1
|
Sepers MD, Woodard CL, Ramandi D, Vecchiarelli HA, Hill MN, Raymond LA. Upregulation of endocannabinoid signaling in vivo restores striatal synaptic plasticity and motor performance in Huntington's disease mice. J Huntingtons Dis 2025:18796397251337021. [PMID: 40275705 DOI: 10.1177/18796397251337021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
BackgroundSynaptic dysfunction underlies early sensorimotor and cognitive deficits in Huntington's disease (HD) and precedes the degeneration of striatal spiny projection neurons and cortical pyramidal neurons. Movement selection and motor learning, which are impaired early in HD, are regulated by connections between the motor cortex, basal ganglia and thalamus. In particular, plasticity at corticostriatal synapses, including endocannabinoid-mediated long-term depression (LTD), is critical for motor learning. Previously, we found impaired endocannabinoid-mediated LTD, induced by high frequency stimulation (HFS) at corticostriatal synapses in brain slice recordings from pre-manifest HD mouse models, which was corrected by JZL184, an inhibitor of endocannabinoid 2-arachidonoyl glycerol (2-AG) degradation.ObjectiveDetermine the effects of in vivo JZL184 administration on YAC128 HD model and wild-type (WT) littermate mice.MethodsJZL184 was administered to mice orally over a 3-week period and their motor function was assessed using several behavioral tasks. In addition, brain tissue was collected from mice in order to quantify changes in endocannabinoid levels and measure HFS-induced plasticity at corticostriatal synapses.ResultsOral administration of JZL184 significantly increased levels of 2-AG in striatal tissue. While JZL184 treatment had no impact on open field behavior, the treatment eliminated the difference in motor learning on the rotarod task between YAC128 and WT mice. Moreover, HFS-induced striatal plasticity in YAC128 mice was normalized to WT levels after JZL184 treatment.ConclusionsThese results suggest a novel target for mitigating early symptoms of HD and support the need for clinical trials of therapies that modulate the endocannabinoid system.
Collapse
Affiliation(s)
- Marja D Sepers
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, Canada
| | - Cameron L Woodard
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel Ramandi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, Canada
- CELL Graduate Program, University of British Columbia, Vancouver, BC, Canada
| | | | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Lynn A Raymond
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, Canada
| |
Collapse
|
2
|
Jiang Z, Zhou Y, Zhou Y, Yang D, Li J, Li Y, Fan Q, Lin J. Exploring the bidirectional causal relationship between Autism Spectrum Disorder and Schizophrenia using Mendelian randomization. Medicine (Baltimore) 2025; 104:e42119. [PMID: 40228250 PMCID: PMC11999464 DOI: 10.1097/md.0000000000042119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Autism Spectrum Disorder (ASD), characterized mainly by stereotyped behaviors and social impairments, affects about one in 100 children worldwide. Schizophrenia (SCZ), a chronic mental illness, affects 1% of the global population. The pathogenesis and specific treatment strategies for ASD and SCZ remain unclear. Previous research has suggested similarities in SCZ and ASD etiology and symptoms. However, no definitive correlation has been confirmed. Therefore, we conducted a Mendelian randomization study to assess the relationship between SCZ and ASD, providing new insights into their etiology and treatment. We used the two-sample Mendelian randomization (TSMR) approach to investigate the bidirectional causal association between SCZ and ASD, employing summary-level genome-wide association studies (GWAS) data. ASD summary data from the IEU GWAS database and SCZ summary data from the Psychiatric Genomics Consortium (PGC) were used as exposure and outcome variables, respectively. Statistical analysis was performed using the TwoSampleMR package in R version 4.3.2, with sensitivity analysis conducted to verify the result's reliability. Based on the results of the MR analysis, we retrieved and analyzed the relevant genetic information from the GWAS Catalog. TSMR analysis revealed higher ASD risk in SCZ (IVW: OR: 1.19, 95% CI: 1.12-1.26, P < .001). Bidirectional MR analysis confirmed a causal relationship between ASD and SCZ (IVW: scz2018clozuk (Clozapine UK), OR: 1.12, 95% CI: 1.04-1.21, P = .003; scz2019asi, OR: 1.14, 95% CI: 1.05-1.23, P = .002). Our study demonstrated a bidirectional relationship between SCZ and ASD in the European population, suggesting that each may induce the onset of the other.
Collapse
Affiliation(s)
- Ziqing Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yiying Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yingxin Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Dongmei Yang
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Jingjun Li
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongchun Li
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qin Fan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jintao Lin
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Jones MR, Haggarty CJ, Petrie GN, Lunge AR, Morrison I, Hill MN, Heilig M, Mayo LM. Endocannabinoid contributions to the perception of socially relevant, affective touch in humans. Neuropsychopharmacology 2025; 50:849-855. [PMID: 39843850 PMCID: PMC11914470 DOI: 10.1038/s41386-025-02053-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/13/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025]
Abstract
Social relationships are central to well-being. A subgroup of afferent nerve fibers, C-tactile (CT) afferents, are primed to respond to affective, socially relevant touch and may mitigate the effects of stress. The endocannabinoid ligand anandamide (AEA) modulates both social reward and stress. We thus hypothesized that AEA levels would be associated with the perceived pleasantness of affective touch in humans. Across two studies, we explored perceptions of affective, socially relevant touch and general affective stimuli. In study 1, adult participants (N = 101) were recruited based on presence (CM+) or absence (CM-) of documented childhood maltreatment (N = 52 CM+; N = 49 CM-). In study 2, healthy individuals were randomized to receive an inhibitor of fatty acid amide hydrolase (FAAH; PF-04457845) to increase AEA levels (n = 16) or placebo (n = 29). Outcomes included self-report ratings of touch pleasantness and intensity, valence and arousal ratings of affective images, and plasma levels of endocannabinoids AEA and 2-AG, cortisol, and oxytocin. In study 1, higher AEA levels were associated with a reduced preference for affective, CT-optimal touch. In study 2, pharmacological elevation of AEA resulted in reduced preference for affective touch. These effects were specific to social processing, as AEA levels were not related to ratings of affective images. In contrast to our hypothesis, elevated AEA was associated with reduced pleasantness ratings of CT-optimal, affective touch. This provides novel, in-human data linking AEA to social processing, adding nuance to the rationale for its use as a potential novel therapeutic target in disordered in social processing.
Collapse
Affiliation(s)
- Madeleine R Jones
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Connor J Haggarty
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Gavin N Petrie
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada
| | - Abigail R Lunge
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada
| | - India Morrison
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Matthew N Hill
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Markus Heilig
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Leah M Mayo
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden.
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
4
|
Bortoletto R, Piscitelli F, Basaldella M, Scipioni C, Comacchio C, Fiorino R, Fornasaro S, Barbieri P, Pagliaro D, Sepulcri O, Fabris M, Curcio F, Balestrieri M, Colizzi M. Assessing the biobehavioral effects of ultramicronized-palmitoylethanolamide monotherapy in autistic adults with different severity levels: a report of two cases. Front Psychiatry 2024; 15:1463849. [PMID: 39502301 PMCID: PMC11536324 DOI: 10.3389/fpsyt.2024.1463849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/13/2024] [Indexed: 11/08/2024] Open
Abstract
Despite promise of its supplementation as both monotherapy and add-on treatment in autism spectrum disorder (ASD), the biobehavioral effects of Palmitoylethanolamide (PEA) in autistic adults have never been explored so far. We discussed the cases of two autistic adults with different degrees of severity (level 1 and level 2) presenting with symptoms of psychic distress, who were treated with ultramicronized-PEA (um-PEA) 600 mg/day monotherapy for a sustained period of 4 months. The level 1 autistic patient showed improved depressive symptoms and social engagement at a 12-week follow-up, in parallel to a tendency toward reduced inflammatory response and enhanced endocannabinoid (eCB) signaling, partially relapsing after um-PEA discontinuation at four months. Opposedly, the level 2 autistic patient exhibited a generally stable psychosocial functioning for the initial 12 weeks, consistent with basically unchanged immune and eCBs levels, abruptly deteriorating and leading to antipsychotic initiation afterwards. No significant side effects were reported in both cases during the observation period. The two cases suggest that um-PEA could be an effective option for the treatment of psychic distress in level 1 autistic adults, warranting further investigation of its age- and level-specificity and of the biological underpinnings of its therapeutic effect in ASD.
Collapse
Affiliation(s)
- Riccardo Bortoletto
- Unit of Psychiatry, Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, National Research Council (CNR), Pozzuoli, Italy
| | - Marta Basaldella
- Unit of Psychiatry, Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Claudia Scipioni
- Unit of Psychiatry, Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Carla Comacchio
- Unit of Psychiatry, Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Roberta Fiorino
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Institute of Clinical Pathology, Friuli Centrale Health University Authority (ASUFC), Udine, Italy
| | - Stefano Fornasaro
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Pierluigi Barbieri
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Daniele Pagliaro
- Unit of Psychiatry, Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Orietta Sepulcri
- Unit of Psychiatry, Friuli Centrale Health University Authority (ASUFC), Udine, Italy
| | - Martina Fabris
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Institute of Clinical Pathology, Friuli Centrale Health University Authority (ASUFC), Udine, Italy
| | - Francesco Curcio
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Institute of Clinical Pathology, Friuli Centrale Health University Authority (ASUFC), Udine, Italy
| | - Matteo Balestrieri
- Unit of Psychiatry, Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Marco Colizzi
- Unit of Psychiatry, Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| |
Collapse
|
5
|
Fuentes JJ, Mayans J, Guarro M, Canosa I, Mestre-Pintó JI, Fonseca F, Torrens M. Peripheral endocannabinoids in major depressive disorder and alcohol use disorder: a systematic review. BMC Psychiatry 2024; 24:551. [PMID: 39118031 PMCID: PMC11308641 DOI: 10.1186/s12888-024-05986-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Major Depressive Disorder (MDD) and Alcohol Use Disorder (AUD) are two high-prevalent conditions where the Endocannabinoid system (ECS) is believed to play an important role. The ECS regulates how different neurotransmitters interact in both disorders, which is crucial for controlling emotions and responses to stress and reward stimuli. Measuring peripheral endocannabinoids (eCBs) in human serum and plasma can help overcome the limitations of detecting endocannabinoid levels in the brain. This systematic review aims to identify levels of peripheral eCBs in patients with MDD and/or AUD and find eCBs to use as diagnostic, prognostic biomarkers, and potential therapeutic targets. METHODS We conducted a systematic literature search according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines from the earliest manuscript until October 22, 2023, in three electronic databases. We included studies of human adults who had a current diagnosis of AUD and/or MDD and evaluated plasma or serum endocannabinoids. We carefully considered known variables that may affect endocannabinoid levels. RESULTS We included 17 articles in this systematic review, which measured peripheral eCBs in 170 AUD and 359 MDD patients. Stressors increase peripheral 2-arachidonyl-glycerol (2-AG) concentrations, and 2-AG may be a particular feature of depression severity and chronicity. Anxiety symptoms are negatively correlated with anandamide (AEA) concentrations, and AEA significantly increases during early abstinence in AUD. Studies suggest a negative correlation between Oleoylethanolamide (OEA) and length of abstinence in AUD patients. They also show a significant negative correlation between peripheral levels of AEA and OEA and fatty acid amide hydrolase (FAAH) activity. Eicosapentaenoylethanolamide (EPEA) is correlated to clinical remission rates in depression. Included studies show known variables such as gender, chronicity, symptom severity, comorbid psychiatric symptoms, length of abstinence in the case of AUD, and stress-inducibility that can affect peripheral eCBs. CONCLUSIONS This systematic review highlights the important role that the ECS plays in MDD and AUD. Peripheral eCBs appear to be useful biomarkers for these disorders, and further research may identify potential therapeutic targets. Using accessible biological samples such as blood in well-designed clinical studies is crucial to develop novel therapies for these disorders.
Collapse
Affiliation(s)
- J J Fuentes
- Mental Health Institute, Hospital del Mar, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallés, Spain
| | - J Mayans
- Department of Psychiatry, Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, Spain
| | - M Guarro
- Mental Health Institute, Hospital del Mar, Barcelona, Spain
| | - I Canosa
- Mental Health Institute, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Research Institute, Barcelona, Spain
| | - J I Mestre-Pintó
- Hospital del Mar Research Institute, Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - F Fonseca
- Mental Health Institute, Hospital del Mar, Barcelona, Spain.
- Hospital del Mar Research Institute, Barcelona, Spain.
