51
|
Segura AG, Mané A, Prohens L, Rodriguez N, Mezquida G, Cuesta MJ, Vieta E, Amoretti S, Lobo A, González-Pinto A, Diaz-Caneja CM, Roldán-Bejarano A, Jimenez E, Baeza I, Legido T, Saiz-Ruiz J, Bernardo M, Mas S. Exploration of cannabis use and polygenic risk scores on the psychotic symptom progression of a FEP cohort. Psychiatry Res 2023; 325:115249. [PMID: 37178502 DOI: 10.1016/j.psychres.2023.115249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/05/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
Cannabis use is highly prevalent in first-episode psychosis (FEP) and plays a critical role in its onset and prognosis, but the genetic underpinnings promoting both conditions are poorly understood. Current treatment strategies for cannabis cessation in FEP are clearly inefficacious. Here, we aimed to characterize the association between cannabis-related polygenic risk scores (PRS) on cannabis use and clinical course after a FEP. A cohort of 249 FEP individuals were evaluated during 12 months. Symptom severity was measured with the Positive and Negative Severity Scale and cannabis use with the EuropASI scale. Individual PRS for lifetime cannabis initiation (PRSCI) and cannabis use disorder (PRSCUD) were constructed. Current cannabis use was associated with increased positive symptoms. Cannabis initiation at younger ages conditioned the 12-month symptom progression. FEP patients with higher cannabis PRSCUD reported increased baseline cannabis use. PRSCI was associated with the course of negative and general symptomatology over follow-up. Cannabis use and symptom progression after a FEP were modulated by cannabis PRS, suggesting that lifetime initiation and use disorders may have partially independent genetic factors. These exploratory results may be the first step to identify those FEP patients more vulnerable to cannabis use and worse outcomes to ultimately develop tailored treatments.
Collapse
Affiliation(s)
- Alex G Segura
- Department of Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Anna Mané
- Hospital del Mar Medical Research Institute, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Llucia Prohens
- Department of Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Natalia Rodriguez
- Department of Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain
| | - Gisela Mezquida
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain; Barcelona Clínic Schizophrenia Unit, Neuroscience Institute Hospital Clínic de Barcelona, Barcelona, Spain
| | - Manuel J Cuesta
- Department of Psychiatry, Complejo Hospitalario de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Eduard Vieta
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain; Hospital Clínic de Barcelona, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Silvia Amoretti
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain; Bipolar and Depressive Disorder Unit, Neuroscience Institute, Hospital Clínic de Barcelona, Barcelona, Spain; Group of Psychiatry, Mental Health and Addictions, Psychiatric Genetics Unit, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Antonio Lobo
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Department of Medicine and Psychiatry, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
| | - Ana González-Pinto
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Hospital Universitario de Alava, Vitoria-Gasteiz, Spain; Instituto de Investigación Sanitaria Bioaraba, Vitoria-Gasteiz, Spain; University of the Basque Country, Vizcaya, Spain
| | - Covadonga M Diaz-Caneja
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; School of Medicine, Universidad Complutense, Madrid, Spain
| | - Alexandra Roldán-Bejarano
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Psychiatry Department, Institut d'Investigació Biomèdica-Sant Pau (IIB-SANTPAU), Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Esther Jimenez
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain; Hospital Clínic de Barcelona, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Immaculada Baeza
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain; Department of Child and Adolescent Psychiatry and Psychology, Clínic Institute of Neurosciences, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Teresa Legido
- Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Jeronimo Saiz-Ruiz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Department of Psychiatry, Universidad de Alcala, Alcalá de Henares, Spain; Hospital Ramon y Cajal, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), Madrid, Spain
| | - Miguel Bernardo
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain; Barcelona Clínic Schizophrenia Unit, Neuroscience Institute Hospital Clínic de Barcelona, Barcelona, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Sergi Mas
- Department of Clinical Foundations, Pharmacology Unit, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPs), Barcelona, Spain.
| |
Collapse
|
52
|
Tuo LJ, Song XY, Zhu YY, He HN, Song YP, Chen DZ, Zheng XM, Zhang H, Xu DX. Gestational folic acid supplement prevents vitamin D deficiency-induced depression-like behavior by reversing cortical DNA hypomethylation in adult offspring. J Steroid Biochem Mol Biol 2023; 231:106313. [PMID: 37075986 DOI: 10.1016/j.jsbmb.2023.106313] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/27/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023]
Abstract
Depression is a common mental disorder with an increasing incidence. Several studies have demonstrated that cortical DNA hypomethylation is associated with depression-like behaviors. This study aims to investigate whether maternal vitamin D deficiency (VDD) induces depression-like behaviors and to explore the effects of folic acid supplement on VDD-induced cortical DNA hypomethylation in adult offspring. Female mice were fed with a VDD diet, beginning at 5 weeks of age and throughout pregnancy. Depression-like behaviors were evaluated, and cortical 5-methylcytosine (5mC) content was detected in adult offspring. Results showed that depression-like behaviors were observed in adult offspring of the VDD group. Cortical Ache and Oxtr mRNAs were upregulated in female offspring of the VDD group. Cortical Cpt1a and Htr1b mRNAs were increased in male offspring of the VDD group. Moreover, cortical 5mC content was reduced in offspring of VDD-fed dams. The additional experiment showed that serum folate and cortical S-adenosylmethionine (SAM) contents were decreased in the offspring of the VDD group. Folic acid supplement attenuated VDD-induced SAM depletion and reversed cortical DNA methylation. Moreover, folic acid supplement attenuated VDD-induced upregulation of depression-related genes. In addition, folic acid supplement alleviated maternal VDD-induced depression-like behaviors in adult offspring. These results suggest that maternal VDD induces depression-like behavior in adult offspring by reducing cortical DNA methylation. The gestational folic acid supplement prevents VDD-induced depression-like behavior by reversing cortical DNA hypomethylation in adult offspring.
Collapse
Affiliation(s)
- Ling-Jin Tuo
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Xiao-Yue Song
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Yan-Yan Zhu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Hong-Ning He
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Ya-Ping Song
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Dao-Zhen Chen
- Department of Clinical Laboratory, Wuxi Maternity and Child Health Care Hospital, Wuxi 214002, China; Laboratory Department of Haidong Second People's Hospital, Haidong, Qinghai 810699, China
| | - Xiao-Min Zheng
- Research Institute for Reproductive Medicine and Genetic Diseases, Wuxi Maternity and Child Health Hospital, Wuxi, 214002, China
| | - Heng Zhang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China; Department of Child Health Care, Wuxi Maternity and Child Health Care Hospital, Wuxi 214002, China.
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
53
|
Siracusa L, Ruberto G, Cristino L. Recent Research on Cannabis sativa L.: Phytochemistry, New Matrices, Cultivation Techniques, and Recent Updates on Its Brain-Related Effects (2018-2023). Molecules 2023; 28:molecules28083387. [PMID: 37110621 PMCID: PMC10146690 DOI: 10.3390/molecules28083387] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/05/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Cannabis sativa L. is a plant that humankind has been using for millennia. The basis of its widespread utilization is its adaptability to so many different climatic conditions, with easy cultivability in numerous diverse environments. Because of its variegate phytochemistry, C. sativa has been used in many sectors, although the discovery of the presence in the plant of several psychotropic substances (e.g., Δ9-tetrahydrocannabinol, THC) caused a drastic reduction of its cultivation and use together with its official ban from pharmacopeias. Fortunately, the discovery of Cannabis varieties with low content of THC as well as the biotechnological development of new clones rich in many phytochemical components endorsed with peculiar and many important bioactivities has demanded the reassessment of these species, the study and use of which are currently experiencing new and important developments. In this review we focus our attention on the phytochemistry, new matrices, suitable agronomic techniques, and new biological activities developed in the five last years.
Collapse
Affiliation(s)
- Laura Siracusa
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Paolo Gaifami, 18, 95126 Catania, CT, Italy
| | - Giuseppe Ruberto
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Paolo Gaifami, 18, 95126 Catania, CT, Italy
| | - Luigia Cristino
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei, 34, 80078 Pozzuoli, NA, Italy
| |
Collapse
|
54
|
Krassnitzer M, Boisvert B, Beiersdorf J, Harkany T, Keimpema E. Resident Astrocytes can Limit Injury to Developing Hippocampal Neurons upon THC Exposure. Neurochem Res 2023; 48:1242-1253. [PMID: 36482034 PMCID: PMC10030412 DOI: 10.1007/s11064-022-03836-1] [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: 10/06/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022]
Abstract
Cannabis legalization prompted the dilemma if plant-derived recreational drugs can have therapeutic potential and, consequently, how to address their regulation and safe distribution. In parallel, the steady worldwide decriminalization of cannabis and the enhanced content of its main psychoactive compound Δ9-tetrahydrocannabinol (THC), exposes populations to increasing amounts of cannabis and THC across all ages. While adverse effects of cannabis during critical stages of fetal neurodevelopment are investigated, these studies center on neurons alone. Thus, a gap of knowledge exists on how intercellular interactions between neighboring cell types, particularly astrocytes and neurons, could modify THC action. Here, we combine transcriptome analysis, transgenic models, high resolution microscopy and live cell imaging to demonstrate that hippocampal astrocytes accumulate in the strata radiatum and lacunosum moleculare of the CA1 subfield, containing particularly sensitive neurons to stressors, upon long term postnatal THC exposure in vivo. As this altered distribution is not dependent on cell proliferation, we propose that resident astrocytes accumulate in select areas to protect pyramidal neurons and their neurite extensions from pathological damage. Indeed, we could recapitulate the neuroprotective effect of astrocytes in vitro, as their physical presence significantly reduced the death of primary hippocampal neurons upon THC exposure (> 5 µM). Even so, astrocytes are also affected by a reduced metabolic readiness to stressors, as reflected by a downregulation of mitochondrial proteins. Thus, we find that astrocytes exert protective functions on local neurons during THC exposure, even though their mitochondrial electron transport chain is disrupted.
Collapse
Affiliation(s)
- Maria Krassnitzer
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Brooke Boisvert
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Johannes Beiersdorf
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Karolinska Institutet, Biomedicum 7D, Solna, Sweden
| | - Erik Keimpema
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
55
|
Carrica LK, Choi CY, Walter FA, Noonan BL, Shi L, Johnson CT, Bradshaw HB, Liang NC, Gulley JM. Effects of combined use of alcohol and delta-9-tetrahydrocannibinol on working memory in Long Evans rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526698. [PMID: 36778500 PMCID: PMC9915622 DOI: 10.1101/2023.02.02.526698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The increase in social acceptance and legalization of cannabis over the last several years is likely to increase the prevalence of its co-use with alcohol. In spite of this, the potential for effects unique to co-use of these drugs, especially in moderate doses, has been studied relatively infrequently. We addressed this in the current study using a laboratory rat model of voluntary drug intake. Periadolescent male and female Long-Evans rats were allowed to orally self-administer ethanol, Î" 9 -tetrahydrocannibinol (THC), both drugs, or their vehicle controls from postnatal day (P) 30 to P47. They were subsequently trained and tested on an instrumental behavior task that assesses attention, working memory and behavioral flexibility. Similar to previous work, consumption of THC reduced both ethanol and saccharin intake in both sexes. Blood samples taken 14h following the final self-administration session revealed that females had higher levels of the THC metabolite THC-COOH. There were modest effects of THC on our delayed matching to position (DMTP) task, with females exhibiting reduced performance compared to their control group or male, drug using counterparts. However, there were no significant effects of co-use of ethanol or THC on DMTP performance, and drug effects were also not apparent in the reversal learning phase of the task when non-matching to position was required as the correct response. These findings are consistent with other published studies in rodent models showing that use of these drugs in low to moderate doses does not significantly impact memory or behavioral flexibility following a protracted abstinence period.
