1
|
Jenkins BW, Moore CF, Jantzie LL, Weerts EM. Prenatal cannabinoid exposure and the developing brain: Evidence of lasting consequences in preclinical rodent models. Neurosci Biobehav Rev 2025; 175:106207. [PMID: 40373945 DOI: 10.1016/j.neubiorev.2025.106207] [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: 02/04/2025] [Revised: 04/18/2025] [Accepted: 05/08/2025] [Indexed: 05/17/2025]
Abstract
Cannabis use by people who are pregnant is increasing. Understanding how prenatal cannabinoid exposure (PCE) affects infants and children is of high public health significance. Epidemiological studies have associated PCE with cognitive symptoms, including impaired learning, memory, attention, and executive control, and affective symptoms, including anxiety, emotional dysregulation, and social impairments, in children, adolescents, and young adults. PCE is also associated with neurobiological changes including decreased corticolimbic white matter and functional connectivity; however, the underlying mechanisms for these persisting effects remain unknown. Rodent models are essential for uncovering the effects of PCE on the developing brain. This review summarizes rodent studies focused on the cognitive and affective behavioral and neurobiological outcomes of PCE. Rodent studies have reported cognitive deficits, including impaired learning, memory, attention, and executive control, and affect-related impairments, including anxiety-like behavior, altered stress coping, social impairments, and anhedonia-like behavior, in adolescent and adult offspring. Studies have also demonstrated that PCE affects several underlying neurotransmitter systems, producing dopamine hyperactivity, glutamate and serotonin hypoactivity, and dysregulating GABA and opioid signaling. Evidence further suggests a marked difference in outcomes between males and females, with males being more susceptible to the enduring effects of PCE. However, studies that investigate female-specific outcomes or sex as a biological variable are scarce. Altogether, rodent studies provide corroborating evidence that PCE produces lasting cognitive and affective impairments underpinned by altered neurobiological mechanisms. Research is critically needed to improve our understanding of the risks associated with cannabis use during pregnancy and effects across the lifespan.
Collapse
Affiliation(s)
- Bryan W Jenkins
- Division of Behavioral Biology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr., Baltimore, MD 21224, USA
| | - Catherine F Moore
- Division of Behavioral Biology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr., Baltimore, MD 21224, USA
| | - Lauren L Jantzie
- Departments of Pediatrics, Neurosurgery, and Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA
| | - Elise M Weerts
- Division of Behavioral Biology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Dr., Baltimore, MD 21224, USA.
| |
Collapse
|
2
|
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
|
3
|
Margolis ET, Gabard‐Durnam LJ. Prenatal influences on postnatal neuroplasticity: Integrating DOHaD and sensitive/critical period frameworks to understand biological embedding in early development. INFANCY 2025; 30:e12588. [PMID: 38449347 PMCID: PMC11647198 DOI: 10.1111/infa.12588] [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: 09/02/2023] [Revised: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
Early environments can have significant and lasting effects on brain, body, and behavior across the lifecourse. Here, we address current research efforts to understand how experiences impact neurodevelopment with a new perspective integrating two well-known conceptual frameworks - the Developmental Origins of Health and Disease (DOHaD) and sensitive/critical period frameworks. Specifically, we consider how prenatal experiences characterized in the DOHaD model impact two key neurobiological mechanisms of sensitive/critical periods for adapting to and learning from the postnatal environment. We draw from both animal and human research to summarize the current state of knowledge on how particular prenatal substance exposures (psychoactive substances and heavy metals) and nutritional profiles (protein-energy malnutrition and iron deficiency) each differentially impact brain circuits' excitation/GABAergic inhibition balance and myelination. Finally, we highlight new research directions that emerge from this integrated framework, including testing how prenatal environments alter sensitive/critical period timing and learning and identifying potential promotional/buffering prenatal exposures to impact postnatal sensitive/critical periods. We hope this integrative framework considering prenatal influences on postnatal neuroplasticity will stimulate new research to understand how early environments have lasting consequences on our brains, behavior, and health.
Collapse
Affiliation(s)
- Emma T. Margolis
- Department of PsychologyNortheastern UniversityBostonMassachusettsUSA
| | | |
Collapse
|
4
|
Castelli V, Lavanco G, Tringali G, D'Amico C, Feo S, Di Bartolomeo M, D'Addario C, Kuchar M, Brancato A, Cannizzaro C. Prenatal THC exposure drives sex-specific alterations in spatial memory and hippocampal excitatory/inhibitory balance in adolescent rats. Biomed Pharmacother 2024; 181:117699. [PMID: 39571245 DOI: 10.1016/j.biopha.2024.117699] [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: 09/13/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 12/21/2024] Open
Abstract
The interaction between the main psychotropic ingredient of Cannabis, Δ⁹- tetrahydrocannabinol (THC), with the endogenous cannabinoid system (ECS) is a critical and underrated issue that deserves utmost attention. The ECS, indeed, contributes to the formation and regulation of excitatory and inhibitory (E/I) neuronal networks that in the hippocampus underly spatial memory. This study explored sex-specific consequences of prenatal exposure to THC in hippocampus-dependent memory and the underlying cellular and molecular contributors of synaptic plasticity and E/I homeostasis. Sprague Dawley dams were exposed to THC (2 mg/kg) or vehicle, from gestational day 5-20. The adolescent progeny of both sexes was tested for: spatial memory retrieval and flexibility in the Barnes Maze; mRNA expression of relevant players of hippocampal synaptic plasticity; density of cholecystokinin-positive basket cells (CCK+BCs) - a major subtype of hippocampal inhibitory interneurons; mRNA expression of the excitatory and inhibitory synaptic proteins neuroligins (Nlgns), as a proxy of synaptic efficiency. Our results show a sex-specific disruption in spatial memory retrieval and flexibility, a male-specific decrease in CCK+BCs density and increase in the expression of markers of neuroplasticity, and consistent changes in the expression of Nlgn-1 and 3 isoforms. Despite a delay in memory retrieval, flexibility of memory was spared in prenatally-THC-exposed female offspring as well as most of the markers of neuroplasticity; a sex-specific increase in CCK+BCs density, and a consistent expression of Nlgn-3 was observed. The current results highlight a major vulnerability to prenatal exposure to THC on memory processing in the male progeny, and sex-specific alterations in the E/I balance and synaptic plasticity.
Collapse
Affiliation(s)
- Valentina Castelli
- University of Palermo, Dept. of Biomedicine, Neuroscience and Advanced Diagnostics, via del Vespro 129, Palermo 90127, 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
| | - 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
| | - Cesare D'Amico
- University of Palermo, Dept. of Biomedicine, Neuroscience and Advanced Diagnostics, via del Vespro 129, Palermo 90127, Italy
| | - Salvatore Feo
- Department of Biological, Chemical and Pharmaceutical Sciences Technologies, University of Palermo, Palermo, Italy; ATEN Center, University of Palermo, Palermo, Italy
| | - Martina Di Bartolomeo
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo 64100, Italy
| | - Claudio D'Addario
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo 64100, Italy; Dept. of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Kuchar
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague, Czechia; Psychedelics Research Centre, National Institute of Mental Health, Prague, Czechia
| | - 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.
| | - Carla Cannizzaro
- University of Palermo, Dept. of Biomedicine, Neuroscience and Advanced Diagnostics, via del Vespro 129, Palermo 90127, Italy
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
McPherson C. Up in Smoke: The Impacts of Marijuana During Pregnancy. Neonatal Netw 2023; 42:222-232. [PMID: 37491043 DOI: 10.1891/nn-2022-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 07/27/2023]
Abstract
Numerous drugs ingested during pregnancy can impact the developing fetus. Although some effects are apparent at birth as overt teratogenicity or profound neonatal withdrawal, others become apparent only after a careful long-term follow-up into childhood. Shifting legal and cultural attitudes toward marijuana have led to increased use during pregnancy. This shift should prompt health care providers to carefully consider the drug's mechanism of action, its interaction with the placenta, and the potential consequences of fetal exposure. The primary psychoactive compound in marijuana is Δ9-tetrahydrocannabinol (THC), which agonizes endogenous cannabinoid receptors. Cannabinoid receptors are present in the fetal brain early in gestation and appear to have an important role in the developing central nervous system. THC crosses the placenta in sufficient quantities to raise concerns about exogenous exposure during fetal development. Robust follow-up studies suggest that marijuana use during pregnancy contributes to suboptimal fetal growth. At school age, heavy prenatal marijuana exposure predicts challenges in executive function (specifically, memory and reasoning) and externalizing behavior (e.g., hyperactivity and inattention). Memory and behavioral problems persist into early adulthood. These challenges coincide with a higher risk of heavy marijuana use in offspring. In concert with a suboptimal environment, young adults may experience a higher risk of global cognitive impairment and/or delinquency. Importantly, these adverse outcomes appear to be mitigated by postnatal factors including home environment. Ongoing studies in the modern era will be vital to enhance our understanding of the mechanisms by which THC impacts the fetus and confirm or refute knowledge regarding long-term impact. This knowledge will inform both health care providers and parents in collaborative decision-making to optimize the outcome of children.
Collapse
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
Sperm DNA methylation alterations from cannabis extract exposure are evident in offspring. Epigenetics Chromatin 2022; 15:33. [PMID: 36085240 PMCID: PMC9463823 DOI: 10.1186/s13072-022-00466-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/26/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Cannabis legalization is expanding and men are the predominant users. We have limited knowledge about how cannabis impacts sperm and whether the effects are heritable.
Results
Whole genome bisulfite sequencing (WGBS) data were generated for sperm of rats exposed to: (1) cannabis extract (CE) for 28 days, then 56 days of vehicle only (~ one spermatogenic cycle); (2) vehicle for 56 days, then 28 days of CE; or (3) vehicle only. Males were then mated with drug-naïve females to produce F1 offspring from which heart, brain, and sperm tissues underwent analyses. There were 3321 nominally significant differentially methylated CpGs in F0 sperm identified via WGBS with select methylation changes validated via bisulfite pyrosequencing. Significant methylation changes validated in F0 sperm of the exposed males at the gene 2-Phosphoxylose Phosphatase 1 (Pxylp1) were also detectable in their F1 sperm but not in controls. Changes validated in exposed F0 sperm at Metastasis Suppressor 1-Like Protein (Mtss1l) were also present in F1 hippocampal and nucleus accumbens (NAc) of the exposed group compared to controls. For Mtss1l, a significant sex-specific relationship between DNA methylation and gene expression was demonstrated in the F1 NAc. Phenotypically, rats born to CSE-exposed fathers exhibited significant cardiomegaly relative to those born to control fathers.
Conclusions
This is the first characterization of the effect of cannabis exposure on the entirety of the rat sperm methylome. We identified CE-associated methylation changes across the sperm methylome, some of which persisted despite a “washout” period. Select methylation changes validated via bisulfite pyrosequencing, and genes associated with methylation changes were involved in early developmental processes. Preconception CE exposure is associated with detectable changes in offspring DNA methylation that are functionally related to changes in gene expression and cardiomegaly.
These results support that paternal preconception exposure to cannabis can influence offspring outcomes.
