1
|
Dempster EL, Wong CCY, Burrage J, Hannon E, Quattrone D, Trotta G, Rodriguez V, Alameda L, Spinazzola E, Tripoli G, Austin-Zimmerman I, Li Z, Gayer-Anderson C, Freeman TP, Johnson EC, Jongsma HE, Stilo S, La Cascia C, Ferraro L, La Barbera D, Lasalvia A, Tosato S, Tarricone I, D'Andrea G, Galatolo M, Tortelli A, Pompili M, Selten JP, de Haan L, Menezes PR, Del Ben CM, Santos JL, Arrojo M, Bobes J, Sanjuán J, Bernardo M, Arango C, Jones PB, Breen G, Mondelli V, Dazzan P, Iyegbe C, Vassos E, Morgan C, Mukherjee D, van Os J, Rutten B, O'Donovan MC, Sham P, Mill J, Murray R, Di Forti M. Methylomic signature of current cannabis use in two first-episode psychosis cohorts. Mol Psychiatry 2025; 30:1277-1286. [PMID: 39406996 PMCID: PMC11919776 DOI: 10.1038/s41380-024-02689-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 03/20/2025]
Abstract
The rising prevalence and legalisation of cannabis worldwide have underscored the need for a comprehensive understanding of its biological impact, particularly on mental health. Epigenetic mechanisms, specifically DNA methylation, have gained increasing recognition as vital factors in the interplay between risk factors and mental health. This study aimed to explore the effects of current cannabis use and high-potency cannabis on DNA methylation in two independent cohorts of individuals experiencing first-episode psychosis (FEP) compared to control subjects. The combined sample consisted of 682 participants (188 current cannabis users and 494 never users). DNA methylation profiles were generated on blood-derived DNA samples using the Illumina DNA methylation array platform. A meta-analysis across cohorts identified one CpG site (cg11669285) in the CAVIN1 gene that showed differential methylation with current cannabis use, surpassing the array-wide significance threshold, and independent of the tobacco-related epigenetic signature. Furthermore, a CpG site localised in the MCU gene (cg11669285) achieved array-wide significance in an analysis of the effect of high-potency (THC = > 10%) current cannabis use. Pathway and regional analyses identified cannabis-related epigenetic variation proximal to genes linked to immune and mitochondrial function, both of which are known to be influenced by cannabinoids. Interestingly, a model including an interaction term between cannabis use and FEP status identified two sites that were significantly associated with current cannabis use with a nominally significant interaction suggesting that FEP status might moderate how cannabis use affects DNA methylation. Overall, these findings contribute to our understanding of the epigenetic impact of current cannabis use and highlight potential molecular pathways affected by cannabis exposure.
Collapse
Affiliation(s)
- Emma L Dempster
- Department of Clinical & Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK.
| | - Chloe C Y Wong
- Department of Social, Genetic and Developmental Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Joe Burrage
- Department of Clinical & Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Eilis Hannon
- Department of Clinical & Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Diego Quattrone
- Department of Social, Genetic and Developmental Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Giulia Trotta
- Department of Social, Genetic and Developmental Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Victoria Rodriguez
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Luis Alameda
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Edoardo Spinazzola
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Giada Tripoli
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Isabelle Austin-Zimmerman
- Department of Social, Genetic and Developmental Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Zhikun Li
- Department of Social, Genetic and Developmental Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Charlotte Gayer-Anderson
- Department of Health Service and Population Research, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Tom P Freeman
- Addiction and Mental Health Group (AIM), Department of Psychology, University of Bath, Bath, UK
| | - Emma C Johnson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah E Jongsma
- Centre for Transcultural Psychiatry 'Veldzicht', Balkbrug, The Netherlands
| | - Simona Stilo
- Department of Mental Health and Addiction Services, ASP Crotone, Crotone, Italy
| | - Caterina La Cascia
- Biomedicine, Neuroscience and Advanced Diagnostic Department, Psychiatry Section, University of Palermo, Palermo, Italy
| | - Laura Ferraro
- Biomedicine, Neuroscience and Advanced Diagnostic Department, Psychiatry Section, University of Palermo, Palermo, Italy
| | - Daniele La Barbera
- Biomedicine, Neuroscience and Advanced Diagnostic Department, Psychiatry Section, University of Palermo, Palermo, Italy
| | - Antonio Lasalvia
- Section of Psychiatry, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Sarah Tosato
- Section of Psychiatry, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Ilaria Tarricone
- Department of Medical and Surgical Science, Psychiatry Unit, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Giuseppe D'Andrea
- Department of Medical and Surgical Science, Psychiatry Unit, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Michela Galatolo
- Department of Medical and Surgical Science, Psychiatry Unit, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | | | - Maurizio Pompili
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Center, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Jean-Paul Selten
- Rivierduinen Institute for Mental Health Care, Leiden, The Netherlands
| | - Lieuwe de Haan
- Early Psychosis Section, Amsterdam UMC, Academic Medical Centre, University of Amsterdam, Meibergdreef 5, 1105, AZ, Amsterdam, The Netherlands
| | - Paulo Rossi Menezes
- Department of Preventive Medicine, Faculdade de Medicina, Universidade of São Paulo, São Paulo, Brazil
| | - Cristina M Del Ben
- Department of Preventive Medicine, Faculdade de Medicina, Universidade of São Paulo, São Paulo, Brazil
| | - Jose Luis Santos
- Department of Psychiatry, Servicio de Psiquiatría Hospital "Virgen de la Luz", Cuenca, Spain
| | - Manuel Arrojo
- Department of Psychiatry, Psychiatric Genetic Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago de Compostela, Santiago, Spain
| | - Julio Bobes
- Department of Medicine, Psychiatry Area, School of Medicine, Universidad de Oviedo, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Oviedo, Spain
| | - Julio Sanjuán
- Department of Psychiatry, School of Medicine, Universidad de Valencia, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Valencia, Spain
| | - Miguel Bernardo
- Barcelona Clinic Schizophrenia Unit, Neuroscience Institute, Hospital Clinic of Barcelona, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Biomedical Research Networking Centre in Mental Health (CIBERSAM), Barcelona, Spain
| | - Celso Arango
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense, IiSGM, CIBERSAM, Madrid, Spain
| | - Peter B Jones
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Gerome Breen
- Department of Social, Genetic and Developmental Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Valeria Mondelli
- Department of Psychological Medicine, Kings College London, London, UK
| | - Paola Dazzan
- Department of Psychological Medicine, Kings College London, London, UK
| | - Conrad Iyegbe
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evangelos Vassos
- Department of Social, Genetic and Developmental Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Craig Morgan
- Department of Psychological Medicine, Kings College London, London, UK
| | - Diptendu Mukherjee
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Jim van Os
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, South Limburg Mental Health Research and Teaching Network, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department Psychiatry, Brain Centre Rudolf Magnus, Utrecht University Medical Centre, Utrecht, The Netherlands
| | - Bart Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, South Limburg Mental Health Research and Teaching Network, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Michael C O'Donovan
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Pak Sham
- Department of Social, Genetic and Developmental Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Psychiatry, the University of Hong Kong, Hong Kong, China
- Centre for Genomic Sciences, Li KaShing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jonathan Mill
- Department of Clinical & Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Robin Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Marta Di Forti
- Department of Social, Genetic and Developmental Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
2
|
Mashayekhi-Sardoo H, Hedayati-Moghadam M, Baghcheghi Y. Hippocampal Apoptosis: Molecular Mechanisms Triggered by Toxic Cannabinoid Exposure: A Narrative Review. Neurotoxicology 2025:S0161-813X(25)00019-1. [PMID: 40024347 DOI: 10.1016/j.neuro.2025.02.006] [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: 01/05/2025] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Hippocampal apoptosis is increasingly recognized as a significant consequence of toxic cannabinoid exposure, with profound implications for cognitive function and mental health. This narrative review comprehensively examines the molecular mechanisms underlying cannabinoid-induced apoptosis, focusing on the interplay of various bioactive compounds and their effects on neuronal integrity. We begin by discussing the key players in cannabinoid biology, followed by a synthesis of findings from animal and clinical studies that highlight the neurotoxic potential of cannabinoids. Central to our analysis are the roles of neuroinflammation and oxidative stress, which exacerbate neuronal damage and contribute to cell death. The activation of cannabinoid receptors, particularly CB1 and CB2, is scrutinized for its dual role in mediating neuroprotective and neurotoxic effects. We explore calcium dysregulation as a critical mechanism that leads to excitotoxicity, mitochondrial dysfunction, and the activation of pro-apoptotic pathways. Additionally, we address the inhibition of anti-apoptotic proteins, induction of endoplasmic reticulum (ER) stress, and disruption of neurotransmitter systems, all of which further facilitate apoptosis in hippocampal neurons. Alterations in neurotrophic factor levels are also examined, as they play a vital role in neuronal survival and plasticity. Ultimately, this review underscores the multifaceted nature of cannabinoid-induced hippocampal apoptosis and calls for further research to elucidate these complex interactions, aiming to inform clinical practices and public health policies regarding cannabinoid use. The findings presented herein highlight the urgent need for a nuanced understanding of the risks associated with cannabinoid exposure, particularly in vulnerable populations.
