1
|
Karatayev O, Collier AD, Targoff SR, Leibowitz SF. Neurological Disorders Induced by Drug Use: Effects of Adolescent and Embryonic Drug Exposure on Behavioral Neurodevelopment. Int J Mol Sci 2024; 25:8341. [PMID: 39125913 PMCID: PMC11313660 DOI: 10.3390/ijms25158341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Clinical studies demonstrate that the risk of developing neurological disorders is increased by overconsumption of the commonly used drugs, alcohol, nicotine and cannabis. These drug-induced neurological disorders, which include substance use disorder (SUD) and its co-occurring emotional conditions such as anxiety and depression, are observed not only in adults but also with drug use during adolescence and after prenatal exposure to these drugs, and they are accompanied by long-lasting disturbances in brain development. This report provides overviews of clinical and preclinical studies, which confirm these adverse effects in adolescents and the offspring prenatally exposed to the drugs and include a more in-depth description of specific neuronal systems, their neurocircuitry and molecular mechanisms, affected by drug exposure and of specific techniques used to determine if these effects in the brain are causally related to the behavioral disturbances. With analysis of further studies, this review then addresses four specific questions that are important for fully understanding the impact that drug use in young individuals can have on future pregnancies and their offspring. Evidence demonstrates that the adverse effects on their brain and behavior can occur: (1) at low doses with short periods of drug exposure during pregnancy; (2) after pre-conception drug use by both females and males; (3) in subsequent generations following the initial drug exposure; and (4) in a sex-dependent manner, with drug use producing a greater risk in females than males of developing SUDs with emotional conditions and female offspring after prenatal drug exposure responding more adversely than male offspring. With the recent rise in drug use by adolescents and pregnant women that has occurred in association with the legalization of cannabis and increased availability of vaping tools, these conclusions from the clinical and preclinical literature are particularly alarming and underscore the urgent need to educate young women and men about the possible harmful effects of early drug use and to seek novel therapeutic strategies that might help to limit drug use in young individuals.
Collapse
Affiliation(s)
| | | | | | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA; (O.K.); (S.R.T.)
| |
Collapse
|
2
|
Peterson CS, Baglot SL, Sallam NA, Mina S, Hill MN, Borgland SL. Oral pre- and early postnatal cannabis exposure disinhibits ventral tegmental area dopamine neuron activity but does not influence cocaine preference in offspring in mice. J Neurosci Res 2024; 102:e25369. [PMID: 39037062 DOI: 10.1002/jnr.25369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 07/23/2024]
Abstract
Cannabis consumption has increased from 1.5% to 2.5% in Canada between 2012 and 2019. Clinical studies have indicated effects of prenatal cannabis exposure on birth weight, substance use, and neurodevelopmental disorders, but are confounded by several difficult to control variables. Animal models allow for examination of the mechanism of cannabis-induced changes in neurodevelopment and behavior, while controlling dose and timing. Several animal models of prenatal cannabis exposure exist which provide varying levels of construct validity, control of dose, and exposure to maternal stress. Using a voluntary oral consumption model, mouse dams received 5 mg/kg Δ9-tetrahydrocannabinol (THC) whole cannabis oil in peanut butter daily from gestational day 1 (GD1) to postnatal day 10 (PD10). At GD1, GD18, PD1, PD10, and PD15, maternal plasma was collected; pup brains were collected from GD18 onward. Pup brains had higher levels of THC and cannabidiol at each time point, each of which persisted in maternal plasma and pup brains past the end of treatment (PD15). Male and female adolescent offspring were examined for changes to ventral tegmental area (VTA) dopamine neuron activity and cocaine-seeking behavior. Prenatal and early postnatal (GD1-PD10) cannabis-exposed male, but not female mice had decreased gamma-aminobutyric acid (GABAergic) input, depolarized resting membrane potential, and increased spontaneous firing of VTA dopamine neurons. Cannabis-exposed offspring showed faster decay of N-methyl-D-aspartate (NMDA) currents in both sexes. However, no differences in cocaine-seeking behavior were noted. These data characterize a voluntary prenatal cannabis exposure model and demonstrates VTA dopamine neuronal activity is disinhibited in offspring.