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| | - M Torrens
- Mental Health Institute, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Research Institute, Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
6
|
Vanhecke D, Bugada V, Steiner R, Polić B, Buch T. Refined tamoxifen administration in mice by encouraging voluntary consumption of palatable formulations. Lab Anim (NY) 2024; 53:205-214. [PMID: 39080504 PMCID: PMC11291282 DOI: 10.1038/s41684-024-01409-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/24/2024] [Indexed: 08/02/2024]
Abstract
Drug administration in preclinical rodent models is essential for research and the development of novel therapies. Compassionate administration methods have been developed, but these are mostly incompatible with water-insoluble drugs such as tamoxifen or do not allow for precise timing or dosing of the drugs. For more than two decades, tamoxifen has been administered by oral gavage or injection to CreERT2-loxP gene-modified mouse models to spatiotemporally control gene expression, with the numbers of such inducible models steadily increasing in recent years. Animal-friendly procedures for accurately administering tamoxifen or other water-insoluble drugs would, therefore, have an important impact on animal welfare. On the basis of a previously published micropipette feeding protocol, we developed palatable formulations to encourage voluntary consumption of tamoxifen. We evaluated the acceptance of the new formulations by mice during training and treatment and assessed the efficacy of tamoxifen-mediated induction of CreERT2-loxP-dependent reporter genes. Both sweetened milk and syrup-based formulations encouraged mice to consume tamoxifen voluntarily, but only sweetened milk formulations were statistically noninferior to oral gavage or intraperitoneal injections in inducing CreERT2-mediated gene expression. Serum concentrations of tamoxifen metabolites, quantified using an in-house-developed cell assay, confirmed the lower efficacy of syrup- as compared to sweetened milk-based formulations. We found dosing with a micropipette to be more accurate than oral gavage or injection, with the added advantage that the method requires little training for the experimenter. The new palatable solutions encourage voluntary consumption of tamoxifen without loss of efficacy compared to oral gavage or injections and thus represent a refined administration method.
Collapse
Affiliation(s)
- Dominique Vanhecke
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Viola Bugada
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Regula Steiner
- Institute of Clinical Chemistry, University and University Hospital of Zurich, Zurich, Switzerland
| | - Bojan Polić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
7
|
Pedrazzi JFC, Hassib L, Ferreira FR, Hallak JC, Del-Bel E, Crippa JA. Therapeutic potential of CBD in Autism Spectrum Disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 177:149-203. [PMID: 39029984 DOI: 10.1016/bs.irn.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental condition characterized by persistent deficits in social communication and interaction, as well as restricted and repetitive patterns of behavior. Despite extensive research, effective pharmacological interventions for ASD remain limited. Cannabidiol (CBD), a non-psychotomimetic compound of the Cannabis sativa plant, has potential therapeutic effects on several neurological and psychiatric disorders. CBD interacts with the endocannabinoid system, a complex cell-signaling system that plays a crucial role in regulating various physiological processes, maintaining homeostasis, participating in social and behavioral processing, and neuronal development and maturation with great relevance to ASD. Furthermore, preliminary findings from clinical trials indicate that CBD may have a modulatory effect on specific ASD symptoms and comorbidities in humans. Interestingly, emerging evidence suggests that CBD may influence the gut microbiota, with implications for the bidirectional communication between the gut and the central nervous system. CBD is a safe drug with low induction of side effects. As it has a multi-target pharmacological profile, it becomes a candidate compound for treating the central symptoms and comorbidities of ASD.
Collapse
Affiliation(s)
- João F C Pedrazzi
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Lucas Hassib
- Department of Mental Health, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Jaime C Hallak
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elaine Del-Bel
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil; National Institute for Science and Technology, Translational Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil; Center for Cannabinoid Research, Mental Health Building, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - José A Crippa
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
8
|
Zhao H, Liu Y, Cai N, Liao X, Tang L, Wang Y. Endocannabinoid Hydrolase Inhibitors: Potential Novel Anxiolytic Drugs. Drug Des Devel Ther 2024; 18:2143-2167. [PMID: 38882045 PMCID: PMC11179644 DOI: 10.2147/dddt.s462785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024] Open
Abstract
Over the past decade, the idea of targeting the endocannabinoid system to treat anxiety disorders has received increasing attention. Previous studies focused more on developing cannabinoid receptor agonists or supplementing exogenous cannabinoids, which are prone to various adverse effects due to their strong pharmacological activity and poor receptor selectivity, limiting their application in clinical research. Endocannabinoid hydrolase inhibitors are considered to be the most promising development strategies for the treatment of anxiety disorders. More recent efforts have emphasized that inhibition of two major endogenous cannabinoid hydrolases, monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH), indirectly activates cannabinoid receptors by increasing endogenous cannabinoid levels in the synaptic gap, circumventing receptor desensitization resulting from direct enhancement of endogenous cannabinoid signaling. In this review, we comprehensively summarize the anxiolytic effects of MAGL and FAAH inhibitors and their potential pharmacological mechanisms, highlight reported novel inhibitors or natural products, and provide an outlook on future directions in this field.
Collapse
Affiliation(s)
- Hongqing Zhao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| | - Yang Liu
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| | - Na Cai
- Outpatient Department, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Xiaolin Liao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| | - Lin Tang
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
- Department of Pharmacy, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Yuhong Wang
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
9
|
Jana A, Nath A, Sen P, Kundu S, Alghamdi BS, Abujamel TS, Saboor M, Woon-Khiong C, Alexiou A, Papadakis M, Alam MZ, Ashraf GM. Unraveling the Endocannabinoid System: Exploring Its Therapeutic Potential in Autism Spectrum Disorder. Neuromolecular Med 2024; 26:20. [PMID: 38744725 PMCID: PMC11093854 DOI: 10.1007/s12017-024-08781-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/04/2024] [Indexed: 05/16/2024]
Abstract
The salient features of autism spectrum disorder (ASD) encompass persistent difficulties in social communication, as well as the presence of restricted and repetitive facets of behavior, hobbies, or pursuits, which are often accompanied with cognitive limitations. Over the past few decades, a sizable number of studies have been conducted to enhance our understanding of the pathophysiology of ASD. Preclinical rat models have proven to be extremely valuable in simulating and analyzing the roles of a wide range of established environmental and genetic factors. Recent research has also demonstrated the significant involvement of the endocannabinoid system (ECS) in the pathogenesis of several neuropsychiatric diseases, including ASD. In fact, the ECS has the potential to regulate a multitude of metabolic and cellular pathways associated with autism, including the immune system. Moreover, the ECS has emerged as a promising target for intervention with high predictive validity. Particularly noteworthy are resent preclinical studies in rodents, which describe the onset of ASD-like symptoms after various genetic or pharmacological interventions targeting the ECS, providing encouraging evidence for further exploration in this area.
Collapse
Affiliation(s)
- Ankit Jana
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Arnab Nath
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Palash Sen
- School of Biosciences, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Swikriti Kundu
- Siksha Bhavana, Visva-Bharati University, Bolpur, West Bengal, 731235, India
| | - Badrah S Alghamdi
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Turki S Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad Saboor
- Department of Medical Laboratory Sciences, College of Health Sciences, and Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Chan Woon-Khiong
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, Greece
- Department of Research & Development, AFNP Med, 1030, Vienna, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohammad Zubair Alam
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, and Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
10
|
Cui Sun M, Otálora-Alcaraz A, Prenderville JA, Downer EJ. Toll-like receptor signalling as a cannabinoid target. Biochem Pharmacol 2024; 222:116082. [PMID: 38438052 DOI: 10.1016/j.bcp.2024.116082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/01/2024] [Accepted: 02/22/2024] [Indexed: 03/06/2024]
Abstract
Toll-like receptors (TLRs) have become a focus in biomedicine and biomedical research given the roles of this unique family of innate immune proteins in immune activation, infection, and autoimmunity. It is evident that TLR dysregulation, and subsequent alterations in TLR-mediated inflammatory signalling, can contribute to disease pathogenesis, and TLR targeted therapies are in development. This review highlights evidence that cannabinoids are key regulators of TLR signalling. Cannabinoids include component of the plant Cannabis sativa L. (C. sativa), synthetic and endogenous ligands, and overall represent a class of compounds whose therapeutic potential and mechanism of action continues to be elucidated. Cannabinoid-based medicines are in the clinic, and are furthermore under intense investigation for broad clinical development to manage symptoms of a range of disorders. In this review, we present an overview of research evidence that signalling linked to a range of TLRs is targeted by cannabinoids, and such cannabinoid mediated effects represent therapeutic avenues for further investigation. First, we provide an overview of TLRs, adaptors and key signalling events, alongside a summary of evidence that TLRs are linked to disease pathologies. Next, we discuss the cannabinoids system and the development of cannabinoid-based therapeutics. Finally, for the bulk of this review, we systematically outline the evidence that cannabinoids (plant-derived cannabinoids, synthetic cannabinoids, and endogenous cannabinoid ligands) can cross-talk with innate immune signalling governed by TLRs, focusing specifically on each member of the TLR family. Cannabinoids should be considered as key regulators of signalling controlled by TLRs, and such regulation should be a major focus in terms of the anti-inflammatory propensity of the cannabinoid system.
Collapse
Affiliation(s)
- Melody Cui Sun
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Almudena Otálora-Alcaraz
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Jack A Prenderville
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Transpharmation Ireland Limited, Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Eric J Downer
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
11
|
Schiavi S, Manduca A, Carbone E, Buzzelli V, Rava A, Feo A, Ascone F, Morena M, Campolongo P, Hill MN, Trezza V. Anandamide and 2-arachidonoylglycerol differentially modulate autistic-like traits in a genetic model of autism based on FMR1 deletion in rats. Neuropsychopharmacology 2023; 48:897-907. [PMID: 36114286 PMCID: PMC10156791 DOI: 10.1038/s41386-022-01454-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/20/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022]
Abstract
Autism spectrum disorder (ASD) has a multifactorial etiology. Major efforts are underway to understand the neurobiological bases of ASD and to develop efficacious treatment strategies. Recently, the use of cannabinoid compounds in children with neurodevelopmental disorders including ASD has received increasing attention. Beyond anecdotal reports of efficacy, however, there is limited current evidence supporting such an intervention and the clinical studies currently available have intrinsic limitations that make the interpretation of the findings challenging. Furthermore, as the mechanisms underlying the beneficial effects of cannabinoid compounds in neurodevelopmental disorders are still largely unknown, the use of drugs targeting the endocannabinoid system remains controversial. Here, we studied the role of endocannabinoid neurotransmission in the autistic-like traits displayed by the recently validated Fmr1-Δexon 8 rat model of autism. Fmr1-Δexon 8 rats showed reduced anandamide levels in the hippocampus and increased 2-arachidonoylglycerol (2-AG) content in the amygdala. Systemic and intra-hippocampal potentiation of anandamide tone through administration of the anandamide hydrolysis inhibitor URB597 ameliorated the cognitive deficits displayed by Fmr1-Δexon 8 rats along development, as assessed through the novel object and social discrimination tasks. Moreover, blockade of amygdalar 2-AG signaling through intra-amygdala administration of the CB1 receptor antagonist SR141716A prevented the altered sociability displayed by Fmr1-Δexon 8 rats. These findings demonstrate that anandamide and 2-AG differentially modulate specific autistic-like traits in Fmr1-Δexon 8 rats in a brain region-specific manner, suggesting that fine changes in endocannabinoid mechanisms contribute to ASD-related behavioral phenotypes.