Collapse
Affiliation(s)
- Habibeh Mashayekhi-Sardoo
- Student Research Committee Jiroft University of Medical Sciences, Jiroft, Iran; Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran; School of Health, Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Mahdiyeh Hedayati-Moghadam
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran; Student Research Committee Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Yousef Baghcheghi
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran; Student Research Committee Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
3
|
Nani JV, Muotri AR, Hayashi MAF. Peering into the mind: unraveling schizophrenia's secrets using models. Mol Psychiatry 2025; 30:659-678. [PMID: 39245692 DOI: 10.1038/s41380-024-02728-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Schizophrenia (SCZ) is a complex mental disorder characterized by a range of symptoms, including positive and negative symptoms, as well as cognitive impairments. Despite the extensive research, the underlying neurobiology of SCZ remain elusive. To overcome this challenge, the use of diverse laboratory modeling techniques, encompassing cellular and animal models, and innovative approaches like induced pluripotent stem cell (iPSC)-derived neuronal cultures or brain organoids and genetically engineered animal models, has been crucial. Immortalized cellular models provide controlled environments for investigating the molecular and neurochemical pathways involved in neuronal function, while iPSCs and brain organoids, derived from patient-specific sources, offer significant advantage in translational research by facilitating direct comparisons of cellular phenotypes between patient-derived neurons and healthy-control neurons. Animal models can recapitulate the different psychopathological aspects that should be modeled, offering valuable insights into the neurobiology of SCZ. In addition, invertebrates' models are genetically tractable and offer a powerful approach to dissect the core genetic underpinnings of SCZ, while vertebrate models, especially mammals, with their more complex nervous systems and behavioral repertoire, provide a closer approximation of the human condition to study SCZ-related traits. This narrative review provides a comprehensive overview of the diverse modeling approaches, critically evaluating their strengths and limitations. By synthesizing knowledge from these models, this review offers a valuable source for researchers, clinicians, and stakeholders alike. Integrating findings across these different models may allow us to build a more holistic picture of SCZ pathophysiology, facilitating the exploration of new research avenues and informed decision-making for interventions.
Collapse
Affiliation(s)
- João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| | - Alysson R Muotri
- Department of Pediatrics and Department of Molecular and Cellular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| |
Collapse
|
4
|
Delgado-Sequera A, Garcia-Mompo C, Gonzalez-Pinto A, Hidalgo-Figueroa M, Berrocoso E. A Systematic Review of the Molecular and Cellular Alterations Induced by Cannabis That May Serve as Risk Factors for Bipolar Disorder. Int J Neuropsychopharmacol 2024; 27:pyae002. [PMID: 38175142 PMCID: PMC10863486 DOI: 10.1093/ijnp/pyae002] [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: 07/26/2023] [Accepted: 01/03/2024] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Cannabis use is a risk factor of psychiatric illness, such as bipolar disorder type-I (BDI). Indeed, cannabis use strongly influences the onset and clinical course of BDI, although the biological mechanisms underlying this interaction remain unknown. Therefore, we have reviewed the biological mechanisms affected by cannabis use that may trigger BD. METHODS A systematic review was carried out of articles in which gene expression was studied in cannabis users or human-derived cells exposed to tetrahydrocannabinol (THC) or cannabidiol (CBD). A second systematic review was then performed to identify articles in which gene expression was studied in BDI samples, highlighting those that described alterations to the same molecular and cellular mechanisms affected by cannabis/THC/CBD. RESULTS The initial search identified 82 studies on cannabis and 962 on BDI. After removing duplicates and applying the inclusion/exclusion criteria, 9 studies into cannabis and 228 on BDI were retained. The molecular and cellular mechanisms altered by cannabis use or THC/CBD exposure were then identified, including neural development and function, cytoskeletal function, cell adhesion, mitochondrial biology, inflammatory related pathways, lipid metabolism, the endocannabinoid system, the hypocretin/orexin system, and apoptosis. Alterations to those activities were also described in 19 of 228 focused on BDI. CONCLUSIONS The biological mechanisms described in this study may be good candidates to the search for diagnostic biomarkers and therapeutic targets for BDI. Because cannabis use can trigger the onset of BD, further studies would be of interest to determine whether they are involved in the early development of the disorder, prompting early treatment.
Collapse
Affiliation(s)
- Alejandra Delgado-Sequera
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Research Unit, Puerta del Mar University Hospital, Cádiz, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Universidad de Cádiz, Cádiz, Spain
| | - Clara Garcia-Mompo
- Department of Medicine, School of Medical Sciences, Universitat Jaume I, Castellón de la Plana, Spain
| | - Ana Gonzalez-Pinto
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain
- Department of Psychiatry, Hospital Universitario de Alava, BIOARABA, UPV/EHU, CIBERSAM, Vitoria-Gasteiz, Spain
| | - Maria Hidalgo-Figueroa
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Research Unit, Puerta del Mar University Hospital, Cádiz, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Universidad de Cádiz, Cádiz, Spain
- Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain
| | - Esther Berrocoso
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Research Unit, Puerta del Mar University Hospital, Cádiz, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Universidad de Cádiz, Cádiz, Spain
- Department of Neuroscience, Universidad de Cádiz, Cádiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain
| |
Collapse
|
5
|
Ganesh S, Lam TT, Garcia-Milian R, D’Souza D, Nairn AC, Elgert K, Eitan E, Ranganathan M. Peripheral signature of altered synaptic integrity in young onset cannabis use disorder: A proteomic study of circulating extracellular vesicles. World J Biol Psychiatry 2023; 24:603-613. [PMID: 36994633 PMCID: PMC10471733 DOI: 10.1080/15622975.2023.2197039] [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: 12/17/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Rates of Cannabis Use Disorder (CUD) are highest amongst young adults. Paucity of brain tissue samples limits the ability to examine the molecular basis of cannabis related neuropathology. Proteomic studies of neuron-derived extracellular vesicles (NDEs) isolated from the biofluids may reveal markers of neuropathology in CUD. METHODS NDEs were extracted using ExoSORT, an immunoaffinity method to enrich NDEs from plasma samples from patients with young onset CUD and matched controls. Differential proteomic profiles were explored with Label Free Quantification (LFQ) mass spectrometry. Selected proteins were validated using orthogonal methods. RESULTS A total of 231 (±10) proteins were identified in NDE preparations from CUD and controls of which 28 were differentially abundant between groups. The difference in abundance of properdin (CFP gene) was statistically significant. SHANK1 (SHANK1 gene), an adapter protein at the post-synaptic density, was nominally depleted in the CUD NDE preparations. CONCLUSION In this pilot study, we noted a decrease in SHANK1 protein, involved in the structural and functional integrity of glutamatergic post-synapse, a potential peripheral signature of CUD neuropathology. The study shows that LFQ mass spectrometry proteomic analysis of NDEs derived from plasma may yield important insights into the synaptic pathology associated with CUD.
