1
|
Chen HT, Mackie K. CB1 and CB2 receptors differentially modulate the cognitive impact of maternal immune activation and perinatal cannabinoid exposure. Behav Brain Res 2025; 485:115543. [PMID: 40113177 PMCID: PMC11986805 DOI: 10.1016/j.bbr.2025.115543] [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: 09/25/2024] [Revised: 02/07/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Maternal immune activation (MIA) commonly arises in response to an infection during pregnancy. MIA elevates cytokine levels, triggering an inflammatory cascade, which may be detrimental to the developing nervous system. Similarly, cannabis use and exposure of the fetus to cannabinoids during pregnancy (PCE) may elicit neuroinflammation and lead to detrimental behavioral outcomes. This is particularly concerning as there has been a notable rise in cannabis use during pregnancy. This study endeavors to examine the interaction between MIA and PCE and elucidate the role of CB1 and CB2 receptors in MIA and PCE outcomes. To this end, we compared the impact of MIA, PCE and MIA+PCE in wildtype, CB1, and CB2 cannabinoid receptor knockout mice of both sexes. PCE was modeled by daily 3 mg/kg THC administration from gestational day 5 (GD5) to postnatal day 10. MIA was modeled by intravenous Poly (I:C) injection at GD16.5. Subsequently, we assessed emotional and cognitive behaviors of adult offspring. Adult male offspring of dams exposed to PCE or MIA were impaired in novel object recognition and the delayed alternation working memory tasks. Interestingly, these behavioral impairments were absent when MIA and PCE were combined. Cannabinoid receptor knockout studies found that CB1 receptors mediated behavioral deficits after PCE. In contrast CB2 receptors were necessary for full expression of MIA-induced behavioral impairments. Although females showed more modest behavioral changes after MIA or PCE, CB1 receptors were required for the PCE deficit and CB2 receptors were required for the MIA deficit also in females. Notably, lack of CB2 receptors in males prevented the "protection" following combined MIA + PCE, while CB1 knockout mice remained protected. Taken together, these results suggest a complex interplay between PCE, MIA and CB1 and CB2 cannabinoid receptors.
Collapse
MESH Headings
- Animals
- Female
- Receptor, Cannabinoid, CB2/metabolism
- Receptor, Cannabinoid, CB2/genetics
- Pregnancy
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB1/genetics
- Male
- Prenatal Exposure Delayed Effects/immunology
- Prenatal Exposure Delayed Effects/metabolism
- Prenatal Exposure Delayed Effects/physiopathology
- Mice
- Mice, Knockout
- Cannabinoids/pharmacology
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Mice, Inbred C57BL
- Dronabinol/pharmacology
- Poly I-C/pharmacology
- Disease Models, Animal
- Memory, Short-Term/physiology
- Memory, Short-Term/drug effects
Collapse
Affiliation(s)
- Han-Ting Chen
- Department of Psychological and Brain Sciences, Indiana University, 702 N Walnut Grove Ave, Bloomington, IN 47405, USA; Gill Institute for Neuroscience, Indiana University, 702 N Walnut Grove Ave, Bloomington, IN 47405, USA
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Indiana University, 702 N Walnut Grove Ave, Bloomington, IN 47405, USA; Gill Institute for Neuroscience, Indiana University, 702 N Walnut Grove Ave, Bloomington, IN 47405, USA.
| |
Collapse
|
2
|
Zamberletti E, Manenti C, Prini P, Gabaglio M, Grimaldi A, Pulze L, Grassi R, Rubino T. Perturbations of CB1 receptor signalling during adolescence impair cortical myelination in female rats. Pharmacol Res 2025; 216:107758. [PMID: 40306605 DOI: 10.1016/j.phrs.2025.107758] [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: 01/15/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/02/2025]
Abstract
The constant increase in cannabis use among adolescents raises concerns about its potential neurobehavioral effects. Adolescence is a critical period for brain development, involving significant changes in grey and white matter. Grey matter decreases as the brain undergoes synaptic pruning, while white matter increases due to myelination. Cannabis use during this developmental window, particularly its active ingredient delta-9-tetrahydrocannabinol (THC), may disrupt these processes, increasing the risk of developing psychiatric disorders later in life. While the impact of THC on grey matter has been explored, the specific role of CB1 receptors as well as the effect of THC exposure in adolescent myelination remain unclear. This study investigates how CB1 receptor blockade and THC exposure during adolescence affect myelination in the prefrontal cortex of female rats. Blocking CB1 receptors during adolescence hindered myelination in the prefrontal cortex. Behaviourally, this disruption in myelin formation was associated with increased risk-taking behaviour. Notably, our data suggest that alterations in the AKT/Hippo/YAP signalling pathway may play a crucial role in mediating these effects. Supporting the involvement of the endocannabinoid system in cortical myelination during adolescence, we found that administering exogenous THC impaired myelin formation only when given during early to mid-adolescence. Moreover, when a more intensive THC exposure protocol was applied during this developmental period, the effects on myelination were long-lasting and persisted into adulthood. Overall, these data support a role for CB1 receptors in shaping cortical myelination in adolescent female rats and show that adolescent exposure to THC might adversely impact this developmental process.
Collapse
Affiliation(s)
- Erica Zamberletti
- Department of Biotechnology and Life Sciences (DBSV) and Neuroscience Center, Università degli Studi dell'Insubria, Busto Arsizio, Italy
| | - Cristina Manenti
- Department of Biotechnology and Life Sciences (DBSV) and Neuroscience Center, Università degli Studi dell'Insubria, Busto Arsizio, Italy
| | - Pamela Prini
- Department of Biotechnology and Life Sciences (DBSV) and Neuroscience Center, Università degli Studi dell'Insubria, Busto Arsizio, Italy
| | - Marina Gabaglio
- Department of Biotechnology and Life Sciences (DBSV) and Neuroscience Center, Università degli Studi dell'Insubria, Busto Arsizio, Italy
| | - Annalisa Grimaldi
- Dept. of Biotechnology and Life Sciences (DBSV), Università degli Studi dell'Insubria, Varese, Italy
| | - Laura Pulze
- Dept. of Biotechnology and Life Sciences (DBSV), Università degli Studi dell'Insubria, Varese, Italy
| | - Riccardo Grassi
- Department of Biotechnology and Life Sciences (DBSV) and Neuroscience Center, Università degli Studi dell'Insubria, Busto Arsizio, Italy
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences (DBSV) and Neuroscience Center, Università degli Studi dell'Insubria, Busto Arsizio, Italy.
| |
Collapse
|
3
|
Gao W, Wang T, Cui J, Huang N, Fan G, Pan T, Jiang C, Wang F, Liu X, Ma L, Le Q. Paternal heroin self-administration in rats increases drug-seeking behavior in male offspring via miR-19b downregulation in the nucleus accumbens. Neuropsychopharmacology 2025:10.1038/s41386-025-02081-8. [PMID: 40057637 DOI: 10.1038/s41386-025-02081-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/07/2025]
Abstract
Accumulating evidence indicates that drug addiction may lead to adaptive behavioral changes in offspring, potentially due to epigenetic modifications in parental germline. However, the underlying mechanisms remain inadequately understood. In this study, we show that paternal heroin self-administration (SA) increased heroin-seeking behavior in the F1 generation, when compared with offspring sired by yoke-infused control males, indicating cross-generational impact of paternal voluntary heroin seeking behavior. Notably, the increase of heroin seeking behavior in offspring was replicated by zygotic microinjection of sperm RNAs derived from sperm of heroin-SA-experienced rats. Analysis of non-coding RNAs in spermatozoa revealed coordinated changes in miRNA content between the nucleus accumbens and spermatozoa. We validated that restoration of miR-19b downregulation in sperm RNA from self-administration-experienced rats, in parallel with its overexpression in the nucleus accumbens of F1 offspring sired by heroin-SA-experienced fathers, reversed the increased heroin SA observed in these F1 offspring. Taken together, our findings suggest in rats that paternal heroin self-administration induces epigenetic changes in both brain and sperm miRNA, with miR-19b downregulation playing a critical role in mediating the epigenetic inheritance of increased heroin self-administration behavior in the F1 generation.
Collapse
Affiliation(s)
- Wenjing Gao
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Tingting Wang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Jian Cui
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Nan Huang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Guangyuan Fan
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Tao Pan
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Changyou Jiang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, 200032, China
| | - Feifei Wang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, 200032, China
| | - Xing Liu
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, 200032, China
| | - Lan Ma
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China.
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, 200032, China.
| | - Qiumin Le
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China.
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, 200032, China.
| |
Collapse
|
4
|
Portillo R, Synova T, Staud F. Effects of prenatal cannabinoid use on the monoamine system in the fetoplacental unit: A systematic review of animal and human studies. Drug Alcohol Depend 2025; 268:112579. [PMID: 39899918 DOI: 10.1016/j.drugalcdep.2025.112579] [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: 09/24/2024] [Revised: 12/04/2024] [Accepted: 01/18/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND The rapid increase in cannabis use during pregnancy-up by 170 % between 2009 and 2016-raises pressing concerns about its effects on fetal health, particularly on the delicate monoamine system within the fetoplacental unit, which is crucial for placental function and neurodevelopment. OBJECTIVE This systematic review explores the impact of prenatal cannabinoid exposure on the monoamine system within the fetoplacental unit, with a focus on its implications for fetal development through the lens of the Developmental Origins of Health and Disease (DOHaD) framework. METHODS A comprehensive search across multiple databases initially retrieved 18,252 papers. After rigorous screening, only 16 animal studies and 4 human studies met the inclusion criteria. Findings were synthesized to evaluate the effects of prenatal cannabis exposure on neurotransmitter regulation, receptor function, and gene expression. RESULTS Although no studies directly addressed the monoamine system in the placenta, animal models revealed significant disruptions in neurotransmitter regulation and neurodevelopmental changes following prenatal cannabis exposure. Human studies suggested potential cognitive and behavioral risks for offspring exposed in utero. CONCLUSION This review exposes a critical gap in the literature on cannabis' effects on the placental monoamine system. While evidence points to notable neurodevelopmental risks, the scarcity of focused research underscores the need for further investigation to fully understand the implications of prenatal cannabis exposure.
Collapse
Affiliation(s)
- Ramon Portillo
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic
| | - Tetiana Synova
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Kralove, Czech Republic.
| |
Collapse
|
5
|
Pham AL, Marquardt AE, Montgomery KR, Sobota KN, McCarthy MM, VanRyzin JW. Timing matters: modeling the effects of gestational cannabis exposure on social behavior and microglia in the developing amygdala. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638714. [PMID: 40027715 PMCID: PMC11870496 DOI: 10.1101/2025.02.17.638714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Cannabis is the most frequently used illicit drug during pregnancy, with use steadily increasing in the United States as legalization and decriminalization expand to more states. Many pregnant individuals use cannabis to reduce adverse symptoms of pregnancy, considering it to be less harmful than other pharmaceuticals or alcohol. The primary psychoactive component of cannabis, delta-9-tetrahydrocannabinol (THC), acts on the endocannabinoid (eCB) system, yet whether it perturbs neural development of the fetus is poorly understood. Previously we have shown that androgen mediated eCB tone in the developing amygdala promotes microglial phagocytosis of newborn astrocytes which has enduring consequences on the neural circuits regulating sex differences in social behavior. Microglia are the resident immune cells of the brain and express both receptors of the eCB system, CB1R and CB2R, making them likely targets of modulation by THC. It is also plausible that exposure to THC at differing gestational timepoints can result in distinct outcomes, as is the case with alcohol exposure. To model human cannabis use during either late or early pregnancy, we exposed rodents to THC either directly during the early postnatal period via intraperitoneal (IP) injection or in utero during the prenatal period via dam IP injection respectively. Here we show that postnatal THC exposure results in sex specific changes in microglial phagocytosis during development as well as social behavior during the juvenile period. Interestingly prenatal exposure to THC resulted in inverse changes to phagocytosis and social behavior. These findings highlight the differential effects of THC exposure across gestation.
Collapse
Affiliation(s)
- Aidan L Pham
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ashley E Marquardt
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pharmacology and Physiology, University of Maryland Medicine - Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD 21201
| | - Kristen R Montgomery
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON Canada
| | - Karina N Sobota
- Department of Pharmacology and Physiology, University of Maryland Medicine - Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD 21201
- Graduate Program in Physiological Sciences and Department of Physiology, State University of Londrina, Londrina, PR Brazil
| | - Margaret M McCarthy
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pharmacology and Physiology, University of Maryland Medicine - Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD 21201
| | - Jonathan W VanRyzin
- Department of Pharmacology and Physiology, University of Maryland Medicine - Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514
| |
Collapse
|
6
|
Chen HT, Mackie K. CB1 and CB2 receptors differentially modulate the cognitive impact of maternal immune activation and perinatal cannabinoid exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.16.623455. [PMID: 39605459 PMCID: PMC11601341 DOI: 10.1101/2024.11.16.623455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Maternal immune activation (MIA) commonly arises in response to an infection during pregnancy. MIA elevates cytokine levels, triggering an inflammatory cascade, which may be detrimental to the developing nervous system. Similarly, cannabis use and exposure of the fetus to cannabinoids during pregnancy (PCE) may elicit neuroinflammation and lead to detrimental behavioral outcomes. This is particularly concerning as there has been a notable rise in cannabis use during pregnancy. This study endeavors to examine the interaction between MIA and PCE and elucidate the role of CB1 and CB2 receptors in MIA and PCE outcomes. To this end, we compared the impact of MIA, PCE and MIA+PCE in wildtype, CB1, and CB2 cannabinoid receptor knockout mice of both sexes. PCE was modeled by daily 3 mg/kg THC administration from gestational day 5 (GD5) to postnatal day 10. MIA was modeled by intravenous Poly (I:C) injection at GD16.5. Subsequently, we assessed emotional and cognitive behaviors of adult offspring. Adult male offspring of dams exposed to PCE or MIA were impaired in novel object recognition and the delayed alternation working memory tasks. Interestingly, these behavioral impairments were absent when MIA and PCE were combined. Cannabinoid receptor knockout studies found that CB1 receptors mediated behavioral deficits after PCE. In contrast CB2 receptors were necessary for full expression of MIA-induced behavioral impairments. Although females showed more modest behavioral changes after MIA or PCE, CB1 receptors were required for the PCE deficit and CB2 receptors were required for the MIA deficit also in females. Notably, lack of CB2 receptors in males prevented the "protection" following combined MIA + PCE, while CB1 knockout mice remained protected. Taken together, these results suggest a complex interplay between PCE, MIA and CB1 and CB2 cannabinoid receptors. Highlights Both PCE and MIA impair cognitive behaviors.Combined PCE and MIA did not affect the cognitive behaviors examined.CB1 receptors are required for deficits after prenatal cannabis exposure.CB2 receptors are required for deficits after maternal immune activation.CB2 receptors are necessary for protection from deficits by combined PCE and MIA.