Collapse
Affiliation(s)
- Lauren K. Carrica
- Department of Psychology, University of Illinois at Urbana-Champaign
| | - Chan Young Choi
- Department of Psychology, University of Illinois at Urbana-Champaign
| | | | - Brynn L. Noonan
- Department of Psychology, University of Illinois at Urbana-Champaign
| | - Linyuan Shi
- Department of Psychology, University of Illinois at Urbana-Champaign
| | - Clare T. Johnson
- Department of Psychological & Brain Science, Indiana University, Bloomington, Indiana
| | - Heather B. Bradshaw
- Department of Psychological & Brain Science, Indiana University, Bloomington, Indiana
| | - Nu-Chu Liang
- Department of Psychology, University of Illinois at Urbana-Champaign
- Neuroscience Program, University of Illinois at Urbana-Champaign
| | - Joshua M. Gulley
- Department of Psychology, University of Illinois at Urbana-Champaign
- Neuroscience Program, University of Illinois at Urbana-Champaign
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign
| |
Collapse
|
56
|
Emerging Roles of Endocannabinoids as Key Lipid Mediators for a Successful Pregnancy. Int J Mol Sci 2023; 24:ijms24065220. [PMID: 36982295 PMCID: PMC10048990 DOI: 10.3390/ijms24065220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
In recent years, Cannabis use/misuse for treating pregnancy-related symptoms and other chronic conditions has increased among pregnant women, favored by decriminalization and/or legalization of its recreational uses in addition to its easy accessibility. However, there is evidence that prenatal Cannabis exposure might have adverse consequences on pregnancy progression and a deleterious impact on proper neurodevelopmental trajectories in the offspring. Maternal Cannabis use could interfere with the complex and finely controlled role performed by the endocannabinoid system in reproductive physiology, impairing multiple gestational processes from blastocyst implantation to parturition, with long-lasting intergenerational effects. In this review, we discuss current clinical and preclinical evidence regarding the role of endocannabinoids in development, function, and immunity of the maternal–fetal interface, focusing on the impact of Cannabis constituents on each of these gestational processes. We also discuss the intrinsic limitations of the available studies and the future perspectives in this challenging research field.
Collapse
|
57
|
Masataka Y, Sugiyama T, Akahoshi Y, Matsumoto T. Risk factors for cannabis use disorders and cannabis psychosis in Japan: Second report of a survey on cannabis-related health problems among community cannabis users using social networking services. Neuropsychopharmacol Rep 2023; 43:85-94. [PMID: 36537061 PMCID: PMC10009416 DOI: 10.1002/npr2.12307] [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: 08/13/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
AIM To determine the risk factors for cannabis use disorders and cannabis psychosis in Japan based on a 2021 online survey among Japanese users of social network services. METHODS The 3142 respondents who had used cannabis within the preceding year were categorized into two groups based on the development of cannabis use disorder and/or cannabis psychosis. Analyses were performed to determine these conditions' risk factors. RESULTS Multivariate analysis revealed that factors significantly associated with cannabis use disorders were "cannabis-use initiation at a young age" (p = 0.004, OR: 0.951, 95% CI [0.920-0.984]), "family history of mental health problems" (p < 0.001, OR: 1.988, 95% CI [1.545-2.556]), "psychiatric disorders preceding cannabis use" (p = 0.039, OR: 1.672, 95% CI [1.026-2.722]), and "use of cannabis products other than dry flower" (p < 0.001, OR: 2.725, 95% CI [1.844-4.026]). Factors significantly associated with cannabis psychosis were "cannabis-use initiation at a young age" (p = 0.011, OR: 0.888, 95% CI [0.811-0.973]) and "family history of mental health problems" (p = 0.002, OR: 2.531, 95% CI [1.400-4. 576]). CONCLUSION Risk factors for problematic cannabis use were cannabis initiation at a young age, pre-cannabis psychiatric disorders, family history of mental health problems, and the use of cannabis products other than dry flower. However, the causal relationship among these factors remains ambivalent.
Collapse
Affiliation(s)
- Yuji Masataka
- General Incorporated Association Green Zone Japan, Saitama, Japan.,Department of Neurology, Kumamoto Seijo Hospital, Kumamoto, Japan
| | - Takeshi Sugiyama
- General Incorporated Association Green Zone Japan, Saitama, Japan
| | - Yoshiyuki Akahoshi
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa, Japan
| | - Toshihiko Matsumoto
- Department of Drug Dependence Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
58
|
Hiraoka D, Makita K, Hamatani S, Tomoda A, Mizuno Y. Effects of prenatal cannabis exposure on developmental trajectory of cognitive ability and brain volumes in the adolescent brain cognitive development (ABCD) study. Dev Cogn Neurosci 2023; 60:101209. [PMID: 36791556 PMCID: PMC9950823 DOI: 10.1016/j.dcn.2023.101209] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/12/2022] [Accepted: 02/04/2023] [Indexed: 02/08/2023] Open
Abstract
Although cannabis use during pregnancy is increasing widely, the effects of cannabis on developmental trajectories, such as whether its effects during pregnancy remain the same between time points or gradually increase, are unclear. This study aimed to examine whether cannabis use during pregnancy affects the process of change in cognition and brain volume. Data from two-time points measured longitudinally were analyzed. We used data from the Adolescent Brain and Cognitive Development Study. Participants included 11,876 children aged 9-11 years participated at baseline, and 10,414 participated at 2-year follow-up from 22 sites across the United States. We explored the associations between prenatal cannabis exposure and cognitive abilities and brain volumes developmental trajectories. Among 11,530 children with valid data for prenatal cannabis exposure, 10,833 had no prenatal cannabis use, and 697 had cannabis use during their pregnancy. There was a significant interaction between time points and cannabis use during pregnancy on visuo-perceptual processing ability (b = -0.019, p = .009) and intracranial volumes (b = -6338.309, p = .009). We found that the effects of exposure to cannabis during pregnancy are not uniform at all times and may gradually become more apparent and magnified as development progresses.
Collapse
Affiliation(s)
- Daiki Hiraoka
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan,The Japan Society for the Promotion of Science, Tokyo, Japan
| | - Kai Makita
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan,Division of Developmental Higher Brain Functions, United Graduate School of Child Development, University of Fukui, Fukui, Japan
| | - Sayo Hamatani
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan,Division of Developmental Higher Brain Functions, United Graduate School of Child Development, University of Fukui, Fukui, Japan,Department of Child and Adolescent Psychological Medicine, University of Fukui Hospital, Fukui, Japan
| | - Akemi Tomoda
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan; Division of Developmental Higher Brain Functions, United Graduate School of Child Development, University of Fukui, Fukui, Japan; Department of Child and Adolescent Psychological Medicine, University of Fukui Hospital, Fukui, Japan.
| | - Yoshifumi Mizuno
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan; Division of Developmental Higher Brain Functions, United Graduate School of Child Development, University of Fukui, Fukui, Japan; Department of Child and Adolescent Psychological Medicine, University of Fukui Hospital, Fukui, Japan.
| |
Collapse
|
59
|
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: 1] [Impact Index Per Article: 1.0] [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
|
60
|
Sadaka AH, Canuel J, Febo M, Johnson CT, Bradshaw HB, Ortiz R, Ciumo F, Kulkarni P, Gitcho MA, Ferris CF. Effects of inhaled cannabis high in Δ9-THC or CBD on the aging brain: A translational MRI and behavioral study. Front Aging Neurosci 2023; 15:1055433. [PMID: 36819730 PMCID: PMC9930474 DOI: 10.3389/fnagi.2023.1055433] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/03/2023] [Indexed: 02/04/2023] Open
Abstract
With the recent legalization of inhaled cannabis for medicinal and recreational use, the elderly represents one of the newest, rapidly growing cohorts of cannabis users. To understand the neurobiological effects of cannabis on the aging brain, 19-20 months old mice were divided into three groups exposed to vaporized cannabis containing ~10% Δ9-THC, ~10% CBD, or placebo for 30 min each day. Voxel based morphometry, diffusion weighted imaging, and resting state functional connectivity data were gathered after 28 days of exposure and following a two-week washout period. Tail-flick, open field, and novel object preference tests were conducted to explore analgesic, anxiolytic, and cognitive effects of cannabis, respectively. Vaporized cannabis high in Δ9-THC and CBD achieved blood levels reported in human users. Mice showed antinociceptive effects to chronic Δ9-THC without tolerance while the anxiolytic and cognitive effects of Δ9-THC waned with treatment. CBD had no effect on any of the behavioral measures. Voxel based morphometry showed a decrease in midbrain dopaminergic volume to chronic Δ9-THC followed but an increase after a two-week washout. Fractional anisotropy values were reduced in the same area by chronic Δ9-THC, suggesting a reduction in gray matter volume. Cannabis high in CBD but not THC increased network strength and efficiency, an effect that persisted after washout. These data would indicate chronic use of inhaled cannabis high in Δ9-THC can be an effective analgesic but not for treatment of anxiety or cognitive decline. The dopaminergic midbrain system was sensitive to chronic Δ9-THC but not CBD showing robust plasticity in volume and water diffusivity prior to and following drug cessation an effect possibly related to the abuse liability of Δ9-THC. Chronic inhaled CBD resulted in enhanced global network connectivity that persisted after drug cessation. The behavioral consequences of this sustained change in brain connectivity remain to be determined.
