodani Y, “Endocannabinoid signaling and synaptic function,” Neuron, 2012; 76(1): pp. 70-81.
  34. Fride E. “The endocannabinoid-CB (1) receptor system in pre- and postnatal life,” European Journal of Pharmacology, 2004; 500: pp. 289-297.
  35. Donvito G, Nass SR, Wilkerson JL, Curry ZA, Schurman LD, Kinsey SG, et al. Lichtman, “The endogenous cannabinoid system: A budding source of targets for treating inflammatory and neuropathic pain,” Neuropsychopharmacology, 2018; 43(1): pp. 52-79.
  36. Campolongo P, Trezza V. “The endocannabinoid system: A key modulator of emotions and cognition,” Frontiers in Behavioral Neuroscience, 2012; 6(73).
  37. Di Marzo V, Goparaju SK, Wang L, Liu J, Bátkai S, Járai Z, et al. “Leptin-regulated endocannabinoids are involved in maintaining food intake,” Nature, 2001; 410(6830): pp. 822-825.
  38. Marsicano G, Lafenetre P. “Roles of the endocannabinoid system in learning and memory,” Current Topics in Behavioral Neurosciences, 2009; 1: pp. 201-230.
  39. Klumpers LE, Thacker DL. “A brief background on cannabis: From plant to medical indications,” Journal of AOAC International, 2019; 102(2): pp. 412-420.
  40. Maroon J, Bost J. “Review of the neurological benefits of phytocannabinoids,” Surgical Neurology International, 2018; 9(91).
  41. Lu D, Potter DE. “Cannabinoids and the Cannabinoid Receptors: An Overview”. In Handbook of Cannabis and Related Pathologies: Biology, Pharmacology, Diagnosis, and Treatment; Elsevier Inc.: Amsterdam, The Netherlands, 2017.
  42. Lu HC, Mackie K. “An Introduction to the Endogenous Cannabinoid System,” Biological Psychiatry, 2016; 79(7): pp. 516-525.
  43. Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, et al. “Isolation and structure of a brain constituent that binds to the cannabinoid receptor,” Science, 1999; 258(5090): pp. 1946-1949.
  44. Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, Itoh K, et al. “2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain,” Biochemical and Biophysical Research Communications, 1995; 215(1): pp. 89-97.
  45. Alves P, Amaral C, Teixeira N, Correia-da-Silva G. “Cannabis sativa: Much more beyond ∆9-tetrahydrocannabinol,” Pharmacological Research, 2020; 157: p. 104822.
  46. Pertwee RG. “The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: Delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin,” British Journal of Pharmacology, 2008; 153(2): pp. 199-215.
  47. Woelfl T, Rohleder C, Mueller JK, Lange B, Reuter A, Schmidt AM, et al. “Effects of Cannabidiol and Delta-9-Tetrahydrocannabinol on Emotion, Cognition, and Attention: A Double-Blind, Placebo-Controlled, Randomized Experimental Trial in Healthy Volunteers,” Frontiers in Psychiatry, 2020; 11: p. 576877.
  48. Chung H, Fierro A, Pessoa-Mahana CD. “Cannabidiol binding and negative allosteric modulation at the cannabinoid type 1 receptor in the presence of delta-9-tetrahydrocannabinol: An in-Silico study,” PLoS One, 2019; 14(7): e0220025.
  49. Laprairie RB, Bagher AM, Kelly MEM, Denovan-Wright EM. “Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor,” British Journal of Pharmacology, 2015; 172(20): pp. 4790-4805.
  50. Tham M, Yilmaz O, Alaverdashvili M, Kelly MEM, Denovan-Wright EM, Laprairie RB. “Allosteric and orthosteric pharmacology of cannabidiol and cannabidiol-dimethylheptyl at the type 1 and type 2 cannabinoid receptors,” British Journal of Pharmacology, 2019; 176(10): pp. 1455-1469.
  51. Thapa D, Cairns EA, Szczesniak AM, Toguri JT, Caldwell MD, Kelly MEM. “The Cannabinoids ∆8THC, CBD, and HU-308 Act via Distinct Receptors to Reduce Corneal Pain and Inflammation,” Cannabis and Cannabinoid Research, 2018; 3(1): pp. 11-20.