Collapse
|
16
|
Dennen CA, Blum K, Bowirrat A, Khalsa J, Thanos PK, Baron D, Badgaiyan RD, Gupta A, Braverman ER, Gold MS. Neurogenetic and Epigenetic Aspects of Cannabinoids. EPIGENOMES 2022; 6:27. [PMID: 36135314 PMCID: PMC9498086 DOI: 10.3390/epigenomes6030027] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 12/04/2022] Open
Abstract
Cannabis is one of the most commonly used and abused illicit drugs in the world today. The United States (US) currently has the highest annual prevalence rate of cannabis consumption in the world, 17.9% in individuals aged 12 or older, and it is on the rise. With increasing cannabis use comes the potential for an increase in abuse, and according to the Substance Abuse and Mental Health Services Administration (SAMHSA), approximately 5.1% of Americans had Cannabis Use Disorder (CUD) in 2020. Research has shown that genetics and epigenetics play a significant role in cannabis use and CUD. In fact, approximately 50-70% of liability to CUD and 40-48% of cannabis use initiation have been found to be the result of genetic factors. Cannabis usage and CUD have also been linked to an increased risk of psychiatric disorders and Reward Deficiency Syndrome (RDS) subsets like schizophrenia, depression, anxiety, and substance use disorder. Comprehension of the genetic and epigenetic aspects of cannabinoids is necessary for future research, treatment plans, and the production of pure cannabinoid compounds, which will be essential for FDA approval. In conclusion, having a better understanding of the epigenetic and genetic underpinnings of cannabis use, CUD, and the endocannabinoid system as a whole will aid in the development of effective FDA-approved treatment therapies and the advancement of personalized medicine.
Collapse
Affiliation(s)
- Catherine A. Dennen
- Department of Family Medicine, Jefferson Health Northeast, Philadelphia, PA 19114, USA
| | - Kenneth Blum
- Division of Nutrigenomics, The Kenneth Blum Behavioral Neurogenetic Institute, Austin, TX 78701, USA
- Division of Addiction Research & Education, Center for Psychiatry, Medicine & Primary Care (Office of the Provost), Graduate College, Western University of Health Sciences, Pomona, CA 91766, USA
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary
- Department of Psychiatry, University of Vermont, Burlington, VT 05405, USA
- Department of Psychiatry, Wright University Boonshoft School of Medicine, Dayton, OH 45324, USA
| | - Abdalla Bowirrat
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Jag Khalsa
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
- Medical Consequences of Drug Abuse and Infections Branch, National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20852, USA
| | - Panayotis K. Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY 14260, USA
| | - David Baron
- Division of Addiction Research & Education, Center for Psychiatry, Medicine & Primary Care (Office of the Provost), Graduate College, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, Icahn School of Medicine Mt Sinai, New York, NY 10029, USA
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, San Antonio, TX 78229, USA
- Long School of Medicine, University of Texas Medical Center, San Antonio, TX 78229, USA
| | - Ashim Gupta
- Future Biologics, Lawrenceville, GA 30043, USA
| | - Eric R. Braverman
- Division of Nutrigenomics, The Kenneth Blum Behavioral Neurogenetic Institute, Austin, TX 78701, USA
| | - Mark S. Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
17
|
Effects of endocannabinoid system modulation on social behaviour: A systematic review of animal studies. Neurosci Biobehav Rev 2022; 138:104680. [PMID: 35513169 DOI: 10.1016/j.neubiorev.2022.104680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/09/2022]
Abstract
There is a clear link between psychiatric disorders and social behaviour, and evidence suggests the involvement of the endocannabinoid system (ECS). A systematic review of preclinical literature was conducted using MEDLINE (PubMed) and PsychINFO databases to examine whether pharmacological and/or genetic manipulations of the ECS alter social behaviours in wildtype (WT) animals or models of social impairment (SIM). Eighty studies were included. Risk of bias (RoB) was assessed using SYRCLE's RoB tool. While some variability was evident, studies most consistently found that direct cannabinoid receptor (CBR) agonism decreased social behaviours in WT animals, while indirect CBR activation via enzyme inhibition or gene-knockout increased social behaviours. Direct and, more consistently, indirect CBR activation reversed social deficits in SIM. These CBR-mediated effects were often sex- and developmental-phase-dependent and blocked by CBR antagonism. Overall, ECS enzyme inhibition may improve social behaviour in SIM, suggesting the potential usefulness of ECS enzyme inhibition as a therapeutic approach for social deficits. Future research should endeavour to elucidate ECS status in neuropsychiatric disorders characterized by social deficits.
Collapse
|
18
|
Maciel IDS, de Abreu GH, Johnson CT, Bonday R, Bradshaw HB, Mackie K, Lu HC. Perinatal CBD or THC Exposure Results in Lasting Resistance to Fluoxetine in the Forced Swim Test: Reversal by Fatty Acid Amide Hydrolase Inhibition. Cannabis Cannabinoid Res 2022; 7:318-327. [PMID: 34182795 PMCID: PMC9225394 DOI: 10.1089/can.2021.0015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Introduction: There is widespread acceptance of cannabis for medical or recreational use across the society, including pregnant women. Concerningly, numerous studies find that the developing central nervous system (CNS) is vulnerable to the detrimental effects of Δ9-tetrahydrocannabinol (THC). In contrast, almost nothing on the consequences of perinatal cannabidiol (CBD) exposure. In this study, we used mice to investigate the adult impact of perinatal cannabinoid exposure (PCE) with THC, CBD, or a 1:1 ratio of THC and CBD on behaviors. Furthermore, the lasting impact of PCE on fluoxetine sensitivity in the forced swim test (FST) was evaluated to probe neurochemical pathways interacting with the endocannabinoid system (ECS). Methods: Pregnant CD1 dams were injected subcutaneously daily with vehicle, 3 mg/kg THC, 3 mg/kg CBD, or 3 mg/kg THC +3 mg/kg CBD from gestational day 5 to postnatal day 10. Mass spectroscopic (MS) analyses were conducted to measure the THC and CBD brain levels in dams and their embryonic progenies. PCE adults were subjected to a battery of behavioral tests: open field arena, sucrose preference test, marble burying test, nestlet shredding test, and FST. Results: MS analysis found substantial levels of THC and CBD in embryonic brains. Our behavioral testing found that PCE females receiving THC or CBD buried significantly more marbles than control mice. Interestingly, PCE males receiving CBD or THC+CBD had significantly increased sucrose preference. While PCE with THC or CBD did not affect FST immobility, PCE with THC or CBD prevented fluoxetine from decreasing immobility in both males and females. Excitingly, fatty acid amide hydrolase (FAAH) inhibition with a dose of URB597 that was behaviorally inactive in the FST rescued fluoxetine efficacy in PCE mice of both sexes. Conclusions: Our data suggest that PCE with either THC, CBD, or THC+CBD alters repetitive and hedonic behaviors in a phytocannabinoid and sex-dependent manner. In addition, PCE with THC or CBD prevents fluoxetine from enhancing coping behavior. The restoration of fluoxetine responsiveness in THC or CBD PCE adults by inhibition of FAAH suggests that PCE causes a lasting reduction of the ECS and that enhancement of anandamide signaling represents a potential treatment for behavioral deficits following PCE.
Collapse
Affiliation(s)
- Izaque de Sousa Maciel
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA
| | - Gabriel H.D. de Abreu
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA.,Program in Neuroscience, Indiana University, Bloomington, Indiana, USA
| | - Claire T. Johnson
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA.,Program in Neuroscience, Indiana University, Bloomington, Indiana, USA
| | - Rida Bonday
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA
| | - Heather B. Bradshaw
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA.,Program in Neuroscience, Indiana University, Bloomington, Indiana, USA
| | - Ken Mackie
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA.,Program in Neuroscience, Indiana University, Bloomington, Indiana, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA.,Program in Neuroscience, Indiana University, Bloomington, Indiana, USA.,Address correspondence to: Hui-Chen Lu, PhD, The Linda and Jack Gill Center for Biomolecular Science, Indiana University, 702 N Walnut Grove Ave, IN 47405, USA,
| |
Collapse
|
19
|
De Genna NM, Willford JA, Richardson GA. Long-term effects of prenatal cannabis exposure: Pathways to adolescent and adult outcomes. Pharmacol Biochem Behav 2022; 214:173358. [PMID: 35216971 PMCID: PMC8911923 DOI: 10.1016/j.pbb.2022.173358] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 02/09/2022] [Accepted: 02/17/2022] [Indexed: 12/09/2022]
Abstract
With the increased prevalence, potency, and acceptability of cannabis use during pregnancy, it is important to understand the developmental effects of prenatal cannabis exposure (PCE). This review discusses methodological considerations for studies of PCE, including the assessment of exposures, covariates, and outcomes, and reviews findings from prospective, longitudinal studies of PCE. There is some evidence for associations between PCE and restricted growth at birth, but not for long-term effects on growth. PCE appears to have subtle yet enduring effects on memory and achievement in children and adolescents. Despite differences in sample demographics and measurement, there are remarkably consistent effects of PCE on externalizing behaviors, such as delinquency and substance use, which persist into adulthood. Longitudinal analyses demonstrate the importance of early cannabis initiation for pathways between PCE and adult functioning, including substance use and abuse, memory deficits, and psychotic symptoms. Animal studies demonstrate direct effects on the development of the brain via activation of endogenous endocannabinoid systems. Cannabis-induced activation of the endocannabinoid system causes alterations in the release of neurotransmitters and the modulation of brain plasticity in neural pathways that underlie cognition, motivation, and behavior regulation. Future research should consider cannabis use before pregnancy, the timing and route of exposure, polysubstance exposures, and inter-generational effects.
Collapse
Affiliation(s)
- Natacha M. De Genna
- University of Pittsburgh School of Medicine, Department of Psychiatry, 3811 O’Hara Street, Pittsburgh, PA 15213,Corresponding author:
| | - Jennifer A. Willford
- Slippery Rock University, Department of Psychology, 1 Morrow Way, Slippery Rock, PA 16057
| | - Gale A. Richardson
- University of Pittsburgh School of Medicine, Department of Psychiatry, 3811 O’Hara Street, Pittsburgh, PA 15213
| |
Collapse
|
20
|
Reid HMO, Snowden TM, Shkolnikov I, Breit KR, Rodriguez C, Thomas JD, Christie BR. Prenatal alcohol and cannabis exposure can have opposing and region-specific effects on parvalbumin interneuron numbers in the hippocampus. Alcohol Clin Exp Res 2021; 45:2246-2255. [PMID: 34523142 PMCID: PMC8642289 DOI: 10.1111/acer.14708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND We recently showed that alcohol and cannabis can interact prenatally, and in a recent review paper, we identified parvalbumin-positive (PV) interneurons in the hippocampus as a potential point of convergence for these teratogens. METHODS A 2 (Ethanol [EtOH], Air) × 2 (tetrahydrocannabinol [THC], Vehicle) design was used to expose pregnant Sprague-Dawley rats to either EtOH or air, in addition to either THC or the inhalant vehicle solution, during gestational days 5-20. Immunohistochemistry was performed to detect PV interneurons in 1 male and 1 female pup from each litter at postnatal day 70. RESULTS Significant between-group and subregion-specific effects were found in the dorsal cornu ammonis 1 (CA1) subfield and the ventral dentate gyrus (DG). In the dorsal CA1 subfield, there was an increase in the number of PV interneurons in both the EtOH and EtOH +THC groups, but a decrease with THC alone. There were fewer changes in interneuron numbers overall in the DG, though there was a sex difference, with a decrease in the number of PV interneurons in the THC-exposed group in males. There was also a greater cell layer volume in the DG in the EtOH +THC group than the control group, and in the CA1 region in the EtOH group compared to the control and THC groups. CONCLUSIONS Prenatal exposure to alcohol and THC differentially affects parvalbumin-positive interneuron numbers in the hippocampus, indicating that both individual and combined exposure can impact the balance of excitation and inhibition in a structure critically involved in learning and memory processes.