Collapse
Affiliation(s)
- Sara Schiavi
- Department of Science, Roma Tre University, Rome, Italy
| | - Antonia Manduca
- Department of Science, Roma Tre University, Rome, Italy
- Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | | | | | | | | | | | - Maria Morena
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Neuropsychopharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
- Departments of Cell Biology and Anatomy & Psychiatry, Hotchkiss Brain Institute and Mathison Center for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Neuropsychopharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Matthew N Hill
- Departments of Cell Biology and Anatomy & Psychiatry, Hotchkiss Brain Institute and Mathison Center for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Viviana Trezza
- Department of Science, Roma Tre University, Rome, Italy.
| |
Collapse
|
12
|
Rong J, Yang Y, Liang M, Zhong H, Li Y, Zhu Y, Sha S, Chen L, Zhou R. Neonatal inflammation increases hippocampal KCC2 expression through methylation-mediated TGF-β1 downregulation leading to impaired hippocampal cognitive function and synaptic plasticity in adult mice. J Neuroinflammation 2023; 20:15. [PMID: 36691035 PMCID: PMC9872321 DOI: 10.1186/s12974-023-02697-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/12/2023] [Indexed: 01/25/2023] Open
Abstract
The mechanisms by which neonatal inflammation leads to cognitive deficits in adulthood remain poorly understood. Inhibitory GABAergic synaptic transmission plays a vital role in controlling learning, memory and synaptic plasticity. Since early-life inflammation has been reported to adversely affect the GABAergic synaptic transmission, the aim of this study was to investigate whether and how neonatal inflammation affects GABAergic synaptic transmission resulting in cognitive impairment. Neonatal mice received a daily subcutaneous injection of lipopolysaccharide (LPS, 50 μg/kg) or saline on postnatal days 3-5. It was found that blocking GABAergic synaptic transmission reversed the deficit in hippocampus-dependent memory or the induction failure of long-term potentiation in the dorsal CA1 in adult LPS mice. An increase of mIPSCs amplitude was further detected in adult LPS mice indicative of postsynaptic potentiation of GABAergic transmission. Additionally, neonatal LPS resulted in the increased expression and function of K+-Cl--cotransporter 2 (KCC2) and the decreased expression of transforming growth factor-beta 1 (TGF-β1) in the dorsal CA1 during adulthood. The local TGF-β1 overexpression improved KCC2 expression and function, synaptic plasticity and memory of adult LPS mice. Adult LPS mice show hypermethylation of TGFb1 promoter and negatively correlate with reduced TGF-β1 transcripts. 5-Aza-deoxycytidine restored the changes in TGFb1 promoter methylation and TGF-β1 expression. Altogether, the results suggest that hypermethylation-induced reduction of TGF-β1 leads to enhanced GABAergic synaptic inhibition through increased KCC2 expression, which is a underlying mechanism of neonatal inflammation-induced hippocampus-dependent memory impairment in adult mice.
Collapse
Affiliation(s)
- Jing Rong
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Yang Yang
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Min Liang
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Haiquan Zhong
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Yingchun Li
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Yichao Zhu
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Sha Sha
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Lei Chen
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Rong Zhou
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| |
Collapse
|
13
|
Khantakova JN, Bondar NP, Sapronova AA, Reshetnikov VV. Delayed effects of neonatal immune activation on brain neurochemistry and hypothalamic-pituitary-adrenal axis functioning. Eur J Neurosci 2022; 56:5931-5951. [PMID: 36156830 DOI: 10.1111/ejn.15831] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/17/2022] [Accepted: 09/15/2022] [Indexed: 12/29/2022]
Abstract
During the postnatal period, the brain is highly sensitive to stress and inflammation, which are hazardous to normal growth and development. There is increasing evidence that inflammatory processes in the early postnatal period increase the risk of psychopathologies and cognitive impairment later in life. On the other hand, there are few studies on the ability of infectious agents to cause long-term neuroinflammation, leading to changes in the hypothalamic-pituitary-adrenal axis functioning and an imbalance in the neurotransmitter system. In this review, we examine short- and long-term effects of neonatal-induced inflammation in rodents on glutamatergic, GABAergic and monoaminergic systems and on hypothalamic-pituitary-adrenal axis activity.
Collapse
Affiliation(s)
- Julia N Khantakova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk, Russia.,Federal State Budgetary Scientific Institution 'Research Institute of Fundamental and Clinical Immunology' (RIFCI), Novosibirsk, Russia
| | - Natalia P Bondar
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk, Russia.,Novosibirsk State University, Novosibirsk, Russia
| | - Anna A Sapronova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Vasiliy V Reshetnikov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk, Russia.,Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
14
|
de Camargo RW, de Novais Júnior LR, da Silva LM, Meneguzzo V, Daros GC, da Silva MG, de Bitencourt RM. Implications of the endocannabinoid system and the therapeutic action of cannabinoids in autism spectrum disorder: A literature review. Pharmacol Biochem Behav 2022; 221:173492. [PMID: 36379443 DOI: 10.1016/j.pbb.2022.173492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/31/2022] [Accepted: 11/09/2022] [Indexed: 11/15/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder, onset in early childhood and associated with cognitive, social, behavioral, and sensory impairments. The pathophysiology is still unclear, and it is believed that genetic and environmental factors are fully capable of influencing ASD, especially cell signaling and microglial functions. Furthermore, the endocannabinoid system (ECS) participates in the modulation of various brain processes and is also involved in the pathophysiological mechanisms of this condition. Due to the health and quality of life impacts of autism for the patient and his/her family and the lack of effective medications, the literature has elucidated the possibility that Cannabis phytocannabinoids act favorably on ASD symptoms, probably through the modulation of neurotransmitters, in addition to endogenous ligands derived from arachidonic acid, metabolizing enzymes and even transporters of the membrane. These findings support the notion that there are links between key features of ASD and ECS due to the favorable actions of cannabidiol (CBD) and other cannabinoids on symptoms related to behavioral and cognitive disorders, as well as deficits in communication and social interaction, hyperactivity, anxiety and sleep disorders. Thus, phytocannabinoids emerge as therapeutic alternatives for ASD.
Collapse
Affiliation(s)
- Rick Wilhiam de Camargo
- Behavioral Neuroscience Laboratory, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil.
| | | | - Larissa Mendes da Silva
- Behavioral Neuroscience Laboratory, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Vicente Meneguzzo
- Behavioral Neuroscience Laboratory, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Guilherme Cabreira Daros
- Behavioral Neuroscience Laboratory, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Marina Goulart da Silva
- Behavioral Neuroscience Laboratory, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | | |
Collapse
|
15
|
Pedrazzi JFC, Ferreira FR, Silva-Amaral D, Lima DA, Hallak JEC, Zuardi AW, Del-Bel EA, Guimarães FS, Costa KCM, Campos AC, Crippa ACS, Crippa JAS. Cannabidiol for the treatment of autism spectrum disorder: hope or hype? Psychopharmacology (Berl) 2022; 239:2713-2734. [PMID: 35904579 DOI: 10.1007/s00213-022-06196-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 07/18/2022] [Indexed: 11/30/2022]
Abstract
RATIONALE Autism spectrum disorder (ASD) is defined as a group of neurodevelopmental disorders whose symptoms include impaired communication and social interaction, restricted and repetitive patterns of behavior, and varying levels of intellectual disability. ASD is observed in early childhood and is one of the most severe chronic childhood disorders in prevalence, morbidity, and impact on society. It is usually accompanied by attention deficit hyperactivity disorder, anxiety, depression, sleep disorders, and epilepsy. The treatment of ASD has low efficacy, possibly because it has a heterogeneous nature, and its neurobiological basis is not clearly understood. Drugs such as risperidone and aripiprazole are the only two drugs available that are recognized by the Food and Drug Administration, primarily for treating the behavioral symptoms of this disorder. These drugs have limited efficacy and a high potential for inducing undesirable effects, compromising treatment adherence. Therefore, there is great interest in exploring the endocannabinoid system, which modulates the activity of other neurotransmitters, has actions in social behavior and seems to be altered in patients with ASD. Thus, cannabidiol (CBD) emerges as a possible strategy for treating ASD symptoms since it has relevant pharmacological actions on the endocannabinoid system and shows promising results in studies related to disorders in the central nervous system. OBJECTIVES Review the preclinical and clinical data supporting CBD's potential as a treatment for the symptoms and comorbidities associated with ASD, as well as discuss and provide information with the purpose of not trivializing the use of this drug.
Collapse
Affiliation(s)
- João F C Pedrazzi
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Frederico R Ferreira
- Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, 21040-900, Brazil
| | - Danyelle Silva-Amaral
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Daniel A Lima
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jaime E C Hallak
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Antônio W Zuardi
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elaine A Del-Bel
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Morphology, Physiology, and Basic Pathology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Karla C M Costa
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Alline C Campos
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana C S Crippa
- Graduate Program in Child and Adolescent Health, Neuropediatric Center of the Hospital of Clinics (CENEP), Federal University of Paraná, Curitiba, Paraná, Brazil
| | - José A S Crippa
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
16
|
Cannabis Use in Autism: Reasons for Concern about Risk for Psychosis. Healthcare (Basel) 2022; 10:healthcare10081553. [PMID: 36011210 PMCID: PMC9407973 DOI: 10.3390/healthcare10081553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/31/2022] [Accepted: 08/14/2022] [Indexed: 01/09/2023] Open
Abstract
Being particularly vulnerable to the pro-psychotic effects of cannabinoid exposure, autism spectrum individuals present with an increased risk of psychosis, which may be passed on to their own children. More specifically, cannabis exposure among autism spectrum individuals seems to exert disruptive epigenetic effects that can be intergenerationally inherited in brain areas which play a critical role in schizophrenia pathophysiology. Additionally, because of such cannabinoid-induced epigenetic effects, autism candidate genes present with bivalent chromatin markings which make them more vulnerable to subsequent disruption, possibly leading to psychosis onset later in life. Thus, findings support a developmental trajectory between autism and psychosis, as per endocannabinoid system modulation. However, such evidence has not received the attention it deserves.
Collapse
|
17
|
Cuskelly A, Hoedt EC, Harms L, Talley NJ, Tadros MA, Keely S, Hodgson DM. Neonatal immune challenge influences the microbiota and behaviour in a sexually dimorphic manner. Brain Behav Immun 2022; 103:232-242. [PMID: 35491004 DOI: 10.1016/j.bbi.2022.04.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/31/2022] [Accepted: 04/26/2022] [Indexed: 10/18/2022] Open
Abstract
There is comorbidity between anxiety disorders and gastrointestinal disorders, with both linked to adverse early life events. The microbiome gut-brain-axis, a bidirectional communication system, is plastic throughout the neonatal period and is a possible mediator of this relationship. Here, we used a well-established neonatal rodent immune activation model to investigate the long-term effect of neonatal lipopolysaccharide (LPS) exposure on adult behaviour and the relationship to microbiome composition. Wistar rats were injected with LPS (0.05 mg/kg) or saline (equivolume) on postnatal days 3 and 5. In adulthood, behavioural tests were performed to assess anxiety-like behaviour, and microbiota sequencing was performed on stool samples. There were distinctly different behavioural phenotypes for LPS-exposed males and females. LPS-exposed males displayed typical anxiety-like behaviours with significantly decreased social interaction (F(1,22) = 7.576, p = 0.009) and increased defecation relative to saline controls (F(1,23) = 8.623, p = 0.005). LPS-exposed females displayed a different behavioural phenotype with significantly increased social interaction (F(1,22) = 6.094, p = 0.018), and exploration (F(1,24) = 6.359, p = 0.015), compared to saline controls. With respect to microbiota profiling data, Bacteroidota was significantly increased for LPS-exposed females (F(1,14) = 4.931p = 0.035) and Proteobacteria was decreased for LPS-exposed rats of both sexes versus controls (F(1,30) = 4.923p = 0.035). Furthermore, alterations in predicted functional pathways for neurotransmitters in faeces were observed with a decrease in the relative abundance of D-glutamine and D-glutamate metabolism in LPS exposed females compared to control females (p < 0.05). This suggests that neonatal immune activation alters both later life behaviour and adult gut microbiota in sex-specific ways. These findings highlight the importance of sex in determining the impact of neonatal immune activation on social behaviour and the gut microbiota.