Collapse
Affiliation(s)
- Colleen S Peterson
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Samantha L Baglot
- Department of Cell Biology and Anatomy, Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nada A Sallam
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt, Cairo University, Cairo, Egypt
| | - Sarah Mina
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Matthew N Hill
- Department of Cell Biology and Anatomy, Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
3
|
Collier AD, Abdulai AR, Leibowitz SF. Utility of the Zebrafish Model for Studying Neuronal and Behavioral Disturbances Induced by Embryonic Exposure to Alcohol, Nicotine, and Cannabis. Cells 2023; 12:2505. [PMID: 37887349 PMCID: PMC10605371 DOI: 10.3390/cells12202505] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
It is estimated that 5% of pregnant women consume drugs of abuse during pregnancy. Clinical research suggests that intake of drugs during pregnancy, such as alcohol, nicotine and cannabis, disturbs the development of neuronal systems in the offspring, in association with behavioral disturbances early in life and an increased risk of developing drug use disorders. After briefly summarizing evidence in rodents, this review focuses on the zebrafish model and its inherent advantages for studying the effects of embryonic exposure to drugs of abuse on behavioral and neuronal development, with an emphasis on neuropeptides known to promote drug-related behaviors. In addition to stimulating the expression and density of peptide neurons, as in rodents, zebrafish studies demonstrate that embryonic drug exposure has marked effects on the migration, morphology, projections, anatomical location, and peptide co-expression of these neurons. We also describe studies using advanced methodologies that can be applied in vivo in zebrafish: first, to demonstrate a causal relationship between the drug-induced neuronal and behavioral disturbances and second, to discover underlying molecular mechanisms that mediate these effects. The zebrafish model has great potential for providing important information regarding the development of novel and efficacious therapies for ameliorating the effects of early drug exposure.
Collapse
Affiliation(s)
| | | | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
4
|
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: 3] [Impact Index Per Article: 3.0] [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
|
5
|
Stankovic IN, Colak D. Prenatal Drugs and Their Effects on the Developing Brain: Insights From Three-Dimensional Human Organoids. Front Neurosci 2022; 16:848648. [PMID: 35401083 PMCID: PMC8990163 DOI: 10.3389/fnins.2022.848648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Decades of research have unequivocally demonstrated that fetal exposure to both recreational and prescription drugs in utero negatively impacts the developing brain. More recently, the application of cutting-edge techniques in neurodevelopmental research has attempted to identify how the fetal brain responds to specific environmental stimuli. Meanwhile, human fetal brain studies still encounter ethical considerations and technical limitations in tissue collection. Human-induced pluripotent stem cell (iPSC)-derived brain organoid technology has emerged as a powerful alternative to examine fetal neurobiology. In fact, human 3D organoid tissues recapitulate cerebral development during the first trimester of pregnancy. In this review, we aim to provide a comprehensive summary of fetal brain metabolic studies related to drug abuse in animal and human models. Additionally, we will discuss the current challenges and prospects of using brain organoids for large-scale metabolomics. Incorporating cutting-edge techniques in human brain organoids may lead to uncovering novel molecular and cellular mechanisms of neurodevelopment, direct novel therapeutic approaches, and raise new exciting questions.
Collapse
Affiliation(s)
- Isidora N. Stankovic
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
- *Correspondence: Isidora N. Stankovic,
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, United States
- Gale & Ira Drukier Institute for Children’s Health, Weill Cornell Medicine, Cornell University, New York, NY, United States
- Dilek Colak,
| |
Collapse
|
6
|
Silva M, Kwok RKH. Use of Computational Toxicology Tools to Predict In Vivo Endpoints Associated with Mode of Action and the Endocannabinoid System: A Case Study with Chlorpyrifos, Chlorpyrifos-oxon and Δ9Tetrahydrocannabinol. Curr Res Toxicol 2022; 3:100064. [PMID: 35243363 PMCID: PMC8860916 DOI: 10.1016/j.crtox.2022.100064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/16/2022] [Accepted: 02/03/2022] [Indexed: 01/04/2023] Open
|
7
|
Fundamentals of the Dynorphins/Kappa Opioid Receptor System: From Distribution to Signaling and Function. Handb Exp Pharmacol 2022; 271:3-21. [PMID: 33754230 PMCID: PMC9013522 DOI: 10.1007/164_2021_433] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This chapter provides a general introduction to the dynorphins (DYNs)/kappa opioid receptor (KOR) system, including DYN peptides, neuroanatomy of the DYNs/KOR system, cellular signaling, and in vivo behavioral effects of KOR activation and inhibition. It is intended to serve as a primer for the book and to provide a basic background for the chapters in the book.