Collapse
|
7
|
Black T, Barnard IL, Baccetto SL, Greba Q, Orvold SN, Austin-Scott FVL, Sanfuego GB, Onofrychuk TJ, Glass AE, Andres RM, Macfarlane LM, Adrian JC, Heidt AL, McElroy DL, Laprairie RB, Howland JG. Differential effects of gestational Cannabis smoke and phytocannabinoid injections on male and female rat offspring behavior. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111241. [PMID: 39765319 DOI: 10.1016/j.pnpbp.2024.111241] [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: 08/02/2024] [Revised: 11/22/2024] [Accepted: 12/29/2024] [Indexed: 01/20/2025]
Abstract
Our understanding of the implications of gestational Cannabis exposure (GCE) remains unclear as Cannabis use increases worldwide. Much of the existing knowledge of the effects of GCE has been gained from preclinical experiments using injections of isolated Δ9-tetrahydrocannabinol (THC) at relatively high doses. Few investigations of the effects of GCE to smoke from the whole Cannabis flower have been conducted, despite this being the most common mode of human consumption. Here, we compared the effects of repeated gestational exposure to high-THC or high-cannabidiol (CBD) Cannabis smoke to i.p. THC or i.p. CBD to those of GCE to high-THC or high-CBD Cannabis smoke on litter health and the offspring. We found that injecting phytocannabinoids generally had a more severe impact on measures of maternal and litter health and produced distinct behavioral phenotypes when compared to offspring from dams treated with high-THC and high-CBD smoke during gestation. GCE to high-THC smoke decreased prepulse inhibition (PPI) and MK-801-induced locomotor activity in female adolescent offspring, which normalized in adulthood. GCE to i.p. THC increased exploratory behavior in the open field test in adolescent offspring of both sexes. GCE had a negative impact on offspring performance in the Identical Stimuli Test and Different Stimuli Test with odors regardless of gestational treatment, sex, or age. CBD (i.p) impaired PPI in both male and female offspring in adulthood and increased time spent in proximity during social interaction for male offspring. There were no effects of GCE in the 5 Choice Serial Reaction Time Task. These data establish distinct behavioral phenotypes in the offspring between smoked and injected GCE, further demonstrating that route and specific phytocannabinoid dose produce differential outcomes across offspring lifespan. Smoked Cannabis is still the most common means of consumption, and more preclinical investigation is needed to determine the effects of smoked Cannabis on developmental trajectories.
Collapse
Affiliation(s)
- Tallan Black
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada; Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ilne L Barnard
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Sarah L Baccetto
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada; Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Quentin Greba
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Spencer N Orvold
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Faith V L Austin-Scott
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Genre B Sanfuego
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Timothy J Onofrychuk
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Aiden E Glass
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Rachel M Andres
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Leah M Macfarlane
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jesse C Adrian
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ashton L Heidt
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Dan L McElroy
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Robert B Laprairie
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada; Department of Pharmacology, College of Medicine, Dalhousie University, Halifax, NS, Canada.
| | - John G Howland
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
8
|
Rodrigues FDS, Jantsch J, de Farias Fraga G, Dias VS, Pereira Medeiros C, Wickert F, Schroder N, Giovernardi M, Guedes RP. Cannabidiol partially rescues behavioral, neuroinflammatory and endocannabinoid dysfunctions stemming from maternal obesity in the adult offspring. Neuropharmacology 2025; 262:110196. [PMID: 39447736 DOI: 10.1016/j.neuropharm.2024.110196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 10/26/2024]
Abstract
Maternal obesity is known to increase the risk of psychiatric disorders, such as anxiety, depression, schizophrenia and autism spectrum disorder in the offspring. While preventive measures are well-documented, practical approaches for addressing the damages once they are already established are limited. We have recently demonstrated the interplay between maternal obesity and treatment with cannabidiol (CBD) on neuroinflammation and peripheral metabolic disturbances during adolescence, however, it is known that both factors tend to vary throughout life. Therefore, here we investigated the potential of CBD to mitigate these alterations in the adult offspring of obese dams. Female Wistar rats were fed a cafeteria diet for 12 weeks prior to mating, and during gestation and lactation. Offspring received CBD (50 mg/kg) for 3 weeks from the 70th day of life. Behavioral tests assessed anxiety-like manifestations and social behavior, while neuroinflammatory and endocannabinoid markers were evaluated in the hypothalamus, prefrontal cortex (PFC) and hippocampus, as well as the biochemical profile in the plasma. CBD treatment attenuated maternal obesity-induced anxiety-like and social behavioral alterations, restoring exacerbated astrocytic and microglial markers in the hypothalamus, PFC and hippocampus of the offspring, as well as endocannabinoid levels in the PFC, with notable sex differences. Additionally, CBD attenuated plasma glucose and lipopolysaccharides (LPS) concentrations in females. These findings underscore the persistent influence of maternal obesity on the offspring's health, encompassing metabolic irregularities and behavioral impairments, as well as the role of the endocannabinoid system in mediating these outcomes across the lifespan.
Collapse
Affiliation(s)
- Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriel de Farias Fraga
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Victor Silva Dias
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Camila Pereira Medeiros
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Wickert
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Nadja Schroder
- Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Marcia Giovernardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Rio Grande do Sul, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Rio Grande do Sul, Brazil.
| |
Collapse
|
9
|
Lau D, Tobin S, Pribiag H, Nakajima S, Fisette A, Matthys D, Franco Flores AK, Peyot ML, Murthy Madiraju SR, Prentki M, Stellwagen D, Alquier T, Fulton S. ABHD6 loss-of-function in mesoaccumbens postsynaptic but not presynaptic neurons prevents diet-induced obesity in male mice. Nat Commun 2024; 15:10652. [PMID: 39681558 DOI: 10.1038/s41467-024-54819-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
α/β-hydrolase domain 6 (ABHD6) is a lipase linked to physiological functions affecting energy metabolism. Brain ABHD6 degrades 2-arachidonoylglycerol and thereby modifies cannabinoid receptor signalling. However, its functional role within mesoaccumbens circuitry critical for motivated behaviour and considerably modulated by endocannabinoids was unknown. Using three viral approaches, we show that control of the nucleus accumbens by neuronal ABHD6 is a key determinant of body weight and reward-directed behaviour in male mice. Contrary to expected outcomes associated with increasing endocannabinoid tone, loss of ABHD6 in nucleus accumbens, but not ventral tegmental area, neurons completely prevents diet-induced obesity, reduces food- and drug-seeking and enhances physical activity without affecting anxiodepressive behaviour. These effects are explained by attenuated inhibitory synaptic transmission onto medium spiny neurons. ABHD6 deletion in nucleus accumbens neurons and dopamine ventral tegmental area neurons produces contrasting effects on effortful responding for food. Intraventricular infusions of an ABHD6 inhibitor also restrain appetite and promote weight loss. Together, these results reveal functional specificity of pre- and post-synaptic mesoaccumbens neuronal ABHD6 to differentially control energy balance and propose ABHD6 inhibition as a potential anti-obesity tool.
Collapse
Affiliation(s)
- David Lau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - Stephanie Tobin
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Horia Pribiag
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University Health Center, Montréal, QC, Canada
| | - Shingo Nakajima
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Alexandre Fisette
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Dominique Matthys
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - Anna Kristyna Franco Flores
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - Marie-Line Peyot
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - S R Murthy Madiraju
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Marc Prentki
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Nutrition, Université de Montréal, Montréal, QC, Canada
| | - David Stellwagen
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, McGill University Health Center, Montréal, QC, Canada
| | - Thierry Alquier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Stephanie Fulton
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.
- Department of Nutrition, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
10
|
Valeria S, Francesco T, Sonia A, Laura VP, Luca C, Marcello S, Roberta L, Patrizia P, Arnau BG, Roberto F, Miriam M. Sex-specific maladaptive responses to acute stress upon in utero THC exposure are mediated by dopamine. Pharmacol Res 2024; 210:107536. [PMID: 39622370 PMCID: PMC7617568 DOI: 10.1016/j.phrs.2024.107536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/04/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024]
Abstract
Cannabis remains by far the most consumed illicit drug in Europe. The availability of more potent cannabis has raised concerns regarding the enhanced health risks associated with its use, particularly among pregnant women. Growing evidence shows that cannabis use during pregnancy increases the risks of child psychopathology. We have previously shown that only male rat offspring prenatally exposed to Δ9-tetrahydrocannabinol (THC), a rat model of prenatal cannabinoid exposure (PCE), display a hyperdopaminergic phenotype associated with a differential susceptibility to acute THC- and stress-mediated effects on sensorimotor gating functions. Here, we explore the contribution of the hypothalamic-pituitary-adrenal (HPA) axis, key regulator of body adaptive stress responses, to the detrimental effects of acute stress on ventral tegmental area (VTA) dopamine neurons and sensorimotor gating function of PCE rats. We report a sex-dependent compromised balance in mRNA levels of genes encoding mineralocorticoid and glucocorticoid receptors in the VTA, alongside with stress-induced pre-pulse inhibition (PPI) impairment. Notably, VTA dopamine neuronal activity is causally linked to the manifestation of stress-dependent deterioration of PPI. Finally, pharmacological manipulations targeting glycogen-synthase-kinase-3-β signaling during postnatal development correct these stress-induced, sex-specific and dopamine-dependent disruption of PPI. Collectively, these results highlight the critical sex-dependent interplay between HPA axis and dopamine system in the regulation of sensorimotor gating functions in rats.
Collapse
Affiliation(s)
- Serra Valeria
- Dept. Biomedical Sciences, Div. Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - Traccis Francesco
- Dept. Biomedical Sciences, Div. Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - Aroni Sonia
- Dept. Biomedical Sciences, Div. Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | | | - Concas Luca
- Dept. Biomedical Sciences, Div. Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - Serra Marcello
- Dept. Biomedical Sciences, Div. Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - Leone Roberta
- Dept. Biomedical Sciences, Div. Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - Porcu Patrizia
- Institute of Neurosciences, National Research Council (C.N.R.), Cagliari, Italy
| | | | - Frau Roberto
- Dept. Biomedical Sciences, Div. Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - Melis Miriam
- Dept. Biomedical Sciences, Div. Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| |
Collapse
|
11
|
Santoni M, Pistis M. Maternal Immune Activation and the Endocannabinoid System: Focus on Two-Hit Models of Schizophrenia. Biol Psychiatry 2024:S0006-3223(24)01783-9. [PMID: 39617194 DOI: 10.1016/j.biopsych.2024.11.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/11/2024] [Accepted: 11/23/2024] [Indexed: 02/05/2025]
Abstract
The devastating effects of the COVID-19 pandemic have underscored the significant threat that infectious diseases pose to our society. Pregnancy represents a period of heightened vulnerability to infections, which can compromise maternal health and increase the risk of neurodevelopmental disorders in offspring. Preclinical and clinical investigations suggest a potential association between maternal immune activation (MIA), which is triggered by viral or bacterial infections, and increased risk for neurodevelopmental disorders such as autism and schizophrenia. Genetic and environmental factors may contribute to the overall risk. Therefore, the two-hit hypothesis of schizophrenia suggests that MIA could act as a first trigger, with subsequent factors, such as stress or drug abuse, exacerbating latent abnormalities. A growing body of research is focused on the interaction between MIA and cannabis use during adolescence, considering the role of the endocannabinoid (eCB) system in neurodevelopment and in neurodevelopmental disorders. The eCB system, crucial for fetal brain development, may be disrupted by MIA, leading to adverse outcomes in adulthood. Recent research indicates the eCB system's significant role in the pathophysiology of neurodevelopmental disorders in preclinical models. However, findings on adolescent cannabinoid exposure in MIA-exposed animals have revealed unexpected complexities, with several studies failing to support the exacerbation of MIA-related abnormalities. In this review, we delve into the functional implications of the eCB system in MIA models, emphasizing the role of 2-AG (2-arachidonoylglycerol) signaling in synaptic plasticity and neuroinflammation and its relevance to the two-hit model of schizophrenia.