  52. Weydt P, Hong S, Witting A, Möller T, Stella N, Kliot M. “Cannabinol delays symptom onset in SOD1 (G93A) transgenic mice without affecting survival,” Amyotrophic Lateral Sclerosis and other Motor Neuron Disorders, 2005; 6(3): pp. 182-184.
  53. Borrelli F, Fasolino I, Romano B, Capasso R, Maiello F, Coppola D, et al. “Beneficial effect of the non-psychotropic plant cannabinoid cannabigerol on experimental inflammatory bowel disease,” Biochemical Pharmacology, 2013; 85(9): pp. 1306-1316.
  54. Udoh M, Santiago M, Devenish S, McGregor IS, Connor M. “Cannabichromene is a cannabinoid CB 2 receptor agonist,” British Journal of Pharmacology, 2019; 176(23): pp. 4537-4547.
  55. Cascio MG, Zamberletti E, Marini P, Parolaro D, Pertwee RG. “The phytocannabinoid, ∆9-tetrahydrocannabivarin, can act through 5-HT1Areceptors to produce antipsychotic effects,” British Journal of Pharmacology, 2015; 172(5): pp. 1305-1318.
  56. Zamberletti E, Rubino T, Parolaro D. “Therapeutic potential of cannabidivarin for epilepsy and autism spectrum disorder,” Pharmacology and Therapeutics, 2021; 226(107878).
  57. Núñez E, Benito C, Pazos MR, Barbachano A, Fajardo O, González S, et al. “Cannabinoid CB2 receptors are expressed by perivascular microglial cells in the human brain: An immunohistochemical study,” Synapse, 2004; 53(4): pp. 208-213.
  58. Komorowska-Müller JA, Schmöle AC. “CB2 Receptor in Microglia: The Guardian of Self-Control,” International Journal of Molecular Sciences, 2020; 22(1): p. 19.
  59. Ho BY, Uezono Y, Takada S, Takase I, Izumi F. “Coupling of the expressed cannabinoid CB1 and CB2 receptors to phospholipase C and G protein-coupled inwardly rectifying K+ channels,” Receptors and Channels, 1999; 6(5): pp. 363-374.
  60. Kerr DM, Downey L, Conboy M, Finn DP, Roche M. “Alterations in the endocannabinoid system in the rat valproic acid model of autism,” Behavioural Brain Research, 2013; 249: pp. 124-132.
  61. Zamberletti E, Gabaglio M, Parolaro D. “The Endocannabinoid System and Autism Spectrum Disorders: Insights from Animal Models,” International Journal of Molecular Sciences, 2017; 18(9): p. 1916.
  62. Pietropaolo S, Bellocchio L, Bouzón-Arnáiz I, Yee BK. “The role of the endocannabinoid system in autism spectrum disorders: Evidence from mouse studies,” Progres in Molecular Biology and Translational Science, 2020; 173: pp.183-208.
  63. Brigida AL, Schultz S, Cascone M, Antonucci N, Siniscalco D. “Endocannabinod Signal Dysregulation in Autism Spectrum Disorders: A Correlation Link between Inflammatory State and Neuro-Immune Alterations,” Internationa Journal of Molecular Sciences, 2017; 18(7): p. 1425.
  64. Zou M, Liu Y, Xie S, Wang L, Li D, Li L, et al. “Alterations of the endocannabinoid system and its therapeutic potential in autism spectrum disorder,” Open Biology, 2021; 11(2): p. 200306.
  65. Siniscalco D, Sapone A, Giordano C, Cirillo A, de Magistris L, Rossi F, et al. “Cannabinoid Receptor Type 2, but not Type 1, is Up-Regulated in Peripheral Blood Mononuclear Cells of Children Affected by Autistic Disorders,” Journal of Autism and Developmental Disorders, 2013; 43(11): pp. 2686-2695.
  66. Karhson DS, Krasinska KM, Dallaire JA, Libove RA, Phillips JM, Chien AS, et al. “Plasma anandamide concentrations are lower in children with autism spectrum disorder,” Molecular Autism, 2018; 9(18).