Collapse
Affiliation(s)
- Hannah M O Reid
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Taylor M Snowden
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Irene Shkolnikov
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Kristen R Breit
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
- Department of Psychology, West Chester University of Pennsylvania, West Chester, Pennsylvania, USA
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA
| | - Cristina Rodriguez
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
| | - Jennifer D Thomas
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Island Medical Program, Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, USA
| |
Collapse
|
21
|
Thomason ME, Palopoli AC, Jariwala NN, Werchan DM, Chen A, Adhikari S, Espinoza-Heredia C, Brito NH, Trentacosta CJ. Miswiring the brain: Human prenatal Δ9-tetrahydrocannabinol use associated with altered fetal hippocampal brain network connectivity. Dev Cogn Neurosci 2021; 51:101000. [PMID: 34388638 PMCID: PMC8363827 DOI: 10.1016/j.dcn.2021.101000] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 01/16/2023] Open
Abstract
Increasing evidence supports a link between maternal prenatal cannabis use and altered neural and physiological development of the child. However, whether cannabis use relates to altered human brain development prior to birth, and specifically, whether maternal prenatal cannabis use relates to connectivity of fetal functional brain systems, remains an open question. The major objective of this study was to identify whether maternal prenatal cannabis exposure (PCE) is associated with variation in human brain hippocampal functional connectivity prior to birth. Prenatal drug toxicology and fetal fMRI data were available in a sample of 115 fetuses [43 % female; mean age 32.2 weeks (SD = 4.3)]. Voxelwise hippocampal connectivity analysis in a subset of age and sex-matched fetuses revealed that PCE was associated with alterations in fetal dorsolateral, medial and superior frontal, insula, anterior temporal, and posterior cingulate connectivity. Classification of group differences by age 5 outcomes suggest that compared to the non-PCE group, the PCE group is more likely to have increased connectivity to regions associated with less favorable outcomes and to have decreased connectivity to regions associated with more favorable outcomes. This is preliminary evidence that altered fetal neural connectome may contribute to neurobehavioral vulnerability observed in children exposed to cannabis in utero.
Collapse
Affiliation(s)
- Moriah E Thomason
- Department of Child and Adolescent Psychiatry, New York University Medical Center, New York, NY, USA; Department of Population Health, New York University Medical Center, New York, NY, USA; Neuroscience Institute, New York University Medical Center, New York, NY, USA.
| | - Ava C Palopoli
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | - Nicki N Jariwala
- Department of Child and Adolescent Psychiatry, New York University Medical Center, New York, NY, USA
| | - Denise M Werchan
- Department of Child and Adolescent Psychiatry, New York University Medical Center, New York, NY, USA
| | - Alan Chen
- Department of Population Health, New York University Medical Center, New York, NY, USA
| | - Samrachana Adhikari
- Department of Population Health, New York University Medical Center, New York, NY, USA
| | - Claudia Espinoza-Heredia
- Department of Child and Adolescent Psychiatry, New York University Medical Center, New York, NY, USA
| | - Natalie H Brito
- Department of Applied Psychology, New York University, New York, NY, USA
| | | |
Collapse
|
22
|
Miller DS, Wright KM. Neuronal Dystroglycan regulates postnatal development of CCK/cannabinoid receptor-1 interneurons. Neural Dev 2021; 16:4. [PMID: 34362433 PMCID: PMC8349015 DOI: 10.1186/s13064-021-00153-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/20/2021] [Indexed: 12/02/2022] Open
Abstract
Background The development of functional neural circuits requires the precise formation of synaptic connections between diverse neuronal populations. The molecular pathways that allow GABAergic interneuron subtypes in the mammalian brain to initially recognize their postsynaptic partners remain largely unknown. The transmembrane glycoprotein Dystroglycan is localized to inhibitory synapses in pyramidal neurons, where it is required for the proper function of CCK+ interneurons. However, the precise temporal requirement for Dystroglycan during inhibitory synapse development has not been examined. Methods In this study, we use NEXCre or Camk2aCreERT2 to conditionally delete Dystroglycan from newly-born or adult pyramidal neurons, respectively. We then analyze forebrain development from postnatal day 3 through adulthood, with a particular focus on CCK+ interneurons. Results In the absence of postsynaptic Dystroglycan in developing pyramidal neurons, presynaptic CCK+ interneurons fail to elaborate their axons and largely disappear from the cortex, hippocampus, amygdala, and olfactory bulb during the first two postnatal weeks. Other interneuron subtypes are unaffected, indicating that CCK+ interneurons are unique in their requirement for postsynaptic Dystroglycan. Dystroglycan does not appear to be required in adult pyramidal neurons to maintain CCK+ interneurons. Bax deletion did not rescue CCK+ interneurons in Dystroglycan mutants during development, suggesting that they are not eliminated by canonical apoptosis. Rather, we observed increased innervation of the striatum, suggesting that the few remaining CCK+ interneurons re-directed their axons to neighboring areas where Dystroglycan expression remained intact. Conclusion Together these findings show that Dystroglycan functions as part of a synaptic partner recognition complex that is required early for CCK+ interneuron development in the forebrain. Supplementary Information The online version contains supplementary material available at 10.1186/s13064-021-00153-1.
Collapse
Affiliation(s)
- Daniel S Miller
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kevin M Wright
- Vollum Institute, Oregon Health & Science University, VIB 3435A, 3181 SW Sam Jackson Park Road, L474, Portland, OR, 97239-3098, USA.
| |
Collapse
|
23
|
Bara A, Ferland JMN, Rompala G, Szutorisz H, Hurd YL. Cannabis and synaptic reprogramming of the developing brain. Nat Rev Neurosci 2021; 22:423-438. [PMID: 34021274 DOI: 10.1038/s41583-021-00465-5] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 02/08/2023]
Abstract
Recent years have been transformational in regard to the perception of the health risks and benefits of cannabis with increased acceptance of use. This has unintended neurodevelopmental implications given the increased use of cannabis and the potent levels of Δ9-tetrahydrocannabinol today being consumed by pregnant women, young mothers and teens. In this Review, we provide an overview of the neurobiological effects of cannabinoid exposure during prenatal/perinatal and adolescent periods, in which the endogenous cannabinoid system plays a fundamental role in neurodevelopmental processes. We highlight impaired synaptic plasticity as characteristic of developmental exposure and the important contribution of epigenetic reprogramming that maintains the long-term impact into adulthood and across generations. Such epigenetic influence by its very nature being highly responsive to the environment also provides the potential to diminish neural perturbations associated with developmental cannabis exposure.
Collapse
Affiliation(s)
- Anissa Bara
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA.,Addiction Institute of Mount Sinai, Mount Sinai, NY, USA.,Friedman Brain Institute, Mount Sinai, NY, USA
| | - Jacqueline-Marie N Ferland
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA.,Addiction Institute of Mount Sinai, Mount Sinai, NY, USA.,Friedman Brain Institute, Mount Sinai, NY, USA
| | - Gregory Rompala
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA.,Addiction Institute of Mount Sinai, Mount Sinai, NY, USA.,Friedman Brain Institute, Mount Sinai, NY, USA
| | - Henrietta Szutorisz
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA.,Addiction Institute of Mount Sinai, Mount Sinai, NY, USA.,Friedman Brain Institute, Mount Sinai, NY, USA
| | - Yasmin L Hurd
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA. .,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA. .,Addiction Institute of Mount Sinai, Mount Sinai, NY, USA. .,Friedman Brain Institute, Mount Sinai, NY, USA.
| |
Collapse
|
24
|
Song CG, Kang X, Yang F, Du WQ, Zhang JJ, Liu L, Kang JJ, Jia N, Yue H, Fan LY, Wu SX, Jiang W, Gao F. Endocannabinoid system in the neurodevelopment of GABAergic interneurons: implications for neurological and psychiatric disorders. Rev Neurosci 2021; 32:803-831. [PMID: 33781002 DOI: 10.1515/revneuro-2020-0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/20/2021] [Indexed: 02/07/2023]
Abstract
In mature mammalian brains, the endocannabinoid system (ECS) plays an important role in the regulation of synaptic plasticity and the functioning of neural networks. Besides, the ECS also contributes to the neurodevelopment of the central nervous system. Due to the increase in the medical and recreational use of cannabis, it is inevitable and essential to elaborate the roles of the ECS on neurodevelopment. GABAergic interneurons represent a group of inhibitory neurons that are vital in controlling neural network activity. However, the role of the ECS in the neurodevelopment of GABAergic interneurons remains to be fully elucidated. In this review, we provide a brief introduction of the ECS and interneuron diversity. We focus on the process of interneuron development and the role of ECS in the modulation of interneuron development, from the expansion of the neural stem/progenitor cells to the migration, specification and maturation of interneurons. We further discuss the potential implications of the ECS and interneurons in the pathogenesis of neurological and psychiatric disorders, including epilepsy, schizophrenia, major depressive disorder and autism spectrum disorder.
Collapse
Affiliation(s)
- Chang-Geng Song
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China.,Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Xin Kang
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Fang Yang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Wan-Qing Du
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Jia-Jia Zhang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Long Liu
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Jun-Jun Kang
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Ning Jia
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Hui Yue
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Lu-Yu Fan
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Sheng-Xi Wu
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Wen Jiang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Fang Gao
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| |
Collapse
|
25
|
Ballaz SJ, Bourin M. Cholecystokinin-Mediated Neuromodulation of Anxiety and Schizophrenia: A "Dimmer-Switch" Hypothesis. Curr Neuropharmacol 2021; 19:925-938. [PMID: 33185164 PMCID: PMC8686311 DOI: 10.2174/1570159x18666201113145143] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/08/2020] [Accepted: 11/10/2020] [Indexed: 11/22/2022] Open
Abstract
Cholecystokinin (CCK), the most abundant brain neuropeptide, is involved in relevant behavioral functions like memory, cognition, and reward through its interactions with the opioid and dopaminergic systems in the limbic system. CCK excites neurons by binding two receptors, CCK1 and CCK2, expressed at low and high levels in the brain, respectively. Historically, CCK2 receptors have been related to the induction of panic attacks in humans. Disturbances in brain CCK expression also underlie the physiopathology of schizophrenia, which is attributed to the modulation by CCK1 receptors of the dopamine flux in the basal striatum. Despite this evidence, neither CCK2 receptor antagonists ameliorate human anxiety nor CCK agonists have consistently shown neuroleptic effects in clinical trials. A neglected aspect of the function of brain CCK is its neuromodulatory role in mental disorders. Interestingly, CCK is expressed in pivotal inhibitory interneurons that sculpt cortical dynamics and the flux of nerve impulses across corticolimbic areas and the excitatory projections to mesolimbic pathways. At the basal striatum, CCK modulates the excitability of glutamate, the release of inhibitory GABA, and the discharge of dopamine. Here we focus on how CCK may reduce rather than trigger anxiety by regulating its cognitive component. Adequate levels of CCK release in the basal striatum may control the interplay between cognition and reward circuitry, which is critical in schizophrenia. Hence, it is proposed that disturbances in the excitatory/ inhibitory interplay modulated by CCK may contribute to the imbalanced interaction between corticolimbic and mesolimbic neural activity found in anxiety and schizophrenia.