Collapse
Affiliation(s)
- Suhas Ganesh
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06520
| | - TuKiet T. Lam
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT, 06520
- Keck Mass Spectrometry & Proteomics Resource, Yale School of Medicine, New Haven, CT, 06510
| | - Rolando Garcia-Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, CT, 06510
| | - Deepak D’Souza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06520
| | - Angus C. Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06520
| | | | | | - Mohini Ranganathan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06520
| |
Collapse
|
6
|
Asher MJ, McMullan HM, Dong A, Li Y, Thayer SA. A Complete Endocannabinoid Signaling System Modulates Synaptic Transmission between Human Induced Pluripotent Stem Cell-Derived Neurons. Mol Pharmacol 2023; 103:100-112. [PMID: 36379717 PMCID: PMC9881009 DOI: 10.1124/molpharm.122.000555] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 10/01/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
The endocannabinoid system (ECS) modulates synaptic function to regulate many aspects of neurophysiology. It adapts to environmental changes and is affected by disease. Thus, the ECS presents an important target for therapeutic development. Despite recent interest in cannabinoid-based treatments, few preclinical studies are conducted in human systems. Human induced pluripotent stem cells (hiPSCs) provide one possible solution to this issue. However, it is not known if these cells have a fully functional ECS. Here, we show that hiPSC-derived neuron/astrocyte cultures exhibit a complete ECS. Using Ca2+ imaging and a genetically encoded endocannabinoid sensor, we demonstrate that they not only respond to exogenously applied cannabinoids but also produce and metabolize endocannabinoids. Synaptically driven [Ca2+]i spiking activity was inhibited (EC50 = 48 ± 13 nM) by the efficacious agonist [R(+)-[2,3-dihydro-5-methyl-3-[(morpholinyl)methyl]pyrolol [1,2,3-de]-1,4-benzoxazin-yl]-(1-naphthalenyl)methanone mesylate] (Win 55,212-2) and by the endogenous ligand 2-arachidonoyl glycerol (2-AG; EC50 = 2.0 ± 0.6 µm). The effects of Win 55212-2 were blocked by a CB1 receptor-selective antagonist. Δ9-Tetrahydrocannabinol acted as a partial agonist, maximally inhibiting synaptic activity by 47 ± 14% (EC50 = 1.4 ± 1.9 µm). Carbachol stimulated 2-AG production in a manner that was independent of Ca2+ and blocked by selective inhibition of diacylglycerol lipase. 2-AG returned to basal levels via a process mediated by monoacylglycerol lipase as indicated by slowed recovery in cultures treated with 4-[Bis(1,3-benzodioxol-5-yl)hydroxymethyl]-1-piperidinecarboxylic acid 4-nitrophenyl ester (JZL 184). Win 55,212-2 markedly desensitized CB1 receptor function following a 1-day exposure, whereas desensitization was incomplete following 7-day treatment with JZL 184. This human cell culture model is well suited for functional analysis of the ECS and as a platform for drug development. SIGNIFICANCE STATEMENT: Despite known differences between the human response to cannabinoids and that of other species, an in vitro human model demonstrating a fully functional endocannabinoid system has not been described. Human induced pluripotent stem cells (hiPSCs) can be obtained from skin samples and then reprogrammed into neurons for use in basic research and drug screening. Here, we show that hiPSC-derived neuronal cultures exhibit a complete endocannabinoid system suitable for mechanistic studies and drug discovery.
Collapse
Affiliation(s)
- Melissa J Asher
- Department of Pharmacology (M.J.A., H.M.M., S.A.T.), Graduate Program in Neuroscience (M.J.A., S.A.T.), and Molecular Pharmacology and Therapeutics Graduate Program (H.M.M., S.A.T.), University of Minnesota Medical School, Minneapolis, Minnesota; State Key Laboratory of Membrane Biology, Peking University School of Life Sciences (A.D., Y.L.), IDG/McGovern Institute for Brain Research (A.D., Y.L.), and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies (A.D., Y.L.), Peking University, Beijing, China; and Chinese Institute for Brain Research, Beijing, China (Y.L.)
| | - Hannah M McMullan
- Department of Pharmacology (M.J.A., H.M.M., S.A.T.), Graduate Program in Neuroscience (M.J.A., S.A.T.), and Molecular Pharmacology and Therapeutics Graduate Program (H.M.M., S.A.T.), University of Minnesota Medical School, Minneapolis, Minnesota; State Key Laboratory of Membrane Biology, Peking University School of Life Sciences (A.D., Y.L.), IDG/McGovern Institute for Brain Research (A.D., Y.L.), and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies (A.D., Y.L.), Peking University, Beijing, China; and Chinese Institute for Brain Research, Beijing, China (Y.L.)
| | - Ao Dong
- Department of Pharmacology (M.J.A., H.M.M., S.A.T.), Graduate Program in Neuroscience (M.J.A., S.A.T.), and Molecular Pharmacology and Therapeutics Graduate Program (H.M.M., S.A.T.), University of Minnesota Medical School, Minneapolis, Minnesota; State Key Laboratory of Membrane Biology, Peking University School of Life Sciences (A.D., Y.L.), IDG/McGovern Institute for Brain Research (A.D., Y.L.), and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies (A.D., Y.L.), Peking University, Beijing, China; and Chinese Institute for Brain Research, Beijing, China (Y.L.)
| | - Yulong Li
- Department of Pharmacology (M.J.A., H.M.M., S.A.T.), Graduate Program in Neuroscience (M.J.A., S.A.T.), and Molecular Pharmacology and Therapeutics Graduate Program (H.M.M., S.A.T.), University of Minnesota Medical School, Minneapolis, Minnesota; State Key Laboratory of Membrane Biology, Peking University School of Life Sciences (A.D., Y.L.), IDG/McGovern Institute for Brain Research (A.D., Y.L.), and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies (A.D., Y.L.), Peking University, Beijing, China; and Chinese Institute for Brain Research, Beijing, China (Y.L.)
| | - Stanley A Thayer
- Department of Pharmacology (M.J.A., H.M.M., S.A.T.), Graduate Program in Neuroscience (M.J.A., S.A.T.), and Molecular Pharmacology and Therapeutics Graduate Program (H.M.M., S.A.T.), University of Minnesota Medical School, Minneapolis, Minnesota; State Key Laboratory of Membrane Biology, Peking University School of Life Sciences (A.D., Y.L.), IDG/McGovern Institute for Brain Research (A.D., Y.L.), and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies (A.D., Y.L.), Peking University, Beijing, China; and Chinese Institute for Brain Research, Beijing, China (Y.L.)
| |
Collapse
|
7
|
Silva JP, Carvalho F. El uso terapéutico del cannabis y los cannabinoides. REVISTA ESPAÑOLA DE DROGODEPENDENCIAS 2022; 47:103-122. [DOI: 10.54108/10031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Los cannabinoides se dirigen principalmente al sistema endocannabinoide (ECS), que surge
como un objetivo terapéutico potencialmente interesante debido a su importante papel en la
modulación de procesos biológicos clave en todo el organismo. Como tal, los cannabinoides
ya se han propuesto como, por ejemplo, antieméticos, agentes antiespásticos, estimulantes del
apetito, antiepilépticos, analgésicos, depresores de la presión intraocular o como agentes para
controlar los trastornos del movimiento en el síndrome de Tourette.
Aquí revisamos las pruebas de investigación disponibles sobre el uso del cannabis y los cannabinoides
para un conjunto de aplicaciones terapéuticas sugeridas, y abordamos algunos de los
riesgos a corto y largo plazo que se han correlacionado con el uso de estas sustancias.