Collapse
Affiliation(s)
- Aymen H. Sadaka
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Justin Canuel
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Marcelo Febo
- Department of Psychiatry and Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - Clare T. Johnson
- Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, IN, United States
| | - Heather B. Bradshaw
- Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, IN, United States
| | - Richard Ortiz
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM, United States
| | - Federica Ciumo
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Praveen Kulkarni
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - Michael A. Gitcho
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, United States
| | - Craig F. Ferris
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
- Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| |
Collapse
|
61
|
Frau R, Melis M. Sex-specific susceptibility to psychotic-like states provoked by prenatal THC exposure: Reversal by pregnenolone. J Neuroendocrinol 2023; 35:e13240. [PMID: 36810840 DOI: 10.1111/jne.13240] [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: 08/01/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Sociocultural attitudes towards cannabis legalization contribute to the common misconception that it is a relatively safe drug and its use during pregnancy poses no risk to the fetus. However, longitudinal studies demonstrate that maternal cannabis exposure results in adverse outcomes in the offspring, with a heightened risk for developing psychopathology. One of the most reported psychiatric outcomes is the proneness to psychotic-like experiences during childhood. How exposure to cannabis during gestation increases psychosis susceptibility in children and adolescents remains elusive. Preclinical research has indicated that in utero exposure to the major psychoactive component of cannabis, delta-9-tetrahydrocannabinol (THC), deranges brain developmental trajectories towards vulnerable psychotic-like endophenotypes later in life. Here, we present how prenatal THC exposure (PCE) deregulates mesolimbic dopamine development predisposing the offspring to schizophrenia-relevant phenotypes, exclusively when exposed to environmental challenges, such as stress or THC. Detrimental effects of PCE are sex-specific because female offspring do not display psychotic-like outcomes upon exposure to these challenges. Moreover, we present how pregnenolone, a neurosteroid that showed beneficial properties on the effects elicited by cannabis intoxication, normalizes mesolimbic dopamine function and rescues psychotic-like phenotypes. We, therefore, suggest this neurosteroid as a safe "disease-modifying" aid to prevent the onset of psychoses in vulnerable individuals. Our findings corroborate clinical evidence and highlight the relevance of early diagnostic screening and preventative strategies for young individuals at risk for mental diseases, such as male PCE offspring.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
- The Guy Everett Laboratory for Neuroscience, University of Cagliari, Cagliari, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| |
Collapse
|
62
|
Iyer P, Watanabe M, Artinger KB. Emerging understanding of the effects of cannabis use during pregnancy. Birth Defects Res 2023; 115:129-132. [PMID: 36181322 DOI: 10.1002/bdr2.2097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 01/26/2023]
Affiliation(s)
- Poorni Iyer
- Staff Toxicologist (Specialist), California EPA-Office of Environmental Health Hazards Assessment, Division of Scientific Programs-Reproductive and Cancer Hazard Assessment Branch, Reproductive Toxicology and Epidemiology Section, Sacramento, California, USA
| | - Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University, School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| | - Kristin B Artinger
- Department of Craniofacial Biology, Co-Director, Biomedical Sciences Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
63
|
Ferland JMN, Ellis RJ, Betts G, Silveira MM, de Firmino JB, Winstanley CA, Hurd YL. Long-Term Outcomes of Adolescent THC Exposure on Translational Cognitive Measures in Adulthood in an Animal Model and Computational Assessment of Human Data. JAMA Psychiatry 2023; 80:66-76. [PMID: 36416863 PMCID: PMC9685552 DOI: 10.1001/jamapsychiatry.2022.3915] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/02/2022] [Indexed: 11/24/2022]
Abstract
Importance Although perceived as relatively harmless and nonaddictive, adolescent cannabis use significantly increases the likelihood of developing cannabis use disorder in adulthood, especially for high-potency cannabis. Risky decision-making is associated with chronic cannabis use, but given confounds of human studies, it remains unclear whether adolescent cannabis exposure and Δ9-tetrahydrocannabinol (THC) potency specifically predicts risky decision-making or influences cognitive response to the drug later in life. Objective To leverage a human data set of cannabis users and a rat model to evaluate the long-term outcomes of adolescent THC exposure on adult decision-making and impulse control. Design, Setting, and Participants This translational rat study tested the link between adolescent THC exposure and adulthood decision-making. A reanalysis of a previously published dataset of human chronic cannabis users was conducted to evaluate decision-making phenotypes. Computational modeling assessed the human and animal results in a single framework. Data were collected from 2017 to 2020 and analyzed from 2020 to 2022. Main Outcomes and Measures Decision-making was measured by the Iowa Gambling Task (IGT) and Rat Gambling Task (rGT). Impulse control was assessed in the rat model. Computational modeling was used to determine reward and punishment learning rates and learning strategy used by cannabis users and THC-exposed rats. Cell-specific molecular measures were conducted in the prefrontal cortex and amygdala. Results Of 37 participants, 24 (65%) were male, and the mean (SD) age was 33.0 (8.3) years. Chronic cannabis users (n = 22; mean [SE] IGT score, -5.182 [1.262]) showed disadvantageous decision-making compared with controls (n = 15; mean [SE] IGT score, 7.133 [2.687]; Cohen d = 1.436). Risky choice was associated with increased reward learning (mean [SE] IGT score: cannabis user, 0.170 [0.018]; control, 0.046 [0.008]; Cohen d = 1.895) and a strategy favoring exploration vs long-term gains (mean [SE] IGT score: cannabis user, 0.088 [0.012]; control, 0.020 [0.002]; Cohen d = 2.218). Rats exposed to high-dose THC but not low-dose THC during adolescence also showed increased risky decision-making (mean [SE] rGT score: vehicle, 46.17 [7.02]; low-dose THC, 69.45 [6.01]; high-dose THC, 21.97 [11.98]; Cohen d = 0.433) and elevated reward learning rates (mean [SE] rGT score: vehicle, 0.17 [0.01]; low-dose THC, 0.10 [0.01]; high-dose THC, 0.24 [0.06]; Cohen d = 1.541) during task acquisition. These animals were also uniquely susceptible to increased cognitive impairments after reexposure to THC in adulthood, which was correlated with even greater reward learning (r = -0.525; P < .001) and a shift in strategy (r = 0.502; P < .001), similar to results seen in human cannabis users. Molecular studies revealed that adolescent THC dose differentially affected cannabinoid-1 receptor messenger RNA expression in the prelimbic cortex and basolateral amygdala in a layer- and cell-specific manner. Further, astrocyte glial fibrillary acidic protein messenger RNA expression associated with cognitive deficits apparent with adult THC reexposure. Conclusions and Relevance In this translational study, high-dose adolescent THC exposure was associated with cognitive vulnerability in adulthood, especially with THC re-exposure. These data also suggest a link between astrocytes and cognition that altogether provides important insights regarding the neurobiological genesis of risky cannabis use that may help promote prevention and treatment efforts.
Collapse
Affiliation(s)
- Jacqueline-Marie N. Ferland
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Randall J. Ellis
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Addiction Institute of Mount Sinai, New York, New York
| | - Graeme Betts
- Djavad Mowafaghian Centre for Brain Health, Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mason M. Silveira
- Djavad Mowafaghian Centre for Brain Health, Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joao Bronze de Firmino
- The Collaborative Advanced Microscopy Laboratories of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Catharine A. Winstanley
- Djavad Mowafaghian Centre for Brain Health, Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yasmin L. Hurd
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Addiction Institute of Mount Sinai, New York, New York
| |
Collapse
|
64
|
Simmons SC, Grecco GG, Atwood BK, Nugent FS. Effects of prenatal opioid exposure on synaptic adaptations and behaviors across development. Neuropharmacology 2023; 222:109312. [PMID: 36334764 PMCID: PMC10314127 DOI: 10.1016/j.neuropharm.2022.109312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
In this review, we focus on prenatal opioid exposure (POE) given the significant concern for the mental health outcomes of children with parents affected by opioid use disorder (OUD) in the view of the current opioid crisis. We highlight some of the less explored interactions between developmental age and sex on synaptic plasticity and associated behavioral outcomes in preclinical POE research. We begin with an overview of the rich literature on hippocampal related behaviors and plasticity across POE exposure paradigms. We then discuss recent work on reward circuit dysregulation following POE. Additional risk factors such as early life stress (ELS) could further influence synaptic and behavioral outcomes of POE. Therefore, we include an overview on the use of preclinical ELS models where ELS exposure during key critical developmental periods confers considerable vulnerability to addiction and stress psychopathology. Here, we hope to highlight the similarity between POE and ELS on development and maintenance of opioid-induced plasticity and altered opioid-related behaviors where similar enduring plasticity in reward circuits may occur. We conclude the review with some of the limitations that should be considered in future investigations. This article is part of the Special Issue on 'Opioid-induced addiction'.
Collapse
Affiliation(s)
- Sarah C Simmons
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Greg G Grecco
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Brady K Atwood
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fereshteh S Nugent
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
65
|
Iezzi D, Caceres-Rodriguez A, Chavis P, Manzoni OJJ. In utero exposure to cannabidiol disrupts select early-life behaviors in a sex-specific manner. Transl Psychiatry 2022; 12:501. [PMID: 36470874 PMCID: PMC9722662 DOI: 10.1038/s41398-022-02271-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/15/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Cannabidiol (CBD), one of the main components of cannabis, is generally considered safe. CBD crosses the placenta and its use during pregnancy is steadily increasing, the impact of gestational CBD's effects on prenatal life and neurodevelopment are poorly understood. Here, we combined behavioral approaches and deep learning analysis to assess the sex-dependent neonatal behavior of CBD exposed progeny. Gestating C57BL6/J dams were exposed daily with vehicle or CBD (3 mg/Kg, s.c.), from gestational day 5 to 18. Body weight, pup ultrasound vocalizations (USVs, PND 10) and homing behavior (PND 13) were quantified in the progeny. Thus, male (but not female) pups from CBD-treated dams gained more weight than sham. There were sex-dependent differences in the coarse characteristics of ultrasonic vocalizations. Prenatally-CBD exposed male pups emitted shorter calls, whereas CBD females made more high frequency calls when compared with their control counterparts. There were significant qualitative changes in the syllabic USV repertoire reflected in call typologies and communication patterns. Finally, the homing behavior test showed that CBD-exposed females presented a greater vulnerability to gestational CBD than males. Only CBD-exposed female pups showed reduced motor and discriminatory abilities. Together the results suggest a sexual divergence in the consequences of in utero CBD exposure on neonates at early developmental ages, which may be predictive of adult psychopathology. Given the extent of cannabis and CBD use worldwide, these findings challenge the idea that CBD is a universally safe compound and reveal the need for additional studies on the effect of perinatal CBD exposure.
Collapse
Affiliation(s)
- Daniela Iezzi
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Cannalab "Cannabinoids Neuroscience Research International Associated Laboratory". INSERM-Aix-Marseille University/Indiana University, Marseille, France
| | - Alba Caceres-Rodriguez
- INMED, INSERM U1249, Marseille, France
- Aix-Marseille University, Marseille, France
- Cannalab "Cannabinoids Neuroscience Research International Associated Laboratory". INSERM-Aix-Marseille University/Indiana University, Marseille, France
| | - Pascale Chavis
- INMED, INSERM U1249, Marseille, France.
- Aix-Marseille University, Marseille, France.
- Cannalab "Cannabinoids Neuroscience Research International Associated Laboratory". INSERM-Aix-Marseille University/Indiana University, Marseille, France.
| | - Olivier J J Manzoni
- INMED, INSERM U1249, Marseille, France.
- Aix-Marseille University, Marseille, France.
- Cannalab "Cannabinoids Neuroscience Research International Associated Laboratory". INSERM-Aix-Marseille University/Indiana University, Marseille, France.
| |
Collapse
|
66
|
Potschka H, Bhatti SFM, Tipold A, McGrath S. Cannabidiol in canine epilepsy. Vet J 2022; 290:105913. [PMID: 36209995 DOI: 10.1016/j.tvjl.2022.105913] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
Abstract
The anticonvulsant effect of cannabidiol (CBD), which has been confirmed by findings from animal models and human trials, has attracted the interest of veterinary practitioners and dog owners. Moreover, social media and public pressure has sparked a renewed awareness of cannabinoids, which have been used for epilepsy since ancient times. Unfortunately, at this moment veterinarians and veterinary neurologists have difficulty prescribing cannabinoids because of the paucity of sound scientific studies. Pharmacokinetic studies in dogs have demonstrated a low oral bioavailability of CBD and a high first-pass effect through the liver. Administering CBD in oil-based formulations and/or with food has been shown to enhance the bioavailability in dogs, rats and humans. Tolerability studies in healthy dogs and dogs with epilepsy have demonstrated that CBD was safe and well tolerated with only mild to moderate adverse effects. In this context, it should be noted that the quality of available CBD varies widely, underscoring the importance of pharmaceutical quality and its control. One clinical trial in dogs with drug-resistant idiopathic epilepsy failed to confirm a difference in response rates between the CBD group and the placebo group, while in another cross-over trial a ≥ 50 % reduction in epileptic seizure frequency was found in six of 14 dogs in the treatment phase, a reduction that was not observed during the placebo phase. Based on the current state of knowledge it is not possible to provide clear-cut recommendations for the use of CBD in canine epilepsy. Randomized controlled canine trials with large sample sizes are needed to determine the range of therapeutic plasma concentrations, develop evidence-based dosing regimens, determine the efficacy of cannabidiol in drug-refractory epilepsy, and explore potential associations between treatment effects and different etiologies, epilepsy types, and drug combinations.