Collapse
Affiliation(s)
- Santiago J. Ballaz
- Address correspondence to this author at the School of Biological Sciences & Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí, Ecuador; Tel: 593 (06) 299 9100, ext. 2626; E-mail:
| | | |
Collapse
|
26
|
Paraíso-Luna J, Aguareles J, Martín R, Ayo-Martín AC, Simón-Sánchez S, García-Rincón D, Costas-Insua C, García-Taboada E, de Salas-Quiroga A, Díaz-Alonso J, Liste I, Sánchez-Prieto J, Cappello S, Guzmán M, Galve-Roperh I. Endocannabinoid signalling in stem cells and cerebral organoids drives differentiation to deep layer projection neurons via CB 1 receptors. Development 2020; 147:226034. [PMID: 33168583 DOI: 10.1242/dev.192161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022]
Abstract
The endocannabinoid (eCB) system, via the cannabinoid CB1 receptor, regulates neurodevelopment by controlling neural progenitor proliferation and neurogenesis. CB1 receptor signalling in vivo drives corticofugal deep layer projection neuron development through the regulation of BCL11B and SATB2 transcription factors. Here, we investigated the role of eCB signalling in mouse pluripotent embryonic stem cell-derived neuronal differentiation. Characterization of the eCB system revealed increased expression of eCB-metabolizing enzymes, eCB ligands and CB1 receptors during neuronal differentiation. CB1 receptor knockdown inhibited neuronal differentiation of deep layer neurons and increased upper layer neuron generation, and this phenotype was rescued by CB1 re-expression. Pharmacological regulation with CB1 receptor agonists or elevation of eCB tone with a monoacylglycerol lipase inhibitor promoted neuronal differentiation of deep layer neurons at the expense of upper layer neurons. Patch-clamp analyses revealed that enhancing cannabinoid signalling facilitated neuronal differentiation and functionality. Noteworthy, incubation with CB1 receptor agonists during human iPSC-derived cerebral organoid formation also promoted the expansion of BCL11B+ neurons. These findings unveil a cell-autonomous role of eCB signalling that, via the CB1 receptor, promotes mouse and human deep layer cortical neuron development.
Collapse
Affiliation(s)
- Juan Paraíso-Luna
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - José Aguareles
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Ricardo Martín
- Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Ane C Ayo-Martín
- Max Planck Institute of Psychiatry, 80804 Munich, Germany.,International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
| | - Samuel Simón-Sánchez
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Daniel García-Rincón
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Carlos Costas-Insua
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Elena García-Taboada
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Adán de Salas-Quiroga
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Javier Díaz-Alonso
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Isabel Liste
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain
| | - José Sánchez-Prieto
- Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | | | - Manuel Guzmán
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| | - Ismael Galve-Roperh
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain.,Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), 28040 Madrid, Spain
| |
Collapse
|
27
|
Beiersdorf J, Hevesi Z, Calvigioni D, Pyszkowski J, Romanov R, Szodorai E, Lubec G, Shirran S, Botting CH, Kasper S, Guy GW, Gray R, Di Marzo V, Harkany T, Keimpema E. Adverse effects of Δ9-tetrahydrocannabinol on neuronal bioenergetics during postnatal development. JCI Insight 2020; 5:135418. [PMID: 33141759 PMCID: PMC7714410 DOI: 10.1172/jci.insight.135418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 10/28/2020] [Indexed: 11/22/2022] Open
Abstract
Ongoing societal changes in views on the medical and recreational roles of cannabis increased the use of concentrated plant extracts with a Δ9-tetrahydrocannabinol (THC) content of more than 90%. Even though prenatal THC exposure is widely considered adverse for neuronal development, equivalent experimental data for young age cohorts are largely lacking. Here, we administered plant-derived THC (1 or 5 mg/kg) to mice daily during P5–P16 and P5–P35 and monitored its effects on hippocampal neuronal survival and specification by high-resolution imaging and iTRAQ proteomics, respectively. We found that THC indiscriminately affects pyramidal cells and both cannabinoid receptor 1+ (CB1R)+ and CB1R– interneurons by P16. THC particularly disrupted the expression of mitochondrial proteins (complexes I–IV), a change that had persisted even 4 months after the end of drug exposure. This was reflected by a THC-induced loss of membrane integrity occluding mitochondrial respiration and could be partially or completely rescued by pH stabilization, antioxidants, bypassed glycolysis, and targeting either mitochondrial soluble adenylyl cyclase or the mitochondrial voltage-dependent anion channel. Overall, THC exposure during infancy induces significant and long-lasting reorganization of neuronal circuits through mechanisms that, in large part, render cellular bioenergetics insufficient to sustain key developmental processes in otherwise healthy neurons. Repeated THC exposure in juvenile mice compromises the limbic circuitry, with life-long impairment to the respiration of neurons.
Collapse
Affiliation(s)
- Johannes Beiersdorf
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Zsofia Hevesi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela Calvigioni
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | | | - Roman Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Edit Szodorai
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Gert Lubec
- Paracelsus Private Medical University, Salzburg, Austria
| | - Sally Shirran
- School of Chemistry, University of St. Andrews, St. Andrews, United Kingdom
| | | | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | | | - Roy Gray
- GW Phamaceuticals, Salisbury, Wiltshire, United Kingdom
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Italy.,Canada Excellence Research Chair, Institut Universitaire de Cardiologie et de Pneumologie de Québec and Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec, Québec, Canada
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.,Department of Neuroscience, Biomedikum D7, Karolinska Institutet, Solna, Sweden
| | - Erik Keimpema
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Perinatal THC exposure via lactation induces lasting alterations to social behavior and prefrontal cortex function in rats at adulthood. Neuropsychopharmacology 2020; 45:1826-1833. [PMID: 32428929 PMCID: PMC7608083 DOI: 10.1038/s41386-020-0716-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/16/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022]
Abstract
Cannabis is the world's most widely abused illicit drug and consumption amongst women during and surrounding the period of pregnancy is increasing. Previously, we have shown that cannabinoid exposure via lactation during the early postnatal period disrupts early developmental trajectories of prefrontal cortex maturation and induces behavioral abnormalities during the first weeks of life in male and female rat progeny. Here, we investigated the lasting consequences of this postnatal cannabinoid exposure on synaptic and behavioral parameters in the adult offspring of ∆9-tetrahydrocannabinol (THC)-treated dams. At adulthood, these perinatally THC-exposed rats exhibits deficits in social discrimination accompanied by an overall augmentation of social exploratory behavior. These behavioral alterations were further correlated with multiple abnormalities in synaptic plasticity in the prefrontal cortex, including lost endocannabinoid-mediated long-term depression (LTD), lost long-term potentiation and augmented mGlu2/3-LTD. Finally, basic parameters of intrinsic excitability at prefrontal cortex pyramidal neurons were similarly altered by the perinatal THC exposure. Thus, perinatal THC exposure via lactation induces lasting deficits in behavior and synaptic function which persist into adulthood life in male and female progeny.
Collapse
|
29
|
Maternal Exposure to the Cannabinoid Agonist WIN 55,12,2 during Lactation Induces Lasting Behavioral and Synaptic Alterations in the Rat Adult Offspring of Both Sexes. eNeuro 2020; 7:ENEURO.0144-20.2020. [PMID: 32868310 PMCID: PMC7540927 DOI: 10.1523/eneuro.0144-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/09/2020] [Accepted: 07/27/2020] [Indexed: 11/30/2022] Open
Abstract
Consumption of cannabis during pregnancy and the lactation period is a rising public health concern (Scheyer et al., 2019). Exposure to synthetic or plant-derived cannabinoids via lactation disrupts the development of GABAergic neurons in the prefrontal cortex (PFC) and alters early-life behaviors (Scheyer et al., 2020b). Recently, additional data revealed that Δ9-tetrahydrocannabinol (THC) perinatal exposure via lactation causes lasting behavioral and neuronal consequences (Scheyer et al., 2020a). Here, the long-term effects in adult offspring of maternal exposure to the synthetic cannabinoid agonist WIN 55,12,2 are reported. The data demonstrate that rats exposed during lactation to WIN display social and motivational deficits at adulthood. These behavioral changes were paralleled by a specific loss of endocannabinoid-mediated long-term depression (eCB-LTD) in the PFC and nucleus accumbens (NAc), while other forms of synaptic plasticity remained intact. Thus, similarly to THC, perinatal WIN exposure via lactation induces behavioral and synaptic abnormalities lasting into adulthood.
Collapse
|
30
|
Smith A, Kaufman F, Sandy MS, Cardenas A. Cannabis Exposure During Critical Windows of Development: Epigenetic and Molecular Pathways Implicated in Neuropsychiatric Disease. Curr Environ Health Rep 2020; 7:325-342. [PMID: 32441004 PMCID: PMC7458902 DOI: 10.1007/s40572-020-00275-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW Cannabis exposure during critical windows of development may have intergenerational physiological consequences disrupting epigenetic programming and marks. This review examines the literature relating to pre-gestational and prenatal cannabinoid exposure and its effect on genes and molecular pathways related to the development of psychiatric disease. RECENT FINDINGS Developmental cannabis exposure alters epigenetic processes with functional gene consequences. These include potentially heritable alterations in genes and molecular pathways critical for brain development and associated with autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), schizophrenia, addiction, and other psychiatric diseases. Cannabis consumption and mental health illness in adolescents and young adults are increasing in the United States (U.S.), and recent studies suggest that cannabis consumption during critical periods of brain development could contribute to mental health illness through epigenetic mechanisms. These findings warrant future studies and consideration by regulators and health communicators.
Collapse
Affiliation(s)
- Anna Smith
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, CA, USA
| | - Farla Kaufman
- Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Sacramento, CA, USA
| | - Martha S Sandy
- Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Sacramento, CA, USA
| | - Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA.
- Center for Computational Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|
31
|
Zamberletti E, Rubino T. Impact of Endocannabinoid System Manipulation on Neurodevelopmental Processes Relevant to Schizophrenia. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2020; 6:616-626. [PMID: 32855107 DOI: 10.1016/j.bpsc.2020.06.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/22/2022]
Abstract
The neurodevelopmental hypothesis of schizophrenia has received much support from epidemiological and neuropathological studies and provides a framework to explain how early developmental abnormalities might manifest as psychosis in early adulthood. According to this theory, the onset of schizophrenia is likely the result of a complex interplay between a genetic predisposition and environmental factors whose respective influence might contribute to the etiology and progression of the disorder. The two most sensitive windows for neurodevelopment are the prenatal/perinatal and the adolescent windows, both of which are characterized by specific processes impinging upon brain structure and functionality, whose alterations may contribute to the onset of schizophrenia. An increasing number of articles suggest the involvement of the endocannabinoid system in the modulation of at least some of these processes, especially in the prenatal/perinatal window. Thus, it is not surprising that disturbing the physiological role of endocannabinoid signaling in these sensitive windows might alter the correct formation of neuronal networks, eventually predisposing to neuropsychiatric diseases later in life. We review the most recent preclinical studies that evaluated the impact of endocannabinoid system modulation in the two sensitive developmental windows on neurodevelopmental processes that possess a specific relevance to schizophrenia.