Collapse
Affiliation(s)
- Michele Santoni
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy; Unit of Clinical Pharmacology, University Hospital, Cagliari, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy; Unit of Clinical Pharmacology, University Hospital, Cagliari, Italy; Neuroscience Institute, National Research Council of Italy, Cagliari, Italy.
| |
Collapse
|
12
|
Luján MÁ, Young-Morrison R, Aroni S, Katona I, Melis M, Cheer JF. Dynamic overrepresentation of accumbal cues in food- and opioid-seeking rats after prenatal THC exposure. SCIENCE ADVANCES 2024; 10:eadq5652. [PMID: 39514650 PMCID: PMC11546747 DOI: 10.1126/sciadv.adq5652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
The increasing prevalence of cannabis use during pregnancy has raised medical concerns, primarily related to Δ9-tetrahydrocannabinol (THC), which readily crosses the placenta and affects fetal brain development. Previous research has identified dopaminergic alterations related to maternal THC consumption. However, the consequences that prenatal cannabis exposure (PCE) has on striatum-based processing during reward pursuit have not been determined. Here, we characterize PCE rats during food or opioid-maintained reward seeking. We find that the supramotivational phenotype of PCE rats is independent of value-based processing and is instead related to augmented reinforcing efficiency of opioid rewards. Our findings reveal that prenatal THC exposure leads to increased cue-evoked dopamine responses and an overrepresentation of effort-driven striatal encoding patterns. Recapitulating clinical findings, drug-related PCE adaptations were more pronounced in males, who showed increased vulnerability for relapse. Collectively, these findings indicate that prenatal THC exposure in male rats engenders a pronounced neurodevelopmental susceptibility to addiction-like disorders.
Collapse
Affiliation(s)
- Miguel Á. Luján
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Reana Young-Morrison
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sonia Aroni
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - István Katona
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Monserrato, Italy
| | - Joseph F. Cheer
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
13
|
Penman SL, Roeder NM, Wang J, Richardson BJ, Freeman-Striegel L, Krayevsky A, Eiden RD, Chakraborty S, Thanos PK. Vaporized Δ9-tetrahydrocannabinol exposure in utero has negative effects on attention in a dose- and sex-dependent manner. Pharmacol Biochem Behav 2024; 242:173808. [PMID: 38914267 DOI: 10.1016/j.pbb.2024.173808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/03/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024]
Abstract
There has been an increasing use of cannabis during pregnancy in recent years. Studies have indicated that THC exposure in utero may increase the risk of attention deficits and memory impairments in adolescence. The goal of the present study is to investigate the effects of vaporized THC exposure during pregnancy on offspring memory and attention performance in early and late adolescence. Pregnant dams were exposed to vaporized THC (10 mg or 40 mg) daily from gestational day 2 until labor. Pups were given either a standard or a high-fat diet at weaning and tested in early and late adolescence in two memory tests, the Novel Object Recognition (NOR) test and the Morris Water Maze (MWM) test, and a test of attention, the Object-Based Attention (OBA) test. Rats exposed to low-dose THC showed significantly decreased object exploration in both the NOR and OBA tests, indicating decreased attention. Object exploration time in OBA was significantly lower in females than males. Additionally, post hoc analysis of MWM tests showed some differences in learning patterns for HD THC offspring in early adolescence, possibly due to diet interaction, but ultimate performance was not impacted. While there are existing studies examining prenatal exposure to THC in rodents, this is the first to our knowledge examining memory and attention in adolescence following vaporized THC exposure in utero, and we find indications that prenatal THC exposure may lead to attention deficits and altered memory performance.
Collapse
Affiliation(s)
- Samantha L Penman
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Nicole M Roeder
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Department of Psychology, University at Buffalo, Buffalo, NY, USA
| | - Jia Wang
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Brittany J Richardson
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Lily Freeman-Striegel
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Alexis Krayevsky
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Rina D Eiden
- Department of Psychology and Social Science Research Institute, The Pennsylvania State University, University Park, PA, USA
| | - Saptarshi Chakraborty
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Department of Psychology, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
14
|
Scheggi S, Concas L, Corsi S, Carta M, Melis M, Frau R. Expanding the therapeutic potential of neuro(active)steroids: a promising strategy for hyperdopaminergic behavioral phenotypes. Neurosci Biobehav Rev 2024; 164:105842. [PMID: 39103066 DOI: 10.1016/j.neubiorev.2024.105842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024]
Abstract
Imbalances in dopamine activity significantly contribute to the pathophysiology of several neuropsychiatric disorders, including addiction, ADHD, schizophrenia, impulse control disorders, and Parkinson's Disease. Neuro(active)steroids, comprising endogenous steroids that finely modulate neuronal activity, are considered crucial regulators of brain function and behavior, with implications in various physiological processes and pathological conditions. Specifically, subclasses of Neuro(active)steroids belonging to the 5α reductase pathway are prominently involved in brain disorders characterized by dopaminergic signaling imbalances. This review highlights the neuromodulatory effects of Neuro(active)steroids on the dopamine system and related aberrant behavioral phenotypes. We critically appraise the role of pregnenolone, progesterone, and allopregnanolone on dopamine signaling. Additionally, we discuss the impact of pharmacological interventions targeting 5α reductase activity in neuropsychiatric conditions characterized by excessive activation of the dopaminergic system, ranging from psychotic (endo)phenotypes and motor complications to decision-making problems and addiction.
Collapse
Affiliation(s)
- Simona Scheggi
- Dept. of Molecular and Developmental Medicine, University of Siena, Italy
| | - Luca Concas
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Sara Corsi
- Dept. of Developmental and Regenerative Neurobiology, Lund University, Sweden
| | - Manolo Carta
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Miriam Melis
- Dept. Of Biomedical Sciences, University of Cagliari, Italy
| | - Roberto Frau
- Dept. Of Biomedical Sciences, University of Cagliari, Italy; Guy Everett Laboratory, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
15
|
DeVuono MV, Nashed MG, Sarikahya MH, Kocsis A, Lee K, Vanin SR, Hudson R, Lonnee EP, Rushlow WJ, Hardy DB, Laviolette SR. Prenatal tetrahydrocannabinol and cannabidiol exposure produce sex-specific pathophysiological phenotypes in the adolescent prefrontal cortex and hippocampus. Neurobiol Dis 2024; 199:106588. [PMID: 38960101 DOI: 10.1016/j.nbd.2024.106588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024] Open
Abstract
Clinical and preclinical evidence has demonstrated an increased risk for neuropsychiatric disorders following prenatal cannabinoid exposure. However, given the phytochemical complexity of cannabis, there is a need to understand how specific components of cannabis may contribute to these neurodevelopmental risks later in life. To investigate this, a rat model of prenatal cannabinoid exposure was utilized to examine the impacts of specific cannabis constituents (Δ9-tetrahydrocannabinol [THC]; cannabidiol [CBD]) alone and in combination on future neuropsychiatric liability in male and female offspring. Prenatal THC and CBD exposure were associated with low birth weight. At adolescence, offspring displayed sex-specific behavioural changes in anxiety, temporal order and social cognition, and sensorimotor gating. These phenotypes were associated with sex and treatment-specific neuronal and gene transcriptional alterations in the prefrontal cortex, and ventral hippocampus, regions where the endocannabinoid system is implicated in affective and cognitive development. Electrophysiology and RT-qPCR analysis in these regions implicated dysregulation of the endocannabinoid system and balance of excitatory and inhibitory signalling in the developmental consequences of prenatal cannabinoids. These findings reveal critical insights into how specific cannabinoids can differentially impact the developing fetal brains of males and females to enhance subsequent neuropsychiatric risk.
Collapse
Affiliation(s)
- Marieka V DeVuono
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada.
| | - Mina G Nashed
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Mohammed H Sarikahya
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Andrea Kocsis
- Dept of Physiology & Pharmacology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Obstetrics & Gynecology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Kendrick Lee
- Dept of Physiology & Pharmacology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Obstetrics & Gynecology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Sebastian R Vanin
- Dept of Physiology & Pharmacology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Obstetrics & Gynecology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Roger Hudson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Eryn P Lonnee
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Walter J Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Psychiatry, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Daniel B Hardy
- Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Physiology & Pharmacology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Obstetrics & Gynecology, University of Western Ontario, London, ON N6A 3K7, Canada; Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute (CHRI), Lawson Health Research Institute, St. Joseph's Health Care, London, ON N6C 2R5, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Psychiatry, University of Western Ontario, London, ON N6A 3K7, Canada; Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute (CHRI), Lawson Health Research Institute, St. Joseph's Health Care, London, ON N6C 2R5, Canada
| |
Collapse
|
16
|
Murru E, Carta G, Manca C, Verce M, Everard A, Serra V, Aroni S, Melis M, Banni S. Impact of prenatal THC exposure on lipid metabolism and microbiota composition in rat offspring. Heliyon 2024; 10:e35637. [PMID: 39170117 PMCID: PMC11336829 DOI: 10.1016/j.heliyon.2024.e35637] [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: 01/08/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Objective Recent studies have demonstrated that prenatal exposure to the psychoactive ingredient of cannabis that is tetrahydrocannabinol (THC) disrupts fatty acid (FA) signaling pathways in the developing brain, potentially linking to psychopathologic consequences. Our research aims to investigate whether changes in midbrain FA metabolism are linked to modifications in peripheral metabolism of FAs and shifts in microbiota composition. Methods In order to model prenatal exposure to THC (PTE) in rats, Sprague Dawley dams were systemically administered with THC (2 mg/kg, s.c.) or vehicle once daily from gestational day 5-20. To evaluate the metabolic impact of PTE in the offspring during preadolescence (postnatal day, PND, 25-28), we analyzed FA profiles and their bioactive metabolites in liver and midbrain tissues, and microbiota alterations. Results Our findings indicate that PTE leads to sex-specific metabolic changes. In both sexes, PTE resulted in increased liver de novo lipogenesis (DNL) and alterations in FA profiles, as well as changes in N-acylethanolamines (NAEs), ligands of peroxisome proliferator-activated receptor alpha (PPAR-α). In females only, PTE influenced gene expression of PPAR-α and fibroblast growth factor 21 (Fgf21). In male offspring only, PTE was associated with significantly reduced fasting glycaemia and with alterations in the levels of midbrain NAEs. Our analysis of the progeny gut microbiota revealed sex-dependent effects of PTE, notably an increased abundance of Ileibacterium in PTE-exposed male offspring, a change previously associated with the long-term effects of a maternal unbalanced diet. Conclusions Our data suggest that in male PTE offspring a reduced fasting glycaemia, resulting from increased liver DNL and the absence of a compensatory effect by Ppar-α and FGF21 on glycemic homeostasis, are associated to alterations in midbrain NAEs ligands of PPAR-α. These metabolic changes within the midbrain, along with Ileibacterium abundance, may partly elucidate the heightened susceptibility to psychopathologic conditions previously observed in male offspring following PTE.
Collapse
Affiliation(s)
- Elisabetta Murru
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Gianfranca Carta
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Claudia Manca
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Marko Verce
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute (WELRI), avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute (WELRI), avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Valeria Serra
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Sonia Aroni
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| |
Collapse
|
17
|
Brouwer A, Carhart‐Harris RL, Raison CL. Psychotomimetic compensation versus sensitization. Pharmacol Res Perspect 2024; 12:e1217. [PMID: 38923845 PMCID: PMC11194300 DOI: 10.1002/prp2.1217] [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: 04/11/2024] [Accepted: 05/08/2024] [Indexed: 06/28/2024] Open
Abstract
It is a paradox that psychotomimetic drugs can relieve symptoms that increase risk of and cooccur with psychosis, such as attention and motivational deficits (e.g., amphetamines), pain (e.g., cannabis) and symptoms of depression (e.g., psychedelics, dissociatives). We introduce the ideas of psychotomimetic compensation and psychotomimetic sensitization to explain this paradox. Psychotomimetic compensation refers to a short-term stressor or drug-induced compensation against stress that is facilitated by engagement of neurotransmitter/modulator systems (endocannabinoid, serotonergic, glutamatergic and dopaminergic) that mediate the effects of common psychotomimetic drugs. Psychotomimetic sensitization occurs after repeated exposure to stress and/or drugs and is evidenced by the gradual intensification and increase of psychotic-like experiences over time. Theoretical and practical implications of this model are discussed.
Collapse
Affiliation(s)
- Ari Brouwer
- Department of Human Development and Family Studies, School of Human EcologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Robin L. Carhart‐Harris
- Department of Neurology and PsychiatryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Charles L. Raison
- Department of Psychiatry, School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Vail Health Behavioral Health Innovation CenterVailColoradoUSA
- Center for the Study of Human HealthEmory UniversityAtlantaGeorgiaUSA
- Department of Spiritual HealthEmory University Woodruff Health Sciences CenterAtlantaGeorgiaUSA
| |
Collapse
|
18
|
Nutor C, Dickerson AS, Hsu T, Al-Jadiri A, Camargo CA, Schweitzer JB, Shuster CL, Karagas MR, Madan JC, Restrepo B, Schmidt RJ, Lugo-Candelas C, Neiderhiser J, Sathyanarayana S, Dunlop AL, Brennan PA. Examining the association between prenatal cannabis exposure and child autism traits: A multi-cohort investigation in the environmental influences on child health outcome program. Autism Res 2024; 17:1651-1664. [PMID: 38953698 PMCID: PMC11341247 DOI: 10.1002/aur.3185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/29/2024] [Indexed: 07/04/2024]
Abstract
This study examined the association between prenatal cannabis exposure and autism spectrum disorder (ASD) diagnoses and traits. A total sample of 11,570 children (ages 1-18; 53% male; 25% Hispanic; 60% White) from 34 cohorts of the National Institutes of Health-funded environmental influences on child health outcomes consortium were included in analyses. Results from generalized linear mixed models replicated previous studies showing that associations between prenatal cannabis exposure and ASD traits in children are not significant when controlling for relevant covariates, particularly tobacco exposure. Child biological sex did not moderate the association between prenatal cannabis exposure and ASD. In a large sample and measuring ASD traits continuously, there was no evidence that prenatal cannabis exposure increases the risk for ASD. This work helps to clarify previous mixed findings by addressing concerns about statistical power and ASD measurement.