Collapse
Affiliation(s)
- Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany.
| | - Sofie F M Bhatti
- Small Animal Department, Small Animal Teaching Hospital, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Andrea Tipold
- Department Small Animal Medicine and Surgery, University of Veterinary Medicine, Hannover, Germany
| | - Stephanie McGrath
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Colorado, USA
| |
Collapse
|
67
|
Peters KZ, Naneix F. The role of dopamine and endocannabinoid systems in prefrontal cortex development: Adolescence as a critical period. Front Neural Circuits 2022; 16:939235. [PMID: 36389180 PMCID: PMC9663658 DOI: 10.3389/fncir.2022.939235] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/14/2022] [Indexed: 01/07/2023] Open
Abstract
The prefrontal cortex plays a central role in the control of complex cognitive processes including action control and decision making. It also shows a specific pattern of delayed maturation related to unique behavioral changes during adolescence and allows the development of adult cognitive processes. The adolescent brain is extremely plastic and critically vulnerable to external insults. Related to this vulnerability, adolescence is also associated with the emergence of numerous neuropsychiatric disorders involving alterations of prefrontal functions. Within prefrontal microcircuits, the dopamine and the endocannabinoid systems have widespread effects on adolescent-specific ontogenetic processes. In this review, we highlight recent advances in our understanding of the maturation of the dopamine system and the endocannabinoid system in the prefrontal cortex during adolescence. We discuss how they interact with GABA and glutamate neurons to modulate prefrontal circuits and how they can be altered by different environmental events leading to long-term neurobiological and behavioral changes at adulthood. Finally, we aim to identify several future research directions to help highlight gaps in our current knowledge on the maturation of these microcircuits.
Collapse
Affiliation(s)
- Kate Zara Peters
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom
| | - Fabien Naneix
- The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom,*Correspondence: Fabien Naneix
| |
Collapse
|
68
|
Cytotoxic Effects of Cannabidiol on Neonatal Rat Cortical Neurons and Astrocytes: Potential Danger to Brain Development. Toxins (Basel) 2022; 14:toxins14100720. [PMID: 36287988 PMCID: PMC9611593 DOI: 10.3390/toxins14100720] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/29/2022] Open
Abstract
The influence of cannabidiol (CBD) on brain development is inadequately understood. Since CBD is considered a non-intoxicating drug, it has attracted great interest concerning its potential medical applicability, including in pregnant women and children. Here, we elucidated the response of perinatal rat cortical neurons and astrocytes to CBD at submicromolar (0.1, 0.5, 1, 5 µM) concentrations attainable in humans. The effect of CBD was concentration- and time-dependent and cell-specific. In neurons, 0.1 µM CBD induced an early and transient change in mitochondrial membrane potential (ΔΨm), ATP depletion, and caspase-8 activation, followed by rapid ATP recovery and progressive activation of caspase-9 and caspase-3/7, resulting in early apoptotic cell death with reduction and shortening of dendrites, cell shrinkage, and chromatin condensation. The decrease in neuronal viability, ATP depletion, and caspase activation due to CBD exposure was prevented by transient receptor potential vanilloid 1 (TRPV1) antagonist. In astrocytes, 0.5 µM CBD caused an immediate short-term dysregulation of ΔΨm, followed by ATP depletion with transient activation of caspase-8 and progressive activation of caspase-9 and caspase-3/7, leading to early apoptosis and subsequent necroptosis. In astrocytes, both TRPV1 and cannabinoid receptor 1 (CB<sub>1</sub>) antagonists protected viability and prevented apoptosis. Given that CBD is a non-intoxicating drug, our results clearly show that this is not the case during critical periods of brain development when it can significantly interfere with the endogenous cannabinoid system.
Collapse
|
69
|
Blum K, Dennen CA, Elman I, Bowirrat A, Thanos PK, Badgaiyan RD, Downs BW, Bagchi D, Baron D, Braverman ER, Gupta A, Green R, McLaughlin T, Barh D, Gold MS. Should Reward Deficiency Syndrome (RDS) Be Considered an Umbrella Disorder for Mental Illness and Associated Genetic and Epigenetic Induced Dysregulation of Brain Reward Circuitry? J Pers Med 2022; 12:1719. [PMID: 36294858 PMCID: PMC9604605 DOI: 10.3390/jpm12101719] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
Reward Deficiency Syndrome (RDS) is defined as a breakdown of reward neurotransmission that results in a wide range of addictive, compulsive, and impulsive behaviors. RDS is caused by a combination of environmental (epigenetic) influences and DNA-based (genetic) neurotransmission deficits that interfere with the normal satisfaction of human physiological drives (i.e., food, water, and sex). An essential feature of RDS is the lack of integration between perception, cognition, and emotions that occurs because of (1) significant dopaminergic surges in motivation, reward, and learning centers causing neuroplasticity in the striato-thalamic-frontal cortical loop; (2) hypo-functionality of the excitatory glutamatergic afferents from the amygdala-hippocampus complex. A large volume of literature regarding the known neurogenetic and psychological underpinnings of RDS has revealed a significant risk of dopaminergic gene polymorphic allele overlap between cohorts of depression and subsets of schizophrenia. The suggestion is that instead of alcohol, opioids, gambling disorders, etc. being endophenotypes, the true phenotype is RDS. Additionally, reward deficiency can result from depleted or hereditary hypodopaminergia, which can manifest as a variety of personality traits and mental/medical disorders that have been linked to genetic studies with dopamine-depleting alleles. The carrying of known DNA antecedents, including epigenetic insults, results in a life-long vulnerability to RDS conditions and addictive behaviors. Epigenetic repair of hypodopaminergia, the causative basis of addictive behaviors, may involve precision DNA-guided therapy achieved by combining the Genetic Addiction Risk Severity (GARS) test with a researched neutraceutical having a number of variant names, including KB220Z. This nutraceutical formulation with pro-dopamine regulatory capabilities has been studied and published in peer-reviewed journals, mostly from our laboratory. Finally, it is our opinion that RDS should be given an ICD code and deserves to be included in the DSM-VI because while the DSM features symptomology, it is equally important to feature etiological roots as portrayed in the RDS model.
Collapse
Affiliation(s)
- Kenneth Blum
- The Kenneth Blum Behavioral & Neurogenetic Institute, LLC., Austin, TX 78701, USA
- Center for Behavioral Health & Sports, Exercise, Psychiatry, Western University Health Sciences, Pomona, CA 91766, USA
- Institute of Psychology, ELTE Eötvös Loránd University, Kazinczy u. 23–27, 1075 Budapest, Hungary
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Catherine A. Dennen
- The Kenneth Blum Behavioral & Neurogenetic Institute, LLC., Austin, TX 78701, USA
- Department of Family Medicine, Jefferson Health Northeast, Philadelphia, PA 19140, USA
| | - Igor Elman
- Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Cambridge Health Alliance, Harvard Medical School, Cambridge, MA 02139, USA
| | - Abdalla Bowirrat
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Panayotis K. Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, Clinical Research Institute on Addictions, University at Buffalo, Buffalo, NY 14203, USA
- Department of Psychology, University at Buffalo, Buffalo, NY 14260, USA
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, Long School of Medicine, University of Texas Medical Center, San Antonio, TX 78229, USA
| | - B. William Downs
- Division of Precision Nutrition, Victory Nutrition International, LLC., Lederoch, PA 19438, USA
| | - Debasis Bagchi
- Division of Precision Nutrition, Victory Nutrition International, LLC., Lederoch, PA 19438, USA
- Department of Pharmaceutical Science, College of Pharmacy & Health Sciences, Texas Southern University, Houston, TX 77004, USA
| | - David Baron
- Center for Behavioral Health & Sports, Exercise, Psychiatry, Western University Health Sciences, Pomona, CA 91766, USA
| | - Eric R. Braverman
- The Kenneth Blum Behavioral & Neurogenetic Institute, LLC., Austin, TX 78701, USA
| | - Ashim Gupta
- Future Biologics, Lawrenceville, GA 30043, USA
| | - Richard Green
- The Kenneth Blum Behavioral & Neurogenetic Institute, LLC., Austin, TX 78701, USA
| | - Thomas McLaughlin
- The Kenneth Blum Behavioral & Neurogenetic Institute, LLC., Austin, TX 78701, USA
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur 721172, India
| | - Mark S. Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
70
|
Niknam Y, Iyer P, Campbell MA, Moran F, Sandy MS, Zeise L. Animal evidence considered in determination of cannabis smoke and Δ 9 -tetrahydrocannabinol as causing reproductive toxicity (developmental endpoint): Part III. Proposed neurodevelopmental mechanisms of action. Birth Defects Res 2022; 114:1169-1185. [PMID: 36125082 DOI: 10.1002/bdr2.2088] [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: 06/01/2022] [Revised: 08/16/2022] [Accepted: 08/28/2022] [Indexed: 11/09/2022]
Abstract
This review summarizes the most common potential pathways of neurodevelopmental toxicity due to perinatal exposure to Δ9 -tetrahydrocannabinol (Δ9 -THC) that lead to behavioral and other adverse outcomes (AOs). This is Part III in a set of reviews highlighting the animal-derived data considered by California's Developmental and Reproductive Toxicant Identification Committee (DARTIC) in 2019. The Hazard Identification Document (HID) provided to the DARTIC included a summary of human, whole animal, and mechanistic data on the neurodevelopmental toxicity of cannabis smoke and Δ9 -THC. The literature search for mechanistic data has been updated through 2020. We focus on mechanistic pathways relating to behavioral and other neurodevelopmental outcomes of perinatal exposure to Δ9 -THC. The endocannabinoid system (EC system) plays a crucial role in many processes involved in neurodevelopment and exposure to Δ9 -THC can alter these processes. Whole animal studies report changes in cognitive ability, behavior, and motor function after prenatal exposure to Δ9 -THC. Findings from mechanistic studies add to this evidence and further provide information regarding the pathways leading to these outcomes. Neuromechanistic studies can bridge the gaps between molecular initiating events and apical neurodevelopmental endpoints caused by a chemical. They offer insight into potential alterations in the same pathways by other chemicals that can also result in AOs. Studies of cannabinoid receptor agonist-induced molecular alterations and provide deep biological plausibility at the mechanistic level for the cognitive, behavioral, and motor impairments observed in animal studies after perinatal exposure to Δ9 -THC.
Collapse
Affiliation(s)
- Yassaman Niknam
- Office of Environmental Health Hazard Assessment (OEHHA)/Reproductive and Cancer Hazard Assessment Branch (RCHAB), California Environmental Protection Agency, Sacramento, California, USA
| | - Poorni Iyer
- Office of Environmental Health Hazard Assessment (OEHHA)/Reproductive and Cancer Hazard Assessment Branch (RCHAB), California Environmental Protection Agency, Sacramento, California, USA
| | - Marlissa A Campbell
- Office of Environmental Health Hazard Assessment (OEHHA)/Reproductive and Cancer Hazard Assessment Branch (RCHAB), California Environmental Protection Agency, Sacramento, California, USA
| | - Francisco Moran
- Office of Environmental Health Hazard Assessment (OEHHA)/Reproductive and Cancer Hazard Assessment Branch (RCHAB), California Environmental Protection Agency, Sacramento, California, USA
| | - Martha S Sandy
- Office of Environmental Health Hazard Assessment (OEHHA)/Reproductive and Cancer Hazard Assessment Branch (RCHAB), California Environmental Protection Agency, Sacramento, California, USA
| | - Lauren Zeise
- Office of Environmental Health Hazard Assessment (OEHHA)/Reproductive and Cancer Hazard Assessment Branch (RCHAB), California Environmental Protection Agency, Sacramento, California, USA
| |
Collapse
|
71
|
Iyer P, Niknam Y, Campbell M, Moran F, Kaufman F, Kim A, Sandy M, Zeise L. Animal evidence considered in determination of cannabis smoke and
Δ
9
‐tetrahydrocannabinol (
Δ
9
‐THC
) as causing reproductive toxicity (developmental endpoint); part
II
. Neurodevelopmental effects. Birth Defects Res 2022; 114:1155-1168. [DOI: 10.1002/bdr2.2084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Poorni Iyer
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Yassaman Niknam
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Marlissa Campbell
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Francisco Moran
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Farla Kaufman
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Allegra Kim
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Martha Sandy
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Lauren Zeise
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| |
Collapse
|
72
|
Sperm DNA methylation alterations from cannabis extract exposure are evident in offspring. Epigenetics Chromatin 2022; 15:33. [PMID: 36085240 PMCID: PMC9463823 DOI: 10.1186/s13072-022-00466-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/26/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Cannabis legalization is expanding and men are the predominant users. We have limited knowledge about how cannabis impacts sperm and whether the effects are heritable.