Collapse
Affiliation(s)
- A Cuskelly
- School of Psychological Sciences, University of Newcastle, Callaghan, NSW, Australia; Viruses, Infection, Immunity, Vaccine and Asthma (VIVA) Program, Hunter Medical Research Institute (HMRI), Newcastle, NSW, Australia.
| | - E C Hoedt
- Viruses, Infection, Immunity, Vaccine and Asthma (VIVA) Program, Hunter Medical Research Institute (HMRI), Newcastle, NSW, Australia; NHMRC Centre of Research Excellence (CRE) in Digestive Health, HMRI, Newcastle, NSW, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
| | - L Harms
- School of Medicine and Public Health, University of Newcastle, New Lambton, NSW, Australia
| | - N J Talley
- Viruses, Infection, Immunity, Vaccine and Asthma (VIVA) Program, Hunter Medical Research Institute (HMRI), Newcastle, NSW, Australia; NHMRC Centre of Research Excellence (CRE) in Digestive Health, HMRI, Newcastle, NSW, Australia; School of Medicine and Public Health, University of Newcastle, New Lambton, NSW, Australia
| | - M A Tadros
- School of Medicine and Public Health, University of Newcastle, New Lambton, NSW, Australia
| | - S Keely
- Viruses, Infection, Immunity, Vaccine and Asthma (VIVA) Program, Hunter Medical Research Institute (HMRI), Newcastle, NSW, Australia; NHMRC Centre of Research Excellence (CRE) in Digestive Health, HMRI, Newcastle, NSW, Australia; School of Medicine and Public Health, University of Newcastle, New Lambton, NSW, Australia
| | - D M Hodgson
- School of Psychological Sciences, University of Newcastle, Callaghan, NSW, Australia; Viruses, Infection, Immunity, Vaccine and Asthma (VIVA) Program, Hunter Medical Research Institute (HMRI), Newcastle, NSW, Australia
| |
Collapse
|
18
|
Effects of endocannabinoid system modulation on social behaviour: A systematic review of animal studies. Neurosci Biobehav Rev 2022; 138:104680. [PMID: 35513169 DOI: 10.1016/j.neubiorev.2022.104680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/09/2022]
Abstract
There is a clear link between psychiatric disorders and social behaviour, and evidence suggests the involvement of the endocannabinoid system (ECS). A systematic review of preclinical literature was conducted using MEDLINE (PubMed) and PsychINFO databases to examine whether pharmacological and/or genetic manipulations of the ECS alter social behaviours in wildtype (WT) animals or models of social impairment (SIM). Eighty studies were included. Risk of bias (RoB) was assessed using SYRCLE's RoB tool. While some variability was evident, studies most consistently found that direct cannabinoid receptor (CBR) agonism decreased social behaviours in WT animals, while indirect CBR activation via enzyme inhibition or gene-knockout increased social behaviours. Direct and, more consistently, indirect CBR activation reversed social deficits in SIM. These CBR-mediated effects were often sex- and developmental-phase-dependent and blocked by CBR antagonism. Overall, ECS enzyme inhibition may improve social behaviour in SIM, suggesting the potential usefulness of ECS enzyme inhibition as a therapeutic approach for social deficits. Future research should endeavour to elucidate ECS status in neuropsychiatric disorders characterized by social deficits.
Collapse
|
19
|
Colizzi M, Bortoletto R, Costa R, Bhattacharyya S, Balestrieri M. The Autism-Psychosis Continuum Conundrum: Exploring the Role of the Endocannabinoid System. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:5616. [PMID: 35565034 PMCID: PMC9105053 DOI: 10.3390/ijerph19095616] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 02/07/2023]
Abstract
Evidence indicates shared physiopathological mechanisms between autism and psychosis. In this regard, the endocannabinoid system has been suggested to modulate neural circuits during the early stage of neurodevelopment, with implications for both autism and psychosis. Nevertheless, such potential common markers of disease have been investigated in both autism and psychosis spectrum disorders, without considering the conundrum of differentiating the two groups of conditions in terms of diagnosis and treatment. Here, we systematically review all human and animal studies examining the endocannabinoid system and its biobehavioral correlates in the association between autism and psychosis. Studies indicate overlapping biobehavioral aberrancies between autism and schizophrenia, subject to correction by modulation of the endocannabinoid system. In addition, common cannabinoid-based pharmacological strategies have been identified, exerting epigenetic effects across genes controlling neural mechanisms shared between autism and schizophrenia. Interestingly, a developmental and transgenerational trajectory between autism and schizophrenia is supported by evidence that exogenous alteration of the endocannabinoid system promotes progression to inheritable psychosis phenotypes in the context of biobehavioral autism vulnerability. However, evidence for a diametral association between autism and psychosis is scant. Several clinical implications follow from evidence of a developmental continuum between autism and psychosis as a function of the endocannabinoid system dysregulation.
Collapse
Affiliation(s)
- Marco Colizzi
- Unit of Psychiatry, Department of Medicine (DAME), University of Udine, 33100 Udine, Italy;
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK;
| | - Riccardo Bortoletto
- Child and Adolescent Neuropsychiatry Unit, Maternal-Child Integrated Care Department, Integrated University Hospital of Verona, 37126 Verona, Italy;
| | - Rosalia Costa
- Community Mental Health Team, Friuli Centrale University Health Service (ASUFC), 33057 Palmanova, Italy;
| | - Sagnik Bhattacharyya
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK;
| | - Matteo Balestrieri
- Unit of Psychiatry, Department of Medicine (DAME), University of Udine, 33100 Udine, Italy;
| |
Collapse
|
20
|
Vecchiarelli HA, Aukema RJ, Hume C, Chiang V, Morena M, Keenan CM, Nastase AS, Lee FS, Pittman QJ, Sharkey KA, Hill MN. Genetic Variants of Fatty Acid Amide Hydrolase Modulate Acute Inflammatory Responses to Colitis in Adult Male Mice. Front Cell Neurosci 2021; 15:764706. [PMID: 34916909 PMCID: PMC8670533 DOI: 10.3389/fncel.2021.764706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Cannabinoids, including cannabis derived phytocannabinoids and endogenous cannabinoids (endocannabinoids), are typically considered anti-inflammatory. One such endocannabinoid is N-arachidonoylethanolamine (anandamide, AEA), which is metabolized by fatty acid amide hydrolase (FAAH). In humans, there is a loss of function single nucleotide polymorphism (SNP) in the FAAH gene (C385A, rs324420), that leads to increases in the levels of AEA. Using a mouse model with this SNP, we investigated how this SNP affects inflammation in a model of inflammatory bowel disease. We administered 2,4,6-trinitrobenzene sulfonic acid (TNBS) intracolonically, to adult male FAAH SNP mice and examined colonic macroscopic tissue damage and myeloperoxidase activity, as well as levels of plasma and amygdalar cytokines and chemokines 3 days after administration, at the peak of colitis. We found that mice possessing the loss of function alleles (AC and AA), displayed no differences in colonic damage or myeloperoxidase activity compared to mice with wild type alleles (CC). In contrast, in plasma, colitis-induced increases in interleukin (IL)-2, leukemia inhibitory factor (LIF), monocyte chemoattractant protein (MCP)-1, and tumor necrosis factor (TNF) were reduced in animals with an A allele. A similar pattern was observed in the amygdala for granulocyte colony stimulating factor (G-CSF) and MCP-1. In the amygdala, the mutant A allele led to lower levels of IL-1α, IL-9, macrophage inflammatory protein (MIP)-1β, and MIP-2 independent of colitis-providing additional understanding of how FAAH may serve as a regulator of inflammatory responses in the brain. Together, these data provide insights into how FAAH regulates inflammatory processes in disease.
Collapse
Affiliation(s)
- Haley A Vecchiarelli
- Neuroscience Graduate Program, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Robert J Aukema
- Neuroscience Graduate Program, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Catherine Hume
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Vincent Chiang
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Maria Morena
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Catherine M Keenan
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Andrei S Nastase
- Neuroscience Graduate Program, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, United States
| | - Quentin J Pittman
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Keith A Sharkey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
21
|
Adcock SJJ. Early Life Painful Procedures: Long-Term Consequences and Implications for Farm Animal Welfare. FRONTIERS IN ANIMAL SCIENCE 2021. [DOI: 10.3389/fanim.2021.759522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Farm animals routinely undergo painful husbandry procedures early in life, including disbudding and castration in calves and goat kids, tail docking and castration in piglets and lambs, and beak trimming in chicks. In rodents, inflammatory events soon after birth, when physiological systems are developing and sensitive to perturbation, can profoundly alter phenotypic outcomes later in life. This review summarizes the current state of research on long-term phenotypic consequences of neonatal painful procedures in rodents and farm animals, and discusses the implications for farm animal welfare. Rodents exposed to early life inflammation show a hypo-/hyper-responsive profile to pain-, fear-, and anxiety-inducing stimuli, manifesting as an initial attenuation in responses that transitions into hyperresponsivity with increasing age or cumulative stress. Neonatal inflammation also predisposes rodents to cognitive, social, and reproductive deficits, and there is some evidence that adverse effects may be passed to offspring. The outcomes of neonatal inflammation are modulated by injury etiology, age at the time of injury and time of testing, sex, pain management, and rearing environment. Equivalent research examining long-term phenotypic consequences of early life painful procedures in farm animals is greatly lacking, despite obvious implications for welfare and performance. Improved understanding of how these procedures shape phenotypes will inform efforts to mitigate negative outcomes through reduction, replacement, and refinement of current practices.
Collapse
|
22
|
De Pol M, Kolla NJ. Endocannabinoid markers in autism spectrum disorder: A scoping review of human studies. Psychiatry Res 2021; 306:114256. [PMID: 34775294 DOI: 10.1016/j.psychres.2021.114256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/29/2021] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social communication deficits and patterns of restrictive and repetitive behavior. Although the neurological underpinnings of ASD remain elusive, the endocannabinoid system (ECS) may play a role in modulating social behavior in ASD. Preclinical studies have suggested that alterations in the ECS result in ASD-like phenotypes, but currently no reviews have examined ECS abnormalities in human studies. This scoping review investigated any evidence of ECS alterations in humans with ASD. A comprehensive literature search was conducted and five studies were eligible for review. Three studies reported a significant reduction of anandamide in ASD compared to controls. Other alterations included decreased 2-arachidonoylglycerol, oleoylethanolamide, and palmitoylethanolamide and elevated diacylglycerol lipase and monoacylglycerol lipase. Some discrepant findings were also noted, which included elevated or reduced CB2 receptor in three studies and elevated or reduced N-acyl phosphatidylethanolamine phospholipase D and fatty acid amide hydrolase in two studies. We conclude from this preliminary investigation that the ECS may be altered in humans with ASD. Potential limitations of the reviewed studies include medication use and psychiatric comorbidities. Further research, such as positron emission tomography studies, are necessary to fully understand the relationship between ECS markers and ASD.
Collapse
Affiliation(s)
- Michelle De Pol
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Nathan J Kolla
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Waypoint Centre for Mental Health Care, Penetanguishene, ON, Canada.
| |
Collapse
|
23
|
Lataliza AAB, de Assis PM, da Rocha Laurindo L, Gonçalves ECD, Raposo NRB, Dutra RC. Antidepressant-like effect of rosmarinic acid during LPS-induced neuroinflammatory model: The potential role of cannabinoid receptors/PPAR-γ signaling pathway. Phytother Res 2021; 35:6974-6989. [PMID: 34709695 DOI: 10.1002/ptr.7318] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/17/2021] [Accepted: 10/13/2021] [Indexed: 01/03/2023]
Abstract
Rosmarinic acid (RA), an ester of caffeic acid and 3, 4-dihydroxyphenyllactic acid, has anti-inflammatory and neuroprotective activities. Herein, this study investigated in silico the drug-likeness and the potential molecular targets to RA. Moreover, it tested the antidepressant-like potential of RA in the lipopolysaccharide (LPS)-induced depression model. RA (MW = 360.31 g/mol) meets the criteria of both Lipinski's rule of five and the Ghose filter. It also attends to relevant pharmacokinetic parameters. Target prediction analysis identified RA's potential targets and biological activities, including the peroxisome proliferator-activated receptor (PPAR) and the cannabinoid receptors CB1 and CB2 . In vivo, RA's acute, repetitive, and therapeutic administration showed antidepressant-like effect since it significantly reduced the immobility time in the tail suspension test and increased grooming time in the splash test. Further, the pretreatment with antagonists of CB1 , CB2 , and PPAR-γ receptors significantly blocked the antidepressant-like effect of RA. Altogether, our findings suggest that cannabinoid receptors/PPAR-γ signaling pathways are involved with the antidepressant-like effect of RA. Moreover, this molecule meets important physicochemical and pharmacokinetic parameters that favor its bioavailability. RA constitutes a promising, innovative, and safe molecule for the pharmacotherapy of major depressive disorder.
Collapse
Affiliation(s)
- Alexandre Augusto Barros Lataliza
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, Araranguá, Brazil
- Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Pollyana Mendonça de Assis
- Center for Research and Innovation in Health Sciences (NUPICS), Faculty of Pharmacy, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Larissa da Rocha Laurindo
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, Araranguá, Brazil
| | - Elaine Cristina Dalazen Gonçalves
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, Araranguá, Brazil
- Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Nádia Rezende Barbosa Raposo
- Center for Research and Innovation in Health Sciences (NUPICS), Faculty of Pharmacy, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Rafael Cypriano Dutra
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, Araranguá, Brazil
- Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
- Laboratory of Neurobiology of Pain and Inflammation, Department of Physiological Sciences, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
24
|
Pietropaolo S, Marsicano G. The role of the endocannabinoid system as a therapeutic target for autism spectrum disorder: Lessons from behavioral studies on mouse models. Neurosci Biobehav Rev 2021; 132:664-678. [PMID: 34813825 DOI: 10.1016/j.neubiorev.2021.11.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 11/02/2021] [Accepted: 11/19/2021] [Indexed: 12/17/2022]
Abstract
Recent years have seen an impressive amount of research devoted to understanding the etiopathology of Autism Spectrum Disorder (ASD) and developing therapies for this syndrome. Because of the lack of biomarkers of ASD, this work has been largely based on the behavioral characterization of rodent models, based on a multitude of genetic and environmental manipulations. Here we highlight how the endocannabinoid system (ECS) has recently emerged within this context of mouse behavioral studies as an etiopathological factor in ASD and a valid potential therapeutic target. We summarize the most recent results showing alterations of the ECS in rodent models of ASD, and demonstrating ASD-like behaviors in mice with altered ECS, induced either by genetic or pharmacological manipulations. We also give a critical overview of the most relevant advances in designing treatments and novel mouse models for ASD targeting the ECS, highlighting the relevance of thorough and innovative behavioral approaches to investigate the mechanisms acting underneath the complex features of ASD.