Collapse
Affiliation(s)
- Erica Zamberletti
- Department of Biotechnology and Life Sciences and Neuroscience Center, University of Insubria, Busto Arsizio, Varese, Italy
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences and Neuroscience Center, University of Insubria, Busto Arsizio, Varese, Italy.
| |
Collapse
|
32
|
Hoffman MC, Hunter SK, D’Alessandro A, Noonan K, Wyrwa A, Freedman R. Interaction of maternal choline levels and prenatal Marijuana's effects on the offspring. Psychol Med 2020; 50:1716-1726. [PMID: 31364525 PMCID: PMC7055467 DOI: 10.1017/s003329171900179x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND This study investigated whether higher maternal choline levels mitigate effects of marijuana on fetal brain development. Choline transported into the amniotic fluid from the mother activates α7-nicotinic acetylcholine receptors on fetal cerebro-cortical inhibitory neurons, whose development is impeded by cannabis blockade of their cannabinoid-1(CB1) receptors. METHODS Marijuana use was assessed during pregnancy from women who later brought their newborns for study. Mothers were informed about choline and other nutrients, but not specifically for marijuana use. Maternal serum choline was measured at 16 weeks gestation. RESULTS Marijuana use for the first 10 weeks gestation or more by 15% of mothers decreased newborns' inhibition of evoked potentials to repeated sounds (d' = 0.55, p < 0.05). This effect was ameliorated if women had higher gestational choline (rs = -0.50, p = 0.011). At 3 months of age, children whose mothers continued marijuana use through their 10th gestational week or more had poorer self-regulation (d' = -0.79, p < 0.05). This effect was also ameliorated if mothers had higher gestational choline (rs = 0.54, p = 0.013). Maternal choline levels correlated with the children's improved duration of attention, cuddliness, and bonding with parents. CONCLUSIONS Prenatal marijuana use adversely affects fetal brain development and subsequent behavioral self-regulation, a precursor to later, more serious problems in childhood. Stopping marijuana use before 10 weeks gestational age prevented these effects. Many mothers refuse to cease use because of familiarity with marijuana and belief in its safety. Higher maternal choline mitigates some of marijuana's adverse effects on the fetus.
Collapse
Affiliation(s)
- M. Camille Hoffman
- Department of Obstetrics and Gynecology, Division of Maternal and Fetal Medicine, University of Colorado Denver School of Medicine, Aurora, Colorado 80045, USA
- Department of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado 80045, USA
| | - Sharon K. Hunter
- Department of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado 80045, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado 80045, USA
| | - Kathleen Noonan
- Department of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado 80045, USA
| | - Anna Wyrwa
- Department of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado 80045, USA
| | - Robert Freedman
- Department of Psychiatry, University of Colorado Denver School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
33
|
Disrupted inhibitory plasticity and homeostasis in Fragile X syndrome. Neurobiol Dis 2020; 142:104959. [PMID: 32512151 PMCID: PMC7959200 DOI: 10.1016/j.nbd.2020.104959] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/28/2020] [Accepted: 06/03/2020] [Indexed: 12/21/2022] Open
Abstract
Fragile X Syndrome (FXS) is a neurodevelopmental disorder instigated by the absence of a key translation regulating protein, Fragile X Mental Retardation Protein (FMRP). The loss of FMRP in the CNS leads to abnormal synaptic development, disruption of critical periods of plasticity, and an overall deficiency in proper sensory circuit coding leading to hyperexcitable sensory networks. However, little is known about how this hyperexcitable environment affects inhibitory synaptic plasticity. Here, we show that in vivo layer 2/3 of the primary somatosensory cortex of the Fmr1 KO mouse exhibits basal hyperexcitability and an increase in neuronal firing rate suppression during whisker activation. This aligns with our in vitro data that indicate an increase in GABAergic spontaneous activity, a faulty mGluR-mediated inhibitory input and impaired inhibitory plasticity processes. Specifically, we find that mGluR activation sensitivity is overall diminished in the Fmr1 KO mouse leading to both a decreased spontaneous inhibitory postsynaptic input to principal cells and a disrupted form of inhibitory long-term depression (I-LTD). These data suggest an adaptive mechanism that acts to homeostatically counterbalance the cortical hyperexcitability observed in FXS.
Collapse
|
34
|
Reid HMO, Lysenko-Martin MR, Snowden TM, Thomas JD, Christie BR. A Systematic Review of the Effects of Perinatal Alcohol Exposure and Perinatal Marijuana Exposure on Adult Neurogenesis in the Dentate Gyrus. Alcohol Clin Exp Res 2020; 44:1164-1174. [PMID: 32246781 PMCID: PMC7905844 DOI: 10.1111/acer.14332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Marijuana and alcohol are both substances that, when used during pregnancy, may have profound effects on the developing fetus. There is evidence to suggest that both drugs have the capacity to affect working memory, one function of the hippocampal formation; however, there is a paucity of data on how perinatal exposure to alcohol or cannabis impacts the process of adult neurogenesis. METHODS This systematic review examines immunohistochemical data from adult rat and mouse models that assess perinatal alcohol or perinatal marijuana exposure. A comprehensive list of search terms was designed and used to search 3 separate databases. All results were imported to Mendeley and screened by 2 authors. Consensus was reached on a set of final papers that met the inclusion criteria, and their results were summarized. RESULTS Twelve papers were identified as relevant, 10 of which pertained to the effects of perinatal alcohol on the adult hippocampus, and 2 pertained to the effects of perinatal marijuana on the adult hippocampus. Cellular proliferation in the dentate gyrus was not affected in adult rats and mice exposed to alcohol perinatally. In general, perinatal alcohol exposure did not have a significant and reliable effect on the maturation and survival of adult born granule neurons in the dentate gyrus. In contrast, interneuron numbers appear to be reduced in the dentate gyrus of adult rats and mice exposed perinatally to alcohol. Perinatal marijuana exposure was also found to reduce inhibitory interneuron numbers in the dentate gyrus. CONCLUSIONS Perinatal alcohol exposure and perinatal marijuana exposure both act on inhibitory interneurons in the hippocampal formation of adult rats. These findings suggest simultaneous perinatal alcohol and marijuana exposure (SAM) may have a dramatic impact on inhibitory processes in the dentate gyrus.
Collapse
Affiliation(s)
- Hannah M O Reid
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
| | - Melanie R Lysenko-Martin
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
| | - Taylor M Snowden
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
| | - Jennifer D Thomas
- Center for Behavioral Teratology, (JDT), San Diego State University, San Diego, California
| | - Brian R Christie
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
- Island Medical Program and Department of Cellular and Physiological Sciences, (BRC), University of British Columbia, Victoria, British Columbia
| |
Collapse
|
35
|
de Salas-Quiroga A, García-Rincón D, Gómez-Domínguez D, Valero M, Simón-Sánchez S, Paraíso-Luna J, Aguareles J, Pujadas M, Muguruza C, Callado LF, Lutz B, Guzmán M, de la Prida LM, Galve-Roperh I. Long-term hippocampal interneuronopathy drives sex-dimorphic spatial memory impairment induced by prenatal THC exposure. Neuropsychopharmacology 2020; 45:877-886. [PMID: 31982904 PMCID: PMC7075920 DOI: 10.1038/s41386-020-0621-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/30/2022]
Abstract
Prenatal exposure to Δ9-tetrahydrocannabinol (THC), the most prominent active constituent of cannabis, alters neurodevelopmental plasticity with a long-term functional impact on adult offspring. Specifically, THC affects the development of pyramidal neurons and GABAergic interneurons via cannabinoid CB1 receptors (CB1R). However, the particular contribution of these two neuronal lineages to the behavioral alterations and functional deficits induced by THC is still unclear. Here, by using conditional CB1R knockout mice, we investigated the neurodevelopmental consequences of prenatal THC exposure in adulthood, as well as their potential sex differences. Adult mice that had been exposed to THC during embryonic development showed altered hippocampal oscillations, brain hyperexcitability, and spatial memory impairment. Remarkably, we found a clear sexual dimorphism in these effects, with males being selectively affected. At the neuronal level, we found a striking interneuronopathy of CCK-containing interneurons in the hippocampus, which was restricted to male progeny. This THC-induced CCK-interneuron reduction was not evident in mice lacking CB1R selectively in GABAergic interneurons, thus pointing to a cell-autonomous THC action. In vivo electrophysiological recordings of hippocampal LFPs revealed alterations in hippocampal oscillations confined to the stratum pyramidale of CA1 in male offspring. In addition, sharp-wave ripples, a major high-frequency oscillation crucial for learning and memory consolidation, were also altered, pointing to aberrant circuitries caused by persistent reduction of CCK+ basket cells. Taken together, these findings provide a mechanistic explanation for the long-term interneuronopathy responsible for the sex-dimorphic cognitive impairment induced by prenatal THC.
Collapse
Affiliation(s)
- Adán de Salas-Quiroga
- Department of Biochemistry and Molecular Biology, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040, Madrid, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049, Madrid, Spain.
| | - Daniel García-Rincón
- 0000 0001 2157 7667grid.4795.fDepartment of Biochemistry and Molecular Biology, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040 Madrid, Spain ,0000 0004 1762 4012grid.418264.dCentro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain
| | - Daniel Gómez-Domínguez
- 0000 0001 2177 5516grid.419043.bInstituto Cajal, CSIC, Avda Dr Arce 37, 28002 Madrid, Spain
| | - Manuel Valero
- 0000 0001 2177 5516grid.419043.bInstituto Cajal, CSIC, Avda Dr Arce 37, 28002 Madrid, Spain
| | - Samuel Simón-Sánchez
- 0000 0001 2157 7667grid.4795.fDepartment of Biochemistry and Molecular Biology, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040 Madrid, Spain ,0000 0004 1762 4012grid.418264.dCentro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain
| | - Juan Paraíso-Luna
- 0000 0001 2157 7667grid.4795.fDepartment of Biochemistry and Molecular Biology, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040 Madrid, Spain ,0000 0004 1762 4012grid.418264.dCentro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain
| | - José Aguareles
- 0000 0001 2157 7667grid.4795.fDepartment of Biochemistry and Molecular Biology, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040 Madrid, Spain ,0000 0004 1762 4012grid.418264.dCentro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain
| | - Mitona Pujadas
- 0000 0004 1767 9005grid.20522.37Integrative Pharmacology and Systems Neuroscience Research Group, Neurosciences Research Program, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Carolina Muguruza
- 0000000121671098grid.11480.3cDepartment of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Luis F. Callado
- 0000000121671098grid.11480.3cDepartment of Pharmacology, University of the Basque Country UPV/EHU and Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Leioa, Spain
| | - Beat Lutz
- grid.410607.4Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Manuel Guzmán
- 0000 0001 2157 7667grid.4795.fDepartment of Biochemistry and Molecular Biology, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040 Madrid, Spain ,0000 0004 1762 4012grid.418264.dCentro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049 Madrid, Spain
| | | | - Ismael Galve-Roperh
- Department of Biochemistry and Molecular Biology, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040, Madrid, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28049, Madrid, Spain.
| |
Collapse
|
36
|
Qi XR, Zhang L. The Potential Role of Gut Peptide Hormones in Autism Spectrum Disorder. Front Cell Neurosci 2020; 14:73. [PMID: 32296309 PMCID: PMC7136424 DOI: 10.3389/fncel.2020.00073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/12/2020] [Indexed: 12/11/2022] Open
Abstract
Gut peptide hormones are one group of secretory factors produced from gastrointestinal endocrine cells with potent functions in modulating digestive functions. In recent decades, they have been found across different brain regions, many of which are involved in autism-related social, emotional and cognitive deficits. Clinical studies have revealed possible correlation between those hormones and autism spectrum disorder pathogenesis. In animal models, gut peptide hormones modulate neurodevelopment, synaptic transmission and neural plasticity, explaining their behavioral relevance. This review article will summarize major findings from both clinical and basic research showing the role of gut peptide hormones in mediating autism-related neurological functions, and their potential implications in autism pathogenesis. The pharmaceutical value of gut hormones in alleviating autism-associated behavioral syndromes will be discussed to provide new insights for future drug development.