Collapse
|
8
|
Allen MC, Moog NK, Buss C, Yen E, Gustafsson HC, Sullivan EL, Graham AM. Co-occurrence of preconception maternal childhood adversity and opioid use during pregnancy: Implications for offspring brain development. Neurotoxicol Teratol 2021; 88:107033. [PMID: 34601061 PMCID: PMC8578395 DOI: 10.1016/j.ntt.2021.107033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/11/2022]
Abstract
Understanding of the effects of in utero opioid exposure on neurodevelopment is a priority given the recent dramatic increase in opioid use among pregnant individuals. However, opioid abuse does not occur in isolation-pregnant individuals abusing opioids often have a significant history of adverse experiences in childhood, among other co-occurring factors. Understanding the specific pathways in which these frequently co-occurring factors may interact and cumulatively influence offspring brain development in utero represents a priority for future research in this area. We highlight maternal history of childhood adversity (CA) as one such co-occurring factor that is more prevalent among individuals using opioids during pregnancy and which is increasingly shown to affect offspring neurodevelopment through mechanisms beginning in utero. Despite the high incidence of CA history in pregnant individuals using opioids, we understand very little about the effects of comorbid prenatal opioid exposure and maternal CA history on fetal brain development. Here, we first provide an overview of current knowledge regarding effects of opioid exposure and maternal CA on offspring neurodevelopment that may occur during gestation. We then outline potential mechanistic pathways through which these factors might have interactive and cumulative influences on offspring neurodevelopment as a foundation for future research in this area.
Collapse
Affiliation(s)
- Madeleine C Allen
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Nora K Moog
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Psychology, Luisenstrasse 57, 10117 Berlin, Germany
| | - Claudia Buss
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Psychology, Luisenstrasse 57, 10117 Berlin, Germany; Development, Health and Disease Research Program, University of California, Irvine, 837 Health Sciences Drive, Irvine, California 92697, United States
| | - Elizabeth Yen
- Department of Pediatrics, Tufts Medical Center, Boston, MA 02111, United States
| | - Hanna C Gustafsson
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Elinor L Sullivan
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States; Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185(th) Ave., Beaverton, OR 97006, United States; Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Alice M Graham
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States.
| |
Collapse
|
9
|
Shah DS, Turner EL, Chroust AJ, Duvall KL, Wood DL, Bailey BA. Marijuana use in opioid exposed pregnancy increases risk of preterm birth. J Matern Fetal Neonatal Med 2021; 35:8456-8461. [PMID: 34582287 DOI: 10.1080/14767058.2021.1980532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND The prevalence of opioid use disorder has increased across the United States, but the rural population of Appalachia has been disproportionately impacted. Concurrently, the slow, but steady progress in the legalization of marijuana may be affecting perception of marijuana use in pregnancy. However, marijuana use in pregnancy has been associated with adverse perinatal outcomes. Concomitant use of opioids and marijuana in pregnancy has not been evaluated. OBJECTIVE The primary aim of the study was to evaluate the association between confirmed marijuana use in late pregnancy and preterm birth in opioid-exposed pregnancies. METHODOLOGY A retrospective chart review was conducted that included all births from July 2011 to June 2016 from 6 delivery hospitals in South-Central Appalachia. Out of 18,732 births, 2368 singleton pregnancies indicated opioid use and met remaining inclusion criteria, with 108 of these mothers testing positive for marijuana at delivery. Independent sample t-test and Chi-Square analyses compared marijuana and non-marijuana exposed groups on maternal and neonatal outcomes. Regression analyses controlled for confounding variables in predicting neonatal abstinence syndrome (NAS), NICU admission, preterm birth, small for gestational age, and low birth weight outcomes as shown in Table 1. RESULTS Neonates born to marijuana-positive women in opioid-exposed pregnancy were more likely to be born preterm, small for gestational age, have low birth weight, and be admitted to NICU. After statistically controlling for parity, marital status, tobacco and benzodiazepine use, preterm birth and low birth weight remained statistically significant with aOR of 2.35 (1.30-4.24) and 2.01 (1.18-3.44), respectively. CONCLUSIONS Maternal use of marijuana in any opioid-exposed pregnancy may increase risk of preterm birth and low-birth weight infants. Prospective studies need to examine the dose and timing of marijuana and opioid use in pregnancy to better delineate perinatal effects. Nonetheless, pregnant women using opioids, including recommended medication assisted treatment for opioid use disorder, should be educated about the risks of concurrent marijuana use during pregnancy and may need to be counseled to abstain from marijuana use during pregnancy for an optimal outcome.