Collapse
Affiliation(s)
| | - Giovanni Marsicano
- INSERM, U1215 NeuroCentre Magendie, 146 rue Léo Saignat, 33077, Bordeaux Cedex, France
| |
Collapse
|
25
|
On the Biomedical Properties of Endocannabinoid Degradation and Reuptake Inhibitors: Pre-clinical and Clinical Evidence. Neurotox Res 2021; 39:2072-2097. [PMID: 34741755 DOI: 10.1007/s12640-021-00424-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/14/2021] [Accepted: 09/28/2021] [Indexed: 10/19/2022]
Abstract
The endocannabinoid system (ECS) is composed of endogenous cannabinoids; components involved in their synthesis, transport, and degradation; and an expansive variety of cannabinoid receptors. Hypofunction or deregulation of the ECS is related to pathological conditions. Consequently, endogenous enhancement of endocannabinoid levels and/or regulation of their metabolism represent promising therapeutic approaches. Several major strategies have been suggested for the modulation of the ECS: (1) blocking endocannabinoids degradation, (2) inhibition of endocannabinoid cellular uptake, and (3) pharmacological modulation of cannabinoid receptors as potential therapeutic targets. Here, we focused in this review on degradation/reuptake inhibitors over cannabinoid receptor modulators in order to provide an updated synopsis of contemporary evidence advancing mechanisms of endocannabinoids as pharmacological tools with therapeutic properties for the treatment of several disorders. For this purpose, we revisited the available literature and reported the latest advances regarding the biomedical properties of fatty acid amide hydrolase and monoacylglycerol lipase inhibitors in pre-clinical and clinical studies. We also highlighted anandamide and 2-arachidonoylglycerol reuptake inhibitors with promising results in pre-clinical studies using in vitro and animal models as an outlook for future research in clinical trials.
Collapse
|
26
|
Ahmed M, Boileau I, Le Foll B, Carvalho AF, Kloiber S. The endocannabinoid system in social anxiety disorder: from pathophysiology to novel therapeutics. ACTA ACUST UNITED AC 2021; 44:81-93. [PMID: 34468550 PMCID: PMC8827369 DOI: 10.1590/1516-4446-2021-1926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Social anxiety disorder (SAD) is a highly prevalent psychiatric disorder that presents with an early age of onset, chronic disease course, and increased risk of psychiatric comorbidity. Current treatment options for SAD are associated with low response rates, suboptimal efficacy, and possible risk of adverse effects. Investigation of new neurobiological mechanisms may aid in the identification of more specific therapeutic targets for the treatment of this disorder. Emerging evidence suggests that the endogenous cannabinoid system, also referred to as the endocannabinoid system (ECS), could play a potential role in the pathophysiology of SAD. This review discusses the known pathophysiological mechanisms of SAD, the potential role of the ECS in this disorder, current drugs targeting the ECS, and the potential of these novel compounds to enhance the therapeutic armamentarium for SAD. Further investigational efforts, specifically in human populations, are warranted to improve our knowledge of the ECS in SAD.
Collapse
Affiliation(s)
- Mashal Ahmed
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Isabelle Boileau
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Bernard Le Foll
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Andre F Carvalho
- Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Innovation in Mental and Physical Health and Clinical Treatment (IMPACT) Strategic Research Centre, Deakin University, Geelong, VIC, Australia, 3216
| | - Stefan Kloiber
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Nezgovorova V, Ferretti CJ, Taylor BP, Shanahan E, Uzunova G, Hong K, Devinsky O, Hollander E. Potential of cannabinoids as treatments for autism spectrum disorders. J Psychiatr Res 2021; 137:194-201. [PMID: 33689997 DOI: 10.1016/j.jpsychires.2021.02.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/22/2021] [Indexed: 01/04/2023]
Abstract
Current treatments for autism spectrum disorders (ASD) are limited in efficacy and are often associated with substantial side effects. These medications typically ameliorate problem behaviors associated with ASD, but do not target core symptom domains. As a result, there is a significant amount of research underway for development of novel experimental therapeutics. Endocannabinoids are arachidonic acid-derived lipid neuromodulators, which, in combination with their receptors and associated metabolic enzymes, constitute the endocannabinoid (EC) system. Cannabinoid signaling may be involved in the social impairment and repetitive behaviors observed in those with ASD. In this review, we discuss a possible role of the EC system in excitatory-inhibitory (E-I) imbalance and immune dysregulation in ASD. Novel treatments for the core symptom domains of ASD are needed and phytocannabinoids could be useful experimental therapeutics for core symptoms and associated domains.
Collapse
Affiliation(s)
- V Nezgovorova
- Autism and Obsessive-Compulsive Spectrum Program, Psychiatry Research Institute at Montefiore- Einstein (PRIME), Albert Einstein College of Medicine, Bronx, New York, USA
| | - C J Ferretti
- Autism and Obsessive-Compulsive Spectrum Program, Psychiatry Research Institute at Montefiore- Einstein (PRIME), Albert Einstein College of Medicine, Bronx, New York, USA
| | - B P Taylor
- Autism and Obsessive-Compulsive Spectrum Program, Psychiatry Research Institute at Montefiore- Einstein (PRIME), Albert Einstein College of Medicine, Bronx, New York, USA
| | - E Shanahan
- Autism and Obsessive-Compulsive Spectrum Program, Psychiatry Research Institute at Montefiore- Einstein (PRIME), Albert Einstein College of Medicine, Bronx, New York, USA
| | - G Uzunova
- Autism and Obsessive-Compulsive Spectrum Program, Psychiatry Research Institute at Montefiore- Einstein (PRIME), Albert Einstein College of Medicine, Bronx, New York, USA
| | - K Hong
- Autism and Obsessive-Compulsive Spectrum Program, Psychiatry Research Institute at Montefiore- Einstein (PRIME), Albert Einstein College of Medicine, Bronx, New York, USA
| | - O Devinsky
- New York University Comprehensive Epilepsy Center, New York, NY, USA
| | - E Hollander
- Autism and Obsessive-Compulsive Spectrum Program, Psychiatry Research Institute at Montefiore- Einstein (PRIME), Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
28
|
Increased Excitatory Synaptic Transmission Associated with Adult Seizure Vulnerability Induced by Early-Life Inflammation in Mice. J Neurosci 2021; 41:4367-4377. [PMID: 33827934 DOI: 10.1523/jneurosci.2667-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 12/27/2022] Open
Abstract
Early-life inflammatory stress increases seizure susceptibility later in life. However, possible sex- and age-specific differences and the associated mechanisms are largely unknown. C57BL/6 mice were bred in house, and female and male pups were injected with lipopolysaccharide (LPS; 100 μg/kg, i.p.) or vehicle control (saline solution) at postnatal day 14 (P14). Seizure threshold was assessed in response to pentylenetetrazol (1% solution, i.v.) in adolescence (∼P40) and adulthood (∼P60). We found that adult, but not adolescent, mice treated with LPS displayed ∼34% lower seizure threshold compared with controls. Females and males showed similar increased seizure susceptibility, suggesting that altered brain excitability was age dependent, but not sex dependent. Whole-cell recordings revealed no differences in excitatory synaptic activity onto CA1 pyramidal neurons from control or neonatally inflamed adolescent mice of either sex. However, adult mice of both sexes previously exposed to LPS displayed spontaneous EPSC frequency approximately twice that of controls, but amplitude was unchanged. Although these changes were not associated with alterations in dendritic spines or in the NMDA/AMPA receptor ratio, they were linked to an increased glutamate release probability from Schaffer collateral, but not temporoammonic pathway. This glutamate increase was associated with reduced activity of presynaptic GABAB receptors and was independent of the endocannabinoid-mediated suppression of excitation. Our new findings demonstrate that early-life inflammation leads to long-term increased hippocampal excitability in adult female and male mice associated with changes in glutamatergic synaptic transmission. These alterations may contribute to enhanced vulnerability of the brain to subsequent pathologic challenges such as epileptic seizures.SIGNIFICANCE STATEMENT Adult physiology has been shown to be affected by early-life inflammation. Our data reveal that early-life inflammation increases excitatory synaptic transmission onto hippocampal CA1 pyramidal neurons in an age-dependent manner through disrupted presynaptic GABAB receptor activity on Schaffer collaterals. This hyperexcitability was seen only in adult, and not in adolescent, animals of either sex. The data suggest a maturation process, independent of sex, in the priming action of early-life inflammation and highlight the importance of studying mature brains to reveal cellular changes associated with early-life interventions.
Collapse
|
29
|
Vecchiarelli HA, Morena M, Keenan CM, Chiang V, Tan K, Qiao M, Leitl K, Santori A, Pittman QJ, Sharkey KA, Hill MN. Comorbid anxiety-like behavior in a rat model of colitis is mediated by an upregulation of corticolimbic fatty acid amide hydrolase. Neuropsychopharmacology 2021; 46:992-1003. [PMID: 33452437 PMCID: PMC8115350 DOI: 10.1038/s41386-020-00939-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/11/2020] [Accepted: 12/06/2020] [Indexed: 01/29/2023]
Abstract
Peripheral inflammatory conditions, including those localized to the gastrointestinal tract, are highly comorbid with psychiatric disorders such as anxiety and depression. These behavioral symptoms are poorly managed by conventional treatments for inflammatory diseases and contribute to quality of life impairments. Peripheral inflammation is associated with sustained elevations in circulating glucocorticoid hormones, which can modulate central processes, including those involved in the regulation of emotional behavior. The endocannabinoid (eCB) system is exquisitely sensitive to these hormonal changes and is a significant regulator of emotional behavior. The impact of peripheral inflammation on central eCB function, and whether this is related to the development of these behavioral comorbidities remains to be determined. To examine this, we employed the trinitrobenzene sulfonic acid-induced model of colonic inflammation (colitis) in adult, male, Sprague Dawley rats to produce sustained peripheral inflammation. Colitis produced increases in behavioral measures of anxiety and elevations in circulating corticosterone. These alterations were accompanied by elevated hydrolytic activity of the enzyme fatty acid amide hydrolase (FAAH), which hydrolyzes the eCB anandamide (AEA), throughout multiple corticolimbic brain regions. This elevation of FAAH activity was associated with broad reductions in the content of AEA, whose decline was driven by central corticotropin releasing factor type 1 receptor signaling. Colitis-induced anxiety was reversed following acute central inhibition of FAAH, suggesting that the reductions in AEA produced by colitis contributed to the generation of anxiety. These data provide a novel perspective for the pharmacological management of psychiatric comorbidities of chronic inflammatory conditions through modulation of eCB signaling.