  67. Aran A, Eylon M, Harel M, Polianski L, Nemirovski A, Tepper S, et al. “Lower circulating endocannabinoid levels in children with autism spectrum disorder,” Moecularl Autism, 2019; 10(2).
  68. Masi A, Demayo MM, Glozier N, Guastella AJ. “An Overview of Autism Spectrum Disorder, Heterogeneity and Treatment Options,” Neuroscience Bulletin, 2017; 33(2): pp. 183-193.
  69. Young N, Findling RL. “An update on pharmacotherapy for autism spectrum disorder in children and adolescents,” Current Opinion in Psychiatry, 2015; 28(2): pp. 91-101.
  70. Poleg S, Golubchik P, Offen D, Weizman A. “Cannabidiol as a suggested candidate for treatment of autism spectrum disorder,” Progress in Neuro-Psychopharmacology and Biological Psychiatry, 2018; 89: pp. 90-96.
  71. Zamberletti E, Gabaglio M, Woolley-Roberts M, Bingham S, Rubino T, Parolaro D. “Cannabidivarin treatment ameliorates autism-like behaviors and restores hippocampal endocannabinoid system and glia alterations induced by prenatal valproic acid exposure in rats,” Frontiers in Cellular Neuroscience, 2019; 13: 367.
  72. De Petrocellis L, Ligresti A, Moriello AS, Allarà M, Bisogno T, Petrosino S, et al. “Effects of cannabinoids and cannabinoid- enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes,” British Journal of Pharmacology, 2011; 163(7): pp. 1479-1494.
  73. Amada N, Yamasaki Y, Williams CM, Whalley BJ. “Cannabidivarin (CBDV) suppresses pentylenetetrazole (PTZ)-induced in- creases in epilepsy-related gene expression,” PeerJ, 2013; 1(e214).
  74. Pagano E, Romano B, Iannotti FA, Parisi OA, D’Armiento M, Pignatiello S, et al. “The non-euphoric phytocannabinoid cannabidivarin counteracts intestinal inflammation in mice and cytokine expression in biopsies from UC pediatric patients,” Pharmacological Research, 2019; 149(104464).
  75. Schultz S, Gould GG, Antonucci N, Brigida AL, Siniscalco D. “Endocannabinoid System Dysregulation from Acetaminophen Use May Lead to Autism Spectrum Disorder: Could Cannabinoid Treatment Be Efficacious?” Molecules, 2021; 26(7): p. 1845.
  76. Loss CM, Teodoro L, Rodrigues GD, Moreira LR, Peres FF, Zuardi AW, et al. “Is Cannabidiol During Neurodevelopment a Promising Therapy for Schizophrenia and Autism Spectrum Disorders?” Frontiers in Pharmacology, 2021; 11(635763).
  77. Zamberletti E, Gabaglio M, Piscitelli F, Brodie JS, Woolley- Roberts M, Barbiero I, et al. “Cannabidivarin completely rescues cognitive deficits and delays neurological and motor defects in male Mecp2 mutant mice,” Journal of Psychopharmacology, 2019; 33(7): pp. 894-907.
  78. Kaplan JS, Stella N, Catterall WA, Westenbroek RE. “Cannabidiol attenuates seizures and social deficits in a mouse model of Dravet syndrome,” Proceedings of the National Academy of Sciences of the United States of America, 2017; 114(42): pp. 11229-11234.
  79. Ben Amar M. “Cannabinoids in medicine: a review of their therapeutic potential,” Journal of Ethnopharmacology, 2006; 105(1-2): pp. 1-25.
  80. Pretzsch CM, Voinescu B, Mendez MA, Wichers R, Ajram L, Ivin G, et al. “The effect of cannabidiol (CBD) on low-frequency activity and functional connectivity in the brain of adults with and without autism spectrum disorder (ASD),” Journal of Psychopharmacology, 2019; 33(9): pp. 1141-1148.