Collapse
Affiliation(s)
- Darshan S Shah
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Emmitt L Turner
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Alyson J Chroust
- Department of Psychology, College of Arts and Sciences, East Tennessee State University, Johnson City, TN, USA
| | - Kathryn L Duvall
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - David L Wood
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Beth A Bailey
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| |
Collapse
|
10
|
Annen K, DomBourian MG. Perceptions on acceptability and reported consumption of marijuana by blood donors prior to donation in the recreational use state of Colorado, USA. Vox Sang 2021; 117:177-184. [PMID: 34291819 DOI: 10.1111/vox.13183] [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: 04/05/2021] [Revised: 06/24/2021] [Accepted: 07/07/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVES Blood donor opinions and behaviours regarding marijuana use are not well known nor is the potential impact to the blood supply. We sought to assess opinions and frequency of marijuana use in proximity to blood donation via a survey of blood donors at a hospital-based blood collection site in a state where recreational marijuana use has been legal since 2012. MATERIALS AND METHODS Blood donors at least 18 years of age who donated between 2014 and 2019 were surveyed electronically, with all responses kept anonymous to encourage engagement and accurate reporting. RESULTS Overall response rate was 8.03% (12,186 surveys sent with 979 responses). Of responding donors, 23.5% indicated that they felt that consuming various forms of marijuana was acceptable prior to blood donation. Marijuana use <72 h prior to blood donation was reported in all demographic groups surveyed except age 18-24 years. Of donors who reported daily marijuana use, 47.4% indicated >20 donations and 52.6% indicated apheresis platelet donation. CONCLUSION Nearly one quarter of responding blood donors feel that marijuana use is acceptable prior to blood donation, and nearly every demographic group surveyed indicated use of marijuana <72 h prior to donation. These results suggest the need for additional research to determine if marijuana-related metabolites in collected blood products negatively impact recipients, particularly vulnerable populations such as children and pregnant women. These results may inform whether changes in donor screening or testing for marijuana use are warranted.
Collapse
Affiliation(s)
- Kyle Annen
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, USA.,Department of Pathology and Laboratory Medicine, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Melkon G DomBourian
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, USA.,Department of Pathology and Laboratory Medicine, Children's Hospital Colorado, Aurora, Colorado, USA
| |
Collapse
|
11
|
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: 82] [Impact Index Per Article: 27.3] [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
|
12
|
Babić Leko M, Hof PR, Šimić G. Alterations and interactions of subcortical modulatory systems in Alzheimer's disease. PROGRESS IN BRAIN RESEARCH 2021; 261:379-421. [PMID: 33785136 DOI: 10.1016/bs.pbr.2020.07.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pathogenesis of Alzheimer's disease (AD) is not fully understood. Here we summarize current knowledge on the involvement of the serotonergic, noradrenergic, dopaminergic, cholinergic, and opioid systems in AD, emphasizing the importance of interactions between the serotonergic and the other subcortical modulatory systems during the progression of AD. In physiological conditions, all neurotransmitter systems function in concert and are interdependent at both the neuroanatomical and molecular levels. Through their early involvement in AD, cognitive and behavioral abilities that rely on their interactions also become disrupted. Considering that serotonin (5HT) regulates the release of noradrenaline (NA), dopamine (DA) and acetylcholine (ACh), any alteration in 5HT levels leads to disturbance of NA, DA, and ACh homeostasis in the brain. One of the earliest pathological changes during the prodromal phase of AD is a decrease of serotonergic transmission throughout the brain, with serotonergic receptors being also affected. Additionally, serotonergic and noradrenergic as well as serotonergic and dopaminergic nuclei are reciprocally interconnected. As the serotonergic dorsal raphe nucleus (DRN) is affected by pathological changes early in AD, and the noradrenergic locus coeruleus (LC) and dopaminergic ventral tegmental area (VTA) exhibit AD-related pathological changes, their connectivity also becomes altered in AD. Such disrupted interactions among neurotransmitter systems in AD can be used in the development of multi-target drugs. Some of the potential AD therapeutics (such as ASS234, RS67333, tropisetron) target multiple neurotransmitter systems to achieve the best possible improvement of cognitive and behavioral deficits observed in AD. Here, we review how serotonergic system interacts with other subcortical modulatory systems (noradrenergic, dopaminergic, cholinergic, and opioid systems) during AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goran Šimić
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia.