Results
Whole genome bisulfite sequencing (WGBS) data were generated for sperm of rats exposed to: (1) cannabis extract (CE) for 28 days, then 56 days of vehicle only (~ one spermatogenic cycle); (2) vehicle for 56 days, then 28 days of CE; or (3) vehicle only. Males were then mated with drug-naïve females to produce F1 offspring from which heart, brain, and sperm tissues underwent analyses. There were 3321 nominally significant differentially methylated CpGs in F0 sperm identified via WGBS with select methylation changes validated via bisulfite pyrosequencing. Significant methylation changes validated in F0 sperm of the exposed males at the gene 2-Phosphoxylose Phosphatase 1 (Pxylp1) were also detectable in their F1 sperm but not in controls. Changes validated in exposed F0 sperm at Metastasis Suppressor 1-Like Protein (Mtss1l) were also present in F1 hippocampal and nucleus accumbens (NAc) of the exposed group compared to controls. For Mtss1l, a significant sex-specific relationship between DNA methylation and gene expression was demonstrated in the F1 NAc. Phenotypically, rats born to CSE-exposed fathers exhibited significant cardiomegaly relative to those born to control fathers.
Conclusions
This is the first characterization of the effect of cannabis exposure on the entirety of the rat sperm methylome. We identified CE-associated methylation changes across the sperm methylome, some of which persisted despite a “washout” period. Select methylation changes validated via bisulfite pyrosequencing, and genes associated with methylation changes were involved in early developmental processes. Preconception CE exposure is associated with detectable changes in offspring DNA methylation that are functionally related to changes in gene expression and cardiomegaly.
These results support that paternal preconception exposure to cannabis can influence offspring outcomes.
Collapse
|
73
|
Zhang LY, Kim AY, Cheer JF. Regulation of glutamate homeostasis in the nucleus accumbens by astrocytic CB1 receptors and its role in cocaine-motivated behaviors. ADDICTION NEUROSCIENCE 2022; 3:100022. [PMID: 36419922 PMCID: PMC9681119 DOI: 10.1016/j.addicn.2022.100022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cannabinoid type 1 receptors (CB1Rs) orchestrate brain reward circuitry and are prevalent neurobiological targets for endocannabinoids and cannabis in the mammalian brain. Decades of histological and electrophysiological studies have established CB1R as presynaptic G-protein coupled receptors (GPCRs) that inhibit neurotransmitter release through retrograde signaling mechanisms. Recent seminal work demonstrates CB1R expression on astrocytes and the pivotal function of glial cells in endocannabinoid-mediated modulation of neuron-astrocyte signaling. Here, we review key facets of CB1R-mediated astroglia regulation of synaptic glutamate transmission in the nucleus accumbens with a specific emphasis on cocaine-directed behaviors.
Collapse
Affiliation(s)
- Lan-Yuan Zhang
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Andrew Y. Kim
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Joseph F. Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States of America
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
74
|
Liang Y, Zhou A, Yoon JY. Machine Learning-Based Quantification of (-)- trans-Δ-Tetrahydrocannabinol from Human Saliva Samples on a Smartphone-Based Paper Microfluidic Platform. ACS OMEGA 2022; 7:30064-30073. [PMID: 36061666 PMCID: PMC9434788 DOI: 10.1021/acsomega.2c03099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/02/2022] [Indexed: 06/15/2023]
Abstract
(-)-trans-Δ-Tetrahydrocannabinol (THC) is a major psychoactive component in cannabis. Despite the recent trends of THC legalization for medical or recreational use in some areas, many THC-driven impairments have been verified. Therefore, convenient, sensitive, quantitative detection of THC is highly needed to improve its regulation and legalization. We demonstrated a biosensor platform to detect and quantify THC with a paper microfluidic chip and a handheld smartphone-based fluorescence microscope. Microfluidic competitive immunoassay was applied with anti-THC-conjugated fluorescent nanoparticles. The smartphone-based fluorescence microscope counted the fluorescent nanoparticles in the test zone, achieving a 1 pg/mL limit of detection from human saliva samples. Specificity experiments were conducted with cannabidiol (CBD) and various mixtures of THC and CBD. No cross-reactivity to CBD was found. Machine learning techniques were also used to quantify the THC concentrations from multiple saliva samples. Multidimensional data were collected by diluting the saliva samples with saline at four different dilutions. A training database was established to estimate the THC concentration from multiple saliva samples, eliminating the sample-to-sample variations. The classification algorithms included k-nearest neighbor (k-NN), decision tree, and support vector machine (SVM), and the SVM showed the best accuracy of 88% in estimating six different THC concentrations. Additional validation experiments were conducted using independent validation sample sets, successfully identifying positive samples at 100% accuracy and quantifying the THC concentration at 80% accuracy. The platform provided a quick, low-cost, sensitive, and quantitative point-of-care saliva test for cannabis.
Collapse
Affiliation(s)
- Yan Liang
- Department
of Chemistry and Biochemistry, The University
of Arizona, Tucson, Arizona 85721, United States
| | - Avory Zhou
- Department
of Biomedical Engineering, The University
of Arizona, Tucson, Arizona 85721, United
States
| | - Jeong-Yeol Yoon
- Department
of Chemistry and Biochemistry, The University
of Arizona, Tucson, Arizona 85721, United States
- Department
of Biomedical Engineering, The University
of Arizona, Tucson, Arizona 85721, United
States
| |
Collapse
|
75
|
Schneider M, Müller CP, Knies AK. Low income and schizophrenia risk: a narrative review. Behav Brain Res 2022; 435:114047. [PMID: 35933046 DOI: 10.1016/j.bbr.2022.114047] [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: 02/25/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/02/2022]
Abstract
Despite decades of research, the precise etiology of schizophrenia is not fully understood. Ample evidence indicates that the disorder derives from a complex interplay of genetic and environmental factors during vulnerable stages of brain maturation. Among the plethora of risk factors investigated, stress, pre- and perinatal insults, and cannabis use have been repeatedly highlighted as crucial environmental risk factors for schizophrenia. Compelling findings from population-based longitudinal studies suggest low income as an additional risk factor for future schizophrenia diagnosis, but underlying mechanisms remain unclear. In this narrative review, we 1) summarize the literature in support of a relationship between low (parental) income and schizophrenia risk, and 2) explore the mediating role of chronic stress, pre- and perinatal factors, and cannabis use as established risk factors for schizophrenia. Our review describes how low income facilitates the occurrence and severity of these established risk factors and thus contributes to schizophrenia liability. The broadest influence of low income was identified for stress, as low income was found to be associated with exposure to a multitude of severe psychological and physiological stressors. This narrative review adds to the growing literature reporting a close relationship between income and mental health.
Collapse
Affiliation(s)
- Miriam Schneider
- Department of Scientific Coordination and Management, Danube Private University, 3500 Krems-Stein, Austria.
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nuremberg, 91054 Erlangen, Germany; Centre for Drug Research, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| | - Andrea K Knies
- Department of Scientific Coordination and Management, Danube Private University, 3500 Krems-Stein, Austria
| |
Collapse
|
76
|
Benevento M, Hökfelt T, Harkany T. Ontogenetic rules for the molecular diversification of hypothalamic neurons. Nat Rev Neurosci 2022; 23:611-627. [PMID: 35906427 DOI: 10.1038/s41583-022-00615-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 11/09/2022]
Abstract
The hypothalamus is an evolutionarily conserved endocrine interface that, among other roles, links central homeostatic control to adaptive bodily responses by releasing hormones and neuropeptides from its many neuronal subtypes. In its preoptic, anterior, tuberal and mammillary subdivisions, a kaleidoscope of magnocellular and parvocellular neuroendocrine command neurons, local-circuit neurons, and neurons that project to extrahypothalamic areas are intermingled in partially overlapping patches of nuclei. Molecular fingerprinting has produced data of unprecedented mass and depth to distinguish and even to predict the synaptic and endocrine competences, connectivity and stimulus selectivity of many neuronal modalities. These new insights support eminent studies from the past century but challenge others on the molecular rules that shape the developmental segregation of hypothalamic neuronal subtypes and their use of morphogenic cues for terminal differentiation. Here, we integrate single-cell RNA sequencing studies with those of mouse genetics and endocrinology to describe key stages of hypothalamus development, including local neurogenesis, the direct terminal differentiation of glutamatergic neurons, transition cascades for GABAergic and GABAergic cell-derived dopamine cells, waves of local neuronal migration, and sequential enrichment in neuropeptides and hormones. We particularly emphasize how transcription factors determine neuronal identity and, consequently, circuit architecture, and whether their deviations triggered by environmental factors and hormones provoke neuroendocrine illnesses.
Collapse
Affiliation(s)
- Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria. .,Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
77
|
Ellis RJ, Bara A, Vargas CA, Frick AL, Loh E, Landry J, Uzamere TO, Callens JE, Martin Q, Rajarajan P, Brennand K, Ramakrishnan A, Shen L, Szutorisz H, Hurd YL. Prenatal Δ 9-Tetrahydrocannabinol Exposure in Males Leads to Motivational Disturbances Related to Striatal Epigenetic Dysregulation. Biol Psychiatry 2022; 92:127-138. [PMID: 34895699 PMCID: PMC8957623 DOI: 10.1016/j.biopsych.2021.09.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cannabis remains one of the most widely abused drugs during pregnancy. In utero exposure to its principal psychoactive component, Δ9-tetrahydrocannabinol (THC), can result in long-term neuropsychiatric risk for the progeny. This study investigated epigenetic signatures underlying these enduring consequences. METHODS Rat dams were exposed daily to THC (0.15 mg/kg) during pregnancy, and adult male offspring were examined for reward and depressive-like behavioral endophenotypes. Using unbiased sequencing approaches, we explored transcriptional and epigenetic profiles in the nucleus accumbens (NAc), a brain area central to reward and emotional processing. An in vitro CRISPR (clustered regularly interspaced short palindromic repeats) activation model coupled with RNA sequencing was also applied to study specific consequences of epigenetic dysregulation, and altered molecular signatures were compared with human major depressive disorder transcriptome datasets. RESULTS Prenatal THC exposure induced increased motivation for food, heightened learned helplessness and anhedonia, and altered stress sensitivity. We identified a robust increase specific to males in the expression of Kmt2a (histone-lysine N-methyltransferase 2A) that targets H3K4 (lysine 4 on histone H3) in cellular chromatin. Normalizing Kmt2a in the NAc rescued the motivational phenotype of prenatally THC-exposed animals. Comparison of RNA- and H3K4me3-sequencing datasets from the NAc of rat offspring with the in vitro model of Kmt2a upregulation revealed overlapping, significant disturbances in pathways that mediate synaptic plasticity. Similar transcriptional alterations were detected in human major depressive disorder. CONCLUSIONS These studies provide direct evidence for the persistent effects of prenatal cannabis exposure on transcriptional and epigenetic deviations in the NAc via Kmt2a dysregulation and associated psychiatric vulnerability.