Collapse
Affiliation(s)
- Xin-Rui Qi
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Li Zhang
- Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| |
Collapse
|
37
|
Pelkey KA, Calvigioni D, Fang C, Vargish G, Ekins T, Auville K, Wester JC, Lai M, Mackenzie-Gray Scott C, Yuan X, Hunt S, Abebe D, Xu Q, Dimidschstein J, Fishell G, Chittajallu R, McBain CJ. Paradoxical network excitation by glutamate release from VGluT3 + GABAergic interneurons. eLife 2020; 9:e51996. [PMID: 32053107 PMCID: PMC7039679 DOI: 10.7554/elife.51996] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/12/2020] [Indexed: 12/18/2022] Open
Abstract
In violation of Dale's principle several neuronal subtypes utilize more than one classical neurotransmitter. Molecular identification of vesicular glutamate transporter three and cholecystokinin expressing cortical interneurons (CCK+VGluT3+INTs) has prompted speculation of GABA/glutamate corelease from these cells for almost two decades despite a lack of direct evidence. We unequivocally demonstrate CCK+VGluT3+INT-mediated GABA/glutamate cotransmission onto principal cells in adult mice using paired recording and optogenetic approaches. Although under normal conditions, GABAergic inhibition dominates CCK+VGluT3+INT signaling, glutamatergic signaling becomes predominant when glutamate decarboxylase (GAD) function is compromised. CCK+VGluT3+INTs exhibit surprising anatomical diversity comprising subsets of all known dendrite targeting CCK+ interneurons in addition to the expected basket cells, and their extensive circuit innervation profoundly dampens circuit excitability under normal conditions. However, in contexts where the glutamatergic phenotype of CCK+VGluT3+INTs is amplified, they promote paradoxical network hyperexcitability which may be relevant to disorders involving GAD dysfunction such as schizophrenia or vitamin B6 deficiency.
Collapse
Affiliation(s)
- Kenneth A Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Daniela Calvigioni
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Calvin Fang
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Geoffrey Vargish
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Tyler Ekins
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Kurt Auville
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Jason C Wester
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Mandy Lai
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Connie Mackenzie-Gray Scott
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Xiaoqing Yuan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Steven Hunt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Daniel Abebe
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Qing Xu
- Center for Genomics and Systems Biology, NYUAbu-DhabiUnited Arab Emirates
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Gordon Fishell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and HarvardCambridgeUnited States
- Department of Neurobiology, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Chris J McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
38
|
Δ9-tetrahydrocannabinol exposure during rat pregnancy leads to symmetrical fetal growth restriction and labyrinth-specific vascular defects in the placenta. Sci Rep 2020; 10:544. [PMID: 31953475 PMCID: PMC6969028 DOI: 10.1038/s41598-019-57318-6] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 12/20/2019] [Indexed: 02/08/2023] Open
Abstract
1 in 5 women report cannabis use during pregnancy, with nausea cited as their primary motivation. Studies show that (-)-△9-tetrahydrocannabinol (Δ9-THC), the major psychoactive ingredient in cannabis, causes fetal growth restriction, though the mechanisms are not well understood. Given the critical role of the placenta to transfer oxygen and nutrients from mother, to the fetus, any compromise in the development of fetal-placental circulation significantly affects maternal-fetal exchange and thereby, fetal growth. The goal of this study was to examine, in rats, the impact of maternal Δ9-THC exposure on fetal development, neonatal outcomes, and placental development. Dams received a daily intraperitoneal injection (i.p.) of vehicle control or Δ9-THC (3 mg/kg) from embryonic (E)6.5 through 22. Dams were allowed to deliver normally to measure pregnancy and neonatal outcomes, with a subset sacrificed at E19.5 for placenta assessment via immunohistochemistry and qPCR. Gestational Δ9-THC exposure resulted in pups born with symmetrical fetal growth restriction, with catch up growth by post-natal day (PND)21. During pregnancy there were no changes to maternal food intake, maternal weight gain, litter size, or gestational length. E19.5 placentas from Δ9-THC-exposed pregnancies exhibited a phenotype characterized by increased labyrinth area, reduced Epcam expression (marker of labyrinth trophoblast progenitors), altered maternal blood space, decreased fetal capillary area and an increased recruitment of pericytes with greater collagen deposition, when compared to vehicle controls. Further, at E19.5 labyrinth trophoblast had reduced glucose transporter 1 (GLUT1) and glucocorticoid receptor (GR) expression in response to Δ9-THC exposure. In conclusion, maternal exposure to Δ9-THC effectively compromised fetal growth, which may be a result of the adversely affected labyrinth zone development. These findings implicate GLUT1 as a Δ9-THC target and provide a potential mechanism for the fetal growth restriction observed in women who use cannabis during pregnancy.
Collapse
|
39
|
Franks AL, Berry KJ, DeFranco DB. Prenatal drug exposure and neurodevelopmental programming of glucocorticoid signalling. J Neuroendocrinol 2020; 32:e12786. [PMID: 31469457 PMCID: PMC6982551 DOI: 10.1111/jne.12786] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/25/2019] [Accepted: 08/27/2019] [Indexed: 12/21/2022]
Abstract
Prenatal neurodevelopment is dependent on precise functioning of multiple signalling pathways in the brain, including those mobilised by glucocorticoids (GC) and endocannabinoids (eCBs). Prenatal exposure to drugs of abuse, including opioids, alcohol, cocaine and cannabis, has been shown to not only impact GC signalling, but also alter functioning of the hypothalamic-pituitary-adrenal (HPA) axis. Such exposures can have long-lasting neurobehavioural consequences, including alterations in the stress response in the offspring. Furthermore, cannabis contains cannabinoids that signal via the eCB pathway, which is linked to some components of GC signalling in the adult brain. Given that GCs are frequently used in pregnancy to prevent complications of prematurity, and also that rates of cannabis use in pregnancy are increasing, the likelihood of foetal co-exposure to these compounds is high and may have additional implications for long-term neurodevelopment. Here, we present a discussion of GC signalling and the HPA axis, as well as the effects of prenatal drug exposure on these pathways and the stress response, and we explore the interactions between GC and EC signalling in the developing brain and potential for neurodevelopmental consequences.
Collapse
Affiliation(s)
- Alexis L Franks
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kimberly J Berry
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Donald B DeFranco
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology and Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
40
|
Manduca A, Servadio M, Melancia F, Schiavi S, Manzoni OJ, Trezza V. Sex-specific behavioural deficits induced at early life by prenatal exposure to the cannabinoid receptor agonist WIN55, 212-2 depend on mGlu5 receptor signalling. Br J Pharmacol 2020; 177:449-463. [PMID: 31658362 PMCID: PMC6989958 DOI: 10.1111/bph.14879] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/04/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Marijuana is the illicit drug most commonly used among pregnant and breastfeeding women. Different studies reported long-term adverse effects induced by in utero exposure to the main component of marijuana, Δ9 -tetrahydrocannabinol (THC), both in rodents and in humans. However, little is known about any potential sex-dependent effects of marijuana consumption during pregnancy on newborns at early developmental ages. EXPERIMENTAL APPROACH We studied the effects of prenatal exposure to the cannabinoid receptor agonist WIN55,212-2 (WIN; 0.5 mg·kg-1 from GD5 to GD20) on the emotional reactivity and cognitive performance of male and female rat offspring from infancy through adolescence and tested the role of mGlu5 receptor signalling in the observed effects. KEY RESULTS Prenatally WIN-exposed male infant pups emitted less isolation-induced ultrasonic vocalizations compared with male control pups, when separated from the dam and siblings and showed increased locomotor activity while females were spared. These effects were normalized when male pups were treated with the positive allosteric modulator of mGlu5 receptor CDPPB. When tested at the prepubertal and pubertal periods, WIN-prenatally exposed rats of both sexes did not show any difference in social play behaviour, anxiety and temporal order memory. CONCLUSIONS AND IMPLICATIONS We reveal a previously undisclosed sexual divergence in the consequences of fetal cannabinoids on newborns at early developmental ages, which is dependent on mGlu5 receptor signalling. These results provide new impetus for the urgent need to investigate the functional and behavioural substrates of prenatal cannabinoid exposure in both the male offspring and the female offspring.
Collapse
Affiliation(s)
- Antonia Manduca
- Department of Science, Section of Biomedical Sciences and TechnologiesUniversity “Roma Tre”RomeItaly
- INSERM, INMEDAix Marseille UniversitéMarseilleFrance
- Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERMIndiana UniversityBloomingtonIndianaUSA
| | - Michela Servadio
- Department of Science, Section of Biomedical Sciences and TechnologiesUniversity “Roma Tre”RomeItaly
| | - Francesca Melancia
- Department of Science, Section of Biomedical Sciences and TechnologiesUniversity “Roma Tre”RomeItaly
| | - Sara Schiavi
- Department of Science, Section of Biomedical Sciences and TechnologiesUniversity “Roma Tre”RomeItaly
| | - Olivier J. Manzoni
- INSERM, INMEDAix Marseille UniversitéMarseilleFrance
- Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERMIndiana UniversityBloomingtonIndianaUSA
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and TechnologiesUniversity “Roma Tre”RomeItaly
| |
Collapse
|
41
|
Nashed MG, Hardy DB, Laviolette SR. Prenatal Cannabinoid Exposure: Emerging Evidence of Physiological and Neuropsychiatric Abnormalities. Front Psychiatry 2020; 11:624275. [PMID: 33519564 PMCID: PMC7841012 DOI: 10.3389/fpsyt.2020.624275] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/14/2020] [Indexed: 12/22/2022] Open
Abstract
Clinical reports of cannabis use prevalence during pregnancy vary widely from 3% to upwards of 35% in North America; this disparity likely owing to underestimates from self-reporting in many cases. The rise in cannabis use is mirrored by increasing global legalization and the overall perceptions of safety, even during pregnancy. These trends are further compounded by a lack of evidence-based policy and guidelines for prenatal cannabis use, which has led to inconsistent messaging by healthcare providers and medically licensed cannabis dispensaries regarding prenatal cannabis use for treatment of symptoms, such as nausea. Additionally, the use of cannabis to self-medicate depression and anxiety during pregnancy is a growing medical concern. This review aims to summarize recent findings of clinical and preclinical data on neonatal outcomes, as well as long-term physiological and neurodevelopmental outcomes of prenatal cannabis exposure. Although many of the outcomes under investigation have produced mixed results, we consider these data in light of the unique challenges facing cannabis research. In particular, the limited longitudinal clinical studies available have not previously accounted for the exponential increase in (-)-Δ9- tetrahydrocannabinol (Δ9-THC; the psychoactive compound in cannabis) concentrations found in cannabis over the past two decades. Polydrug use and the long-term effects of individual cannabis constituents [Δ9-THC vs. cannabidiol (CBD)] are also understudied, along with sex-dependent outcomes. Despite these limitations, prenatal cannabis exposure has been linked to low birth weight, and emerging evidence suggests that prenatal exposure to Δ9-THC, which crosses the placenta and impacts placental development, may have wide-ranging physiological and neurodevelopmental consequences. The long-term effects of these changes require more rigorous investigation, though early reports suggest Δ9-THC increases the risk of cognitive impairment and neuropsychiatric disease, including psychosis, depression, anxiety, and sleep disorders. In light of the current trends in the perception and use of cannabis during pregnancy, we emphasize the social and medical imperative for more rigorous investigation of the long-term effects of prenatal cannabis exposure.