| |
Collapse
|
13
|
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: 42] [Impact Index Per Article: 10.5] [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
|
14
|
Roncero C, Valriberas-Herrero I, Mezzatesta-Gava M, Villegas JL, Aguilar L, Grau-López L. Cannabis use during pregnancy and its relationship with fetal developmental outcomes and psychiatric disorders. A systematic review. Reprod Health 2020; 17:25. [PMID: 32066469 PMCID: PMC7027300 DOI: 10.1186/s12978-020-0880-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/10/2020] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION This study analyze factors associated to cannabis use in pregnant women, its perceived availability, its risk perception and the relationship between prenatal exposure to cannabis and developmental and mental disorders. OBJECTIVES We present a review of the literature on cannabis use among pregnant women. The objective is to analyze factors associated to cannabis use during pregnancy and assess the potential effects of prenatal exposure to cannabis on the development of the fetus and the mental health of those exposed. METHODS Systematic review of studies on the maternal use of cannabis and the relationship between early exposure and the development of psychiatric disorders in the PubMed database until July 2018 in English and Spanish with the following keywords: Marijuana, Cannabinoids, Mental disorders, Pregnancy, Prenatal Cannabis Exposure, Risk factors. RESULTS The use of cannabis among pregnant women is frequent but it has not been extensively researched. Prenatal exposure to cannabis may be associated with affective symptoms and ADHD. CONCLUSIONS Mental healthcare professionals who treat women during their fertile life need to be able to explain the relationship between prenatal exposure to cannabis and the presence of developmental and mental disorders.
Collapse
Affiliation(s)
- Carlos Roncero
- Psychiatric Service, University of Salamanca Health Care Complex, Salamanca, Spain. .,Institute of Biomedicine of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain.
| | - Isabel Valriberas-Herrero
- Psychiatric Service, University of Salamanca Health Care Complex, Salamanca, Spain.,Institute of Biomedicine of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Marcela Mezzatesta-Gava
- Multidisciplinary Unit of Autism Spectrum Disorder (UnimTEA), Mental Health Department, Sant Joan de Deu Hospital, Barcelona, Spain
| | - José L Villegas
- Psychiatric Service, University of Salamanca Health Care Complex, Salamanca, Spain.,Institute of Biomedicine of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Lourdes Aguilar
- Psychiatric Service, University of Salamanca Health Care Complex, Salamanca, Spain.,Institute of Biomedicine of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Lara Grau-López
- Addiction and Dual Diagnosis Unit, Psychiatric Service, Vall d'Hebron University Hospital, CIBERSAM, Barcelona, Spain.,Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute, Barcelona, Spain.,Department of Psychiatry and Forensic Medicine, Autonomus University of Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
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: 8.0] [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
|
16
|
Goldfarb SS, Stanwood GD, Flynn HA, Graham DL. Developmental opioid exposures: Neurobiological underpinnings, behavioral impacts, and policy implications. Exp Biol Med (Maywood) 2019; 245:131-137. [PMID: 31630569 DOI: 10.1177/1535370219883601] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The devastating impact of opioid abuse and dependence on the individual, family, and society are well known but extremely difficult to combat. During pregnancy, opioid drugs and withdrawal also affect fetal brain development and newborn neural functions, in addition to maternal effects. Neonatal Abstinence Syndrome/Neonatal Opioid Withdrawal Syndrome (NAS/NOWS) rates have drastically increased in the US in the past decade. Solutions to this complex problem must be multi-faceted, which would be greatly enhanced by a translational, multidisciplinary understanding. Therefore, this mini-review incorporates biomedical, clinical, and policy aspects of opioid use during pregnancy. We review the known roles for endogenous opioids in mediating circuit formation and function in the developing brain, discuss how exogenous opioid drug use and addiction impact these processes in animal models and humans, and discuss the implications of these data on public policy. We suggest that some current policy initiatives produce unintended harm on both mothers and their children and delineate recommendations for how legislation could better contribute to addiction recovery and increase neural resilience in affected children. Impact statement Opioid abuse is a critical epidemic affecting individuals, families, and communities. This mini-review summarizes current literature on the impact of opioid drugs—including prescription pain relievers and illicit opioids—on neurobiological and neurobehavioral development. Using concepts related to the medical model of addiction as a brain disease, we review the public policy implications of these data and identify needs for future investigations.