Collapse
Affiliation(s)
- Randall J. Ellis
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA,Addiction Institute of Mount Sinai, New York, NY, USA
| | - Anissa Bara
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA,Friedman Brain Institute, Department of Psychiatry, New York, NY, USA
| | - Claudia A. Vargas
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA
| | - Amy L. Frick
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA
| | - Eddie Loh
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA
| | - Joseph Landry
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA,Addiction Institute of Mount Sinai, New York, NY, USA
| | - Teddy O. Uzamere
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA,Addiction Institute of Mount Sinai, New York, NY, USA
| | - James E. Callens
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA,Addiction Institute of Mount Sinai, New York, NY, USA
| | - Qammarah Martin
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA,Addiction Institute of Mount Sinai, New York, NY, USA
| | - Prashanth Rajarajan
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA
| | - Kristen Brennand
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA,Addiction Institute of Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA
| | - Li Shen
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, Department of Neuroscience, New York, NY, USA
| | - Henrietta Szutorisz
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Yasmin L Hurd
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Addiction Institute of Mount Sinai, New York, New York.
| |
Collapse
|
78
|
Little R, D'Mello D. A Cannabinoid Hypothesis of Schizophrenia: Pathways to Psychosis. INNOVATIONS IN CLINICAL NEUROSCIENCE 2022; 19:38-43. [PMID: 36204167 PMCID: PMC9507146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
BACKGROUND Observations regarding psychostimulant and psychedelic drug-induced psychotic states led to the dopamine, serotonin, and glutamate hypotheses of schizophrenia. Expanding knowledge about the endocannabinoid system and the impact of exogenous cannabinoids on the brain and behavior have elucidated several putative pathways to cannabis-induced psychosis. OBJECTIVE The purpose of the present article was to describe these pathways and propose a cannabinoid hypothesis of schizophrenia. MAIN POINTS The endocannabinoid system was reviewed. Evidence regarding the effect of delta 9-tetrahydrocannabinol (THC) on the brain was described. A connection between cannabis use and first-episode psychosis was elucidated. CONCLUSION Understanding the putative pathways to cannabis-induced psychosis might lead to targeted therapeutic interventions and prevention of schizophrenia in susceptible individuals.
Collapse
Affiliation(s)
| | - Dale D'Mello
- Dr. D'Mello is Associate Professor Emeritus with the Department of Psychiatry at Michigan State University in East Lansing, Michigan
| |
Collapse
|
79
|
Crume TL, Powers S, Dufford AJ, Kim P. Cannabis and Pregnancy: Factors Associated with Cannabis Use Among Pregnant Women and the Consequences for Offspring Neurodevelopment and Early Postpartum Parenting Behavior. CURRENT ADDICTION REPORTS 2022. [DOI: 10.1007/s40429-022-00419-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
80
|
Ishiguro H, Kibret BG, Horiuchi Y, Onaivi ES. Potential Role of Cannabinoid Type 2 Receptors in Neuropsychiatric and Neurodegenerative Disorders. Front Psychiatry 2022; 13:828895. [PMID: 35774086 PMCID: PMC9237241 DOI: 10.3389/fpsyt.2022.828895] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 05/02/2022] [Indexed: 12/12/2022] Open
Abstract
The endocannabinoid system (ECS) is composed of the two canonical receptor subtypes; type-1 cannabinoid (CB1R) and type 2 receptor (CB2R), endocannabinoids (eCBs) and enzymes responsible for the synthesis and degradation of eCBs. Recently, with the identification of additional lipid mediators, enzymes and receptors, the expanded ECS called the endocannabinoidome (eCBome) has been identified and recognized. Activation of CB1R is associated with a plethora of physiological effects and some central nervous system (CNS) side effects, whereas, CB2R activation is devoid of such effects and hence CB2Rs might be utilized as potential new targets for the treatment of different disorders including neuropsychiatric disorders. Previous studies suggested that CB2Rs were absent in the brain and they were considered as peripheral receptors, however, recent studies confirmed the presence of CB2Rs in different brain regions. Several studies have now focused on the characterization of its physiological and pathological roles. Studies done on the role of CB2Rs as a therapeutic target for treating different disorders revealed important putative role of CB2R in neuropsychiatric disorders that requires further clinical validation. Here we provide current insights and knowledge on the potential role of targeting CB2Rs in neuropsychiatric and neurodegenerative disorders. Its non-psychoactive effect makes the CB2R a potential target for treating CNS disorders; however, a better understanding of the fundamental pharmacology of CB2R activation is essential for the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- Hiroki Ishiguro
- Department of Clinical Genetics, Graduate School of Medical Science, University of Yamanashi, Kofu, Japan
- Department of Neuropsychiatry, Graduate School of Medical Science, University of Yamanashi, Kofu, Japan
| | - Berhanu Geresu Kibret
- Department of Biology, College of Science and Health, William Paterson University, Wayne, NJ, United States
| | - Yasue Horiuchi
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Emmanuel S. Onaivi
- Department of Biology, College of Science and Health, William Paterson University, Wayne, NJ, United States
| |
Collapse
|
81
|
Augustin SM, Lovinger DM. Synaptic changes induced by cannabinoid drugs and cannabis use disorder. Neurobiol Dis 2022; 167:105670. [DOI: 10.1016/j.nbd.2022.105670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/10/2022] [Accepted: 02/21/2022] [Indexed: 10/19/2022] Open
|
82
|
Betz LT, Penzel N, Kambeitz J. A network approach to relationships between cannabis use characteristics and psychopathology in the general population. Sci Rep 2022; 12:7163. [PMID: 35504926 PMCID: PMC9065088 DOI: 10.1038/s41598-022-11092-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 04/08/2022] [Indexed: 11/27/2022] Open
Abstract
Cannabis use characteristics, such as earlier initiation and frequent use, have been associated with an increased risk for developing psychotic experiences and psychotic disorders. However, little is known how these characteristics relate to specific aspects of sub-clinical psychopathology in the general population. Here, we explore the relationships between cannabis use characteristics and psychopathology in a large general population sample (N = 2,544, mean age 29.2 years, 47% women) by employing a network approach. This allows for the identification of unique associations between two cannabis use characteristics (lifetime cumulative frequency of cannabis use, age of cannabis use initiation), and specific psychotic experiences and affective symptoms, while controlling for early risk factors (childhood trauma, urban upbringing). We found particularly pronounced unique positive associations between frequency of cannabis use and specific delusional experiences (persecutory delusions and thought broadcasting). Age of cannabis use initiation was negatively related to visual hallucinatory experiences and irritability, implying that these experiences become more likely the earlier use is initiated. Earlier initiation, but not lifetime frequency of cannabis use, was related to early risk factors. These findings suggest that cannabis use characteristics may contribute differentially to risk for specific psychotic experiences and affective symptoms in the general population.
Collapse
Affiliation(s)
- Linda T Betz
- Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany.
| | - Nora Penzel
- Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University, Munich, Germany
- Group of Psychiatric Neuroscience, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - Joseph Kambeitz
- Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
83
|
Cioffredi LA, Anderson H, Loso H, East J, Nguyen P, Garavan H, Potter A. Prenatal cannabis exposure predicts attention problems, without changes on fMRI in adolescents. Neurotoxicol Teratol 2022; 91:107089. [PMID: 35314358 PMCID: PMC9136933 DOI: 10.1016/j.ntt.2022.107089] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVES We hypothesized that prenatal cannabis exposure (PCE) would be associated with increased attention problems and altered neurocognition in young adolescents. METHODS Data were obtained from the Adolescent Brain Cognitive Development (ABCD study®), a cohort of approximately 12,000 children. Presence or absence of PCE after knowledge of pregnancy was measured by caregiver report. All participants with PCE (N = 224) were included and compared to two control groups; those matched on tobacco and alcohol exposure and those without prenatal tobacco or alcohol exposures. Outcomes were measured with the ABCD baseline assessment when participants were 9-10 years old and included attention, internalizing, externalizing and total problems scales on the Child Behavior Checklist (CBCL). Teacher reports were used when available. Mixed effects modeling assessed the association between PCE and outcomes controlling for parental psychopathology, prematurity and socioeconomic status. For participants with available data, patterns of brain activity during three fMRI tasks (the Stop Signal Task measuring response inhibition, the Monetary Incentive Delay (MID) task measuring reward processing and the EN-Back task measuring working memory) were analyzed using Permutation Analyses of the Linear Model. RESULTS Compared to both control groups, participants with PCE had significantly higher attention problems, externalizing, and total problem scores. PCE did not impact cognitive performance or patterns of brain activation during fMRI tasks. CONCLUSIONS There are long-term associations between PCE and early adolescent attention and behavioral problems. These are not reflected in cognitive performance or task fMRI measures, a finding that is consistent with reports that fewer than half of children with ADHD have any specific cognitive deficit (Nigg et al., 2005; Willcutt et al., 2005). The young age of the sample may also relate to this finding and future investigation of neurodevelopmental trajectories of youth with PCE is warranted.
Collapse
Affiliation(s)
- Leigh-Anne Cioffredi
- Larner College of Medicine at the University of Vermont, Department of Pediatrics, USA.
| | - Hillary Anderson
- Larner College of Medicine at the University of Vermont, Department of Pediatrics, USA
| | - Hannah Loso
- Larner College of Medicine at the University of Vermont, Department of Psychiatry, USA
| | - James East
- Larner College of Medicine at the University of Vermont, Department of Radiology, USA
| | - Philip Nguyen
- Larner College of Medicine at the University of Vermont, Department of Psychiatry, USA
| | - Hugh Garavan
- Larner College of Medicine at the University of Vermont, Department of Psychiatry, USA
| | - Alexandra Potter
- Larner College of Medicine at the University of Vermont, Department of Psychiatry, USA
| |
Collapse
|
84
|
Barger MK. Systematic Reviews to Inform Practice, May/June 2022. J Midwifery Womens Health 2022; 67:403-409. [PMID: 35522134 DOI: 10.1111/jmwh.13368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Mary K Barger
- Midwifery researcher and consultant, San Diego, California
| |
Collapse
|
85
|
Cossart R, Garel S. Step by step: cells with multiple functions in cortical circuit assembly. Nat Rev Neurosci 2022; 23:395-410. [DOI: 10.1038/s41583-022-00585-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2022] [Indexed: 12/23/2022]
|
86
|
Lawson J, O'Brien T, Graham M, Renaud E, Jones D, Freeman J, Lawn N, Martin JH. Expert advice for prescribing cannabis medicines for patients with epilepsy-drawn from the Australian clinical experience. Br J Clin Pharmacol 2022; 88:3101-3113. [PMID: 35261078 PMCID: PMC9311726 DOI: 10.1111/bcp.15262] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/18/2022] [Accepted: 01/26/2022] [Indexed: 12/26/2022] Open
Abstract
There is international interest for consensus advice for prescribers working in the field of drug resistant epilepsy intending to trial potential therapies that are nonregistered or off‐label. Cannabinoids are one such therapy. In 2017, the New South Wales State Government (Australia) set up a cannabinoid prescribing guidance service for a wide variety of indications, based on known pharmacology together with the relevant new literature as it became available. Increasing interest in cannabis medicines use outside this State over the following 5 years together with a paucity of registration‐standard clinical trials, lack of information around dosing issues, drug interactions and biological plausibility meant there remained a large unmet need for such advice. To address the unmet need in epilepsy, and until medicines were registered or regulator quality data were available, it was agreed to bring together a working group comprising paediatric and adult epilepsy specialists, clinical pharmacists., clinical pharmacologists and cannabis researchers from across Australia to develop interim consensus advice for prescribers. Although interim, this consensus advice addresses much of the current practice gap by providing an informed overview of the different cannabis medicines currently available for use in the treatment of epilepsy in paediatric and adult settings, with information on dose, drug interactions, toxicity, type of seizure and frequency of symptom relief. As such it supplements the limited evidence currently available from clinical trials with experience from front‐line practice. It is expected that this consensus advice will be updated as new evidence emerges and will provide guidance for a subsequent Guideline.