Collapse
Affiliation(s)
- Mina G Nashed
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Daniel B Hardy
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada.,Department of Obstetrics & Gynecology, University of Western Ontario, London, ON, Canada
| | - Steven R Laviolette
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada.,Department of Psychiatry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
42
|
Philippot G, Forsberg E, Tahan C, Viberg H, Fredriksson R. A Single δ 9-Tetrahydrocannabinol (THC) Dose During Brain Development Affects Markers of Neurotrophy, Oxidative Stress, and Apoptosis. Front Pharmacol 2019; 10:1156. [PMID: 31636565 PMCID: PMC6787269 DOI: 10.3389/fphar.2019.01156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/09/2019] [Indexed: 11/13/2022] Open
Abstract
δ9-tetrahydrocannabinol (THC) is one of the most used drugs during pregnancy and lactation and efficiently crosses the placental and blood–brain barriers. Despite the recent legalization initiatives worldwide, the adverse outcome pathway (AOP) of THC following exposure during brain development is incompletely understood. We have previously reported that a single injection of THC on postnatal day (PND) 10 altered adult spontaneous behavior and habituation rates in adult mice. Similar behavioral alterations have been reported following PND 10 exposure to the commonly used over-the-counter analgesic acetaminophen (AAP; also known as paracetamol); as both THC and AAP interact with the endocannabinoid system, we hypothesize that this system might be involved in the AOP of both these pharmaceuticals/drugs. Here, we report that a single THC dose on PND 10 decreased transcript levels of tropomyosin receptor kinase b (Trkb) 24 h after exposure in both the frontal and parietal cortex, and in the hippocampus in mice. An increase in the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1) ratio were also found in both the parietal cortex and hippocampus following neonatal exposure to THC. In addition, THC exposure increased transcript levels of cannabinoid receptor type 1 (Cb1r) in the parietal cortex and increased the apoptosis regulator BAX in the frontal cortex. This study is important for mainly 3 reasons: 1) we are starting to get information on the developmental neurotoxic AOP of PND 10 exposure to THC, where we suggest that transcriptional changes of the neurotrophic receptor Trkb are central, 2) our PND 10 exposure model provides information relevant to human exposure and 3) since PND 10 exposure to AAP also decreased Trkb transcript levels, we suggest THC and AAP may share key events in their respective AOP through endocannabinoid-mediated alterations of the brain-derived neurotrophic factor (BDNF)-TRKB signaling pathway.
Collapse
Affiliation(s)
- Gaëtan Philippot
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Erica Forsberg
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| | - Caroline Tahan
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| | - Henrik Viberg
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
43
|
Hurd YL, Manzoni OJ, Pletnikov MV, Lee FS, Bhattacharyya S, Melis M. Cannabis and the Developing Brain: Insights into Its Long-Lasting Effects. J Neurosci 2019; 39:8250-8258. [PMID: 31619494 PMCID: PMC6794936 DOI: 10.1523/jneurosci.1165-19.2019] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 12/24/2022] Open
Abstract
The recent shift in sociopolitical debates and growing liberalization of cannabis use across the globe has raised concern regarding its impact on vulnerable populations, such as pregnant women and adolescents. Epidemiological studies have long demonstrated a relationship between developmental cannabis exposure and later mental health symptoms. This relationship is especially strong in people with particular genetic polymorphisms, suggesting that cannabis use interacts with genotype to increase mental health risk. Seminal animal research directly linked prenatal and adolescent exposure to delta-9-tetrahydrocannabinol, the major psychoactive component of cannabis, with protracted effects on adult neural systems relevant to psychiatric and substance use disorders. In this article, we discuss some recent advances in understanding the long-term molecular, epigenetic, electrophysiological, and behavioral consequences of prenatal, perinatal, and adolescent exposure to cannabis/delta-9-tetrahydrocannabinol. Insights are provided from both animal and human studies, including in vivo neuroimaging strategies.
Collapse
Affiliation(s)
- Yasmin L Hurd
- Department of Psychiatry and Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029,
| | - Olivier J Manzoni
- Aix Marseille University, Institut National de la Santé et de la Recherche Médicale, Institut de neurobiologie de la méditerranée, 13273 Marseille, France, and Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, Institut National de la Santé et de la Recherche Médicale, 13273 Marseille, France
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Francis S Lee
- Department of Psychiatry, Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, New York, New York 10065
| | - Sagnik Bhattacharyya
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom, and
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Cagliari, Italy
| |
Collapse
|
44
|
Scheyer AF, Melis M, Trezza V, Manzoni OJJ. Consequences of Perinatal Cannabis Exposure. Trends Neurosci 2019; 42:871-884. [PMID: 31604585 DOI: 10.1016/j.tins.2019.08.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/10/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022]
Abstract
Cannabis exposure during the perinatal period results in varied and significant consequences in affected offspring. The prevalence of detrimental outcomes of perinatal cannabis exposure is likely to increase in tandem with the broadening of legalization and acceptance of the drug. As such, it is crucial to highlight the immediate and protracted consequences of cannabis exposure on pre- and postnatal development. Here, we identify lasting changes in neurons' learning flexibility (synaptic plasticity) and epigenetic misregulation in animal models of perinatal cannabinoid exposure (using synthetic cannabinoids or active components of the cannabis plant), in addition to significant alterations in social behavior and executive functions. These findings are supported by epidemiological data indicating similar behavioral outcomes throughout life in human offspring exposed to cannabis during pregnancy. Further, we indicate important lingering questions regarding accurate modeling of perinatal cannabis exposure as well as the need for sex- and age-dependent outcome measures in future studies.
Collapse
Affiliation(s)
- Andrew F Scheyer
- INMED, INSERM U1249, Marseille, France; Aix-Marseille University, Provence, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Aix-Marseille University, Provence, France/Indiana University, Bloomington, IN, USA
| | - Miriam Melis
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy; National Institute of Neuroscience, Cagliari, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University 'Roma Tre', Rome, Italy
| | - Olivier J J Manzoni
- INMED, INSERM U1249, Marseille, France; Aix-Marseille University, Provence, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Aix-Marseille University, Provence, France/Indiana University, Bloomington, IN, USA.
| |
Collapse
|
45
|
Leung MCK, Silva MH, Palumbo AJ, Lohstroh PN, Koshlukova SE, DuTeaux SB. Adverse outcome pathway of developmental neurotoxicity resulting from prenatal exposures to cannabis contaminated with organophosphate pesticide residues. Reprod Toxicol 2019; 85:12-18. [PMID: 30668982 DOI: 10.1016/j.reprotox.2019.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/07/2018] [Accepted: 01/14/2019] [Indexed: 01/11/2023]
Abstract
There is growing concern that increased use of medical and recreational cannabis may result in increased exposure to contaminants on the cannabis, such as pesticides. Several states are moving towards implementing robust regulation of the sales, cultivation, and manufacture of cannabis products. However, there are challenges with creating health-protective regulations in an industry that, to date, has been largely unregulated. The focus of this publication is a theoretical examination of what may happen when women are exposed pre-conceptually or during pregnancy to cannabis contaminated with pesticides. We propose an adverse outcome pathway of concomitant prenatal exposure to cannabinoids and the organophosphate pesticide chlorpyrifos by curating what we consider to be the key events at the molecular, cellular, and tissue levels that result in developmental neurotoxicity. The implications of this adverse outcome pathway underscore the need to elucidate the potential developmental neurotoxicity that may result from prenatal exposure to pesticide-contaminated cannabis.
Collapse
Affiliation(s)
- Maxwell C K Leung
- Department of Pesticide Regulation, California Environmental Protection Agency, Sacramento, 1001 I Street, Sacramento, CA 95812, United States.
| | - Marilyn H Silva
- Department of Pesticide Regulation, California Environmental Protection Agency, Sacramento, 1001 I Street, Sacramento, CA 95812, United States
| | - Amanda J Palumbo
- Department of Pesticide Regulation, California Environmental Protection Agency, Sacramento, 1001 I Street, Sacramento, CA 95812, United States
| | - Peter N Lohstroh
- Department of Pesticide Regulation, California Environmental Protection Agency, Sacramento, 1001 I Street, Sacramento, CA 95812, United States
| | - Svetlana E Koshlukova
- Department of Pesticide Regulation, California Environmental Protection Agency, Sacramento, 1001 I Street, Sacramento, CA 95812, United States
| | - Shelley B DuTeaux
- Department of Pesticide Regulation, California Environmental Protection Agency, Sacramento, 1001 I Street, Sacramento, CA 95812, United States
| |
Collapse
|
46
|
The Role of AMPARs in the Maturation and Integration of Caudal Ganglionic Eminence-Derived Interneurons into Developing Hippocampal Microcircuits. Sci Rep 2019; 9:5435. [PMID: 30931998 PMCID: PMC6443733 DOI: 10.1038/s41598-019-41920-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/19/2019] [Indexed: 12/25/2022] Open
Abstract
In the hippocampal CA1, caudal ganglionic eminence (CGE)-derived interneurons are recruited by activation of glutamatergic synapses comprising GluA2-containing calcium-impermeable AMPARs and exert inhibitory regulation of the local microcircuit. However, the role played by AMPARs in maturation of the developing circuit is unknown. We demonstrate that elimination of the GluA2 subunit (GluA2 KO) of AMPARs in CGE-derived interneurons, reduces spontaneous EPSC frequency coupled to a reduction in dendritic glutamatergic synapse density. Removal of GluA1&2&3 subunits (GluA1-3 KO) in CGE-derived interneurons, almost completely eliminated sEPSCs without further reducing synapse density, but increased dendritic branching. Moreover, in GluA1-3 KOs, the number of interneurons invading the hippocampus increased in the early postnatal period but converged with WT numbers later due to increased apoptosis. However, the CCK-containing subgroup increased in number, whereas the VIP-containing subgroup decreased. Both feedforward and feedback inhibitory input onto pyramidal neurons was decreased in GluA1-3 KO. These combined anatomical, synaptic and circuit alterations, were accompanied with a wide range of behavioural abnormalities in GluA1-3 KO mice compared to GluA2 KO and WT. Thus, AMPAR subunits differentially contribute to numerous aspects of the development and maturation of CGE-derived interneurons and hippocampal circuitry that are essential for normal behaviour.