  81. Pretzsch CM, Freyberg J, Voinescu B, Lythgoe D, Horder J, Mendez MA, et al. “Effects of cannabidiol on brain excitation and inhibition systems; a randomised placebo-controlled single dose trial during magnetic resonance spectroscopy in adults with and without autism spectrum disorder,” Neuropsychopharmacology, 2019; 44(8): pp. 1398-1405.
  82. Pretzsch CM, Voinescu B, Lythgoe D, Horder J, Mendez MA, Wichers R, et al. “Effects of cannabidivarin (CBDV) on brain excitation and inhibition systems in adults with and without autism spectrum disorder (ASD): A single dose trial during magnetic resonance spectroscopy,” Translational Psychiatry, 2019; 9(1): p. 313.
  83. Narayanan A, White CA, Saklayen S, Scaduto MJ, Carpenter AL, Abduljalil A, et al. “Effect of propranolol on functional connectivity in autism spectrum disorder: a pilot study,” Brain Imaging and Behavior, 2010; 4(2): pp. 189-197.
  84. Gordon I, Jack A, Pretzsch CM, Vander Wyk B, Leckman JF, Feldman R, et al. “Intranasal oxytocin enhances connectivity in the neural circuitry supporting social motivation and Social Perception in Children with Autism,” Scientific Reports, 2016; 6(35054).
  85. Ashwin C, Baron-Cohen S, Wheelwright S, Riordan M, Bullmore ET. “Differential activation of the amygdala and the ‘social brain’ during fearful face-processing in Asperger Syndrome,” Neuropsychologia, 2006; 45(1): 14.
  86. Daly EM, Deeley Q, Ecker C, Craig M, Hallahan B, Murphy C, et al. “Serotonin and the neural processing of facial emotions in adults with autism: An fMRI study using acute tryptophan depletion,” Archives of General Psychiatry, 2012; 69(10): pp. 1003-1013.
  87. Cordes D, Haughton VM, Arfanakis K, Carew JD, Turski PA, Moritz CH, et al. “Frequencies contributing to functional connectivity in the cerebral cortex in “resting-state” data,” AJNR American Journal of Neuroradiology, 2001; 22(7): pp. 1326-1333.
  88. Friston KJ, Frith CD, Liddle PF, Frackowiak RS. “Functional connectivity: The principal-component analysis of large (PET) data sets,” Journal of Cerebral Blood Flow and Metabolism, 1993; 13(1): pp. 5-14.
  89. Bejjani A, O’Neill J, Kim JA, Frew AJ, Yee VW, Ly R, et al. “Elevated glutamatergic compounds in pregenual anterior cingulate in pediatric autism spectrum disorder demonstrated by 1H MRS and 1H MRSI,” PLoS One, 2012; 7(7): e38786.
  90. Hegarty JP, Weber DJ, Cirstea CM, Beversdorf DQ. “Cerebro-cerebellar functional connectivity is associated with cerebellar excitation-inhibition balance in autism spectrum disorder,” Journal of Autism and Developmental Disorders, 2018; 48(10): pp. 3460-3473.
  91. Oblak AL, Gibbs TT, Blatt GJ. “Reduced GABA(A) receptors and benzodiazepine binding sites in the posterior cingulate cortex and fusiform gyrus in autism,” Brain Research, 2011; 1380: pp. 218-228.
  92. Yip J, Soghomonian JJ, Blatt GJ. “Decreased GAD67 mRNA levels in cerebellar Purkinje cells in autism: Pathophysiological implications,” Acta Neuropathologica, 2007; 113(5): pp. 559-568.
  93. Andrews DS, Avino TA, Gudbrandsen M, Daly E, Marquand A, Murphy CM, et al. “In vivo evidence of reduced integrity of the gray-white matter boundary in autism spectrum disorder,” Cerebral Cortex, 2017; 27(2): pp. 877-887.
  94. Barnea-Goraly N, Kwon H, Menon V, Eliez S, Lotspeich L, Reiss AL. “White matter structure in autism: Preliminary evidence from diffusion tensor imaging,” Biological Psychiatry, 2004; 55(3): pp. 323-326.
  95. Noriuchi M, Kikuchi Y, Yoshiura T, Kira R, Shigeto H, Hara T, et al. “Altered white matter fractional anisotropy and social impairment in children with autism spectrum disorder,” Brain Research, 2010; 1362: pp. 141-149.