Collapse
Affiliation(s)
- Chaela Nutor
- Department of Psychology, Emory University, Atlanta, Georgia, USA
| | - Aisha S Dickerson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Tingju Hsu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Aseel Al-Jadiri
- Institute for Child Development, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Carlos A Camargo
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julie B Schweitzer
- School of Medicine, University of California, Sacramento, California, USA
| | - Coral L Shuster
- Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA
| | | | - Juliette C Madan
- Department of Pediatrics, Psychiatry & Epidemiology, Children's Hospital at Dartmouth, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Bibiana Restrepo
- Department of Pediatrics, University of California Davis School of Medicine, MIND Institute, Sacramento, California, USA
| | - Rebecca J Schmidt
- Department of Public Health Sciences and the MIND Institute, University of California Davis School of Medicine, Davis, California, USA
| | | | - Jenae Neiderhiser
- Department of Psychology, Pennsylvania State University, University Park, Pennsylvania, USA
| | | | - Anne L Dunlop
- Department of Gynecology and Obstetrics, Emory University, Atlanta, Georgia, USA
| | | |
Collapse
|
19
|
Karatayev O, Collier AD, Targoff SR, Leibowitz SF. Neurological Disorders Induced by Drug Use: Effects of Adolescent and Embryonic Drug Exposure on Behavioral Neurodevelopment. Int J Mol Sci 2024; 25:8341. [PMID: 39125913 PMCID: PMC11313660 DOI: 10.3390/ijms25158341] [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: 06/18/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Clinical studies demonstrate that the risk of developing neurological disorders is increased by overconsumption of the commonly used drugs, alcohol, nicotine and cannabis. These drug-induced neurological disorders, which include substance use disorder (SUD) and its co-occurring emotional conditions such as anxiety and depression, are observed not only in adults but also with drug use during adolescence and after prenatal exposure to these drugs, and they are accompanied by long-lasting disturbances in brain development. This report provides overviews of clinical and preclinical studies, which confirm these adverse effects in adolescents and the offspring prenatally exposed to the drugs and include a more in-depth description of specific neuronal systems, their neurocircuitry and molecular mechanisms, affected by drug exposure and of specific techniques used to determine if these effects in the brain are causally related to the behavioral disturbances. With analysis of further studies, this review then addresses four specific questions that are important for fully understanding the impact that drug use in young individuals can have on future pregnancies and their offspring. Evidence demonstrates that the adverse effects on their brain and behavior can occur: (1) at low doses with short periods of drug exposure during pregnancy; (2) after pre-conception drug use by both females and males; (3) in subsequent generations following the initial drug exposure; and (4) in a sex-dependent manner, with drug use producing a greater risk in females than males of developing SUDs with emotional conditions and female offspring after prenatal drug exposure responding more adversely than male offspring. With the recent rise in drug use by adolescents and pregnant women that has occurred in association with the legalization of cannabis and increased availability of vaping tools, these conclusions from the clinical and preclinical literature are particularly alarming and underscore the urgent need to educate young women and men about the possible harmful effects of early drug use and to seek novel therapeutic strategies that might help to limit drug use in young individuals.
Collapse
Affiliation(s)
| | | | | | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA; (O.K.); (S.R.T.)
| |
Collapse
|
20
|
Navarro D, Gasparyan A, Navarrete F, Manzanares J. Fetal Cannabinoid Syndrome: Behavioral and Brain Alterations of the Offspring Exposed to Dronabinol during Gestation and Lactation. Int J Mol Sci 2024; 25:7453. [PMID: 39000559 PMCID: PMC11242182 DOI: 10.3390/ijms25137453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
This study establishes a fetal cannabinoid syndrome model to evaluate the effects of high doses of dronabinol (synthetic THC) during pregnancy and lactation on behavioral and brain changes in male and female progeny and their susceptibility to alcohol consumption. Female C57BL/6J mice received dronabinol (10 mg/kg/12 h, p.o.) from gestational day 5 to postnatal day 21. On the weaning day, the offspring were separated by sex, and on postnatal day 60, behavioral and neurobiological changes were analyzed. Mice exposed to dronabinol exhibited increased anxiogenic and depressive-like behaviors and cognitive impairment. These behaviors were associated with neurodevelopment-related gene and protein expression changes, establishing, for the first time, an association among behavioral changes, cognitive impairment, and neurobiological alterations. Exposure to dronabinol during pregnancy and lactation disrupted the reward system, leading to increased motivation to consume alcohol in the offspring. All these modifications exhibited sex-dependent patterns. These findings reveal the pronounced adverse effects on fetal neurodevelopment resulting from cannabis use during pregnancy and lactation and strongly suggest the need to prevent mothers who use cannabis in this period from the severe and permanent side effects on behavior and brain development that may occur in their children.
Collapse
Affiliation(s)
- Daniela Navarro
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
21
|
Peterson CS, Baglot SL, Sallam NA, Mina S, Hill MN, Borgland SL. Oral pre- and early postnatal cannabis exposure disinhibits ventral tegmental area dopamine neuron activity but does not influence cocaine preference in offspring in mice. J Neurosci Res 2024; 102:e25369. [PMID: 39037062 DOI: 10.1002/jnr.25369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 07/23/2024]
Abstract
Cannabis consumption has increased from 1.5% to 2.5% in Canada between 2012 and 2019. Clinical studies have indicated effects of prenatal cannabis exposure on birth weight, substance use, and neurodevelopmental disorders, but are confounded by several difficult to control variables. Animal models allow for examination of the mechanism of cannabis-induced changes in neurodevelopment and behavior, while controlling dose and timing. Several animal models of prenatal cannabis exposure exist which provide varying levels of construct validity, control of dose, and exposure to maternal stress. Using a voluntary oral consumption model, mouse dams received 5 mg/kg Δ9-tetrahydrocannabinol (THC) whole cannabis oil in peanut butter daily from gestational day 1 (GD1) to postnatal day 10 (PD10). At GD1, GD18, PD1, PD10, and PD15, maternal plasma was collected; pup brains were collected from GD18 onward. Pup brains had higher levels of THC and cannabidiol at each time point, each of which persisted in maternal plasma and pup brains past the end of treatment (PD15). Male and female adolescent offspring were examined for changes to ventral tegmental area (VTA) dopamine neuron activity and cocaine-seeking behavior. Prenatal and early postnatal (GD1-PD10) cannabis-exposed male, but not female mice had decreased gamma-aminobutyric acid (GABAergic) input, depolarized resting membrane potential, and increased spontaneous firing of VTA dopamine neurons. Cannabis-exposed offspring showed faster decay of N-methyl-D-aspartate (NMDA) currents in both sexes. However, no differences in cocaine-seeking behavior were noted. These data characterize a voluntary prenatal cannabis exposure model and demonstrates VTA dopamine neuronal activity is disinhibited in offspring.
Collapse
Affiliation(s)
- Colleen S Peterson
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Samantha L Baglot
- Department of Cell Biology and Anatomy, Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nada A Sallam
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt, Cairo University, Cairo, Egypt
| | - Sarah Mina
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Matthew N Hill
- Department of Cell Biology and Anatomy, Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
22
|
Nutor C, Dunlop A, Sadler O, Brennan PA. Prenatal Cannabis Use and Offspring Autism-Related Behaviors: Examining Maternal Stress as a Moderator in a Black American Cohort. J Autism Dev Disord 2024; 54:2355-2367. [PMID: 37097527 PMCID: PMC10127191 DOI: 10.1007/s10803-023-05982-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 04/26/2023]
Abstract
Prenatal cannabis use and maternal stress have been proposed as risk factors for autism spectrum disorder (ASD). Black mothers and mothers of lower socioeconomic status (SES) may be especially likely to experience high levels of stress. This study examined the impact of prenatal cannabis use and maternal stress (i.e., prenatal distress, racial discrimination, and lower SES) on child ASD-related behaviors in a sample of 172 Black mother-child pairs. We found that prenatal stress was significantly associated with ASD-related behaviors. Prenatal cannabis use did not predict ASD-related behaviors and did not interact with maternal stress to predict ASD-related behaviors. These findings replicate previous work on prenatal stress-ASD associations and add to the limited literature on prenatal cannabis-ASD associations in Black samples.
Collapse
Affiliation(s)
- C Nutor
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA.
| | - A Dunlop
- Department of Gynecology and Obstetrics, Emory University, 1365 E Clifton Rd NE, Atlanta, GA, 30322, USA
| | - O Sadler
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA
| | - P A Brennan
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA
| |
Collapse
|
23
|
Luján MÁ, Young-Morrison R, Aroni S, Katona I, Melis M, Cheer J. Dynamic Overrepresentation of Accumbal Cues in Food- and Opioid-Seeking Rats after Prenatal THC Exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592839. [PMID: 38766015 PMCID: PMC11100737 DOI: 10.1101/2024.05.06.592839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The increasing prevalence of cannabis use during pregnancy has raised significant medical concerns, primarily related to the presence of Δ9-tetrahydrocannabinol (THC), which readily crosses the placenta and impacts fetal brain development. Previous research has identified midbrain dopaminergic neuronal alterations related to maternal THC consumption. However, the enduring consequences that prenatal cannabis exposure (PCE) has on striatum-based processing during voluntary reward pursuit have not been specifically determined. Here, we characterize PCE rats during food (palatable pellets) or opioid (remifentanyl)-maintained reward seeking. We find that the supra motivational phenotype of PCE rats is independent of value-based processing and is instead related to augmented reinforcing efficiency of opioid rewards. Our findings reveal that in utero THC exposure leads to increased cue-evoked dopamine release responses and an overrepresentation of cue-aligned, effort-driven striatal patterns of encoding. Recapitulating findings in humans, drug-related neurobiological adaptations of PCE were more pronounced in males, who similarly showed increased vulnerability for relapse. Collectively, these findings indicate that prenatal THC exposure in male rats engenders a pronounced neurodevelopmental susceptibility to addiction-like disorders later in life.
Collapse
|
24
|
Riyahi J, Taslimi Z, Gelfo F, Petrosini L, Haghparast A. Trans-generational effects of parental exposure to drugs of abuse on offspring memory functions. Neurosci Biobehav Rev 2024; 160:105644. [PMID: 38548003 DOI: 10.1016/j.neubiorev.2024.105644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/10/2024] [Accepted: 03/22/2024] [Indexed: 04/01/2024]
Abstract
Recent evidence reported that parental-derived phenotypes can be passed on to the next generations. Within the inheritance of epigenetic characteristics allowing the transmission of information related to the ancestral environment to the offspring, the specific case of the trans-generational effects of parental drug addiction has been extensively studied. Drug addiction is a chronic disorder resulting from complex interactions among environmental, genetic, and drug-related factors. Repeated exposures to drugs induce epigenetic changes in the reward circuitry that in turn mediate enduring changes in brain function. Addictive drugs can exert their effects trans-generally and influence the offspring of addicted parents. Although there is growing evidence that shows a wide range of behavioral, physiological, and molecular phenotypes in inter-, multi-, and trans-generational studies, transmitted phenotypes often vary widely even within similar protocols. Given the breadth of literature findings, in the present review, we restricted our investigation to learning and memory performances, as examples of the offspring's complex behavioral outcomes following parental exposure to drugs of abuse, including morphine, cocaine, cannabinoids, nicotine, heroin, and alcohol.
Collapse
Affiliation(s)
- Javad Riyahi
- Department of Cognitive and Behavioral Science and Technology in Sport, Faculty of Sport Sciences and Health, Shahid Beheshti University, Tehran, Iran
| | - Zahra Taslimi
- Behavioral Disorders and Substance Abuse Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Fertility and Infertility Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Francesca Gelfo
- IRCCS Santa Lucia Foundation, Rome, Italy; Department of Human Sciences, Guglielmo Marconi University, Rome, Italy
| | | | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran; Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Baddenhausen S, Lutz B, Hofmann C. Cannabinoid type-1 receptor signaling in dopaminergic Engrailed-1 expressing neurons modulates motivation and depressive-like behavior. Front Mol Neurosci 2024; 17:1379889. [PMID: 38660383 PMCID: PMC11042029 DOI: 10.3389/fnmol.2024.1379889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/21/2024] [Indexed: 04/26/2024] Open
Abstract
The endocannabinoid system comprises highly versatile signaling functions within the nervous system. It is reported to modulate the release of several neurotransmitters, consequently affecting the activity of neuronal circuits. Investigations have highlighted its roles in numerous processes, including appetite-stimulating characteristics, particularly for palatable food. Moreover, endocannabinoids are shown to fine-tune dopamine-signaled processes governing motivated behavior. Specifically, it has been demonstrated that excitatory and inhibitory inputs controlled by the cannabinoid type 1 receptor (CB1) regulate dopaminergic neurons in the mesocorticolimbic pathway. In the present study, we show that mesencephalic dopaminergic (mesDA) neurons in the ventral tegmental area (VTA) express CB1, and we investigated the consequences of specific deletion of CB1 in cells expressing the transcription factor Engrailed-1 (En1). To this end, we validated a new genetic mouse line EN1-CB1-KO, which displays a CB1 knockout in mesDA neurons beginning from their differentiation, as a tool to elucidate the functional contribution of CB1 in mesDA neurons. We revealed that EN1-CB1-KO mice display a significantly increased immobility time and shortened latency to the first immobility in the forced swim test of adult mice. Moreover, the maximal effort exerted to obtain access to chocolate-flavored pellets was significantly reduced under a progressive ratio schedule. In contrast, these mice do not differ in motor skills, anhedonia- or anxiety-like behavior compared to wild-type littermates. Taken together, these findings suggest a depressive-like or despair behavior in an inevitable situation and a lack of motivation to seek palatable food in EN1-CB1-KO mice, leading us to propose that CB1 plays an important role in the physiological functions of mesDA neurons. In particular, our data suggest that CB1 directly modifies the mesocorticolimbic pathway implicated in depressive-like/despair behavior and motivation. In contrast, the nigrostriatal pathway controlling voluntary movement seems to be unaffected.