Encontramos escasas pruebas científicas que apoyen el uso de productos basados en el cannabis
para la mayoría de las aplicaciones sugeridas, así como ninguna necesidad médica no satisfecha
que no esté ya abordada por los medicamentos existentes (algunos basados en cannabinoides)
en el mercado. En este escenario, los riesgos potenciales asociados al uso crónico de estas sustancias
pueden disuadir su uso médico.
Collapse
|
8
|
Chandy M, Obal D, Wu JC. Elucidating effects of environmental exposure using human-induced pluripotent stem cell disease modeling. EMBO Mol Med 2022; 14:e13260. [PMID: 36285490 PMCID: PMC9641419 DOI: 10.15252/emmm.202013260] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 11/15/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are a powerful modeling system for medical discovery and translational research. To date, most studies have focused on the potential for iPSCs for regenerative medicine, drug discovery, and disease modeling. However, iPSCs are also a powerful modeling system to investigate the effects of environmental exposure on the cardiovascular system. With the emergence of e-cigarettes, air pollution, marijuana use, opioids, and microplastics as novel cardiovascular risk factors, iPSCs have the potential for elucidating the effects of these toxins on the body using conventional two-dimensional (2D) arrays and more advanced tissue engineering approaches with organoid and other three-dimensional (3D) models. The effects of these environmental factors may be enhanced by genetic polymorphisms that make some individuals more susceptible to the effects of toxins. iPSC disease modeling may reveal important gene-environment interactions that exacerbate cardiovascular disease and predispose some individuals to adverse outcomes. Thus, iPSCs and gene-editing techniques could play a pivotal role in elucidating the mechanisms of gene-environment interactions and understanding individual variability in susceptibility to environmental effects.
Collapse
Affiliation(s)
- Mark Chandy
- Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCAUSA
- Department of MedicineWestern UniversityLondonONCanada
- Department of Physiology and PharmacologyWestern UniversityLondonONCanada
| | - Detlef Obal
- Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCAUSA
- Department of Anesthesiology, Perioperative, and Pain MedicineStanford UniversityStanfordCAUSA
| | - Joseph C Wu
- Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCAUSA
- Department of Medicine, Division of Cardiovascular MedicineStanford University School of MedicineStanfordCAUSA
| |
Collapse
|
9
|
Schrott R, Greeson KW, King D, Crow KMS, Easley CA, Murphy SK. Cannabis alters DNA methylation at maternally imprinted and autism candidate genes in spermatogenic cells. Syst Biol Reprod Med 2022; 68:357-369. [PMID: 35687495 PMCID: PMC10032331 DOI: 10.1080/19396368.2022.2073292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 04/18/2022] [Accepted: 04/23/2022] [Indexed: 10/18/2022]
Abstract
Cannabis use in the United States is increasing, with highest consumption among men at their peak reproductive years. We previously demonstrated widespread changes in sperm DNA methylation with cannabis exposure in humans and rats, including genes important in neurodevelopment. Here, we use an in vitro human spermatogenesis model to recapitulate chronic cannabis use and assess DNA methylation at imprinted and autism spectrum disorder (ASD) candidate genes in spermatogonial stem cell (SSC)- and spermatid-like cells. Methylation at maternally imprinted genes SGCE and GRB10 was significantly altered in SSC- and spermatid-like cells, respectively, while PEG3 was significantly differentially methylated in spermatid-like cells. Two of ten randomly selected ASD candidate genes, HCN1 and NR4A2, had significantly altered methylation with cannabis exposure in SSC-like cells. These results support our findings in human cohorts and provide a new tool with which to gain mechanistic insights into the association between paternal cannabis use and risk of ASD in offspring.
Collapse
Affiliation(s)
- Rose Schrott
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, 27701, USA
- Integrated Toxicology and Environmental Health Program, Nicholas School of the Environment, Duke University, Durham, NC, 27701, USA
| | - Katherine W. Greeson
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA, 30602, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA
| | - Dillon King
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, 27701, USA
- Integrated Toxicology and Environmental Health Program, Nicholas School of the Environment, Duke University, Durham, NC, 27701, USA
| | - Krista M. Symosko Crow
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA, 30602, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA
| | - Charles A. Easley
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA, 30602, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, 30602, USA
| | - Susan K. Murphy
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, 27701, USA
- Integrated Toxicology and Environmental Health Program, Nicholas School of the Environment, Duke University, Durham, NC, 27701, USA
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
The cytokines interleukin-6 and interferon-α induce distinct microglia phenotypes. J Neuroinflammation 2022; 19:96. [PMID: 35429976 PMCID: PMC9013466 DOI: 10.1186/s12974-022-02441-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Background Elevated production of the cytokines interleukin (IL)-6 or interferon (IFN)-α in the central nervous system (CNS) is implicated in the pathogenesis of neurological diseases such as neuromyelitis optica spectrum disorders or cerebral interferonopathies, respectively. Transgenic mice with CNS-targeted chronic production of IL-6 (GFAP-IL6) or IFN-α (GFAP-IFN) recapitulate important clinical and pathological features of these human diseases. The activation of microglia is a prominent manifestation found both in the human diseases and in the transgenic mice, yet little is known about how this contributes to disease pathology. Methods Here, we used a combination of ex vivo and in situ techniques to characterize the molecular, cellular and transcriptomic phenotypes of microglia in GFAP-IL6 versus GFAP-IFN mice. In addition, a transcriptomic meta-analysis was performed to compare the microglia response from GFAP-IL6 and GFAP-IFN mice to the response of microglia in a range of neurodegenerative and neuroinflammatory disorders. Results We demonstrated that microglia show stimulus-specific responses to IL-6 versus IFN-α in the brain resulting in unique and extensive molecular and cellular adaptations. In GFAP-IL6 mice, microglia proliferated, had shortened, less branched processes and elicited transcriptomic and molecular changes associated with phagocytosis and lipid processing. In comparison, microglia in the brain of GFAP-IFN mice exhibited increased proliferation and apoptosis, had larger, hyper-ramified processes and showed transcriptomic and surface marker changes associated with antigen presentation and antiviral response. Further, a transcriptomic meta-analysis revealed that IL-6 and IFN-α both contribute to the formation of a core microglia response in animal models of neurodegenerative and neuroinflammatory disorders, such as Alzheimer’s disease, tauopathy, multiple sclerosis and lipopolysaccharide-induced endotoxemia. Conclusions Our findings demonstrate that microglia responses to IL-6 and IFN-α are highly stimulus-specific, wide-ranging and give rise to divergent phenotypes that modulate microglia responses in neuroinflammatory and neurodegenerative diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02441-x.
Collapse
|
13
|
Association of cumulative early medical factors with autism and autistic symptoms in a population-based twin sample. Transl Psychiatry 2022; 12:73. [PMID: 35194015 PMCID: PMC8863884 DOI: 10.1038/s41398-022-01833-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 12/03/2022] Open
Abstract
Although highly heritable, environment also contributes to the etiology of autism spectrum disorder (ASD), with several specific environmental factors previously suggested. A registry-linked population-based twin cohort of 15,701 pairs (586 individuals with an ASD diagnosis), was established within the Child and Adolescent Twin Study in Sweden. Participants were evaluated for autistic symptoms at age 9 using the Autism-Tics, ADHD and other Comorbidities parental interview. A series of binary cut-offs indicated whether participants scored over various ASD symptom percentiles. Three early medical factors previously associated with ASD, beyond familial confounding (low birth weight, congenital malformations and perinatal hypoxia), were summed up creating an individual cumulative exposure load. A series of unconditional logistic regressions between all individuals and conditional regressions within twin pairs were performed for each outcome and exposure level. Between all individuals increasing cumulative early exposure loads were associated with increasing risk of ASD diagnosis (OR 3.33 (95%CI 1.79-6.20) for three exposures) and autistic symptoms (ranging from OR 2.12 (1.57-2.86) for three exposures at the 55th symptom percentile cut-off to OR 3.39 (2.2-5.24) at the 95th). Within twin pairs, the association between three exposures and an ASD diagnosis remained similar, but not statistically significant (OR 2.39 (0.62-9.24)). Having a higher load of early cumulative exposure was consistently associated with autistic symptoms after adjusting for familial confounding and sex (OR 3.45 (1.66-7.15) to OR 7.36 (1.99-27.18)). This study gives support to the cumulative stress hypothesis of ASD, and the dimensional model regarding environmental exposures, after adjustment for familial confounding.