Collapse
Affiliation(s)
- Samantha S Goldfarb
- Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Gregg D Stanwood
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.,Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Heather A Flynn
- Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, Tallahassee, FL 32306, USA.,Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Devon L Graham
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.,Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
17
|
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: 20.6] [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
|
18
|
Pinky PD, Bloemer J, Smith WD, Moore T, Hong H, Suppiramaniam V, Reed MN. Prenatal cannabinoid exposure and altered neurotransmission. Neuropharmacology 2019; 149:181-194. [PMID: 30771373 DOI: 10.1016/j.neuropharm.2019.02.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/18/2019] [Accepted: 02/12/2019] [Indexed: 11/26/2022]
Abstract
Marijuana is one of the most commonly used illicit drugs worldwide. In addition, use of synthetic cannabinoids is increasing, especially among adolescents and young adults. Although human studies have shown that the use of marijuana during pregnancy leads to adverse behavioral effects, such as deficiencies in attention and executive function in affected offspring, the rate of marijuana use among pregnant women is steadily increasing. Various aspects of human behavior including emotion, learning, and memory are dependent on complex interactions between multiple neurotransmitter systems that are especially vulnerable to alterations during the developmental period. Thus, exploration of neurotransmitter changes in response to prenatal cannabinoid exposure is crucial to develop an understanding of how homeostatic imbalance and various long-term neurobehavioral deficits manifest following the abuse of marijuana or other synthetic cannabinoids during pregnancy. Current literature confirms that vast alterations to neurotransmitter systems are present following prenatal cannabinoid exposure, and many of these alterations within the brain are region specific, time-dependent, and sexually dimorphic. In this review, we aim to provide a summary of observed changes to various neurotransmitter systems following cannabinoid exposure during pregnancy and to draw possible correlations to reported behavioral alterations in affected offspring.
Collapse
Affiliation(s)
- Priyanka D Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Warren D Smith
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Timothy Moore
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| | - Hao Hong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA.
| | - Miranda N Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA.
| |
Collapse
|
19
|
Endocannabinoid System and Alcohol Abuse Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1162:89-127. [PMID: 31332736 DOI: 10.1007/978-3-030-21737-2_6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Δ9-tetrahydrocannabinol (Δ9-THC), the primary active component in Cannabis sativa preparations such as hashish and marijuana, signals by binding to cell surface receptors. Two types of receptors have been cloned and characterized as cannabinoid (CB) receptors. CB1 receptors (CB1R) are ubiquitously present in the central nervous system (CNS) and are present in both inhibitory interneurons and excitatory neurons at the presynaptic terminal. CB2 receptors (CB2R) are demonstrated in microglial cells, astrocytes, and several neuron subpopulations and are present in both pre- and postsynaptic terminals. The majority of studies on these receptors have been conducted in the past two and half decades after the identification of the molecular constituents of the endocannabinoid (eCB) system that started with the characterization of CB1R. Subsequently, the seminal discovery was made, which suggested that alcohol (ethanol) alters the eCB system, thus establishing the contribution of the eCB system in the motivation to consume ethanol. Several preclinical studies have provided evidence that CB1R significantly contributes to the motivational and reinforcing properties of ethanol and that the chronic consumption of ethanol alters eCB transmitters and CB1R expression in the brain nuclei associated with addiction pathways. Additionally, recent seminal studies have further established the role of the eCB system in the development of ethanol-induced developmental disorders, such as fetal alcohol spectrum disorders (FASD). These results are augmented by in vitro and ex vivo studies, showing that acute and chronic treatment with ethanol produces physiologically relevant alterations in the function of the eCB system during development and in the adult stage. This chapter provides a current and comprehensive review of the literature concerning the role of the eCB system in alcohol abuse disorders (AUD).