Collapse
Affiliation(s)
- Sarah Baddenhausen
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
| | - Clementine Hofmann
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
26
|
Moore BF. Prenatal Exposure to Cannabis: Effects on Childhood Obesity and Cardiometabolic Health. Curr Obes Rep 2024; 13:154-166. [PMID: 38172481 PMCID: PMC10933144 DOI: 10.1007/s13679-023-00544-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE OF REVIEW To consolidate information on the obesogenic and cardiometabolic effects of prenatal exposure to cannabis. RECENT FINDINGS A PubMed search strategy updated from January 1, 2014, through 14 June 2023, produced a total of 47 epidemiologic studies and 12 animal studies. Prenatal exposure to cannabis is consistently associated with small for gestational age and low birth weight. After birth, these offspring gain weight rapidly and have increased adiposity and higher glucose (fat mass percentage) in childhood. More preclinical and prospective studies are needed to deepen our understanding of whether these associations vary by sex, dose, timing, and composition of cannabis (e.g., ratio of delta-Δ9-tetrahydrocannabinol [Δ9-THC] to cannabidiol [CBD]). Addressing these gaps may help to solidify causality and identify intervention strategies. Based on the available data, clinicians and public health officials should continue to caution against cannabis use during pregnancy to limit its potential obesogenic and adverse cardiometabolic effects on the offspring.
Collapse
Affiliation(s)
- Brianna F Moore
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA.
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, 1890 N Revere Ct, Aurora, 80045, CO, USA.
| |
Collapse
|
27
|
Türkoğlu Ö, Ertuğrul A. The Role of Cannabis in the Development of Psychosis. TURK PSIKIYATRI DERGISI = TURKISH JOURNAL OF PSYCHIATRY 2024; 35:234-244. [PMID: 39224996 PMCID: PMC11375744 DOI: 10.5080/u27122] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cannabis is known to cause psychotic disorders, and the increasing use of cannabis constitutes an important health problem. Growing evidence that cannabis causes the development of psychosis has led to an increase in the number of studies in this field. This review aims to clarify the role of cannabis use in the development of psychosis, discuss the current literature about the underlying neurobiological mechanisms. For this purpose PubMed was searched for the keywords "cannabis use, psychosis, schizophrenia, endocannabinoid system, pathophysiology, neurobiology"; the articles published in the last 10 years were reviewed. Epidemiological studies showed that cannabis use starting at an earlier age is associated with an increased risk of psychosis, this risk is more pronounced in people with genetic predisposition and increases with heavy and high potency cannabis use. Studies showed that the endocannabinoid system, which plays a role in nervous system development and functions as a homeostatic regulator in physiological processes, is affected by cannabis use during critical periods of development like adolescence; cannabis use affects physiological processes such as synaptic pruning due to the effects of this system on neurotransmitters like glutamate and dopamine leading to long-term behavioral and psychological consequences. Additionally, evidence that dysfunctions in the endocannabinoid system play a role in the etiology of schizophrenia suggests that cannabis affects the disease process by worsening existing dysfunctions in this system. Understanding the relationship between cannabis use and the development of psychosis and underlying neurobiological mechanisms will help to identify new treatment targets, and develop appropriate preventive approaches. Keywords: Cannabis Abuse, Psychotic Disorders, Schizophrenia, Endocannabinoids, Neurobiology.
Collapse
|
28
|
Black T, Baccetto SL, Barnard IL, Finch E, McElroy DL, Austin-Scott FVL, Greba Q, Michel D, Zagzoog A, Howland JG, Laprairie RB. Characterization of cannabinoid plasma concentration, maternal health, and cytokine levels in a rat model of prenatal Cannabis smoke exposure. Sci Rep 2023; 13:21070. [PMID: 38030657 PMCID: PMC10687022 DOI: 10.1038/s41598-023-47861-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/19/2023] [Indexed: 12/01/2023] Open
Abstract
Cannabis sativa has gained popularity as a "natural substance", leading many to falsely assume that it is not harmful. This assumption has been documented amongst pregnant mothers, many of whom consider Cannabis use during pregnancy as benign. The purpose of this study was to validate a Cannabis smoke exposure model in pregnant rats by determining the plasma levels of cannabinoids and associated metabolites in the dams after exposure to either Cannabis smoke or injected cannabinoids. Maternal and fetal cytokine and chemokine profiles were also assessed after exposure. Pregnant Sprague-Dawley rats were treated daily from gestational day 6-20 with either room air, i.p. vehicle, inhaled high-Δ9-tetrahydrocannabinol (THC) (18% THC, 0.1% cannabidiol [CBD]) smoke, inhaled high-CBD (0.7% THC, 13% CBD) smoke, 3 mg/kg i.p. THC, or 10 mg/kg i.p. CBD. Our data reveal that THC and CBD, but not their metabolites, accumulate in maternal plasma after repeated exposures. Injection of THC or CBD was associated with fewer offspring and increased uterine reabsorption events. For cytokines and chemokines, injection of THC or CBD up-regulated several pro-inflammatory cytokines compared to control or high-THC smoke or high-CBD smoke in placental and fetal brain tissue, whereas smoke exposure was generally associated with reduced cytokine and chemokine concentrations in placental and fetal brain tissue compared to controls. These results support existing, but limited, knowledge on how different routes of administration contribute to inconsistent manifestations of cannabinoid-mediated effects on pregnancy. Smoked Cannabis is still the most common means of human consumption, and more preclinical investigation is needed to determine the effects of smoke inhalation on developmental and behavioural trajectories.
Collapse
Affiliation(s)
- Tallan Black
- College of Pharmacy and Nutrition, University of Saskatchewan, 3B36, Health Sciences Building, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Health Sciences Building, 107 Wiggins Rd, Saskatoon, SK, S7N 5E5, Canada
| | - Sarah L Baccetto
- College of Pharmacy and Nutrition, University of Saskatchewan, 3B36, Health Sciences Building, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Health Sciences Building, 107 Wiggins Rd, Saskatoon, SK, S7N 5E5, Canada
| | - Ilne L Barnard
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Health Sciences Building, 107 Wiggins Rd, Saskatoon, SK, S7N 5E5, Canada
| | - Emma Finch
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Health Sciences Building, 107 Wiggins Rd, Saskatoon, SK, S7N 5E5, Canada
| | - Dan L McElroy
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Health Sciences Building, 107 Wiggins Rd, Saskatoon, SK, S7N 5E5, Canada
| | - Faith V L Austin-Scott
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Health Sciences Building, 107 Wiggins Rd, Saskatoon, SK, S7N 5E5, Canada
| | - Quentin Greba
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Health Sciences Building, 107 Wiggins Rd, Saskatoon, SK, S7N 5E5, Canada
| | - Deborah Michel
- College of Pharmacy and Nutrition, University of Saskatchewan, 3B36, Health Sciences Building, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Ayat Zagzoog
- College of Pharmacy and Nutrition, University of Saskatchewan, 3B36, Health Sciences Building, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - John G Howland
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Health Sciences Building, 107 Wiggins Rd, Saskatoon, SK, S7N 5E5, Canada.
| | - Robert B Laprairie
- College of Pharmacy and Nutrition, University of Saskatchewan, 3B36, Health Sciences Building, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
- Department of Pharmacology, College of Medicine, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
29
|
Sandini TM, Onofrychuk TJ, Roebuck AJ, Hammond SA, Udenze D, Hayat S, Herdzik MA, McElroy DL, Orvold SN, Greba Q, Laprairie RB, Howland JG. Repeated Exposure to High-THC Cannabis Smoke during Gestation Alters Sex Ratio, Behavior, and Amygdala Gene Expression of Sprague Dawley Rat Offspring. eNeuro 2023; 10:ENEURO.0100-23.2023. [PMID: 37957008 PMCID: PMC10687874 DOI: 10.1523/eneuro.0100-23.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/21/2023] Open
Abstract
Because of the legalization of Cannabis in many jurisdictions and the trend of increasing Δ9-tetrahydrocannabinol (THC) content in Cannabis products, an urgent need exists to understand the impact of Cannabis use during pregnancy on fetal neurodevelopment and behavior. To this end, we exposed female Sprague Dawley rats to Cannabis smoke daily from gestational day 6 to 20 or room air. Maternal reproductive parameters, offspring behavior, and gene expression in the offspring amygdala were assessed. Body temperature was decreased in dams following smoke exposure and more fecal boli were observed in the chambers before and after smoke exposure in dams exposed to smoke. Maternal weight gain, food intake, gestational length, litter number, and litter weight were not altered by exposure to Cannabis smoke. A significant increase in the male-to-female ratio was noted in the Cannabis-exposed litters. In adulthood, male and female Cannabis smoke-exposed offspring explored the inner zone of an open field significantly less than control offspring. Gestational Cannabis smoke exposure did not affect behavior on the elevated plus maze test or social interaction test in the offspring. Cannabis offspring were better at visual pairwise discrimination and reversal learning tasks conducted in touchscreen-equipped operant conditioning chambers. Analysis of gene expression in the adult amygdala using RNA sequencing revealed subtle changes in genes related to development, cellular function, and nervous system disease in a subset of the male offspring. These results demonstrate that repeated exposure to high-THC Cannabis smoke during gestation alters maternal physiological parameters, sex ratio, and anxiety-like behaviors in the adulthood offspring.
Collapse
Affiliation(s)
- Thaisa M Sandini
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Timothy J Onofrychuk
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Andrew J Roebuck
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
- School of Liberal Arts, Yukon University, Whitehorse, Yukon Territory Y1A 5K4, Canada
| | - S Austin Hammond
- Global Institute for Food Security, Saskatoon, Saskatchewan S7N 4L8, Canada
| | - Daniel Udenze
- Next Generation Sequencing Facility, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Shahina Hayat
- Deparment of Oncology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Melissa A Herdzik
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Dan L McElroy
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Spencer N Orvold
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Quentin Greba
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Robert B Laprairie
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - John G Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| |
Collapse
|
30
|
Pinky PD, Bloemer J, Smith WD, Du Y, Heslin RT, Setti SE, Pfitzer JC, Chowdhury K, Hong H, Bhattacharya S, Dhanasekaran M, Dityatev A, Reed MN, Suppiramaniam V. Prenatal Cannabinoid Exposure Elicits Memory Deficits Associated with Reduced PSA-NCAM Expression, Altered Glutamatergic Signaling, and Adaptations in Hippocampal Synaptic Plasticity. Cells 2023; 12:2525. [PMID: 37947603 PMCID: PMC10648717 DOI: 10.3390/cells12212525] [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: 06/21/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 11/12/2023] Open
Abstract
Cannabis is now one of the most commonly used illicit substances among pregnant women. This is particularly concerning since developmental exposure to cannabinoids can elicit enduring neurofunctional and cognitive alterations. This study investigates the mechanisms of learning and memory deficits resulting from prenatal cannabinoid exposure (PCE) in adolescent offspring. The synthetic cannabinoid agonist WIN55,212-2 was administered to pregnant rats, and a series of behavioral, electrophysiological, and immunochemical studies were performed to identify potential mechanisms of memory deficits in the adolescent offspring. Hippocampal-dependent memory deficits in adolescent PCE animals were associated with decreased long-term potentiation (LTP) and enhanced long-term depression (LTD) at hippocampal Schaffer collateral-CA1 synapses, as well as an imbalance between GluN2A- and GluN2B-mediated signaling. Moreover, PCE reduced gene and protein expression of neural cell adhesion molecule (NCAM) and polysialylated-NCAM (PSA-NCAM), which are critical for GluN2A and GluN2B signaling balance. Administration of exogenous PSA abrogated the LTP deficits observed in PCE animals, suggesting PSA mediated alterations in GluN2A- and GluN2B- signaling pathways may be responsible for the impaired hippocampal synaptic plasticity resulting from PCE. These findings enhance our current understanding of how PCE affects memory and how this process can be manipulated for future therapeutic purposes.
Collapse
Affiliation(s)
- Priyanka D. Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697, USA
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Department of Pharmaceutical and Biomedical Sciences, Touro College of Pharmacy, New York, NY 10036, USA
| | - Warren D. Smith
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Yifeng Du
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Ryan T. Heslin
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Sharay E. Setti
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Jeremiah C. Pfitzer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Kawsar Chowdhury
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Hao Hong
- Key Laboratory of Neuropsychiatric Diseases, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
- Keck Graduate Institute, School of Pharmacy and Health Sciences, Claremont Colleges, Claremont, CA 91711, USA
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 37075 Magdeburg, Germany
| | - Alexander Dityatev
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 37075 Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, 39106 Magdeburg, Germany
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
31
|
Collier AD, Abdulai AR, Leibowitz SF. Utility of the Zebrafish Model for Studying Neuronal and Behavioral Disturbances Induced by Embryonic Exposure to Alcohol, Nicotine, and Cannabis. Cells 2023; 12:2505. [PMID: 37887349 PMCID: PMC10605371 DOI: 10.3390/cells12202505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
It is estimated that 5% of pregnant women consume drugs of abuse during pregnancy. Clinical research suggests that intake of drugs during pregnancy, such as alcohol, nicotine and cannabis, disturbs the development of neuronal systems in the offspring, in association with behavioral disturbances early in life and an increased risk of developing drug use disorders. After briefly summarizing evidence in rodents, this review focuses on the zebrafish model and its inherent advantages for studying the effects of embryonic exposure to drugs of abuse on behavioral and neuronal development, with an emphasis on neuropeptides known to promote drug-related behaviors. In addition to stimulating the expression and density of peptide neurons, as in rodents, zebrafish studies demonstrate that embryonic drug exposure has marked effects on the migration, morphology, projections, anatomical location, and peptide co-expression of these neurons. We also describe studies using advanced methodologies that can be applied in vivo in zebrafish: first, to demonstrate a causal relationship between the drug-induced neuronal and behavioral disturbances and second, to discover underlying molecular mechanisms that mediate these effects. The zebrafish model has great potential for providing important information regarding the development of novel and efficacious therapies for ameliorating the effects of early drug exposure.