Collapse
Affiliation(s)
- Haley A. Vecchiarelli
- grid.22072.350000 0004 1936 7697Neuroscience Graduate Program, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB T2N4N1 Canada
| | - Maria Morena
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Psychiatry, University of Calgary, Calgary, AB T2N4N1 Canada
| | - Catherine M. Keenan
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N4N1 Canada
| | - Vincent Chiang
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Psychiatry, University of Calgary, Calgary, AB T2N4N1 Canada
| | - Kaitlyn Tan
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Psychiatry, University of Calgary, Calgary, AB T2N4N1 Canada
| | - Min Qiao
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Psychiatry, University of Calgary, Calgary, AB T2N4N1 Canada
| | - Kira Leitl
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Psychiatry, University of Calgary, Calgary, AB T2N4N1 Canada
| | - Alessia Santori
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Psychiatry, University of Calgary, Calgary, AB T2N4N1 Canada
| | - Quentin J. Pittman
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N4N1 Canada
| | - Keith A. Sharkey
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N4N1 Canada
| | - Matthew N. Hill
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N4N1 Canada ,grid.22072.350000 0004 1936 7697Department of Psychiatry, University of Calgary, Calgary, AB T2N4N1 Canada
| |
Collapse
|
30
|
Giacobbe J, Marrocu A, Di Benedetto MG, Pariante CM, Borsini A. A systematic, integrative review of the effects of the endocannabinoid system on inflammation and neurogenesis in animal models of affective disorders. Brain Behav Immun 2021; 93:353-367. [PMID: 33383145 DOI: 10.1016/j.bbi.2020.12.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/09/2022] Open
Abstract
The endocannabinoid (eCB) system is considered relevant in the pathophysiology of affective disorders, and a potential therapeutic target, as its hypoactivity is considered an important risk factor of depression. However, the biological mechanisms whereby the eCB system affects mood remain elusive. Through a systematic review, thirty-seven articles were obtained from the PubMed/Medline, Web of Science, Embase, PsychInfo, and CINAHL databases, investigating the role of the eCB system on the immune system and neurogenesis, as well as resulting behavioural effects in rodent models of affective disorders. Overall, activation of the eCB system appears to decrease depressive-like behaviour and to be anti-inflammatory, while promoting neuro- and synaptogenesis in various models. Activation of cannabinoid receptors (CBRs) is shown to be crucial in improving depressive-like and anxiety-like behaviour, although cannabidiol administration suggests a role of additional mechanisms. CB1R signalling, as well as fatty acid amide hydrolase (FAAH) inhibition, are associated with decreased pro-inflammatory cytokines. Moreover, activation of CBRs is required for neurogenesis, which is also upregulated by FAAH inhibitors. This review is the first to assess the association between the eCB system, immune system and neurogenesis, alongside behavioural outcomes, across rodent models of affective disorders. We confirm the therapeutic potential of eCB system activation in depression and anxiety, highlighting immunoregulation as an important mechanism whereby dysfunctional behaviour and neurogenesis can be improved.
Collapse
Affiliation(s)
- Juliette Giacobbe
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, United Kingdom
| | - Alessia Marrocu
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, United Kingdom
| | - Maria Grazia Di Benedetto
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, United Kingdom; Biological Psychiatry Unit, IRCCS Fatebenefratelli S. Giovanni di Dio, Brescia, Italy
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, United Kingdom
| | - Alessandra Borsini
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, United Kingdom.
| |
Collapse
|
31
|
Zou M, Liu Y, Xie S, Wang L, Li D, Li L, Wang F, Zhang Y, Xia W, Sun C, Wu L. Alterations of the endocannabinoid system and its therapeutic potential in autism spectrum disorder. Open Biol 2021; 11:200306. [PMID: 33529552 PMCID: PMC8061688 DOI: 10.1098/rsob.200306] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of developmental disabilities, the aetiology of which remains elusive. The endocannabinoid (eCB) system modulates neurotransmission and neuronal plasticity. Evidence points to the involvement of this neuromodulatory system in the pathophysiology of ASD. We investigated whether there is a disruption to the eCB system in ASD and whether pharmacological modulation of the eCB system might offer therapeutic potential. We examined three major components of the eCB system—endogenous cannabinoids, their receptors and associated enzymes—in ASD children as well as in the valproic acid (VPA) induced animal model in autism. Furthermore, we specifically increased 2-arachidonoylglycerol (2-AG) levels by administering JZL184, a selective inhibitor of monoacylglycerol lipase which is the hydrolytic enzyme for 2-AG, to examine ASD-like behaviours in VPA-induced rats. Results showed that autistic children and VPA-induced rats exhibited reduced eCB content, increased degradation of enzymes and upregulation of CBRs. We found that repetitive and stereotypical behaviours, hyperactivity, sociability, social preference and cognitive functioning improved after acute and chronic JZL184 treatment. The major efficacy of JZL184 was observed after administration of a dosage regimen of 3 mg kg−1, which affected both the eCB system and ASD-like behaviours. In conclusion, a reduced eCB signalling was observed in autistic children and in the ASD animal model, and boosting 2-AG could ameliorate ASD-like phenotypes in animals. Collectively, the results suggested a novel approach to ASD treatment.
Collapse
Affiliation(s)
- Mingyang Zou
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Yu Liu
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Shu Xie
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Luxi Wang
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Dexin Li
- Department of Children Psychology, Zhuhai Maternal and Child Health Care Hospital, Zhuhai 519001, People's Republic of China
| | - Ling Li
- Office of Leading Group for Control and Prevention of Major Diseases and Infectious diseases, Dezhou Center for Disease Control and Prevention, Dezhou 253011, People's Republic of China
| | - Feng Wang
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Yujue Zhang
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Wei Xia
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Caihong Sun
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Lijie Wu
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin 150081, People's Republic of China
| |
Collapse
|
32
|
Borikar SP, Dongare SI, Danao KR. Reversal of lipopolysaccharide-induced learning and memory deficits by agmatine in mice. Int J Neurosci 2021; 132:621-632. [PMID: 33089716 DOI: 10.1080/00207454.2020.1830086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
MATERIALS AND METHODS Learning and memory functions in animals were evaluated by using Novel object recognition (NOR) and Morris water maze (MWM) tests. Following 7 days of LPS administration, animals were subjected to NOR test on Day-8 and MWM test on Days-9 to 13 for the assessment of recognition and spatial learning and memory, respectively. RESULTS LPS administration produced significant deficits in recognition and spatial memory in mice after seven days of LPS administration. In LPS pre-treated mice, agmatine treatment on Day-8 resulted in the increased exploration to the novel object. Agmatine treatment (Day 8-12) in mice showed reduction in the escape latency and time spent in the target quadrant (probe trial) in the MWM test. However, co-administration of agmatine with LPS in mice for 7 days showed higher discrimination index in NOR test on Day-8. This co-administration also decreased escape latency and time spent in the target quadrant in MWM test on Days 9-13 as compared to LPS control group. CONCLUSION Results implies the protective and curative effects of agmatine against LPS-induced loss of memory functions in experimental animals.HighlightsSubchronic but not acute lipopolysaccharides induce memory deficitsLipopolysaccharides impairs recognition and spatial memory in mice.Agmatine prevents lipopolysaccharides-induced loss of memory.Agmatine reverses deficits in learning and memory by lipopolysaccharides.
Collapse
Affiliation(s)
- Sachin P Borikar
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research Shirpur, Dist-Dhule, Maharashtra, India
| | - Shruti I Dongare
- Department of Pharmaceutics, Gurunanak College of Pharmacy, Nagpur, Maharashtra, India
| | - Kishor R Danao
- Department of Pharmaceutical Chemistry, Dadasaheb Balpande College of Pharmacy, Nagpur, Maharashtra, India
| |
Collapse
|
33
|
Portugalov A, Akirav I. Do Adolescent Exposure to Cannabinoids and Early Adverse Experience Interact to Increase the Risk of Psychiatric Disorders: Evidence from Rodent Models. Int J Mol Sci 2021; 22:ijms22020730. [PMID: 33450928 PMCID: PMC7828431 DOI: 10.3390/ijms22020730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/30/2022] Open
Abstract
There have been growing concerns about the protracted effects of cannabis use in adolescents on emotion and cognition outcomes, motivated by evidence of growing cannabis use in adolescents, evidence linking cannabis use to various psychiatric disorders, and the increasingly perceived notion that cannabis is harmless. At the same time, studies suggest that cannabinoids may have therapeutic potential against the impacts of stress on the brain and behavior, and that young people sometimes use cannabinoids to alleviate feelings of depression and anxiety (i.e., “self-medication”). Exposure to early adverse life events may predispose individuals to developing psychopathology in adulthood, leading researchers to study the causality between early life factors and cognitive and emotional outcomes in rodent models and to probe the underlying mechanisms. In this review, we aim to better understand the long-term effects of cannabinoids administered in sensitive developmental periods (mainly adolescence) in rodent models of early life stress. We suggest that the effects of cannabinoids on emotional and cognitive function may vary between different sensitive developmental periods. This could potentially affect decisions regarding the use of cannabinoids in clinical settings during the early stages of development and could raise questions regarding educating the public as to potential risks associated with cannabis use.
Collapse
Affiliation(s)
- Anna Portugalov
- Department of Psychology, School of Psychological Sciences, University of Haifa, 3498838 Haifa, Israel;
- The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, 3498838 Haifa, Israel
| | - Irit Akirav
- Department of Psychology, School of Psychological Sciences, University of Haifa, 3498838 Haifa, Israel;
- The Integrated Brain and Behavior Research Center (IBBR), University of Haifa, 3498838 Haifa, Israel
- Correspondence:
| |
Collapse
|
34
|
Su T, Yan Y, Li Q, Ye J, Pei L. Endocannabinoid System Unlocks the Puzzle of Autism Treatment via Microglia. Front Psychiatry 2021; 12:734837. [PMID: 34744824 PMCID: PMC8568770 DOI: 10.3389/fpsyt.2021.734837] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/27/2021] [Indexed: 01/15/2023] Open
Abstract
Autism spectrum disorder (ASD) is a serious neurodevelopmental disorder and characterized by early childhood-onset impairments in social interaction and communication, restricted and repetitive patterns of behavior or interests. So far there is no effective treatment for ASD, and the pathogenesis of ASD remains unclear. Genetic and epigenetic factors have been considered to be the main cause of ASD. It is known that endocannabinoid and its receptors are widely distributed in the central nervous system, and provide a positive and irreversible change toward a more physiological neurodevelopment. Recently, the endocannabinoid system (ECS) has been found to participate in the regulation of social reward behavior, which has attracted considerable attention from neuroscientists and neurologists. Both animal models and clinical studies have shown that the ECS is a potential target for the treatment of autism, but the mechanism is still unknown. In the brain, microglia express a complete ECS signaling system. Studies also have shown that modulating ECS signaling can regulate the functions of microglia. By comprehensively reviewing previous studies and combining with our recent work, this review addresses the effects of targeting ECS on microglia, and how this can contribute to maintain the positivity of the central nervous system, and thus improve the symptoms of autism. This will provide insights for revealing the mechanism and developing new treatment strategies for autism.
Collapse
Affiliation(s)
- Tangfeng Su
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Yan
- Department of Neurology, People's Hospital of Dongxihu District, Wuhan, China
| | - Qiang Li
- Exchange, Development and Service Center for Science and Technology Talents, The Ministry of Science and Technology, Beijing, China
| | - Jiacai Ye
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Lei Pei
- Collaborative Innovation Center for Brain Science, The Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, China.,Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Anesthesiology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
35
|
Smith CJ, Bilbo SD. Sickness and the Social Brain: Love in the Time of COVID. Front Psychiatry 2021; 12:633664. [PMID: 33692712 PMCID: PMC7937950 DOI: 10.3389/fpsyt.2021.633664] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
As a highly social species, inclusion in social networks and the presence of strong social bonds are critical to our health and well-being. Indeed, impaired social functioning is a component of numerous neuropsychiatric disorders including depression, anxiety, and substance use disorder. During the current COVID-19 pandemic, our social networks are at risk of fracture and many are vulnerable to the negative consequences of social isolation. Importantly, infection itself leads to changes in social behavior as a component of "sickness behavior." Furthermore, as in the case of COVID-19, males and females often differ in their immunological response to infection, and, therefore, in their susceptibility to negative outcomes. In this review, we discuss the many ways in which infection changes social behavior-sometimes to the benefit of the host, and in some instances for the sake of the pathogen-in species ranging from eusocial insects to humans. We also explore the neuroimmune mechanisms by which these changes in social behavior occur. Finally, we touch upon the ways in which the social environment (group living, social isolation, etc.) shapes the immune system and its ability to respond to challenge. Throughout we emphasize how males and females differ in their response to immune activation, both behaviorally and physiologically.
Collapse
Affiliation(s)
- Caroline J Smith
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States
| |
Collapse
|
36
|
Folkes OM, Báldi R, Kondev V, Marcus DJ, Hartley ND, Turner BD, Ayers JK, Baechle JJ, Misra MP, Altemus M, Grueter CA, Grueter BA, Patel S. An endocannabinoid-regulated basolateral amygdala-nucleus accumbens circuit modulates sociability. J Clin Invest 2020; 130:1728-1742. [PMID: 31874107 DOI: 10.1172/jci131752] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 12/18/2019] [Indexed: 12/24/2022] Open
Abstract
Deficits in social interaction (SI) are a core symptom of autism spectrum disorders (ASDs); however, treatments for social deficits are notably lacking. Elucidating brain circuits and neuromodulatory signaling systems that regulate sociability could facilitate a deeper understanding of ASD pathophysiology and reveal novel treatments for ASDs. Here we found that in vivo optogenetic activation of the basolateral amygdala-nucleus accumbens (BLA-NAc) glutamatergic circuit reduced SI and increased social avoidance in mice. Furthermore, we found that 2-arachidonoylglycerol (2-AG) endocannabinoid signaling reduced BLA-NAc glutamatergic activity and that pharmacological 2-AG augmentation via administration of JZL184, a monoacylglycerol lipase inhibitor, blocked SI deficits associated with in vivo BLA-NAc stimulation. Additionally, optogenetic inhibition of the BLA-NAc circuit markedly increased SI in the Shank3B-/- mouse, an ASD model with substantial SI impairment, without affecting SI in WT mice. Finally, we demonstrated that JZL184 delivered systemically or directly to the NAc also normalized SI deficits in Shank3B-/- mice, while ex vivo JZL184 application corrected aberrant NAc excitatory and inhibitory neurotransmission and reduced BLA-NAc-elicited feed-forward inhibition of NAc neurons in Shank3B-/- mice. These data reveal circuit-level and neuromodulatory mechanisms regulating social function relevant to ASDs and suggest 2-AG augmentation could reduce social deficits via modulation of excitatory and inhibitory neurotransmission in the NAc.