Collapse
|
20
|
Age as a factor in stress and alcohol interactions: A critical role for the kappa opioid system. Alcohol 2018; 72:9-18. [PMID: 30322483 DOI: 10.1016/j.alcohol.2017.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/27/2017] [Accepted: 10/06/2017] [Indexed: 12/18/2022]
Abstract
The endogenous kappa opioid system has primarily been shown to be involved with a state of dysphoria and aversion. Stress and exposure to drugs of abuse, particularly alcohol, can produce similar states of unease and anxiety, implicating the kappa opioid system as a target of stress and alcohol. Numerous behavioral studies have demonstrated reduced sensitivity to manipulations of the kappa opioid system in early life relative to adulthood, and recent reports have shown that the kappa opioid system is functionally different across ontogeny. Given the global rise in early-life stress and alcohol consumption, understanding how the kappa opioid system responds and adapts to stress and/or alcohol exposure differently in early life and adulthood is imperative. Therefore, the objective of this review is to highlight and discuss studies examining the impact of early-life stress and/or alcohol on the kappa opioid system, with focus on the documented neuroadaptations that may contribute to future vulnerability to stress and/or increase the risk of relapse. We first provide a brief summary of the importance of studying the effects of stress and alcohol during early life (prenatal, neonatal/juvenile, and adolescence). We then discuss the literature on the effects of stress or alcohol during early life and adulthood on the kappa opioid system. Finally, we discuss the few studies that have shown interactions between stress and alcohol on the kappa opioid system and provide some discussion about the need for studies investigating the development of the kappa opioid system.
Collapse
|
21
|
Ryan SA, Ammerman SD, O’Connor ME, Gonzalez L, Patrick SW, Quigley J, Walker LR, Meek JY, Johnston M, Stellwagen L, Thomas J, Ware J. Marijuana Use During Pregnancy and Breastfeeding: Implications for Neonatal and Childhood Outcomes. Pediatrics 2018; 142:peds.2018-1889. [PMID: 30150209 DOI: 10.1542/peds.2018-1889] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Marijuana is one of the most widely used substances during pregnancy in the United States. Emerging data on the ability of cannabinoids to cross the placenta and affect the development of the fetus raise concerns about both pregnancy outcomes and long-term consequences for the infant or child. Social media is used to tout the use of marijuana for severe nausea associated with pregnancy. Concerns have also been raised about marijuana use by breastfeeding mothers. With this clinical report, we provide data on the current rates of marijuana use among pregnant and lactating women, discuss what is known about the effects of marijuana on fetal development and later neurodevelopmental and behavioral outcomes, and address implications for education and policy.