Collapse
|
14
|
Pugsley K, Scherer SW, Bellgrove MA, Hawi Z. Environmental exposures associated with elevated risk for autism spectrum disorder may augment the burden of deleterious de novo mutations among probands. Mol Psychiatry 2022; 27:710-730. [PMID: 34002022 PMCID: PMC8960415 DOI: 10.1038/s41380-021-01142-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 12/11/2022]
Abstract
Although the full aetiology of autism spectrum disorder (ASD) is unknown, familial and twin studies demonstrate high heritability of 60-90%, indicating a predominant role of genetics in the development of the disorder. The genetic architecture of ASD consists of a complex array of rare and common variants of all classes of genetic variation usually acting additively to augment individual risk. The relative contribution of heredity in ASD persists despite selective pressures against the classic autistic phenotype; a phenomenon thought to be explained, in part, by the incidence of spontaneous (or de novo) mutations. Notably, environmental exposures attributed as salient risk factors for ASD may play a causal role in the emergence of deleterious de novo variations, with several ASD-associated agents having significant mutagenic potential. To explore this hypothesis, this review article assesses published epidemiological data with evidence derived from assays of mutagenicity, both in vivo and in vitro, to determine the likely role such agents may play in augmenting the genetic liability in ASD. Broadly, these exposures were observed to elicit genomic alterations through one or a combination of: (1) direct interaction with genetic material; (2) impaired DNA repair; or (3) oxidative DNA damage. However, the direct contribution of these factors to the ASD phenotype cannot be determined without further analysis. The development of comprehensive prospective birth cohorts in combination with genome sequencing is essential to forming a causal, mechanistic account of de novo mutations in ASD that links exposure, genotypic alterations, and phenotypic consequences.
Collapse
Affiliation(s)
- Kealan Pugsley
- grid.1002.30000 0004 1936 7857Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC Australia
| | - Stephen W. Scherer
- grid.42327.300000 0004 0473 9646The Centre for Applied Genomics and Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, ON Canada
| | - Mark A. Bellgrove
- grid.1002.30000 0004 1936 7857Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC Australia
| | - Ziarih Hawi
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
15
|
Molecular Findings Guiding the Modulation of the Endocannabinoid System as a Potential Target to Treat Schizophrenia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1400:89-103. [DOI: 10.1007/978-3-030-97182-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
16
|
Gelernter J, Polimanti R. Genetics of substance use disorders in the era of big data. Nat Rev Genet 2021; 22:712-729. [PMID: 34211176 PMCID: PMC9210391 DOI: 10.1038/s41576-021-00377-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
Substance use disorders (SUDs) are conditions in which the use of legal or illegal substances, such as nicotine, alcohol or opioids, results in clinical and functional impairment. SUDs and, more generally, substance use are genetically complex traits that are enormously costly on an individual and societal basis. The past few years have seen remarkable progress in our understanding of the genetics, and therefore the biology, of substance use and abuse. Various studies - including of well-defined phenotypes in deeply phenotyped samples, as well as broadly defined phenotypes in meta-analysis and biobank samples - have revealed multiple risk loci for these common traits. A key emerging insight from this work establishes a biological and genetic distinction between quantity and/or frequency measures of substance use (which may involve low levels of use without dependence), versus symptoms related to physical dependence.
Collapse
Affiliation(s)
- Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA.
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA.
| | - Renato Polimanti
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| |
Collapse
|
17
|
Unterholzner J, Millischer V, Wotawa C, Sawa A, Lanzenberger R. Making Sense of Patient-Derived iPSCs, Transdifferentiated Neurons, Olfactory Neuronal Cells, and Cerebral Organoids as Models for Psychiatric Disorders. Int J Neuropsychopharmacol 2021; 24:759-775. [PMID: 34216465 PMCID: PMC8538891 DOI: 10.1093/ijnp/pyab037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 05/30/2021] [Accepted: 07/02/2021] [Indexed: 11/17/2022] Open
Abstract
The improvement of experimental models for disorders requires a constant approximation towards the dysregulated tissue. In psychiatry, where an impairment of neuronal structure and function is assumed to play a major role in disease mechanisms and symptom development, this approximation is an ongoing process implicating various fields. These include genetic, animal, and post-mortem studies. To test hypotheses generated through these studies, in vitro models using non-neuronal cells such as fibroblasts and lymphocytes have been developed. For brain network disorders, cells with neuronal signatures would, however, represent a more adequate tissue. Considering the limited accessibility of brain tissue, research has thus turned towards neurons generated from induced pluripotent stem cells as well as directly induced neurons, cerebral organoids, and olfactory neuroepithelium. Regarding the increasing importance and amount of research using these neuronal cells, this review aims to provide an overview of all these models to make sense of the current literature. The development of each model system and its use as a model for the various psychiatric disorder categories will be laid out. Also, advantages and limitations of each model will be discussed, including a reflection on implications and future perspectives.
Collapse
Affiliation(s)
- Jakob Unterholzner
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Vincent Millischer
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria,Neurogenetics Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Christoph Wotawa
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Akira Sawa
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA,Departments of Psychiatry, Neuroscience, Biomedical Engineering and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria,Correspondence: Prof. Rupert Lanzenberger, MD, PD, NEUROIMAGING LABS (NIL) - PET, MRI, EEG, TMS & Chemical Lab, Department of Psychiatry and Psychotherapy, Medical University of Vienna, Waehringer Guertel 18–20, 1090 Vienna, Austria ()
| |
Collapse
|
18
|
Abstract
OBJECTIVES Cannabis is a known teratogen. Data availability addressing both major congenital anomalies and cannabis use allowed us to explore their geospatial relationships. METHODS Data for the years 1998 to 2009 from Canada Health and Statistics Canada was analyzed in R. Maps have been drawn and odds ratios, principal component analysis, correlation matrices, least squares regression and geospatial regression analyses have been conducted using the R packages base, dplyr, epiR, psych, ggplot2, colorplaner and the spml and spreml functions from package splm. RESULTS Mapping showed cannabis use was more common in the northern Territories of Canada in the Second National Survey of Cannabis Use 2018. Total congenital anomalies, all cardiovascular defects, orofacial clefts, Downs syndrome and gastroschisis were all found to be more common in these same regions and rose as a function of cannabis exposure. When Canada was dichotomized into high and low cannabis use zones by Provinces v Territories the Territories had a higher rate of total congenital anomalies 450.026 v 390.413 (O.R. = 1.16 95%C.I. 1.08-1.25, P = 0.000058; attributable fraction in exposed 13.25%, 95%C.I. 7.04-19.04%). In geospatial analysis in a spreml spatial error model cannabis was significant both alone as a main effect (P < 2.0 × 10) and in all its first and second order interactions with both tobacco and opioids from P < 2.0 × 10. CONCLUSION These results show that the northern Territories of Canada share a higher rate of cannabis use together with elevated rates of total congenital anomalies, all cardiovascular defects, Down's syndrome and gastroschisis. This is the second report of a significant association between cannabis use and both total defects and all cardiovascular anomalies and the fourth published report of a link with Downs syndrome and thereby direct major genotoxicity. The correlative relationships described in this paper are confounded by many features of social disadvantage in Canada's northern territories. However, in the context of a similar broad spectrum of defects described both in animals and in epidemiological reports from Hawaii, Colorado, USA and Australia they are cause for particular concern and indicate further research.