Collapse
Affiliation(s)
- Oakleigh M Folkes
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Pharmacology and
| | - Rita Báldi
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Veronika Kondev
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - David J Marcus
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Nolan D Hartley
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Brandon D Turner
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jade K Ayers
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jordan J Baechle
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maya P Misra
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Megan Altemus
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Carrie A Grueter
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Brad A Grueter
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sachin Patel
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Pharmacology and.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
37
|
Pandina G, Ring RH, Bangerter A, Ness S. Current Approaches to the Pharmacologic Treatment of Core Symptoms Across the Lifespan of Autism Spectrum Disorder. Psychiatr Clin North Am 2020; 43:629-645. [PMID: 33126999 DOI: 10.1016/j.psc.2020.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There are no approved medications for autism spectrum disorder (ASD) core symptoms. However, given the significant clinical need, children and adults with ASD are prescribed medication off label for core or associated conditions, sometimes based on limited evidence for effectiveness. Recent developments in the understanding of biologic basis of ASD have led to novel targets with potential to impact core symptoms, and several clinical trials are underway. Heterogeneity in course of development, co-occurring conditions, and age-related treatment response variability hampers study outcomes. Novel measures and approaches to ASD clinical trial design will help in development of effective pharmacologic treatments.
Collapse
Affiliation(s)
- Gahan Pandina
- Janssen Research & Development, LLC, 1125 Trenton Harbouron Road, Titusville, NJ 08560, USA.
| | | | - Abigail Bangerter
- Janssen Research & Development, LLC, 1125 Trenton Harbouron Road, Titusville, NJ 08560, USA
| | - Seth Ness
- Janssen Research & Development, LLC, 1125 Trenton Harbouron Road, Titusville, NJ 08560, USA
| |
Collapse
|
38
|
Neonatal immune challenge induces female-specific changes in social behavior and somatostatin cell number. Brain Behav Immun 2020; 90:332-345. [PMID: 32860938 PMCID: PMC7556772 DOI: 10.1016/j.bbi.2020.08.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/20/2022] Open
Abstract
Decreases in social behavior are a hallmark aspect of acute "sickness behavior" in response to infection. However, immune insults that occur during the perinatal period may have long-lasting consequences for adult social behavior by impacting the developmental organization of underlying neural circuits. Microglia, the resident immune cells of the central nervous system, are sensitive to immune stimulation and play a critical role in the developmental sculpting of neural circuits, making them likely mediators of this process. Here, we investigated the impact of a postnatal day (PND) 4 lipopolysaccharide (LPS) challenge on social behavior in adult mice. Somewhat surprisingly, neonatal LPS treatment decreased sociability in adult female, but not male mice. LPS-treated females also displayed reduced social interaction and social memory in a social discrimination task as compared to saline-treated females. Somatostatin (SST) interneurons within the anterior cingulate cortex (ACC) have recently been suggested to modulate a variety of social behaviors. Interestingly, the female-specific changes in social behavior observed here were accompanied by an increase in SST interneuron number in the ACC. Finally, these changes in social behavior and SST cell number do not appear to depend on microglial inflammatory signaling, because microglia-specific genetic knock-down of myeloid differentiation response protein 88 (MyD88; the removal of which prevents LPS from increasing proinflammatory cytokines such as TNFα and IL-1β) did not prevent these LPS-induced changes. This study provides novel evidence for enduring effects of neonatal immune activation on social behavior and SST interneurons in females, largely independent of microglial inflammatory signaling.
Collapse
|
39
|
Zajkowska Z, Borsini A, Nikkheslat N, Russell A, Romano GF, Tomassi S, Hepgul N, Forton D, Agarwal K, Hotopf M, Mondelli V, Zunszain P, Pariante CM. Differential effect of interferon-alpha treatment on AEA and 2-AG levels. Brain Behav Immun 2020; 90:248-258. [PMID: 32860939 PMCID: PMC7575143 DOI: 10.1016/j.bbi.2020.08.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/14/2020] [Accepted: 08/23/2020] [Indexed: 12/31/2022] Open
Abstract
The endocannabinoid (eCB) system is one of the key players in immunoregulation, and reduced activity of the eCB system has been linked with depressive-like behaviours in animal studies and depression in clinical samples. There is a well-established link between immune activation and depression, such as following the administration of the pro-inflammatory cytokine, interferon-α (IFN-α), used to treat hepatitis C viral (HCV) infection. However, the role of peripheral endocannabinoids (eCBs), anandamide (AEA) and 2-arachidonoylglycerol (2-AG), following immunotherapy with IFN-α and in IFN-α -induced depression, have not been examined yet. In this study, we investigated whether circulating AEA and 2-AG were modified by treatment with IFN-α and whether they were involved in the development of IFN-α-induced depression. We also explored whether circulating eCBs were associated with peripheral cytokines during and after IFN-α treatment. We measured serum concentrations of AEA and 2-AG using High Performance Liquid Chromatography with Tandem Mass Spectrometry, and serum concentrations of cytokines using Meso Scale Discovery electrochemiluminescence V-PLEX assay, in 70 patients with HCV infection and 41 healthy subjects. We assessed HCV patients at baseline, IFN-α-treatment weeks (TW) 4 and 24, end of treatment (END) and at six months follow-up (FU). We assessed depression using M.I.N.I. International Neuropsychiatric Interview. We found a different pattern of change in peripheral AEA and 2-AG during and after IFN-α treatment. Whilst 2-AG increased earlier in immunotherapy (TW4), remained elevated throughout treatment, and reduced at six months follow-up (FU), AEA increased later in treatment (TW24) and remained elevated six months post-treatment. We also found that baseline levels of AEA were lower in HCV patients compared with healthy controls, whereas there were no differences in 2-AG levels. Interestingly, AEA, but not 2-AG, was significantly, negatively correlated with interleukin (IL)-2 and IL-17a at six months follow-up. We did not find any difference in both eCBs between patients with and without IFN-α-induced depression, at any time point. Our findings suggest that AEA and 2-AG are involved in different stages of immunoregulation following IFN-α treatment, where AEA might be involved in chronic inflammation. Lack of association between peripheral eCBs and IFN-α-induced depression suggests that different biological mechanisms may underpin inflammation-induced depression compared with classic "psychiatric" depression, or that any changes in the eCB system in depression may not be captured by peripheral AEA and 2-AG.
Collapse
Affiliation(s)
- Zuzanna Zajkowska
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK.
| | - Alessandra Borsini
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Naghmeh Nikkheslat
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Alice Russell
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Graziella F Romano
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Simona Tomassi
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Nilay Hepgul
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Daniel Forton
- Department of Gastroenterology & Hepatology, St George's University of London, UK
| | - Kosh Agarwal
- Institute of Liver Studies, King's College Hospital, UK
| | - Matthew Hotopf
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK; National Institute for Health Research (NIHR), Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, UK
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK; National Institute for Health Research (NIHR), Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, UK
| | - Patricia Zunszain
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK; National Institute for Health Research (NIHR), Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, UK
| |
Collapse
|
40
|
Druggable Targets in Endocannabinoid Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1274:177-201. [PMID: 32894511 DOI: 10.1007/978-3-030-50621-6_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cannabis and cannabinoid-based extracts have long been utilized for their perceived therapeutic value, and support for the legalization of cannabis for medicinal purposes continues to increase worldwide. Since the discovery of Δ9-tetrahydrocannabinol (THC) as the primary psychoactive component of cannabis over 50 years ago, substantial effort has been directed toward detection of endogenous mediators of cannabinoid activity. The discovery of anandamide and 2-arachidonoylglycerol as two endogenous lipid mediators of cannabinoid-like effects (endocannabinoids) has inspired exponential growth in our understanding of this essential pathway, as well as the pathological conditions that result from dysregulated endocannabinoid signaling. This review examines current knowledge of the endocannabinoid system including metabolic enzymes involved in biosynthesis and degradation and their receptors, and evaluates potential druggable targets for therapeutic intervention.
Collapse
|
41
|
van Egmond N, Straub VM, van der Stelt M. Targeting Endocannabinoid Signaling: FAAH and MAG Lipase Inhibitors. Annu Rev Pharmacol Toxicol 2020; 61:441-463. [PMID: 32867595 DOI: 10.1146/annurev-pharmtox-030220-112741] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Inspired by the medicinal properties of the plant Cannabis sativa and its principal component (-)-trans-Δ9-tetrahydrocannabinol (THC), researchers have developed a variety of compounds to modulate the endocannabinoid system in the human brain. Inhibitors of fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), which are the enzymes responsible for the inactivation of the endogenous cannabinoids anandamide and 2-arachidonoylglycerol, respectively, may exert therapeutic effects without inducing the adverse side effects associated with direct cannabinoid CB1 receptor stimulation by THC. Here we review the FAAH and MAGL inhibitors that have reached clinical trials, discuss potential caveats, and provide an outlook on where the field is headed.
Collapse
Affiliation(s)
- Noëlle van Egmond
- Department of Molecular Physiology, Leiden University, 2333 CC Leiden, The Netherlands;
| | - Verena M Straub
- Department of Molecular Physiology, Leiden University, 2333 CC Leiden, The Netherlands;
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden University, 2333 CC Leiden, The Netherlands;
| |
Collapse
|
42
|
PiDose: an open-source system for accurate and automated oral drug administration to group-housed mice. Sci Rep 2020; 10:11584. [PMID: 32665577 PMCID: PMC7360602 DOI: 10.1038/s41598-020-68477-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/25/2020] [Indexed: 12/31/2022] Open
Abstract
Drug treatment studies in laboratory mice typically employ manual administration methods such as injection or gavage, which can be time-consuming to perform over long periods and cause substantial stress in animals. These stress responses may mask or enhance treatment effects, increasing the risk of false positive or negative results and decreasing reliability. To address the lack of an automated method for drug treatment in group-housed mice, we have developed PiDose, a home-cage attached device that weighs individual animals and administers a daily dosage of drug solution based on each animal’s bodyweight through their drinking water. Group housed mice are identified through the use of RFID tagging and receive both regular water and drug solution drops by licking at a spout within the PiDose module. This system allows animals to be treated over long periods (weeks to months) in a fully automated fashion, with high accuracy and minimal experimenter interaction. PiDose is low-cost and fully open-source and should prove useful for researchers in both translational and basic research.
Collapse
|
43
|
Pandina G, Ring RH, Bangerter A, Ness S. Current Approaches to the Pharmacologic Treatment of Core Symptoms Across the Lifespan of Autism Spectrum Disorder. Child Adolesc Psychiatr Clin N Am 2020; 29:301-317. [PMID: 32169264 DOI: 10.1016/j.chc.2019.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There are no approved medications for autism spectrum disorder (ASD) core symptoms. However, given the significant clinical need, children and adults with ASD are prescribed medication off label for core or associated conditions, sometimes based on limited evidence for effectiveness. Recent developments in the understanding of biologic basis of ASD have led to novel targets with potential to impact core symptoms, and several clinical trials are underway. Heterogeneity in course of development, co-occurring conditions, and age-related treatment response variability hampers study outcomes. Novel measures and approaches to ASD clinical trial design will help in development of effective pharmacologic treatments.