Collapse
Affiliation(s)
| | | | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
32
|
Blum K, Ashford JW, Kateb B, Sipple D, Braverman E, Dennen CA, Baron D, Badgaiyan R, Elman I, Cadet JL, Thanos PK, Hanna C, Bowirrat A, Modestino EJ, Yamamoto V, Gupta A, McLaughlin T, Makale M, Gold MS. Dopaminergic dysfunction: Role for genetic & epigenetic testing in the new psychiatry. J Neurol Sci 2023; 453:120809. [PMID: 37774561 DOI: 10.1016/j.jns.2023.120809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/02/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023]
Abstract
Reward Deficiency Syndrome (RDS), particularly linked to addictive disorders, costs billions of dollars globally and has resulted in over one million deaths in the United States (US). Illicit substance use has been steadily rising and in 2021 approximately 21.9% (61.2 million) of individuals living in the US aged 12 or older had used illicit drugs in the past year. However, only 1.5% (4.1 million) of these individuals had received any substance use treatment. This increase in use and failure to adequately treat or provide treatment to these individuals resulted in 106,699 overdose deaths in 2021 and increased in 2022. This article presents an alternative non-pharmaceutical treatment approach tied to gene-guided therapy, the subject of many decades of research. The cornerstone of this paradigm shift is the brain reward circuitry, brain stem physiology, and neurotransmitter deficits due to the effects of genetic and epigenetic insults on the interrelated cascade of neurotransmission and the net release of dopamine at the Ventral Tegmental Area -Nucleus Accumbens (VTA-NAc) reward site. The Genetic Addiction Risk Severity (GARS) test and pro-dopamine regulator nutraceutical KB220 were combined to induce "dopamine homeostasis" across the brain reward circuitry. This article aims to encourage four future actionable items: 1) the neurophysiologically accurate designation of, for example, "Hyperdopameism /Hyperdopameism" to replace the blaming nomenclature like alcoholism; 2) encouraging continued research into the nature of dysfunctional brainstem neurotransmitters across the brain reward circuitry; 3) early identification of people at risk for all RDS behaviors as a brain check (cognitive testing); 4) induction of dopamine homeostasis using "precision behavioral management" along with the coupling of GARS and precision Kb220 variants; 5) utilization of promising potential treatments include neuromodulating modalities such as Transmagnetic stimulation (TMS) and Deep Brain Stimulation(DBS), which target different areas of the neural circuitry involved in addiction and even neuroimmune agents like N-acetyl-cysteine.
Collapse
Affiliation(s)
- Kenneth Blum
- Division of Addiction Research & Education, Center for Exercise, Sports and Mental Health, Western University Health Sciences, Pomona, CA, USA; The Kenneth Blum Behavioral & Neurogenetic Institute, LLC., Austin, TX, USA; Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel, Israel.
| | - J Wesson Ashford
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA, USA; War Related Illness & Injury Study Center, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Babak Kateb
- Brain Mapping Foundation, Los Angeles, CA, USA; National Center for Nanobioelectronic, Los Angeles, CA, USA; Brain Technology and Innovation Park, Los Angeles, CA, USA
| | | | - Eric Braverman
- The Kenneth Blum Behavioral & Neurogenetic Institute, LLC., Austin, TX, USA
| | - Catherine A Dennen
- Department of Family Medicine, Jefferson Health Northeast, Philadelphia, PA, USA
| | - David Baron
- Division of Addiction Research & Education, Center for Exercise, Sports and Mental Health, Western University Health Sciences, Pomona, CA, USA
| | - Rajendra Badgaiyan
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, San Antonio, TX, USA; Long School of Medicine, University of Texas Medical Center, San Antonio, TX, USA
| | - Igor Elman
- Center for Pain and the Brain (PAIN Group), Department of Anesthesiology, Critical Care & Pain Medicine, Boston Children's Hospital, Waltham, MA, USA; Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH National Institute on Drug Abuse, Bethesda, MD, USA
| | - Panayotis K Thanos
- Department of Psychology & Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Colin Hanna
- Department of Psychology & Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Abdalla Bowirrat
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel, Israel
| | | | - Vicky Yamamoto
- Brain Mapping Foundation, Los Angeles, CA, USA; National Center for Nanobioelectronic, Los Angeles, CA, USA; Brain Technology and Innovation Park, Los Angeles, CA, USA; Society for Brain Mapping and Therapeutics, Los Angeles, CA, USA; USC-Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | - Thomas McLaughlin
- Division of Reward Deficiency Research, Reward Deficiency Syndrome Clinics of America, Austin, TX, USA
| | - Mlan Makale
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
| | - Mark S Gold
- Department of Psychiatry, Washington College of Medicine, St. Louis, MO, USA
| |
Collapse
|
33
|
Sarikahya MH, Cousineau SL, De Felice M, Szkudlarek HJ, Wong KKW, DeVuono MV, Lee K, Rodríguez-Ruiz M, Gummerson D, Proud E, Ng THJ, Hudson R, Jung T, Hardy DB, Yeung KKC, Schmid S, Rushlow W, Laviolette SR. Prenatal THC exposure induces long-term, sex-dependent cognitive dysfunction associated with lipidomic and neuronal pathology in the prefrontal cortex-hippocampal network. Mol Psychiatry 2023; 28:4234-4250. [PMID: 37525013 DOI: 10.1038/s41380-023-02190-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 08/02/2023]
Abstract
With increasing maternal cannabis use, there is a need to investigate the lasting impact of prenatal exposure to Δ9-tetrahydrocannabinol (THC), the main psychotropic compound in cannabis, on cognitive/memory function. The endocannabinoid system (ECS), which relies on polyunsaturated fatty acids (PUFAs) to function, plays a crucial role in regulating prefrontal cortical (PFC) and hippocampal network-dependent behaviors essential for cognition and memory. Using a rodent model of prenatal cannabis exposure (PCE), we report that male and female offspring display long-term deficits in various cognitive domains. However, these phenotypes were associated with highly divergent, sex-dependent mechanisms. Electrophysiological recordings revealed hyperactive PFC pyramidal neuron activity in both males and females, but hypoactivity in the ventral hippocampus (vHIPP) in males, and hyperactivity in females. Further, cortical oscillatory activity states of theta, alpha, delta, beta, and gamma bandwidths were strongly sex divergent. Moreover, protein expression analyses at postnatal day (PD)21 and PD120 revealed primarily PD120 disturbances in dopamine D1R/D2 receptors, NMDA receptor 2B, synaptophysin, gephyrin, GAD67, and PPARα selectively in the PFC and vHIPP, in both regions in males, but only the vHIPP in females. Lastly, using matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS), we identified region-, age-, and sex-specific deficiencies in specific neural PUFAs, namely docosahexaenoic acid (DHA) and arachidonic acid (ARA), and related metabolites, in the PFC and hippocampus (ventral/dorsal subiculum, and CA1 regions). This study highlights several novel, long-term and sex-specific consequences of PCE on PFC-hippocampal circuit dysfunction and the potential role of specific PUFA signaling abnormalities underlying these pathological outcomes.
Collapse
Affiliation(s)
- Mohammed H Sarikahya
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Samantha L Cousineau
- Departments of Chemistry and Biochemistry, Western University, London, Ontario, N6A 3K7, Canada
| | - Marta De Felice
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Hanna J Szkudlarek
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Karen K W Wong
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Marieka V DeVuono
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Kendrick Lee
- Departments of Physiology and Pharmacology and Obstetrics and Gynaecology, Western University, London, Ontario, N6A 5C1, Canada
- Children's Health Research Institute, St. Josephs Health Care,, London, Ontario, N6C 2R5, Canada
| | - Mar Rodríguez-Ruiz
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Dana Gummerson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Emma Proud
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Tsun Hay Jason Ng
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Roger Hudson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Tony Jung
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Daniel B Hardy
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
- Departments of Physiology and Pharmacology and Obstetrics and Gynaecology, Western University, London, Ontario, N6A 5C1, Canada
- Children's Health Research Institute, St. Josephs Health Care,, London, Ontario, N6C 2R5, Canada
| | - Ken K-C Yeung
- Departments of Chemistry and Biochemistry, Western University, London, Ontario, N6A 3K7, Canada
| | - Susanne Schmid
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
- Department of Psychology, Western University, London, Ontario, N6A 3K7, Canada
| | - Walter Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
- Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, N6C 2R5, Canada
- Department of Psychiatry, Western University, London, Ontario, N6A 3K7, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada.
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada.
- Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, N6C 2R5, Canada.
- Department of Psychiatry, Western University, London, Ontario, N6A 3K7, Canada.
| |
Collapse
|
34
|
Motamedi S, Amleshi RS, Javar BA, Shams P, Kohlmeier KA, Shabani M. Cannabis during pregnancy: A way to transfer an impairment to later life. Birth Defects Res 2023; 115:1327-1344. [PMID: 37318343 DOI: 10.1002/bdr2.2207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/11/2023] [Accepted: 05/20/2023] [Indexed: 06/16/2023]
Abstract
Epidemiological studies examining the influence of cannabis across the lifespan show that exposure to cannabis during gestation or during the perinatal period is associated with later-life mental health issues that manifest during childhood, adolescence, and adulthood. The risk of later-life negative outcomes following early exposure is particularly high in persons who have specific genetic variants, implying that cannabis usage interacts with genetics to heighten mental health risks. Prenatal and perinatal exposure to psychoactive components has been shown in animal research to be associated with long-term effects on neural systems relevant to psychiatric and substance use disorders. The long-term molecular, epigenetic, electrophysiological, and behavioral consequences of prenatal and perinatal exposure to cannabis are discussed in this article. Animal and human studies, as well as in vivo neuroimaging methods, are used to provide insights into the changes induced in the brain by cannabis. Here, based on the literature from both animal models and humans, it can be concluded that prenatal cannabis exposure alters the developmental route of several neuronal regions with correlated functional consequences evidenced as changes in social behavior and executive functions throughout life.
Collapse
Affiliation(s)
- Sina Motamedi
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Reza Saboori Amleshi
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Behnoush Akbari Javar
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
- Health Foresight and Innovation Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Parisa Shams
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
35
|
Castelli V, Lavanco G, D’Amico C, Feo S, Tringali G, Kuchar M, Cannizzaro C, Brancato A. CBD enhances the cognitive score of adolescent rats prenatally exposed to THC and fine-tunes relevant effectors of hippocampal plasticity. Front Pharmacol 2023; 14:1237485. [PMID: 37583903 PMCID: PMC10424934 DOI: 10.3389/fphar.2023.1237485] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/20/2023] [Indexed: 08/17/2023] Open
Abstract
Introduction: An altered neurodevelopmental trajectory associated with prenatal exposure to ∆-9-tetrahydrocannabinol (THC) leads to aberrant cognitive processing through a perturbation in the effectors of hippocampal plasticity in the juvenile offspring. As adolescence presents a unique window of opportunity for "brain reprogramming", we aimed at assessing the role of the non-psychoactive phytocannabinoid cannabidiol (CBD) as a rescue strategy to temper prenatal THC-induced harm. Methods: To this aim, Wistar rats prenatally exposed to THC (2 mg/kg s.c.) or vehicle (gestational days 5-20) were tested for specific indexes of spatial and configural memory in the reinforcement-motivated Can test and in the aversion-driven Barnes maze test during adolescence. Markers of hippocampal excitatory plasticity and endocannabinoid signaling-NMDAR subunits NR1 and 2A-, mGluR5-, and their respective scaffold proteins PSD95- and Homer 1-; CB1R- and the neuromodulatory protein HINT1 mRNA levels were evaluated. CBD (40 mg/kg i.p.) was administered to the adolescent offspring before the cognitive tasks. Results: The present results show that prenatal THC impairs hippocampal memory functions and the underlying synaptic plasticity; CBD is able to mitigate cognitive impairment in both reinforcement- and aversion-related tasks and the neuroadaptation of hippocampal excitatory synapses and CB1R-related signaling. Discussion: While this research shows CBD potential in dampening prenatal THC-induced consequences, we point out the urgency to curb cannabis use during pregnancy in order to avoid detrimental bio-behavioral outcomes in the offspring.