Collapse
Affiliation(s)
- John Lawson
- Sydney Children's Hospital Randwick, Neurology; University of New South Wales - Randwick Campus, School of Women and Children's Health, Sydney, New South Wales, Australia.,Australian Centre for Cannabis Clinical and Research Excellence, Australia
| | - Terry O'Brien
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Myfanwy Graham
- Australian Centre for Cannabis Clinical and Research Excellence, Australia.,Centre for Drug Repurposing and Medicines Research, Clinical Pharmacology, Hunter Medical Research Institute, University of Newcastle, Australia
| | - Elianne Renaud
- Australian Centre for Cannabis Clinical and Research Excellence, Australia.,Centre for Drug Repurposing and Medicines Research, Clinical Pharmacology, Hunter Medical Research Institute, University of Newcastle, Australia
| | - Dean Jones
- University of Technology, Sydney, New South Wales, Australia
| | - Jeremy Freeman
- Murdoch Children's Research Institute; The Royal Children's Hospital Melbourne, Western Australian Adult Epilepsy service in Perth, Western Australia
| | | | - Jennifer H Martin
- Australian Centre for Cannabis Clinical and Research Excellence, Australia.,Centre for Drug Repurposing and Medicines Research, Clinical Pharmacology, Hunter Medical Research Institute, University of Newcastle, Australia
| |
Collapse
|
87
|
De Genna NM, Willford JA, Richardson GA. Long-term effects of prenatal cannabis exposure: Pathways to adolescent and adult outcomes. Pharmacol Biochem Behav 2022; 214:173358. [PMID: 35216971 PMCID: PMC8911923 DOI: 10.1016/j.pbb.2022.173358] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 02/09/2022] [Accepted: 02/17/2022] [Indexed: 12/09/2022]
Abstract
With the increased prevalence, potency, and acceptability of cannabis use during pregnancy, it is important to understand the developmental effects of prenatal cannabis exposure (PCE). This review discusses methodological considerations for studies of PCE, including the assessment of exposures, covariates, and outcomes, and reviews findings from prospective, longitudinal studies of PCE. There is some evidence for associations between PCE and restricted growth at birth, but not for long-term effects on growth. PCE appears to have subtle yet enduring effects on memory and achievement in children and adolescents. Despite differences in sample demographics and measurement, there are remarkably consistent effects of PCE on externalizing behaviors, such as delinquency and substance use, which persist into adulthood. Longitudinal analyses demonstrate the importance of early cannabis initiation for pathways between PCE and adult functioning, including substance use and abuse, memory deficits, and psychotic symptoms. Animal studies demonstrate direct effects on the development of the brain via activation of endogenous endocannabinoid systems. Cannabis-induced activation of the endocannabinoid system causes alterations in the release of neurotransmitters and the modulation of brain plasticity in neural pathways that underlie cognition, motivation, and behavior regulation. Future research should consider cannabis use before pregnancy, the timing and route of exposure, polysubstance exposures, and inter-generational effects.
Collapse
Affiliation(s)
- Natacha M. De Genna
- University of Pittsburgh School of Medicine, Department of Psychiatry, 3811 O’Hara Street, Pittsburgh, PA 15213,Corresponding author:
| | - Jennifer A. Willford
- Slippery Rock University, Department of Psychology, 1 Morrow Way, Slippery Rock, PA 16057
| | - Gale A. Richardson
- University of Pittsburgh School of Medicine, Department of Psychiatry, 3811 O’Hara Street, Pittsburgh, PA 15213
| |
Collapse
|
88
|
Dos(e)Age: Role of Dose and Age in the Long-Term Effect of Cannabinoids on Cognition. Molecules 2022; 27:molecules27041411. [PMID: 35209200 PMCID: PMC8876668 DOI: 10.3390/molecules27041411] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022] Open
Abstract
Cannabis is still the most widely used illicit drug around the world. While its use has always been prevalent among adolescents, recent evidence suggests that its consumption is also increasing among other population groups, such as pregnant women and aged people. Given the known impact of cannabis on brain development and behavior, it is important to dissect the possible long-term impact of its use across different age groups, especially on measures of cognitive performance. Animal models of cannabinoid exposure have represented a fundamental tool to characterize the long-lasting consequences of cannabinoids on cognitive performance and helped to identify possible factors that could modulate cannabinoids effects in the long term, such as the age of exposure and doses administered. This scoping review was systematically conducted using PubMed and includes papers published from 2015 to December 2021 that examined the effects of cannabinoids, either natural or synthetic, on cognitive performance in animal models where exposure occurred in the prenatal period, during adolescence, or in older animals. Overall, available data clearly point to a crucial role of age in determining the long-term effect of cannabinoid on cognition, highlighting possible detrimental consequences during brain development (prenatal and adolescent exposure) and beneficial outcomes in old age. In contrast, despite the recent advances in the field, it appears difficult to clearly establish a possible role of dosage in the effects of cannabinoids on cognition, especially when the adolescent period is taken into account.
Collapse
|
89
|
Garzón M, Chan J, Mackie K, Pickel VM. Prefrontal cortical distribution of muscarinic M2 and cannabinoid-1 (CB1) receptors in adult male mice with or without chronic adolescent exposure to Δ9-tetrahydrocannabinol. Cereb Cortex 2022; 32:5420-5437. [PMID: 35151230 PMCID: PMC9712711 DOI: 10.1093/cercor/bhac024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 12/27/2022] Open
Abstract
Chronic adolescent administration of marijuana's major psychoactive compound, ∆9-tetrahydrocannabinol (Δ9-THC), produces adaptive changes in adult social and cognitive functions sustained by prelimbic prefrontal cortex (PL-PFC). Memory and learning processes in PL-PFC neurons can be regulated through cholinergic muscarinic-2 receptors (M2R) and modulated by activation of cannabinoid-1 receptors (CB1Rs) targeted by Δ9-THC. Thus, chronic exposure to Δ9-THC during adolescence may alter the expression and/or distribution of M2Rs in PL-PFC neurons receiving CB1R terminals. We tested this hypothesis by using electron microscopic dual CB1R and M2R immunolabeling in adult C57BL/6 J male mice that had received vehicle or escalating dose of Δ9-THC through adolescence. In vehicle controls, CB1R immunolabeling was mainly localized to axonal profiles virtually devoid of M2R but often apposing M2R-immunoreactive dendrites and dendritic spines. The dendrites received inputs from CB1R-labeled or unlabeled terminals, whereas spines received asymmetric synapses exclusively from axon terminals lacking CB1Rs. Adolescent Δ9-THC significantly increased plasmalemmal M2R-immunogold density exclusively in large dendrites receiving input from CB1R-labeled terminals. In contrast, cytoplasmic M2R-immunogold density decreased in small spines of the Δ9-THC-treated adult mice. We conclude that Δ9-THC engagement of CB1Rs during adolescence increases M2R plasmalemmal accumulation in large proximal dendrites and decreases M2R cytoplasmic expression in small spines of PL-PFC.
Collapse
Affiliation(s)
- Miguel Garzón
- Corresponding author: Department of Anatomy, Histology and Neuroscience, Medical School, Autónoma University of Madrid, Arzobispo Morcillo 4, 28029 Madrid, Spain.
| | - June Chan
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Ken Mackie
- Linda and Jack Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Virginia M Pickel
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| |
Collapse
|
90
|
Beggiato S, Ieraci A, Zuccarini M, Di Iorio P, Schwarcz R, Ferraro L. Alterations in rat prefrontal cortex kynurenic acid levels are involved in the enduring cognitive dysfunctions induced by tetrahydrocannabinol exposure during the adolescence. Front Psychiatry 2022; 13:996406. [PMID: 36483135 PMCID: PMC9722723 DOI: 10.3389/fpsyt.2022.996406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Cannabis abuse during adolescence is a risk factor for cognitive impairments in psychiatric disorders later in life. To date, the possible causal relationship between cannabinoids, kynurenic acid (KYNA; i.e., a neuroactive metabolite of tryptophan degradation) and cognition has not been investigated in adolescence. Early exposure to delta 9-tetrahydrocannabinol (THC; i.e., the main psychotropic component of cannabis) causes enduring cognitive deficits, which critically involve impaired glutamatergic function in the prefrontal cortex (PFC). In addition, prenatal cannabis exposure results in enduring increases in PFC KYNA levels. Based on these findings, the effects of chronic THC exposure in rats, during another critical period of neurodevelopment particularly sensitive to perturbation by exogenous stimuli, such as adolescence, have been investigated. METHODS Male Wistar rats were chronically treated with vehicle or ascending intraperitoneal (i.p.) doses of THC starting on postnatal day (PND) 35 until PND 45. In adulthood (PND 75), cognitive assessment (Y-maze) and extracellular KYNA/glutamate levels were measured in the PFC by in vivo microdialysis, before and after a challenge with KYN (5 mg/kg i.p., the biological precursor of KYNA). By using the selective, brain-penetrable KAT II inhibitor PF-04859989, we then examined whether blockade of KYNA neosynthesis prevents the cognitive impairment. RESULTS Compared to vehicle-treated controls, extracellular basal KYNA levels were higher in the PFC of adult rats chronically exposed to THC in adolescence (p < 0.01). No changes were observed in extracellular glutamate levels. Following a challenge with KYN, extracellular KYNA levels similarly increased in both groups (i.e., vehicle- and THC-treated; p < 0.001 and p < 0.01, respectively). Chronic adolescent THC exposure negatively affected short-term memory (reduced spontaneous alternation), in adult animals (p < 0.001), while PF-04859989 (30 mg/kg i.p.) restored the cognitive impairment (p < 0.05). DISCUSSION We propose that the observed alterations in PFC KYNA signaling might be involved in the cognitive dysfunction induced by the exposure to THC during the adolescence. In the translational realm, these experiments raise the prospect of prevention of KYNA neosynthesis as a possible novel approach to counteract some of the detrimental long-term effects of adolescence cannabis use.
Collapse
Affiliation(s)
- Sarah Beggiato
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy.,Department of Life Sciences and Biotechnologies, University of Ferrara, Ferrara, Italy
| | - Alessandro Ieraci
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy.,Department of Theoretical and Applied Science, eCampus University, Novedrate, Italy
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Robert Schwarcz
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Luca Ferraro
- Department of Life Sciences and Biotechnologies, University of Ferrara, Ferrara, Italy.,Laboratory for the Technology of Advanced Therapies (LTTA Centre), University of Ferrara, Ferrara, Italy
| |
Collapse
|
91
|
Le AA, Quintanilla J, Amani M, Piomelli D, Lynch G, Gall CM. Persistent sexually dimorphic effects of adolescent THC exposure on hippocampal synaptic plasticity and episodic memory in rodents. Neurobiol Dis 2022; 162:105565. [PMID: 34838664 DOI: 10.1016/j.nbd.2021.105565] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/04/2021] [Accepted: 11/23/2021] [Indexed: 01/01/2023] Open
Abstract
There is evidence that cannabis use during adolescence leads to memory and cognitive problems in young adulthood but little is known about effects of early life cannabis exposure on synaptic operations that are critical for encoding and organizing information. We report here that a 14-day course of daily Δ9-tetrahydrocannabinol treatments administered to adolescent rats and mice (aTHC) leads to profound but selective deficits in synaptic plasticity in two axonal systems in female, and to lesser extent male, hippocampus as assessed in adulthood. Adolescent-THC exposure did not alter basic synaptic transmission (input/output curves) and had only modest effects on frequency facilitation. Nevertheless, aTHC severely impaired the endocannabinoid-dependent long-term potentiation in the lateral perforant path in females of both species, and in male mice; this was reliably associated with impaired acquisition of a component of episodic memory that depends on lateral perforant path function. Potentiation in the Schaffer-commissural (S-C) projection to field CA1 was disrupted by aTHC treatment in females only and this was associated with both a deficit in estrogen effects on S-C synaptic responses and impairments to CA1-dependent spatial (object location) memory. In all the results demonstrate sexually dimorphic and projection system-specific effects of aTHC exposure that could underlie discrete effects of early life cannabinoid usage on adult cognitive function. Moreover they suggest that some of the enduring, sexually dimorphic effects of cannabis use reflect changes in synaptic estrogen action.