  96. Ben Bashat D, Kronfeld-Duenias V, Zachor DA, Ekstein PM, Hendler T, Tarrasch R, et al. “Accelerated maturation of white matter in young children with autism: A high b value DWI study,” Neuroimage, 2007; 37(1): pp. 40-47.
  97. Kleinhans NM, Richards T, Sterling L, Stegbauer KC, Mahurin R, Johnson lC, et al. “Abnormal functional connectivity in autism spectrum disorders during face processing,” Brain, 2008; 131(Pt4): pp.1000-1012.
  98. Perlman SB, Hudac CM, Pegors T, Minshew NJ, Pelphrey KA. “Experimental manipulation of face-evoked activity in the fusiform gyrus of individuals with autism,” Social Neuroscience, 2011; 6(1): pp. 22-30.
  99. Buxbaum JB, Silverman JM, Smith CJ, Greenberg DA, Kilifarski M, Reichert J, et al. “Association between a GABRB3 polymorphism and autism,” Molecular Psychiatry, 2002; 7(3): pp. 311-316.
  100. Myers JFM, Evans CJ, Kalk NJ, Edden RAE, Lingford-Hughes AR. “Measurement of GABA using J-difference edited H-1-MRS following modulation of synaptic GABA concentration with tiagabine,” Synapse, 2014; 68(8): pp. 355-362.
  101. Stagg CJ, Bestmann S, Constantinescu AO, Moreno LM, Allman C, Mekle R, et al. “Relationship between physiological measures of excitability and levels of glutamate and GABA in the human motor cortex,” Journal of Physioliogy, 2011; 589(Pt 23): pp. 5845-5585.
  102. Santana N, Bortolozzi A, Serrats J, Mengod G, Artigas F. “Expression of serotonin1A and serotonin2A receptors in pyramidal and GABAergic neurons of the rat pre-frontal cortex,” Cerebral Cortex, 2004; 14(10): pp. 1100-1109.
  103. Russo EB, Burnett A, Hall B, Parker KK. “Agonistic properties of cannabidiol at 5-HT1a receptors,” Neurochemical Research, 2005; 30(8): pp. 1037-1043.
  104. Veenstra-VanderWeele J, Muller CL, Iwamoto H, Sauer JE, Owens WA, Shah CR, et al. “Autism gene variant causes hyperserotonemia, serotonin receptor hyper- sensitivity, social impairment and repetitive behavior,” Proceedings of the National Academy of Sciences of the United States of America, 2012; 109(14): pp. 5469-5474.
  105. D’Mello AM, Stoodley CJ. “Cerebro-cerebellar circuits in autism spectrum disorder,” Frontiers in Neuroscience, 2015; 9(408).
  106. Calabresi P, Picconi B, Tozzi A, Ghiglieri V. “Interaction between basal ganglia and limbic circuits in learning and memory processes,” Parkinsonism and Related Disorders, 2016; 22(Suppl 1): S65-S68.
  107. Delmonte JH Balsters, McGrath J, Fitzgerald J, Brennan S, Fagan AJ, Gallagher L. “Social and monetary reward processing in autism spectrum disorders,” Molecular Autism, 2012; 3(1,7).
  108. Musella A, De Chiara V, Rossi S, Prosperetti C, Bernardi G, Maccarrone M, et al. “TRPV1 channels facilitate glutamate transmission in the striatum,” Molecular and Cellular Neuroscience, 2009; 40(1): pp. 89-97.
  109. Kunert-Keil C, Bisping F, Kruger J, Brinkmeier H. “Tissue-specific expression of TRP channel genes in the mouse and its variation in three different mouse strains,” BMC Genomics, 2006; 7(159).
  110. Marrone MC, Morabito A, Giustizieri M, Chiurchiù V, Leuti A, Mattioli M, et al. “TRPV1 channels are critical brain inflammation detectors and neuropathic pain biomarkers in mice,” Nature Communications, 2017; 8(15292).
logo

Subscribe to newsletter

© 2020. All rights reserved.

TOP