Collapse
Affiliation(s)
- Valentina Castelli
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Gianluca Lavanco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties of Excellence “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Cesare D’Amico
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies and ATEN Center, University of Palermo, Palermo, Italy
| | - Salvatore Feo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies and ATEN Center, University of Palermo, Palermo, Italy
| | - Giuseppe Tringali
- Pharmacology Section, Department of Healthcare Surveillance and Bioethics, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome, Italy
| | - Martin Kuchar
- Forensic Laboratory of Biologically Active Compounds, Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague, Czechia
- Psychedelics Research Centre, National Institute of Mental Health, Prague, Czechia
| | - Carla Cannizzaro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Brancato
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties of Excellence “G. D’Alessandro”, University of Palermo, Palermo, Italy
| |
Collapse
|
36
|
Swenson K. Cannabis for morning sickness: areas for intervention to decrease cannabis consumption during pregnancy. J Cannabis Res 2023; 5:22. [PMID: 37330589 DOI: 10.1186/s42238-023-00184-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 04/30/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND Cannabis use during pregnancy is increasing, with 19-22% of patients testing positive at delivery in Colorado and California. Patients report using cannabis to alleviate their nausea and vomiting, anxiety, and pain. However, preclinical and clinical data highlight harmful effects to offspring physiology and behavior following fetal cannabis exposure. This narrative review identifies potential areas for intervention to decrease cannabis consumption during pregnancy. METHODS A combination of keywords, including "cannabis", "cannabis", "weed", "pregnancy", "morning sickness", "child protective services", and "budtender" were searched in databases such as PubMed and Google Scholar, as well as in social media forums, governmental webpages, and other publicly available sources. RESULTS The literature search identified several areas for intervention to reduce cannabis use during pregnancy, including physician and pharmacist training, engagement with pregnant patients, regulation of dispensary workers, and the role of child protective services. DISCUSSION This comprehensive review identifies multiple areas for improvement to benefit pregnant patients. Recommendations are independent and can be implemented simultaneously by the identified groups. Limitations of this research includes the relatively limited availability of data focused specifically on cannabis consumption during pregnancy and the complexity of the sociopolitical field of substance use during pregnancy. CONCLUSIONS Cannabis consumption during pregnancy is increasing and causes harm to the developing fetus. To educate pregnant patients about these risks, we must address the gaps in education from multiple contact points.
Collapse
Affiliation(s)
- Karli Swenson
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, 12800 E 19th Avenue, RC1 North MS 8313, Aurora, CO, 80045, USA.
| |
Collapse
|
37
|
Gao W, Pan T, Fan G, Cui J, Wang T, Huang N, Jiang C, Ma L, Wang F, Liu X, Le Q. Enhanced heroin analgesic effect in male offspring of sires who self-administered heroin. Front Pharmacol 2023; 14:1211897. [PMID: 37388448 PMCID: PMC10303812 DOI: 10.3389/fphar.2023.1211897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/30/2023] [Indexed: 07/01/2023] Open
Abstract
Introduction: A growing body of evidence suggests that parental substance abuse, even prior to conception, may induce phenotypic changes in offspring. Parental opioid exposure has been shown to affect developmental processes, induce memory deficits, and lead to psycho-emotional disorders in offspring. However, how parental, especially paternal, chronic drug exposure affects offspring remains unexplored. Methods: Adult male rats were subjected to 31 days of heroin self-administration followed by mating with naïve females. Litter size and body weight of F1 offspring were recorded. Object-based attention tests, cocaine self-administration tests, and hot plate tests were used to test for potential effects of chronic paternal heroin seeking on cognition, reward, or analgesic sensitivity in the offspring. Results: Body weight and litter size of the heroin F1 generation were not altered compared to the saline F1 generation. Furthermore, paternal chronic heroin self-administration experience had no significant effect on object-based attention tests or cocaine self-administration behavior in either sex. However, in the hot plate test, although no difference in basal latency was found between the two groups in either sex, a significant increase in the analgesic effect of heroin was observed in the male heroin F1 generation. Conclusions: Taken together, these data provide evidence that paternal chronic heroin self-administration experience could sex-dimorphically increase the analgesic effect of heroin in male offspring, but had no significant effect on response to cocaine reinforcement or attentional behavior.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lan Ma
- *Correspondence: Qiumin Le, ; Lan Ma,
| | | | | | - Qiumin Le
- *Correspondence: Qiumin Le, ; Lan Ma,
| |
Collapse
|
38
|
Haney M, Vallée M, Fabre S, Collins Reed S, Zanese M, Campistron G, Arout CA, Foltin RW, Cooper ZD, Kearney-Ramos T, Metna M, Justinova Z, Schindler C, Hebert-Chatelain E, Bellocchio L, Cathala A, Bari A, Serrat R, Finlay DB, Caraci F, Redon B, Martín-García E, Busquets-Garcia A, Matias I, Levin FR, Felpin FX, Simon N, Cota D, Spampinato U, Maldonado R, Shaham Y, Glass M, Thomsen LL, Mengel H, Marsicano G, Monlezun S, Revest JM, Piazza PV. Signaling-specific inhibition of the CB 1 receptor for cannabis use disorder: phase 1 and phase 2a randomized trials. Nat Med 2023; 29:1487-1499. [PMID: 37291212 PMCID: PMC10287566 DOI: 10.1038/s41591-023-02381-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 05/01/2023] [Indexed: 06/10/2023]
Abstract
Cannabis use disorder (CUD) is widespread, and there is no pharmacotherapy to facilitate its treatment. AEF0117, the first of a new pharmacological class, is a signaling-specific inhibitor of the cannabinoid receptor 1 (CB1-SSi). AEF0117 selectively inhibits a subset of intracellular effects resulting from Δ9-tetrahydrocannabinol (THC) binding without modifying behavior per se. In mice and non-human primates, AEF0117 decreased cannabinoid self-administration and THC-related behavioral impairment without producing significant adverse effects. In single-ascending-dose (0.2 mg, 0.6 mg, 2 mg and 6 mg; n = 40) and multiple-ascending-dose (0.6 mg, 2 mg and 6 mg; n = 24) phase 1 trials, healthy volunteers were randomized to ascending-dose cohorts (n = 8 per cohort; 6:2 AEF0117 to placebo randomization). In both studies, AEF0117 was safe and well tolerated (primary outcome measurements). In a double-blind, placebo-controlled, crossover phase 2a trial, volunteers with CUD were randomized to two ascending-dose cohorts (0.06 mg, n = 14; 1 mg, n = 15). AEF0117 significantly reduced cannabis' positive subjective effects (primary outcome measurement, assessed by visual analog scales) by 19% (0.06 mg) and 38% (1 mg) compared to placebo (P < 0.04). AEF0117 (1 mg) also reduced cannabis self-administration (P < 0.05). In volunteers with CUD, AEF0117 was well tolerated and did not precipitate cannabis withdrawal. These data suggest that AEF0117 is a safe and potentially efficacious treatment for CUD.ClinicalTrials.gov identifiers: NCT03325595 , NCT03443895 and NCT03717272 .
Collapse
Affiliation(s)
- Margaret Haney
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Monique Vallée
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | | | - Stephanie Collins Reed
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | | | | | - Caroline A Arout
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Richard W Foltin
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | - Ziva D Cooper
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
- University of California, Los Angeles, Los Angeles, CA, USA
| | - Tonisha Kearney-Ramos
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | | | - Zuzana Justinova
- Behavioral Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, USA
| | - Charles Schindler
- Behavioral Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, USA
| | | | - Luigi Bellocchio
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | - Adeline Cathala
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | | | | | - David B Finlay
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Filippo Caraci
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
- Department of Drug and Health Sciences, University of Catania, Italy, and Oasi Research Institute-IRCCS, Unit of Translational Neuropharmacology, Troina, Italy
| | - Bastien Redon
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
- Basic Neuroscience Department, Université de Genève, Genève, Switzerland
| | - Elena Martín-García
- Laboratory of Neuropharmacology, Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain
| | - Arnau Busquets-Garcia
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Programme, IMIM Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Isabelle Matias
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | - Frances R Levin
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY, USA
| | | | - Nicolas Simon
- Aix Marseille Univ, APHM, INSERM, IRD, SESSTIM, Hop Sainte Marguerite, Service de Pharmacologie Clinique, Marseille, France
| | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | | | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain
| | - Yavin Shaham
- Behavioral Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, USA
| | - Michelle Glass
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | | | | | | | | | | | | |
Collapse
|
39
|
Corsi S, Scheggi S, Pardu A, Braccagni G, Caruso D, Cioffi L, Diviccaro S, Gentile M, Fanni S, Stancampiano R, Gambarana C, Melcangi RC, Frau R, Carta M. Pregnenolone for the treatment of L-DOPA-induced dyskinesia in Parkinson's disease. Exp Neurol 2023; 363:114370. [PMID: 36878398 DOI: 10.1016/j.expneurol.2023.114370] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/02/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023]
Abstract
Growing preclinical and clinical evidence highlights neurosteroid pathway imbalances in Parkinson's Disease (PD) and L-DOPA-induced dyskinesias (LIDs). We recently reported that 5α-reductase (5AR) inhibitors dampen dyskinesias in parkinsonian rats; however, unraveling which specific neurosteroid mediates this effect is critical to optimize a targeted therapy. Among the 5AR-related neurosteroids, striatal pregnenolone has been shown to be increased in response to 5AR blockade and decreased after 6-OHDA lesions in the rat PD model. Moreover, this neurosteroid rescued psychotic-like phenotypes by exerting marked antidopaminergic activity. In light of this evidence, we investigated whether pregnenolone might dampen the appearance of LIDs in parkinsonian drug-naïve rats. We tested 3 escalating doses of pregnenolone (6, 18, 36 mg/kg) in 6-OHDA-lesioned male rats and compared the behavioral, neurochemical, and molecular outcomes with those induced by the 5AR inhibitor dutasteride, as positive control. The results showed that pregnenolone dose-dependently countered LIDs without affecting L-DOPA-induced motor improvements. Post-mortem analyses revealed that pregnenolone significantly prevented the increase of validated striatal markers of dyskinesias, such as phospho-Thr-34 DARPP-32 and phospho-ERK1/2, as well as D1-D3 receptor co-immunoprecipitation in a fashion similar to dutasteride. Moreover, the antidyskinetic effect of pregnenolone was paralleled by reduced striatal levels of BDNF, a well-established factor associated with the development of LIDs. In support of a direct pregnenolone effect, LC/MS-MS analyses revealed that striatal pregnenolone levels strikingly increased after the exogenous administration, with no significant alterations in downstream metabolites. All these data suggest pregnenolone as a key player in the antidyskinetic properties of 5AR inhibitors and highlight this neurosteroid as an interesting novel tool to target LIDs in PD.
Collapse
Affiliation(s)
- Sara Corsi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Simona Scheggi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Alessandra Pardu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Giulia Braccagni
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Lucia Cioffi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Silvia Diviccaro
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Mauro Gentile
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Silvia Fanni
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy; Basal Ganglia Pathophysiology Unit, Department Experimental Medical Science, Lund University, Sweden
| | | | - Carla Gambarana
- Department of Molecular and Developmental Medicine, University of Siena, Siena, SI, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, MI, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy; "Guy Everett Laboratory", Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy.
| |
Collapse
|
40
|
Bockmann EC, Brito R, Madeira LF, da Silva Sampaio L, de Melo Reis RA, França GR, Calaza KDC. The Role of Cannabinoids in CNS Development: Focus on Proliferation and Cell Death. Cell Mol Neurobiol 2023; 43:1469-1485. [PMID: 35925507 PMCID: PMC11412427 DOI: 10.1007/s10571-022-01263-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/15/2022] [Indexed: 11/26/2022]
Abstract
The active principles of Cannabis sativa are potential treatments for several diseases, such as pain, seizures and anorexia. With the increase in the use of cannabis for medicinal purposes, a more careful assessment of the possible impacts on embryonic development becomes necessary. Surveys indicate that approximately 3.9% of pregnant women use cannabis in a recreational and/or medicinal manner. However, although the literature has already described the presence of endocannabinoid system components since the early stages of CNS development, many of their physiological effects during this stage have not yet been established. Moreover, it is still uncertain how the endocannabinoid system can be altered in terms of cell proliferation and cell fate, neural migration, neural differentiation, synaptogenesis and particularly cell death. In relation to cell death in the CNS, knowledge about the effects of cannabinoids is scarce. Thus, the present work aims to review the role of the endocannabinoid system in different aspects of CNS development and discuss possible side effects or even opportunities for treating some conditions in the development of this tissue.
Collapse
Affiliation(s)
- Eduardo Cosendey Bockmann
- Instituto de Biologia, Departamento de Neurobiologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Rafael Brito
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Lucianne Fragel Madeira
- Instituto de Biologia, Departamento de Neurobiologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Luzia da Silva Sampaio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Augusto de Melo Reis
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme Rapozeiro França
- Departamento de Ciências Fisiológicas, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Karin da Costa Calaza
- Instituto de Biologia, Departamento de Neurobiologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
41
|
Serra V, Aroni S, Bortolato M, Frau R, Melis M. Endocannabinoid-dependent decrease of GABAergic transmission on dopaminergic neurons is associated with susceptibility to cocaine stimulant effects in pre-adolescent male MAOA hypomorphic mice exposed to early life stress. Neuropharmacology 2023; 233:109548. [PMID: 37080337 DOI: 10.1016/j.neuropharm.2023.109548] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/04/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Vulnerability to cocaine use disorder depends upon a combination of genetic and environmental risk factors. While early life adversity is a critical environmental vulnerability factor for drug misuse, allelic variants of the monoamine oxidase A (MAOA) gene have been shown to moderate its influence on the risk of drug-related problems. However, data on the interactions between MAOA variants and early life stress (ES) with respect to predisposition to cocaine abuse are limited. Here, we show that a mouse model capturing the interaction of genetic (low-activity alleles of the Maoa gene; MAOANeo) and environmental (i.e., ES) vulnerability factors displays an increased sensitivity to repeated in vivo cocaine psychomotor stimulant actions associated with a reduction of GABAA receptor-mediated inhibition of dopamine neurons of the ventral tegmental area (VTA). Depolarization-induced suppression of inhibition (DSI), a 2-arachidonoylglycerol (2AG)-dependent form of short-term plasticity, also becomes readily expressed by dopamine neurons from male MAOANeo ES mice repeatedly treated with cocaine. The activation of either dopamine D2 or CB1 receptors contributes to cocaine-induced DSI expression, decreased GABA synaptic efficacy, and hyperlocomotion. Next, in vivo pharmacological enhancement of 2AG signaling during repeated cocaine exposure occludes its actions both in vivo and ex vivo. This data extends our knowledge of the multifaceted sequelae imposed by this gene-environment interaction in VTA dopamine neurons of male pre-adolescent mice and contributes to our understanding of neural mechanisms of vulnerability for early onset cocaine use.