Collapse
|
47
|
Trexler KR, Nass SR, Crowe MS, Gross JD, Jones MS, McKitrick AW, Siderovski DP, Kinsey SG. Novel behavioral assays of spontaneous and precipitated THC withdrawal in mice. Drug Alcohol Depend 2018; 191:14-24. [PMID: 30071445 PMCID: PMC6404969 DOI: 10.1016/j.drugalcdep.2018.05.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 05/15/2018] [Accepted: 05/28/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND A subset of cannabis users develop some degree of Cannabis Use Disorder (CUD). Although behavioral therapy has some success in treating CUD, many users relapse, often citing altered sleep, mood, and irritability. Preclinical animal tests of cannabinoid withdrawal focus primarily on somatic-related behaviors precipitated by a cannabinoid receptor antagonist. The goal of the present study was to develop novel cannabinoid withdrawal assays that are either antagonist-precipitated or spontaneously induced by abstinence. METHODS C57BL/6 J mice were repeatedly administered the phytocannabinoid Δ9-tetrahydrocannabinol (THC; 1, 10 or 50 mg/kg, s.c.), the synthetic cannabinoid receptor agonist JWH-018 (1 mg/kg, s.c.), or vehicle (1:1:18 parts ethanol:Kolliphor EL:saline, s.c.) for 6 days. Withdrawal was precipitated with the cannabinoid receptor inverse agonist rimonabant (3 mg/kg, i.p.) or elicited via abstinence (i.e., spontaneous withdrawal), and putative stress-related behavior was scored. Classic somatic signs of cannabinoid withdrawal were also quantified. RESULTS Precipitated THC withdrawal significantly increased plasma corticosterone. Precipitated withdrawal from either THC or JWH-018 suppressed marble burying, increased struggling in the tail suspension test, and elicited somatic withdrawal behaviors. The monoacylglycerol lipase inhibitor JZL184 attenuated somatic precipitated withdrawal but had no effect on marble burying or struggling. Spontaneous THC or JWH-018 withdrawal-induced paw tremors, head twitches, and struggled in the tail suspension test after 24-48 h abstinence. JZL184 or THC attenuated these spontaneous withdrawal-induced behaviors. CONCLUSION Outcomes from tail suspension and marble burying tests reveal that THC withdrawal is multifaceted, eliciting and suppressing behaviors in these tests, in addition to inducing well-documented somatic signs of withdrawal.
Collapse
Affiliation(s)
| | - Sara R. Nass
- Department of Psychology, West Virginia University; Department of Pharmacology & Toxicology, Virginia Commonwealth University
| | - Molly S. Crowe
- Department of Psychology, West Virginia University; Department of Physiology & Biophysics, Virginia Commonwealth University
| | - Joshua D. Gross
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University
| | | | | | - David P. Siderovski
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University
| | | |
Collapse
|
48
|
Bara A, Manduca A, Bernabeu A, Borsoi M, Serviado M, Lassalle O, Murphy M, Wager-Miller J, Mackie K, Pelissier-Alicot AL, Trezza V, Manzoni OJ. Sex-dependent effects of in utero cannabinoid exposure on cortical function. eLife 2018; 7:e36234. [PMID: 30201092 PMCID: PMC6162091 DOI: 10.7554/elife.36234] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/15/2018] [Indexed: 12/11/2022] Open
Abstract
Cannabinoids can cross the placenta, thus may interfere with fetal endocannabinoid signaling during neurodevelopment, causing long-lasting deficits. Despite increasing reports of cannabis consumption during pregnancy, the protracted consequences of prenatal cannabinoid exposure (PCE) remain incompletely understood. Here, we report sex-specific differences in behavioral and neuronal deficits in the adult progeny of rat dams exposed to low doses of cannabinoids during gestation. In males, PCE reduced social interaction, ablated endocannabinoid long-term depression (LTD) and heightened excitability of prefrontal cortex pyramidal neurons, while females were spared. Group 1 mGluR and endocannabinoid signaling regulate emotional behavior and synaptic plasticity. Notably, sex-differences following PCE included levels of mGluR1/5 and TRPV1R mRNA. Finally, positive allosteric modulation of mGlu5 and enhancement of anandamide levels restored LTD and social interaction in PCE adult males. Together, these results highlight marked sexual differences in the effects of PCE and introduce strategies for reversing detrimental effects of PCE.
Collapse
Affiliation(s)
- Anissa Bara
- Aix Marseille University, INSERM, INMEDMarseilleFrance
- Cannalab, Cannabinoids Neuroscience Research International Associated LaboratoryIndiana UniversityIndianaUnited States
| | - Antonia Manduca
- Aix Marseille University, INSERM, INMEDMarseilleFrance
- Cannalab, Cannabinoids Neuroscience Research International Associated LaboratoryIndiana UniversityIndianaUnited States
- Section of Biomedical Sciences and Technologies, Department of ScienceUniversity Roma TreRomeItaly
| | - Axel Bernabeu
- Aix Marseille University, INSERM, INMEDMarseilleFrance
- Cannalab, Cannabinoids Neuroscience Research International Associated LaboratoryIndiana UniversityIndianaUnited States
- APHMCHU Conception, Service de PsychiatrieMarseilleFrance
| | - Milene Borsoi
- Aix Marseille University, INSERM, INMEDMarseilleFrance
- Cannalab, Cannabinoids Neuroscience Research International Associated LaboratoryIndiana UniversityIndianaUnited States
| | - Michela Serviado
- Section of Biomedical Sciences and Technologies, Department of ScienceUniversity Roma TreRomeItaly
| | - Olivier Lassalle
- Aix Marseille University, INSERM, INMEDMarseilleFrance
- Cannalab, Cannabinoids Neuroscience Research International Associated LaboratoryIndiana UniversityIndianaUnited States
| | - Michelle Murphy
- Cannalab, Cannabinoids Neuroscience Research International Associated LaboratoryIndiana UniversityIndianaUnited States
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonUnited States
- Gill CentreIndiana UniversityBloomingtonUnited States
| | - Jim Wager-Miller
- Cannalab, Cannabinoids Neuroscience Research International Associated LaboratoryIndiana UniversityIndianaUnited States
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonUnited States
- Gill CentreIndiana UniversityBloomingtonUnited States
| | - Ken Mackie
- Cannalab, Cannabinoids Neuroscience Research International Associated LaboratoryIndiana UniversityIndianaUnited States
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonUnited States
- Gill CentreIndiana UniversityBloomingtonUnited States
| | - Anne-Laure Pelissier-Alicot
- Aix Marseille University, INSERM, INMEDMarseilleFrance
- Cannalab, Cannabinoids Neuroscience Research International Associated LaboratoryIndiana UniversityIndianaUnited States
- APHMCHU Conception, Service de PsychiatrieMarseilleFrance
- APHMCHU Timone Adultes, Service de Médecine LégaleMarseilleFrance
| | - Viviana Trezza
- Section of Biomedical Sciences and Technologies, Department of ScienceUniversity Roma TreRomeItaly
| | - Olivier J Manzoni
- Aix Marseille University, INSERM, INMEDMarseilleFrance
- Cannalab, Cannabinoids Neuroscience Research International Associated LaboratoryIndiana UniversityIndianaUnited States
| |
Collapse
|
49
|
Huizenga MN, Fureman BE, Soltesz I, Stella N. Proceedings of the Epilepsy Foundation's 2017 Cannabinoids in Epilepsy Therapy Workshop. Epilepsy Behav 2018; 85:237-242. [PMID: 29908905 PMCID: PMC6372237 DOI: 10.1016/j.yebeh.2018.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 11/19/2022]
Affiliation(s)
- Megan N Huizenga
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC, United States.
| | - Brandy E Fureman
- Research and New Therapies, Epilepsy Foundation of America, Landover, MD, United States
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University, Stanford, CA, United States
| | - Nephi Stella
- Department of Pharmacology, University of Washington, Seattle, WA, United States; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States
| |
Collapse
|
50
|
Kotzadimitriou D, Nissen W, Paizs M, Newton K, Harrison PJ, Paulsen O, Lamsa K. Neuregulin 1 Type I Overexpression Is Associated with Reduced NMDA Receptor-Mediated Synaptic Signaling in Hippocampal Interneurons Expressing PV or CCK. eNeuro 2018; 5:ENEURO.0418-17.2018. [PMID: 29740596 PMCID: PMC5938717 DOI: 10.1523/eneuro.0418-17.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/25/2018] [Accepted: 02/28/2018] [Indexed: 11/21/2022] Open
Abstract
Hypofunction of N-methyl-d-aspartate receptors (NMDARs) in inhibitory GABAergic interneurons is implicated in the pathophysiology of schizophrenia (SZ), a heritable disorder with many susceptibility genes. However, it is still unclear how SZ risk genes interfere with NMDAR-mediated synaptic transmission in diverse inhibitory interneuron populations. One putative risk gene is neuregulin 1 (NRG1), which signals via the receptor tyrosine kinase ErbB4, itself a schizophrenia risk gene. The type I isoform of NRG1 shows increased expression in the brain of SZ patients, and ErbB4 is enriched in GABAergic interneurons expressing parvalbumin (PV) or cholecystokinin (CCK). Here, we investigated ErbB4 expression and synaptic transmission in interneuronal populations of the hippocampus of transgenic mice overexpressing NRG1 type I (NRG1tg-type-I mice). Immunohistochemical analyses confirmed that ErbB4 was coexpressed with either PV or CCK in hippocampal interneurons, but we observed a reduced number of ErbB4-immunopositive interneurons in the NRG1tg-type-I mice. NMDAR-mediated currents in interneurons expressing PV (including PV+ basket cells) or CCK were reduced in NRG1tg-type-I mice compared to their littermate controls. We found no difference in AMPA receptor-mediated currents. Optogenetic activation (5 pulses at 20 Hz) of local glutamatergic fibers revealed a decreased NMDAR-mediated contribution to disynaptic GABAergic inhibition of pyramidal cells in the NRG1tg-type-I mice. GABAergic synaptic transmission from either PV+ or CCK+ interneurons, and glutamatergic transmission onto pyramidal cells, did not significantly differ between genotypes. The results indicate that synaptic NMDAR-mediated signaling in hippocampal interneurons is sensitive to chronically elevated NGR1 type I levels. This may contribute to the pathophysiological consequences of increased NRG1 expression in SZ.
Collapse
Affiliation(s)
| | - Wiebke Nissen
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Melinda Paizs
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, 6720, Hungary
| | - Kathryn Newton
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Paul J. Harrison
- Department of Psychiatry, University of Oxford, and Oxford Health NHS Foundation Trust, Oxford, UK
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Karri Lamsa
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, 6720, Hungary
| |
Collapse
|