Collapse
|
19
|
Tsatsanis A, McCorkindale AN, Wong BX, Patrick E, Ryan TM, Evans RW, Bush AI, Sutherland GT, Sivaprasadarao A, Guennewig B, Duce JA. The acute phase protein lactoferrin is a key feature of Alzheimer's disease and predictor of Aβ burden through induction of APP amyloidogenic processing. Mol Psychiatry 2021; 26:5516-5531. [PMID: 34400772 PMCID: PMC8758478 DOI: 10.1038/s41380-021-01248-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/17/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023]
Abstract
Amyloidogenic processing of the amyloid precursor protein (APP) forms the amyloid-β peptide (Aβ) component of pathognomonic extracellular plaques of AD. Additional early cortical changes in AD include neuroinflammation and elevated iron levels. Activation of the innate immune system in the brain is a neuroprotective response to infection; however, persistent neuroinflammation is linked to AD neuropathology by uncertain mechanisms. Non-parametric machine learning analysis on transcriptomic data from a large neuropathologically characterised patient cohort revealed the acute phase protein lactoferrin (Lf) as the key predictor of amyloid pathology. In vitro studies showed that an interaction between APP and the iron-bound form of Lf secreted from activated microglia diverted neuronal APP endocytosis from the canonical clathrin-dependent pathway to one requiring ADP ribosylation factor 6 trafficking. By rerouting APP recycling to the Rab11-positive compartment for amyloidogenic processing, Lf dramatically increased neuronal Aβ production. Lf emerges as a novel pharmacological target for AD that not only modulates APP processing but provides a link between Aβ production, neuroinflammation and iron dysregulation.
Collapse
Affiliation(s)
- Andrew Tsatsanis
- grid.5335.00000000121885934The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.9909.90000 0004 1936 8403Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, West Yorkshire UK
| | - Andrew N. McCorkindale
- grid.1013.30000 0004 1936 834XFaculty of Medicine and Health, Charles Perkins Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW Australia
| | - Bruce X. Wong
- grid.5335.00000000121885934The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.9909.90000 0004 1936 8403Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, West Yorkshire UK ,grid.1008.90000 0001 2179 088XMelbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC Australia
| | - Ellis Patrick
- grid.1013.30000 0004 1936 834XFaculty of Science, School of Mathematics and Statistics, University of Sydney, Camperdown, NSW Australia
| | - Tim M. Ryan
- grid.1008.90000 0001 2179 088XMelbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC Australia
| | - Robert W. Evans
- grid.7728.a0000 0001 0724 6933School of Engineering and Design, Brunel University, London, UK
| | - Ashley I. Bush
- grid.1008.90000 0001 2179 088XMelbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC Australia
| | - Greg T. Sutherland
- grid.1013.30000 0004 1936 834XFaculty of Medicine and Health, Charles Perkins Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW Australia
| | - Asipu Sivaprasadarao
- grid.9909.90000 0004 1936 8403Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, West Yorkshire UK
| | - Boris Guennewig
- grid.1013.30000 0004 1936 834XFaculty of Medicine and Health, Brain and Mind Centre and School of Medical Sciences, The University of Sydney, Camperdown, NSW Australia
| | - James A. Duce
- grid.5335.00000000121885934The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.9909.90000 0004 1936 8403Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, West Yorkshire UK ,grid.1008.90000 0001 2179 088XMelbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC Australia
| |
Collapse
|
20
|
Paraíso-Luna J, Aguareles J, Martín R, Ayo-Martín AC, Simón-Sánchez S, García-Rincón D, Costas-Insua C, García-Taboada E, de Salas-Quiroga A, Díaz-Alonso J, Liste I, Sánchez-Prieto J, Cappello S, Guzmán M, Galve-Roperh I. Endocannabinoid signalling in stem cells and cerebral organoids drives differentiation to deep layer projection neurons via CB 1 receptors. Development 2020; 147:226034. [PMID: 33168583 DOI: 10.1242/dev.192161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022]
Abstract
The endocannabinoid (eCB) system, via the cannabinoid CB1 receptor, regulates neurodevelopment by controlling neural progenitor proliferation and neurogenesis. CB1 receptor signalling in vivo drives corticofugal deep layer projection neuron development through the regulation of BCL11B and SATB2 transcription factors. Here, we investigated the role of eCB signalling in mouse pluripotent embryonic stem cell-derived neuronal differentiation. Characterization of the eCB system revealed increased expression of eCB-metabolizing enzymes, eCB ligands and CB1 receptors during neuronal differentiation. CB1 receptor knockdown inhibited neuronal differentiation of deep layer neurons and increased upper layer neuron generation, and this phenotype was rescued by CB1 re-expression. Pharmacological regulation with CB1 receptor agonists or elevation of eCB tone with a monoacylglycerol lipase inhibitor promoted neuronal differentiation of deep layer neurons at the expense of upper layer neurons. Patch-clamp analyses revealed that enhancing cannabinoid signalling facilitated neuronal differentiation and functionality. Noteworthy, incubation with CB1 receptor agonists during human iPSC-derived cerebral organoid formation also promoted the expansion of BCL11B+ neurons. These findings unveil a cell-autonomous role of eCB signalling that, via the CB1 receptor, promotes mouse and human deep layer cortical neuron development.
Collapse
Affiliation(s)
- Juan Paraíso-Luna
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - José Aguareles
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Ricardo Martín
- Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Ane C Ayo-Martín
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.,International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
| | - Samuel Simón-Sánchez
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Daniel García-Rincón
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Carlos Costas-Insua
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Elena García-Taboada
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Adán de Salas-Quiroga
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Javier Díaz-Alonso
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Isabel Liste
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain
| | - José Sánchez-Prieto
- Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | | | - Manuel Guzmán
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Ismael Galve-Roperh
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| |
Collapse
|
21
|
Miranda CC, Barata T, Vaz SH, Ferreira C, Quintas A, Bekman EP. hiPSC-Based Model of Prenatal Exposure to Cannabinoids: Effect on Neuronal Differentiation. Front Mol Neurosci 2020; 13:119. [PMID: 32733202 PMCID: PMC7357827 DOI: 10.3389/fnmol.2020.00119] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022] Open
Abstract
Phytocannabinoids are psychotropic substances ofcannabis with the ability to bind endocannabinoid (eCB) receptors that regulate synaptic activity in the central nervous system (CNS). Synthetic cannabinoids (SCs) are synthetic analogs of Δ9-tetrahydrocannabinol (Δ9-THC), the psychotropic compound of cannabis, acting as agonists of eCB receptor CB1. SC is an easily available and popular alternative to cannabis, and their molecular structure is always changing, increasing the hazard for the general population. The popularity of cannabis and its derivatives may lead, and often does, to a child's exposure to cannabis both in utero and through breastfeeding by a drug-consuming mother. Prenatal exposure to cannabis has been associated with an altered rate of mental development and significant changes in nervous system functioning. However, the understanding of mechanisms of its action on developing the human CNS is still lacking. We investigated the effect of continuous exposure to cannabinoids on developing human neurons, mimicking the prenatal exposure by drug-consuming mother. Two human induced pluripotent stem cells (hiPSC) lines were induced to differentiate into neuronal cells and exposed for 37 days to cannabidiol (CBD), Δ9-THC, and two SCs, THJ-018 and EG-018. Both Δ9-THC and SC, at 10 μM, promote precocious neuronal and glial differentiation, while CBD at the same concentration is neurotoxic. Neurons exposed to Δ9-THC and SC show abnormal functioning of voltage-gated calcium channels when stimulated by extracellular potassium. In sum, all studied substances have a profound impact on the developing neurons, highlighting the importance of thorough research on the impact of prenatal exposure to natural and SC.