Collapse
Affiliation(s)
- Valeria Serra
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy
| | - Sonia Aroni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, 84112, USA
| | - Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy.
| |
Collapse
|
42
|
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
|
43
|
Rouzer SK, Gutierrez J, Larin KV, Miranda RC. Alcohol & cannabinoid co-use: Implications for impaired fetal brain development following gestational exposure. Exp Neurol 2023; 361:114318. [PMID: 36627039 PMCID: PMC9892278 DOI: 10.1016/j.expneurol.2023.114318] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
Alcohol and marijuana are two of the most consumed psychoactive substances by pregnant people, and independently, both substances have been associated with lifelong impacts on fetal neurodevelopment. Importantly, individuals of child-bearing age are increasingly engaging in simultaneous alcohol and cannabinoid (SAC) use, which amplifies each drug's pharmacodynamic effects and increases craving for both substances. However, to date, investigations of prenatal polysubstance use are notably limited in both human and non-human populations. In this review paper, we will address what is currently known about combined exposure to these substances, both directly and prenatally, and identify shared prenatal targets from single-exposure paradigms that may highlight susceptible neurobiological mechanisms for future investigation and therapeutic intervention. Finally, we conclude this manuscript by discussing factors that we feel are essential in the consideration and experimental design of future preclinical SAC studies.
Collapse
Affiliation(s)
- Siara Kate Rouzer
- Department of Neuroscience & Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX 77807, United States.
| | - Jessica Gutierrez
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, United States
| | - Kirill V Larin
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, United States
| | - Rajesh C Miranda
- Department of Neuroscience & Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX 77807, United States
| |
Collapse
|
44
|
Santoni M, Sagheddu C, Serra V, Mostallino R, Castelli MP, Pisano F, Scherma M, Fadda P, Muntoni AL, Zamberletti E, Rubino T, Melis M, Pistis M. Maternal immune activation impairs endocannabinoid signaling in the mesolimbic system of adolescent male offspring. Brain Behav Immun 2023; 109:271-284. [PMID: 36746342 DOI: 10.1016/j.bbi.2023.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/09/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023] Open
Abstract
Prenatal infections can increase the risk of developing psychiatric disorders such as schizophrenia in the offspring, especially when combined with other postnatal insults. Here, we tested, in a rat model of prenatal immune challenge by the viral mimic polyriboinosinic-polyribocytidilic acid, whether maternal immune activation (MIA) affects the endocannabinoid system and endocannabinoid-mediated modulation of dopamine functions. Experiments were performed during adolescence to assess i) the behavioral endophenotype (locomotor activity, plus maze, prepulse inhibition of startle reflex); ii) the locomotor activity in response to Δ9-Tetrahydrocannabinol (THC) and iii) the properties of ventral tegmental area (VTA) dopamine neurons in vivo and their response to THC; iv) endocannabinoid-mediated synaptic plasticity in VTA dopamine neurons; v) the expression of cannabinoid receptors and enzymes involved in endocannabinoid synthesis and catabolism in mesolimbic structures and vi) MIA-induced neuroinflammatory scenario evaluated by measurements of levels of cytokine and neuroinflammation markers. We revealed that MIA offspring displayed an altered locomotor activity in response to THC, a higher bursting activity of VTA dopamine neurons and a lack of response to cumulative doses of THC. Consistently, MIA adolescence offspring showed an enhanced 2-arachidonoylglycerol-mediated synaptic plasticity and decreased monoacylglycerol lipase activity in mesolimbic structures. Moreover, they displayed a higher expression of cyclooxygenase 2 (COX-2) and ionized calcium-binding adaptor molecule 1 (IBA-1), associated with latent inflammation and persistent microglia activity. In conclusion, we unveiled neurobiological mechanisms whereby inflammation caused by MIA influences the proper development of endocannabinoid signaling that negatively impacts the dopamine system, eventually leading to psychotic-like symptoms in adulthood.
Collapse
Affiliation(s)
- Michele Santoni
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Claudia Sagheddu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Valeria Serra
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Rafaela Mostallino
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Maria Paola Castelli
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Francesco Pisano
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Maria Scherma
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Paola Fadda
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy
| | - Anna Lisa Muntoni
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy
| | - Erica Zamberletti
- Department of Biotechnology and Life Sciences and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy; Unit of Clinical Pharmacology, University Hospital, Cagliari, Italy.
| |
Collapse
|
45
|
Mulligan MK, Hamre KM. Influence of prenatal cannabinoid exposure on early development and beyond. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2023; 3:10981. [PMID: 38389825 PMCID: PMC10880766 DOI: 10.3389/adar.2023.10981] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2024]
Abstract
Public perception surrounding whether cannabis use is harmful during pregnancy often diverges greatly from the recommendations of doctors and healthcare providers. In contrast to the medical guidance of abstinence before, during, and after pregnancy, many women of reproductive age believe cannabis use during pregnancy is associated with little potential harm. Legalization and social cues support public perceptions that cannabis use during pregnancy is safe. Moreover, pregnant women may consider cannabis to be a safe alternative for treating pregnancy related ailments, including morning sickness. Compounding the problem is a lack of medical and federal guidance on safe, low, or high-risk levels of cannabis use. These issues mirror the continuing debate surrounding alcohol use and health, in particular, whether there are safe or lower risk levels of alcohol consumption during pregnancy. Clinical studies to date suffer from several limitations. First, most human studies are correlative in nature, meaning that causal associations cannot be made between in utero cannabis exposure and health and behavioral outcomes later in life. Due to obvious ethical constraints, it is not possible to randomly assign pregnant mothers to cannabis or other drug exposure conditions-a requirement needed to establish causality. In addition, clinical studies often lack quantitative information on maternal exposure (i.e., dose, frequency, and duration), include a small number of individuals, lack replication of outcome measures across cohorts, rely on self-report to establish maternal drug use, and suffer from unmeasured or residual confounding factors. Causal associations between maternal cannabis exposure and offspring outcomes are possible in preclinical cohorts but there is a large amount of heterogeneity across study designs and developmental differences between rodents and humans may limit translatability. In this review, we summarize research from human and preclinical models to provide insight into potential risks associated with prenatal cannabinoid exposure (PCE). Finally, we highlight gaps in knowledge likely to contribute to the growing divide between medical guidance and public attitudes regarding cannabis use during pregnancy.
Collapse
Affiliation(s)
- Megan K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States
| | - Kristin M Hamre
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States
| |
Collapse
|
46
|
Prenatal Exposure to Δ9-Tetrahydrocannabinol Affects Hippocampus-Related Cognitive Functions in the Adolescent Rat Offspring: Focus on Specific Markers of Neuroplasticity. Pharmaceutics 2023; 15:pharmaceutics15020692. [PMID: 36840014 PMCID: PMC9963541 DOI: 10.3390/pharmaceutics15020692] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Previous evidence suggests that prenatal exposure to THC (pTHC) derails the neurodevelopmental trajectories towards a vulnerable phenotype for impaired emotional regulation and limbic memory. Here we aimed to investigate pTHC effect on hippocampus-related cognitive functions and markers of neuroplasticity in adolescent male offspring. Wistar rats were exposed to THC (2 mg/kg) from gestational day 5 to 20 and tested for spatial memory, object recognition memory and reversal learning in the reinforce-motivated Can test and in the aversion-driven Barnes maze test; locomotor activity and exploration, anxiety-like behaviour, and response to natural reward were assessed in the open field, elevated plus maze, and sucrose preference tests, respectively. The gene expression levels of NMDA NR1-2A subunits, mGluR5, and their respective scaffold proteins PSD95 and Homer1, as well as CB1R and the neuromodulatory protein HINT1, were measured in the hippocampus. pTHC offspring exhibited deficits in spatial and object recognition memory and reversal learning, increased locomotor activity, increased NR1-, decreased NR2A- and PSD95-, increased mGluR5- and Homer1-, and augmented CB1R- and HINT1-hippocampal mRNA levels. Our data shows that pTHC is associated with specific impairment in spatial cognitive processing and effectors of hippocampal neuroplasticity and suggests novel targets for future pharmacological challenges.
Collapse
|
47
|
Peng H, Li H, Wei Y, Zhang R, Chang X, Meng L, Wang K, He Q, Duan T. Effects of prenatal exposure to THC on hippocampal neural development in offspring. Toxicol Lett 2023; 374:48-56. [PMID: 36529297 DOI: 10.1016/j.toxlet.2022.12.007] [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: 11/07/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Cannabis use is a worldwide issue with the development of legalization. Prenatal exposure to Δ9-tetrahydrocannabinol (THC), the main psychoactive component of cannabis, is related to affect fetal nervous system development. In our present study, we administered THC to pregnant mice from gestational day 5.5-12.5. Differences in neuronal cell composition and organization between the two groups were found by staining sections of the offspring hippocampus at PND21. In addition, RNA-seq of hippocampal tissue also suggested differences in gene expression due to THC treatment, especially significant enrichment to neurogenesis and neural differentiation. Subsequently, the effect of THC treatment on the proliferation and differentiation capacity of neural stem cells (NSCs) was confirmed. Based on the RNA-seq results, we selected the differentially expressed transcription factor MEF2C for validation. The effect of THC treatment on NSCs differentiation was found to be regulated by knocking down the expression of MEF2C in NSCs. Considering that THC is an agonist of cannabinoid receptor (CB1R), the differentiation outcome of NSC after THC treatment was significantly rescued, by pretreating with the CB1R inhibitor Rimonabant. Notably, pretreatment with Rimonabant restored the expression of MEF2C. Taken together, the present results suggested that THC regulated the MEF2C pathway through CB1R and had an impact on hippocampal neurodevelopment.
Collapse
Affiliation(s)
- Hao Peng
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Han Li
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yingying Wei
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ruonan Zhang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinwen Chang
- Department of Assisted Reproductive Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lulu Meng
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qizhi He
- Department of Pathology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Tao Duan
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
48
|
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: 5] [Impact Index Per Article: 2.5] [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
|
49
|
Martinez Ramirez CE, Ruiz-Pérez G, Stollenwerk TM, Behlke C, Doherty A, Hillard CJ. Endocannabinoid signaling in the central nervous system. Glia 2023; 71:5-35. [PMID: 36308424 PMCID: PMC10167744 DOI: 10.1002/glia.24280] [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: 02/01/2022] [Revised: 09/02/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022]
Abstract
It is hard to overestimate the influence of the endocannabinoid signaling (ECS) system on central nervous system (CNS) function. In the 40 years since cannabinoids were found to trigger specific cell signaling cascades, studies of the ECS system continue to cause amazement, surprise, and confusion! CB1 cannabinoid receptors are expressed widely in the CNS and regulate cell-cell communication via effects on the release of both neurotransmitters and gliotransmitters. CB2 cannabinoid receptors are difficult to detect in the CNS but seem to "punch above their weight" as compounds targeting these receptors have significant effects on inflammatory state and behavior. Positive and negative allosteric modulators for both receptors have been identified and examined in preclinical studies. Concentrations of the endocannabinoid ligands, N-arachidonoylethanolamine and 2-arachidonoylglycerol (2-AG), are regulated by a combination of enzymatic synthesis and degradation and inhibitors of these processes are available and making their way into clinical trials. Importantly, ECS regulates many essential brain functions, including regulation of reward, anxiety, inflammation, motor control, and cellular development. While the field is on the cusp of preclinical discoveries providing impactful clinical and therapeutic insights into many CNS disorders, there is still much to be learned about this remarkable and versatile modulatory system.
Collapse
Affiliation(s)
- César E Martinez Ramirez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Gonzalo Ruiz-Pérez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Todd M Stollenwerk
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Christina Behlke
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ashley Doherty
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Cecilia J Hillard
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
50
|
Cáceres D, Ochoa M, González-Ortiz M, Bravo K, Eugenín J. Effects of Prenatal Cannabinoids Exposure upon Placenta and Development of Respiratory Neural Circuits. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:199-232. [PMID: 37466775 DOI: 10.1007/978-3-031-32554-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Cannabis use has risen dangerously during pregnancy in the face of incipient therapeutic use and a growing perception of safety. The main psychoactive compound of the Cannabis sativa plant is the phytocannabinoid delta-9-tetrahydrocannabinol (A-9 THC), and its status as a teratogen is controversial. THC and its endogenous analogues, anandamide (AEA) and 2-AG, exert their actions through specific receptors (eCBr) that activate intracellular signaling pathways. CB1r and CB2r, also called classic cannabinoid receptors, together with their endogenous ligands and the enzymes that synthesize and degrade them, constitute the endocannabinoid system. This system is distributed ubiquitously in various central and peripheral tissues. Although the endocannabinoid system's most studied role is controlling the release of neurotransmitters in the central nervous system, the study of long-term exposure to cannabinoids on fetal development is not well known and is vital for understanding environmental or pathological embryo-fetal or postnatal conditions. Prenatal exposure to cannabinoids in animal models has induced changes in placental and embryo-fetal organs. Particularly, cannabinoids could influence both neural and nonneural tissues and induce embryo-fetal pathological conditions in critical processes such as neural respiratory control. This review aims at the acute and chronic effects of prenatal exposure to cannabinoids on placental function and the embryo-fetal neurodevelopment of the respiratory pattern. The information provided here will serve as a theoretical framework to critically evaluate the teratogen effects of the consumption of cannabis during pregnancy.
Collapse
Affiliation(s)
- Daniela Cáceres
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Martín Ochoa
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Marcelo González-Ortiz
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
| | - Karina Bravo
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Facultad de Ingeniería, Universidad Autónoma de Chile, Providencia, Chile
| | - Jaime Eugenín
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|