Collapse
Affiliation(s)
- Aliza A Le
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA 92697, United States of America
| | - Julian Quintanilla
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA 92697, United States of America
| | - Mohammad Amani
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA 92697, United States of America
| | - Daniele Piomelli
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA 92697, United States of America
| | - Gary Lynch
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA 92697, United States of America; Departments of Psychiatry & Human Behavior, University of California, Irvine, CA 92868, United States of America.
| | - Christine M Gall
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA 92697, United States of America; Departments of Neurobiology & Behavior, University of California, Irvine, CA 92697, United States of America.
| |
Collapse
|
92
|
Dufford AJ, Spann M, Scheinost D. How prenatal exposures shape the infant brain: Insights from infant neuroimaging studies. Neurosci Biobehav Rev 2021; 131:47-58. [PMID: 34536461 DOI: 10.1016/j.neubiorev.2021.09.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/30/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
Brain development during the prenatal period is rapid and unparalleled by any other time during development. Biological systems undergoing rapid development are at higher risk for disorganizing influences. Therefore, certain prenatal exposures impact brain development, increasing risk for negative neurodevelopmental outcome. While prenatal exposures have been associated with cognitive and behavioral outcomes later in life, the underlying macroscopic brain pathways remain unclear. Here, we review magnetic resonance imaging (MRI) studies investigating the association between prenatal exposures and infant brain development focusing on prenatal exposures via maternal physical health factors, maternal mental health factors, and maternal drug and medication use. Further, we discuss the need for studies to consider multiple prenatal exposures in parallel and suggest future directions for this body of research.
Collapse
Affiliation(s)
| | - Marisa Spann
- Columbia University Irving Medical Center, 622 West 168th Street, New York, NY, 10032, USA
| | - Dustin Scheinost
- Child Study Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, USA; Department of Statistics and Data Science, Yale University, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| |
Collapse
|
93
|
Ostlund BD, Pérez-Edgar KE, Shisler S, Terrell S, Godleski S, Schuetze P, Eiden RD. Prenatal substance exposure and maternal hostility from pregnancy to toddlerhood: Associations with temperament profiles at 16 months of age. Dev Psychopathol 2021; 33:1566-1583. [PMID: 35095214 PMCID: PMC8794013 DOI: 10.1017/s0954579421001000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We investigated whether infant temperament was predicted by level of and change in maternal hostility, a putative transdiagnostic vulnerability for psychopathology, substance use, and insensitive parenting. A sample of women (N = 247) who were primarily young, low-income, and had varying levels of substance use prenatally (69 nonsmokers, 81 tobacco-only smokers, and 97 tobacco and marijuana smokers) reported their hostility in the third trimester of pregnancy and at 2, 9, and 16 months postpartum, and their toddler's temperament and behavior problems at 16 months. Maternal hostility decreased from late pregnancy to 16 months postpartum. Relative to pregnant women who did not use substances, women who used both marijuana and tobacco prenatally reported higher levels of hostility while pregnant and exhibited less change in hostility over time. Toddlers who were exposed to higher levels of prenatal maternal hostility were more likely to be classified in temperament profiles that resemble either irritability or inhibition, identified via latent profile analysis. These two profiles were each associated with more behavior problems concurrently, though differed in their association with competence. Our results underscore the utility of transdiagnostic vulnerabilities in understanding the intergenerational transmission of psychopathology risk and are discussed in regards to the Research Domain Criteria (RDoC) framework.
Collapse
Affiliation(s)
- Brendan D. Ostlund
- Department of Psychology, The Pennsylvania State University, University Park, USA
| | | | - Shannon Shisler
- Research Institute on Addictions, University at Buffalo, State University of New York, Buffalo, USA
| | - Sarah Terrell
- Department of Human Development and Family Studies, The Pennsylvania State University, University Park, USA
| | - Stephanie Godleski
- Department of Psychology, Rochester Institute of Technology, Rochester, USA
| | - Pamela Schuetze
- Department of Psychology, State University of New York, Buffalo, USA
| | - Rina D. Eiden
- Department of Psychology, The Pennsylvania State University, University Park, USA
| |
Collapse
|
94
|
Placenta keeps the score of maternal cannabis use and child anxiety. Proc Natl Acad Sci U S A 2021; 118:2118394118. [PMID: 34789581 DOI: 10.1073/pnas.2118394118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2021] [Indexed: 11/18/2022] Open
|
95
|
Kiguchi N, Ko MC. Potential therapeutic targets for the treatment of opioid abuse and pain. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:335-371. [PMID: 35341570 PMCID: PMC10948018 DOI: 10.1016/bs.apha.2021.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although μ-opioid peptide (MOP) receptor agonists are effective analgesics available in clinical settings, their serious adverse effects put limits on their use. The marked increase in abuse and misuse of prescription opioids for pain relief and opioid overdose mortality in the past decade has seriously impacted society. Therefore, safe analgesics that produce potent analgesic effects without causing MOP receptor-related adverse effects are needed. This review highlights the potential therapeutic targets for the treatment of opioid abuse and pain based on available evidence generated through preclinical studies and clinical trials. To ameliorate the abuse-related effects of opioids, orexin-1 receptor antagonists and mixed nociceptin/MOP partial agonists have shown promising results in translational aspects of animal models. There are several promising non-opioid targets for selectively inhibiting pain-related responses, including nerve growth factor inhibitors, voltage-gated sodium channel inhibitors, and cannabinoid- and nociceptin-related ligands. We have also discussed several emerging and novel targets. The current medications for opioid abuse are opioid receptor-based ligands. Although neurobiological studies in rodents have discovered several non-opioid targets, there is a translational gap between rodents and primates. Given that the neuroanatomical aspects underlying opioid abuse and pain are different between rodents and primates, it is pivotal to investigate the functional profiles of these non-opioid compounds compared to those of clinically used drugs in non-human primate models before initiating clinical trials. More pharmacological studies of the functional efficacy, selectivity, and tolerability of these newly discovered compounds in non-human primates will accelerate the development of effective medications for opioid abuse and pain.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan.
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
96
|
de Melo Reis RA, Isaac AR, Freitas HR, de Almeida MM, Schuck PF, Ferreira GC, Andrade-da-Costa BLDS, Trevenzoli IH. Quality of Life and a Surveillant Endocannabinoid System. Front Neurosci 2021; 15:747229. [PMID: 34776851 PMCID: PMC8581450 DOI: 10.3389/fnins.2021.747229] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/01/2021] [Indexed: 12/11/2022] Open
Abstract
The endocannabinoid system (ECS) is an important brain modulatory network. ECS regulates brain homeostasis throughout development, from progenitor fate decision to neuro- and gliogenesis, synaptogenesis, brain plasticity and circuit repair, up to learning, memory, fear, protection, and death. It is a major player in the hypothalamic-peripheral system-adipose tissue in the regulation of food intake, energy storage, nutritional status, and adipose tissue mass, consequently affecting obesity. Loss of ECS control might affect mood disorders (anxiety, hyperactivity, psychosis, and depression), lead to drug abuse, and impact neurodegenerative (Alzheimer's, Parkinson, Huntington, Multiple, and Amyotrophic Lateral Sclerosis) and neurodevelopmental (autism spectrum) disorders. Practice of regular physical and/or mind-body mindfulness and meditative activities have been shown to modulate endocannabinoid (eCB) levels, in addition to other players as brain-derived neurotrophic factor (BDNF). ECS is involved in pain, inflammation, metabolic and cardiovascular dysfunctions, general immune responses (asthma, allergy, and arthritis) and tumor expansion, both/either in the brain and/or in the periphery. The reason for such a vast impact is the fact that arachidonic acid, a precursor of eCBs, is present in every membrane cell of the body and on demand eCBs synthesis is regulated by electrical activity and calcium shifts. Novel lipid (lipoxins and resolvins) or peptide (hemopressin) players of the ECS also operate as regulators of physiological allostasis. Indeed, the presence of cannabinoid receptors in intracellular organelles as mitochondria or lysosomes, or in nuclear targets as PPARγ might impact energy consumption, metabolism and cell death. To live a better life implies in a vigilant ECS, through healthy diet selection (based on a balanced omega-3 and -6 polyunsaturated fatty acids), weekly exercises and meditation therapy, all of which regulating eCBs levels, surrounded by a constructive social network. Cannabidiol, a diet supplement has been a major player with anti-inflammatory, anxiolytic, antidepressant, and antioxidant activities. Cognitive challenges and emotional intelligence might strengthen the ECS, which is built on a variety of synapses that modify human behavior. As therapeutically concerned, the ECS is essential for maintaining homeostasis and cannabinoids are promising tools to control innumerous targets.
Collapse
Affiliation(s)
- Ricardo Augusto de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alinny Rosendo Isaac
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hércules Rezende Freitas
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana Macedo de Almeida
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Fernanda Schuck
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Costa Ferreira
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Isis Hara Trevenzoli
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
97
|
Peters KZ, Zlebnik NE, Cheer JF. Cannabis exposure during adolescence: A uniquely sensitive period for neurobiological effects. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:95-120. [PMID: 34801175 DOI: 10.1016/bs.irn.2021.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Adolescence is a crucial developmental period where neural circuits are refined and the brain is especially vulnerable to external insults. The endocannabinoid (eCB) system undergoes changes during adolescence which affect the way in which it modulates the development of other systems, in particular dopamine circuits, which show protracted development into adolescence. Given the rise of cannabis use by adolescents and young people, as well as variants containing increasingly higher concentrations of THC, it is now crucial to understand the unique effects of adolescent exposure to cannabis on the developing brain and it might shape future adult vulnerabilities to conditions such as psychosis, schizophrenia, addiction and more. Here we discuss the development of the eCB system across the lifespan, how CB1 receptors modulate dopamine release and potential neurobiological and behavioral effects of adolescent THC exposure on the developing brain such as alterations in excitatory/inhibitory balance during this developmental period.
Collapse
Affiliation(s)
- K Z Peters
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States; Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom.
| | - N E Zlebnik
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - J F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
98
|
Irrera N, Bitto A, Sant’Antonio E, Lauro R, Musolino C, Allegra A. Pros and Cons of the Cannabinoid System in Cancer: Focus on Hematological Malignancies. Molecules 2021; 26:molecules26133866. [PMID: 34202812 PMCID: PMC8270322 DOI: 10.3390/molecules26133866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/09/2021] [Accepted: 06/20/2021] [Indexed: 11/23/2022] Open
Abstract
The endocannabinoid system (ECS) is a composite cell-signaling system that allows endogenous cannabinoid ligands to control cell functions through the interaction with cannabinoid receptors. Modifications of the ECS might contribute to the pathogenesis of different diseases, including cancers. However, the use of these compounds as antitumor agents remains debatable. Pre-clinical experimental studies have shown that cannabinoids (CBs) might be effective for the treatment of hematological malignancies, such as leukemia and lymphoma. Specifically, CBs may activate programmed cell death mechanisms, thus blocking cancer cell growth, and may modulate both autophagy and angiogenesis. Therefore, CBs may have significant anti-tumor effects in hematologic diseases and may synergistically act with chemotherapeutic agents, possibly also reducing chemoresistance. Moreover, targeting ECS might be considered as a novel approach for the management of graft versus host disease, thus reducing some symptoms such as anorexia, cachexia, fatigue, anxiety, depression, and neuropathic pain. The aim of the present review is to collect the state of the art of CBs effects on hematological tumors, thus focusing on the essential topics that might be useful before moving into the clinical practice.
Collapse
Affiliation(s)
- Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (N.I.); (A.B.); (R.L.)
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (N.I.); (A.B.); (R.L.)
| | | | - Rita Lauro
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (N.I.); (A.B.); (R.L.)
| | - Caterina Musolino
- Department of Human Pathology in Adulthood and Childhood, University of Messina, 98125 Messina, Italy;
| | - Alessandro Allegra
- Department of Human Pathology in Adulthood and Childhood, University of Messina, 98125 Messina, Italy;
- Correspondence: ; Tel.: +390902212364
| |
Collapse
|