Collapse
Affiliation(s)
- Cláudia C Miranda
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisbon, Lisbona, Portugal
| | - Tiago Barata
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisbon, Lisbona, Portugal
| | - Sandra H Vaz
- Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Carla Ferreira
- Molecular Pathology and Forensic Biochemistry Laboratory, CiiEM, Campus Universitário Quinta da Granja, Monte da Caparica, Caparica, Portugal.,Forensic and Psychological Sciences Laboratory Egas Moniz, Campus Universitário Quinta da Granja, Monte da Caparica, Caparica, Portugal
| | - Alexandre Quintas
- Molecular Pathology and Forensic Biochemistry Laboratory, CiiEM, Campus Universitário Quinta da Granja, Monte da Caparica, Caparica, Portugal.,Forensic and Psychological Sciences Laboratory Egas Moniz, Campus Universitário Quinta da Granja, Monte da Caparica, Caparica, Portugal
| | - Evguenia P Bekman
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisbon, Lisbona, Portugal.,Instituto de Medicina Molecular, João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Lisbon, Portugal
| |
Collapse
|
22
|
Powell SK, O'Shea CP, Shannon SR, Akbarian S, Brennand KJ. Investigation of Schizophrenia with Human Induced Pluripotent Stem Cells. ADVANCES IN NEUROBIOLOGY 2020; 25:155-206. [PMID: 32578147 DOI: 10.1007/978-3-030-45493-7_6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Schizophrenia is a chronic and severe neuropsychiatric condition manifested by cognitive, emotional, affective, perceptual, and behavioral abnormalities. Despite decades of research, the biological substrates driving the signs and symptoms of the disorder remain elusive, thus hampering progress in the development of treatments aimed at disease etiologies. The recent emergence of human induced pluripotent stem cell (hiPSC)-based models has provided the field with a highly innovative approach to generate, study, and manipulate living neural tissue derived from patients, making possible the exploration of fundamental roles of genes and early-life stressors in disease-relevant cell types. Here, we begin with a brief overview of the clinical, epidemiological, and genetic aspects of the condition, with a focus on schizophrenia as a neurodevelopmental disorder. We then highlight relevant technical advancements in hiPSC models and assess novel findings attained using hiPSC-based approaches and their implications for disease biology and treatment innovation. We close with a critical appraisal of the developments necessary for both further expanding knowledge of schizophrenia and the translation of new insights into therapeutic innovations.
Collapse
Affiliation(s)
- Samuel K Powell
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Callan P O'Shea
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sara Rose Shannon
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Schahram Akbarian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristen J Brennand
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
23
|
García S, Martín Giménez VM, Mocayar Marón FJ, Reiter RJ, Manucha W. Melatonin and cannabinoids: mitochondrial-targeted molecules that may reduce inflammaging in neurodegenerative diseases. Histol Histopathol 2020; 35:789-800. [PMID: 32154907 DOI: 10.14670/hh-18-212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Generally, the development and progression of neurodegenerative diseases are associated with advancing age, so they are usually diagnosed in late adulthood. A primary mechanism underlying the onset of neurodegenerative diseases is neuroinflammation. Based on this background, the concept of "neuroinflammaging" has emerged. In this deregulated neuroinflammatory process, a variety of immune cells participate, especially glial cells, proinflammatory cytokines, receptors, and subcellular organelles including mitochondria, which are mainly responsible for maintaining redox balance at the cellular level. Senescence and autophagic processes also play a crucial role in the neuroinflammatory disease associated with aging. Of particular interest, melatonin, cannabinoids, and the receptors of both molecules which are closely related, exert beneficial effects on the neuroinflammatory processes that precede the onset of neurodegenerative pathologies such as Parkinson's and Alzheimer's diseases. Some of these neuroprotective effects are fundamentally related to its anti-inflammatory and antioxidative actions at the mitochondrial level due to the strategic functions of this organelle. The aim of this review is to summarize the most recent advances in the study of neuroinflammation and neurodegeneration associated with age and to consider the use of new mitochondrial therapeutic targets related to the endocannabinoid system and the pineal gland.
Collapse
Affiliation(s)
- Sebastián García
- Institute of Pharmacology, Department of Pathology, School of Medical Sciences, Cuyo National University, Mendoza, Argentina.,Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Council of Scientific and Technological Research (CONICET), Mendoza, Argentina
| | - Virna Margarita Martín Giménez
- Institute of Research in Chemical Sciences, School of Chemical and Technological Sciences, Cuyo Catholic University, San Juan, Argentina
| | - Feres José Mocayar Marón
- Institute of Pharmacology, Department of Pathology, School of Medical Sciences, Cuyo National University, Mendoza, Argentina.,Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Council of Scientific and Technological Research (CONICET), Mendoza, Argentina
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science at San Antonio, San Antonio, TX, USA
| | - Walter Manucha
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Council of Scientific and Technological Research (CONICET), Mendoza, Argentina.,Institute of Pharmacology, Department of Pathology, School of Medical Sciences, Cuyo National University, Mendoza, Argentina.
| |
Collapse
|
24
|
McCutcheon RA, Krystal JH, Howes OD. Dopamine and glutamate in schizophrenia: biology, symptoms and treatment. World Psychiatry 2020; 19:15-33. [PMID: 31922684 PMCID: PMC6953551 DOI: 10.1002/wps.20693] [Citation(s) in RCA: 353] [Impact Index Per Article: 70.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glutamate and dopamine systems play distinct roles in terms of neuronal signalling, yet both have been proposed to contribute significantly to the pathophysiology of schizophrenia. In this paper we assess research that has implicated both systems in the aetiology of this disorder. We examine evidence from post-mortem, preclinical, pharmacological and in vivo neuroimaging studies. Pharmacological and preclinical studies implicate both systems, and in vivo imaging of the dopamine system has consistently identified elevated striatal dopamine synthesis and release capacity in schizophrenia. Imaging of the glutamate system and other aspects of research on the dopamine system have produced less consistent findings, potentially due to methodological limitations and the heterogeneity of the disorder. Converging evidence indicates that genetic and environmental risk factors for schizophrenia underlie disruption of glutamatergic and dopaminergic function. However, while genetic influences may directly underlie glutamatergic dysfunction, few genetic risk variants directly implicate the dopamine system, indicating that aberrant dopamine signalling is likely to be predominantly due to other factors. We discuss the neural circuits through which the two systems interact, and how their disruption may cause psychotic symptoms. We also discuss mechanisms through which existing treatments operate, and how recent research has highlighted opportunities for the development of novel pharmacological therapies. Finally, we consider outstanding questions for the field, including what remains unknown regarding the nature of glutamate and dopamine function in schizophrenia, and what needs to be achieved to make progress in developing new treatments.
Collapse
Affiliation(s)
- Robert A McCutcheon
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
- South London and Maudsley Foundation NHS Trust, Maudsley Hospital, London, UK
| | - John H Krystal
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Oliver D Howes
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
- South London and Maudsley Foundation NHS Trust, Maudsley Hospital, London, UK
| |
Collapse
|
25
|
Shum C, Dutan L, Annuario E, Warre-Cornish K, Taylor SE, Taylor RD, Andreae LC, Buckley NJ, Price J, Bhattacharyya S, Srivastava DP. Δ 9-tetrahydrocannabinol and 2-AG decreases neurite outgrowth and differentially affects ERK1/2 and Akt signaling in hiPSC-derived cortical neurons. Mol Cell Neurosci 2020; 103:103463. [PMID: 31917333 DOI: 10.1016/j.mcn.2019.103463] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 12/20/2019] [Accepted: 12/30/2019] [Indexed: 12/26/2022] Open
Abstract
Endocannabinoids regulate different aspects of neurodevelopment. In utero exposure to the exogenous psychoactive cannabinoid Δ9-tetrahydrocannabinol (Δ9-THC), has been linked with abnormal cortical development in animal models. However, much less is known about the actions of endocannabinoids in human neurons. Here we investigated the effect of the endocannabinoid 2-arachidonoyl glycerol (2AG) and Δ9-THC on the development of neuronal morphology and activation of signaling kinases, in cortical neurons derived from human induced pluripotent stem cells (hiPSCs). Our data indicate that the cannabinoid type 1 receptor (CB1R), but not the cannabinoid 2 receptor (CB2R), GPR55 or TRPV1 receptors, is expressed in young, immature hiPSC-derived cortical neurons. Consistent with previous reports, 2AG and Δ9-THC negatively regulated neurite outgrowth. Interestingly, acute exposure to both 2AG and Δ9-THC inhibited phosphorylation of serine/threonine kinase extracellular signal-regulated protein kinases (ERK1/2), whereas Δ9-THC also reduced phosphorylation of Akt (aka PKB). Moreover, the CB1R inverse agonist SR 141716A attenuated the decrease in neurite outgrowth and ERK1/2 phosphorylation induced by 2AG and Δ9-THC. Taken together, our data suggest that hiPSC-derived cortical neurons express CB1Rs and are responsive to exogenous cannabinoids. Thus, hiPSC-neurons may represent a good cellular model for investigating the role of the endocannabinoid system in regulating cellular processes in developing human neurons.