Collapse
Affiliation(s)
- Gahan Pandina
- Janssen Research & Development, LLC, 1125 Trenton Harbouron Road, Titusville, NJ 08560, USA.
| | | | - Abigail Bangerter
- Janssen Research & Development, LLC, 1125 Trenton Harbouron Road, Titusville, NJ 08560, USA
| | - Seth Ness
- Janssen Research & Development, LLC, 1125 Trenton Harbouron Road, Titusville, NJ 08560, USA
| |
Collapse
|
44
|
Gonczarowska N, Tomaz C, Caixeta FV, Malcher-Lopes R, Barros M, Nishijo H, Maior RS. CB1 receptor antagonism in capuchin monkeys alters social interaction and aversive memory extinction. Psychopharmacology (Berl) 2019; 236:3413-3419. [PMID: 31250073 DOI: 10.1007/s00213-019-05305-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 06/11/2019] [Indexed: 12/13/2022]
Abstract
RATIONALE The endocannabinoid system (eCS) is an important modulator of social anxiety and social reward, as well as memory functions. OBJECTIVES The present study evaluated the role of eCS in social interactions and aversive memory extinction in capuchin monkeys (Sapajus spp.) by blocking the cannabinoid type 1 receptor (CB1r). METHODS In experiment 1, spontaneous social and non-social behaviors of five capuchin males, each one living in triads with two other females, were observed after AM251 treatment (vehicle, 0.3, 1.0, and 3.0 mg/kg; i.m.). In experiment 2, seven male capuchin monkeys were trained to reach for a reward inside a wooden box. After training, they were given either vehicle or a 3.0-mg/kg i.m. dose of AM251 before a single aversive encounter with a live snake in the box. The latency to return to reach the reward inside the box in subsequent trials was measured. RESULTS The 3.0-mg/kg dose significantly increased the time spent performing self-directed behaviors, while decreasing that of social interactions. No changes were observed in vigilance or locomotion. AM251 increased the latency to reach the reward after the aversive encounter. CONCLUSION Taken together, these results suggest that CB1r antagonism induces social deficits without increasing anxiety levels and impairs the extinction of aversive memories. This behavioral profile in monkeys underscores the potential involvement of eCS signaling in the deficits observed in autism spectrum disorders.
Collapse
Affiliation(s)
- Natália Gonczarowska
- Primate Center, Institute of Biology, University of Brasília, Brasilia, DF, Brazil.,Laboratory of Neuroscience and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasilia, DF, 70910-900, Brazil
| | - Carlos Tomaz
- Primate Center, Institute of Biology, University of Brasília, Brasilia, DF, Brazil.,Laboratory of Neuroscience and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasilia, DF, 70910-900, Brazil.,Laboratory of Neuroscience & Behavior, Universidade Ceuma, São Luis, MA, Brazil
| | - Fabio V Caixeta
- Laboratory of Neuroscience and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasilia, DF, 70910-900, Brazil
| | - Renato Malcher-Lopes
- Laboratory of Neuroscience and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasilia, DF, 70910-900, Brazil
| | - Marilia Barros
- Primate Center, Institute of Biology, University of Brasília, Brasilia, DF, Brazil.,Department of Pharmacy, School of Health Sciences, University of Brasilia, Brasilia, DF, Brazil
| | - Hisao Nishijo
- System Emotional Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, 2630, Japan
| | - Rafael S Maior
- Primate Center, Institute of Biology, University of Brasília, Brasilia, DF, Brazil. .,Laboratory of Neuroscience and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasilia, DF, 70910-900, Brazil.
| |
Collapse
|
45
|
Araujo DJ, Tjoa K, Saijo K. The Endocannabinoid System as a Window Into Microglial Biology and Its Relationship to Autism. Front Cell Neurosci 2019; 13:424. [PMID: 31619967 PMCID: PMC6759510 DOI: 10.3389/fncel.2019.00424] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/03/2019] [Indexed: 11/17/2022] Open
Abstract
Microglia are the resident, innate immune cells of the central nervous system (CNS) and are critical in managing CNS injuries and infections. Microglia also maintain CNS homeostasis by influencing neuronal development, viability, and function. However, aberrant microglial activity and phenotypes are associated with CNS pathology, including autism spectrum disorder (ASD). Thus, improving our knowledge of microglial regulation could provide insights into the maintenance of CNS homeostasis as well as the prevention and treatment of ASD. Control of microglial activity is in part overseen by small, lipid-derived molecules known as endogenous cannabinoids (endocannabinoids). Endocannabinoids are one component of the endocannabinoid system (ECS), which also includes the enzymes that metabolize these ligands, in addition to cannabinoid receptor 1 (CB1) and 2 (CB2). Interestingly, increased ECS signaling leads to an anti-inflammatory, neuroprotective phenotype in microglia. Here, we review the literature and propose that ECS signaling represents a largely untapped area for understanding microglial biology and its relationship to ASD, with special attention paid to issues surrounding the use of recreational cannabis (marijuana). We also discuss major questions within the field and suggest directions for future research.
Collapse
Affiliation(s)
- Daniel John Araujo
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Karensa Tjoa
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Kaoru Saijo
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA, United States.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
46
|
Perkins AE, Varlinskaya EI, Deak T. From adolescence to late aging: A comprehensive review of social behavior, alcohol, and neuroinflammation across the lifespan. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 148:231-303. [PMID: 31733665 DOI: 10.1016/bs.irn.2019.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The passage of time dictates the pace at which humans and other organisms age but falls short of providing a complete portrait of how environmental, lifestyle and underlying biological processes contribute to senescence. Two fundamental features of the human experience that change dramatically across the lifespan include social interactions and, for many, patterns of alcohol consumption. Rodent models show great utility for understanding complex interactions among aging, social behavior and alcohol use and abuse, yet little is known about the neural changes in late aging that contribute to the natural decline in social behavior. Here, we posit that aging-related neuroinflammation contributes to the insipid loss of social motivation across the lifespan, an effect that is exacerbated by patterns of repeated alcohol consumption observed in many individuals. We provide a comprehensive review of (i) neural substrates crucial for the expression of social behavior under non-pathological conditions; (ii) unique developmental/lifespan vulnerabilities that may contribute to the divergent effects of low-and high-dose alcohol exposure; and (iii) aging-associated changes in neuroinflammation that may sit at the intersection between social processes and alcohol exposure. In doing so, we provide an overview of correspondence between lifespan/developmental periods between common rodent models and humans, give careful consideration to model systems used to aptly probe social behavior, identify points of coherence between human and animal models, and point toward a multitude of unresolved issues that should be addressed in future studies. Together, the combination of low-dose and high-dose alcohol effects serve to disrupt the normal development and maintenance of social relationships, which are critical for both healthy aging and quality of life across the lifespan. Thus, a more complete understanding of neural systems-including neuroinflammatory processes-which contribute to alcohol-induced changes in social behavior will provide novel opportunities and targets for promoting healthy aging.
Collapse
Affiliation(s)
- Amy E Perkins
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| | - Elena I Varlinskaya
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States.
| |
Collapse
|
47
|
Late Brain Involvement after Neonatal Immune Activation. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9573248. [PMID: 31467920 PMCID: PMC6699266 DOI: 10.1155/2019/9573248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/07/2019] [Indexed: 01/18/2023]
Abstract
The neonatal immune system is still immature, which makes it more susceptible to the infectious agents. Neonatal immune activation is associated with increased permeability of the blood-brain barrier, causing an inflammatory cascade in the CNS and altering behavioral and neurochemical parameters. One of the hypotheses that has been studied is that neuroinflammation may be involved in neurodegenerative processes, such as Alzheimer's disease (AD). We evaluate visuospatial memory, cytokines levels, and the expression of tau and GSK-3β proteins in hippocampus and cortex of animals exposed to neonatal endotoxemia. C57BL/6 mice aging two days received a single injection of subcutaneous lipopolysaccharide (LPS). At 60,120, and 180 days of age, visual-spatial memory was evaluated and the hippocampus and cortex were dissected to evaluate the cytokines levels and expression of tau and GSK-3β proteins. The animals exposed to LPS in the neonatal period present with visuospatial memory impairment at 120 and 180 days of age. Here there was an increase of TNF-α and IL-1β levels in the hippocampus and cortex only at 60 days of age. Here there was an increase in the expression of GSK-3β in hippocampus of the animals at 60, 120, and 180 days of age. In the cortex, this increase occurred in the 120 and 180 days of age. Tau protein expression was high in hippocampus and cortex at 120 days of age and in hippocampus at 180 days of age. The data observed show that neonatal immune activation may be associated with visuospatial memory impairment, neuroinflammation, and increased expression of GSK-3β and Tau proteins in the long term.
Collapse
|
48
|
|
49
|
Early Life Inflammation Increases CA1 Pyramidal Neuron Excitability in a Sex and Age Dependent Manner through a Chloride Homeostasis Disruption. J Neurosci 2019; 39:7244-7259. [PMID: 31308096 DOI: 10.1523/jneurosci.2973-18.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 05/31/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023] Open
Abstract
Early life, systemic inflammation causes long-lasting changes in behavior. To unmask possible mechanisms associated with this phenomenon, we asked whether the intrinsic membrane properties in hippocampal neurons were altered as a consequence of early life inflammation. C57BL/6 mice were bred in-house and both male and female pups from multiple litters were injected with lipopolysaccharide (LPS; 100 μg/kg, i.p.) or vehicle at postnatal day (P)14, and kept until adolescence (P35-P45) or adulthood (P60-P70), when brain slices were prepared for whole-cell and perforated-patch recordings from CA1 hippocampal pyramidal neurons. In neurons of adult male mice pretreated with LPS, the number of action potentials elicited by depolarizing current pulses was significantly increased compared with control neurons, concomitant with increased input resistance, and a lower action potential threshold. Although these changes were not associated with changes in relevant sodium channel expression or differences in capacitance or dendritic architecture, they were linked to a mechanism involving intracellular chloride overload, revealed through a depolarized GABA reversal potential and increased expression of the chloride transporter, NKCC1. In contrast, no significant changes were observed in neurons of adult female mice pretreated with LPS, nor in adolescent mice of either sex. These data uncover a potential mechanism involving neonatal inflammation-induced plasticity in chloride homeostasis, which may contribute to early life inflammation-induced behavioral alterations.SIGNIFICANCE STATEMENT Early life inflammation results in long-lasting changes in many aspects of adult physiology. In this paper we reveal that a brief exposure to early life peripheral inflammation with LPS increases excitability in hippocampal neurons in a sex- and age-dependent manner through a chloride homeostasis disruption. As this hyperexcitability was only seen in adult males, and not in adult females or adolescent animals of either sex, it raises the possibility of a hormonal interaction with early life inflammation. Furthermore, as neonatal inflammation is a normal feature of early life in most animals, as well as humans, these findings may be very important for the development of animal models of disease that more appropriately resemble the human condition.
Collapse
|
50
|
Sokol DK, Maloney B, Westmark CJ, Lahiri DK. Novel Contribution of Secreted Amyloid-β Precursor Protein to White Matter Brain Enlargement in Autism Spectrum Disorder. Front Psychiatry 2019; 10:165. [PMID: 31024350 PMCID: PMC6469489 DOI: 10.3389/fpsyt.2019.00165] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/06/2019] [Indexed: 12/27/2022] Open
Abstract
The most replicated neuroanatomical finding in autism is the tendency toward brain overgrowth, especially in younger children. Research shows that both gray and white matter are enlarged. Proposed mechanisms underlying brain enlargement include abnormal inflammatory and neurotrophic signals that lead to excessive, aberrant dendritic connectivity via disrupted pruning and cell adhesion, and enlargement of white matter due to excessive gliogenesis and increased myelination. Amyloid-β protein precursor (βAPP) and its metabolites, more commonly associated with Alzheimer's disease (AD), are also dysregulated in autism plasma and brain tissue samples. This review highlights findings that demonstrate how one βAPP metabolite, secreted APPα, and the ADAM family α-secretases, may lead to increased brain matter, with emphasis on increased white matter as seen in autism. sAPPα and the ADAM family α-secretases contribute to the anabolic, non-amyloidogenic pathway, which is in contrast to the amyloid (catabolic) pathway known to contribute to Alzheimer disease. The non-amyloidogenic pathway could produce brain enlargement via genetic mechanisms affecting mRNA translation and polygenic factors that converge on molecular pathways (mitogen-activated protein kinase/MAPK and mechanistic target of rapamycin/mTOR), promoting neuroinflammation. A novel mechanism linking the non-amyloidogenic pathway to white matter enlargement is proposed: α-secretase and/or sAPPα, activated by ERK receptor signaling activates P13K/AKt/mTOR and then Rho GTPases favoring myelination via oligodendrocyte progenitor cell (OPC) activation of cofilin. Applying known pathways in AD to autism should allow further understanding and provide options for new drug targets.
Collapse
Affiliation(s)
- Deborah K. Sokol
- Pediatrics Section, Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Bryan Maloney
- Indiana Alzheimers Disease Center, Department of Psychiatry, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, United States
| | - Debomoy K. Lahiri
- Indiana Alzheimers Disease Center, Department of Psychiatry, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|