Collapse
Affiliation(s)
- Carole Shum
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King's College London, London SE5 8AF, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Lucia Dutan
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King's College London, London SE5 8AF, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Emily Annuario
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King's College London, London SE5 8AF, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Katherine Warre-Cornish
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King's College London, London SE5 8AF, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Samuel E Taylor
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK; Centre for Developmental Neurobiology, King's College London, London, UK
| | - Ruth D Taylor
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK; Centre for Developmental Neurobiology, King's College London, London, UK
| | - Laura C Andreae
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK; Centre for Developmental Neurobiology, King's College London, London, UK
| | | | - Jack Price
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King's College London, London SE5 8AF, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK; National Institute for Biological Standards and Control, South Mimms, UK
| | | | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King's College London, London SE5 8AF, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
26
|
Reece AS, Hulse GK. Impacts of cannabinoid epigenetics on human development: reflections on Murphy et. al. 'cannabinoid exposure and altered DNA methylation in rat and human sperm' epigenetics 2018; 13: 1208-1221. Epigenetics 2019; 14:1041-1056. [PMID: 31293213 PMCID: PMC6773386 DOI: 10.1080/15592294.2019.1633868] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent data from the Kollins lab (‘Cannabinoid exposure and altered DNA methylation in rat and human sperm’ Epigenetics 2018; 13: 1208–1221) indicated epigenetic effects of cannabis use on sperm in man parallel those in rats and showed substantial shifts in both hypo- and hyper-DNA methylation with the latter predominating. This provides one likely mechanism for the transgenerational transmission of epigenomic instability with sperm as the vector. It therefore contributes important pathophysiological insights into the probable mechanisms underlying the epidemiology of prenatal cannabis exposure potentially explaining diverse features of cannabis-related teratology including effects on the neuraxis, cardiovasculature, immune stimulation, secondary genomic instability and carcinogenesis related to both adult and pediatric cancers. The potentially inheritable and therefore multigenerational nature of these defects needs to be carefully considered in the light of recent teratological and neurobehavioural trends in diverse jurisdictions such as the USA nationally, Hawaii, Colorado, Canada, France and Australia, particularly relating to mental retardation, age-related morbidity and oncogenesis including inheritable cancerogenesis. Increasing demonstrations that the epigenome can respond directly and in real time and retain memories of environmental exposures of many kinds implies that the genome-epigenome is much more sensitive to environmental toxicants than has been generally realized. Issues of long-term multigenerational inheritance amplify these concerns. Further research particularly on the epigenomic toxicology of many cannabinoids is also required.
Collapse
Affiliation(s)
- Albert Stuart Reece
- Division of Psychiatry, University of Western Australia , Crawley , Western Australia Australia.,School of Medical and Health Sciences, Edith Cowan University , Joondalup , Western Australia , Australia
| | - Gary Kenneth Hulse
- Division of Psychiatry, University of Western Australia , Crawley , Western Australia Australia.,School of Medical and Health Sciences, Edith Cowan University , Joondalup , Western Australia , Australia
| |
Collapse
|
27
|
Zhou M, Du W, Salvador-Carulla L, Glasgow N. Adverse drug event-related hospitalisation in persons with neurodevelopmental disorders: a state-wide retrospective cohort study. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2019; 63:429-440. [PMID: 30609156 DOI: 10.1111/jir.12586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/06/2018] [Accepted: 12/01/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Little is known about the sociodemographic and clinical characteristics of adverse drug events (ADEs) in patients with neurodevelopmental disorders (NDD). OBJECTIVE The objective of this study was to describe and compare the demographic details of people with and without NDD hospitalised due to ADEs. METHODS The all-inclusive New South Wales Admitted Patient Data Collection from 2001 to 2014 was employed to identify ADE-related hospitalisations in patients with NDD using the International Classification of Diseases 10th revision Australian modification codes. We derived case sets specific to different clinical groups and patient characteristics and compared proportional differences between patients with and without intellectual disability using chi squared tests. RESULTS A total of 2173 patients with NDD were admitted for acute care of ADEs, accounting for 0.7% of all ADE-related hospitalisations. Hospitalised ADEs among patients with NDD increased by twofold over the 14-year study period. Psychotropic medications and opioid analgesic medications were leading causes of ADE-related hospitalisations in patients with NDD. Compared with their counterparts, patients with NDD were younger, experienced more socio-economic disadvantage and less private insurance coverage, suffered with less severe but different co-morbid clinical conditions and incurred more challenges in the acute hospital care setting. CONCLUSION Although the pattern of ADE-related hospitalisations in patients with NDD differed from that in patients without NDD, there is a lack of targeted healthcare programmes to meet their special needs. This study suggests the need for countermeasures in primary healthcare settings to reduce the burden of ADEs in this vulnerable group.
Collapse
Affiliation(s)
- M Zhou
- Research School of Population Health, Australian National University, Canberra, ACT, Australia
- Pharmacy department, People's Hospital of Xinjiang, Urumqi, China
| | - W Du
- Department of Health Services Research and Policy, Research School of Population Health, Australian National University, Canberra, Australia
| | - L Salvador-Carulla
- Centre of Mental Health Research, Research School of Population Health, Australian National University, Canberra, Australia
| | - N Glasgow
- Department of Health Services Research and Policy, Research School of Population Health, Australian National University, Canberra, Australia
| |
Collapse
|
28
|
Hoffmann A, Ziller M, Spengler D. Childhood-Onset Schizophrenia: Insights from Induced Pluripotent Stem Cells. Int J Mol Sci 2018; 19:E3829. [PMID: 30513688 PMCID: PMC6321410 DOI: 10.3390/ijms19123829] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/21/2018] [Accepted: 11/27/2018] [Indexed: 01/25/2023] Open
Abstract
Childhood-onset schizophrenia (COS) is a rare psychiatric disorder characterized by earlier onset, more severe course, and poorer outcome relative to adult-onset schizophrenia (AOS). Even though, clinical, neuroimaging, and genetic studies support that COS is continuous to AOS. Early neurodevelopmental deviations in COS are thought to be significantly mediated through poorly understood genetic risk factors that may also predispose to long-term outcome. In this review, we discuss findings from induced pluripotent stem cells (iPSCs) that allow the generation of disease-relevant cell types from early brain development. Because iPSCs capture each donor's genotype, case/control studies can uncover molecular and cellular underpinnings of COS. Indeed, recent studies identified alterations in neural progenitor and neuronal cell function, comprising dendrites, synapses, electrical activity, glutamate signaling, and miRNA expression. Interestingly, transcriptional signatures of iPSC-derived cells from patients with COS showed concordance with postmortem brain samples from SCZ, indicating that changes in vitro may recapitulate changes from the diseased brain. Considering this progress, we discuss also current caveats from the field of iPSC-based disease modeling and how to proceed from basic studies to improved diagnosis and treatment of COS.
Collapse
Affiliation(s)
- Anke Hoffmann
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| | - Michael Ziller
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| | - Dietmar Spengler
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany.
| |
Collapse
|