Collapse
Affiliation(s)
- Sheryl A. Ryan
- Department of Pediatrics, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Seth D. Ammerman
- Division of Adolescent Medicine, Department of Pediatrics, Stanford University and Teen Health Van, Stanford Children’s Health, Palo Alto, California
| | - Mary E. O’Connor
- Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado; and
- Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Tan KZ, Cunningham AM, Joshi A, Oei JL, Ward MC. Expression of kappa opioid receptors in developing rat brain - Implications for perinatal buprenorphine exposure. Reprod Toxicol 2018; 78:81-89. [PMID: 29635048 DOI: 10.1016/j.reprotox.2018.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 04/03/2018] [Accepted: 04/06/2018] [Indexed: 12/19/2022]
Abstract
Buprenorphine, a mu opioid receptor partial agonist and kappa opioid receptor (KOR) antagonist, is an emerging therapeutic agent for maternal opioid dependence in pregnancy and neonatal abstinence syndrome. However, the endogenous opioid system plays a critical role in modulating neurodevelopment and perinatal buprenorphine exposure may detrimentally influence this. To identify aspects of neurodevelopment vulnerable to perinatal buprenorphine exposure, we defined KOR protein expression and its cellular associations in normal rat brain from embryonic day 16 to postnatal day 23 with double-labelling immunohistochemistry. KOR was expressed on neural stem and progenitor cells (NSPCs), choroid plexus epithelium, subpopulations of cortical neurones and oligodendrocytes, and NSPCs and subpopulations of neurones in postnatal hippocampus. These distinct patterns of KOR expression suggest several pathways vulnerable to perinatal buprenorphine exposure, including proliferation, neurogenesis and neurotransmission. We thus suggest the cautious use of buprenorphine in both mothers and infants until its impact on neurodevelopment is better defined.
Collapse
Affiliation(s)
- Kathleen Z Tan
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Randwick, NSW 2031, Australia
| | - Anne M Cunningham
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Randwick, NSW 2031, Australia; Westfield Research Laboratories, Sydney Children's Hospital, High Street, Randwick, NSW 2031, Australia.
| | - Anjali Joshi
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Randwick, NSW 2031, Australia; Westfield Research Laboratories, Sydney Children's Hospital, High Street, Randwick, NSW 2031, Australia
| | - Ju Lee Oei
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Randwick, NSW 2031, Australia; The Royal Hospital for Women, Barker Street, Randwick, NSW 2031, Australia
| | - Meredith C Ward
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Randwick, NSW 2031, Australia; The Royal Hospital for Women, Barker Street, Randwick, NSW 2031, Australia; Westfield Research Laboratories, Sydney Children's Hospital, High Street, Randwick, NSW 2031, Australia.
| |
Collapse
|
23
|
Melis M, Frau R, Kalivas PW, Spencer S, Chioma V, Zamberletti E, Rubino T, Parolaro D. New vistas on cannabis use disorder. Neuropharmacology 2017; 124:62-72. [PMID: 28373077 DOI: 10.1016/j.neuropharm.2017.03.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 02/06/2023]
Abstract
Cannabis sativa preparations are the most consumed illicit drugs for recreational purposes worldwide, and the number of people seeking treatment for cannabis use disorder has dramatically increased in the last decades. Due to the recent decriminalization or legalization of cannabis use in the Western Countries, we may predict that the number of people suffering from cannabis use disorder will increase. Despite the increasing number of cannabis studies over the past two decades, we have gaps of scientific knowledge pertaining to the neurobiological consequences of long-term cannabis use. Moreover, no specific treatments for cannabis use disorders are currently available. In this review, we explore new research that may help fill these gaps. We discuss and provide a solution to the experimental limitation of a lack of rodent models of THC self-administration, and the importance this model can play in understanding the neurobiology of relapse and in providing a biological rationale for potential therapeutic targets. We also focus our attention on glial cells, commenting on recent preclinical evidence suggesting that alterations in microglia and astrocytes might contribute to the detrimental effects associated with cannabis abuse. Finally, due to the worrisome prevalence rates of cannabis use during pregnancy, we highlight the associations between cannabis use disorders during pregnancy and congenital disorders, describing the possible neuronal basis of vulnerability at molecular and circuit level. This article is part of the Special Issue entitled "A New Dawn in Cannabinoid Neurobiology".
Collapse
Affiliation(s)
- Miriam Melis
- Dept. of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - Roberto Frau
- Dept. of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, SC, USA
| | - Sade Spencer
- Department of Neuroscience, Medical University of South Carolina, SC, USA
| | - Vivian Chioma
- Department of Neuroscience, Medical University of South Carolina, SC, USA
| | - Erica Zamberletti
- Dept. of Biotechnology and Life Sciences, University of Insubria, Busto Arsizio (VA), Italy
| | - Tiziana Rubino
- Dept. of Biotechnology and Life Sciences, University of Insubria, Busto Arsizio (VA), Italy
| | - Daniela Parolaro
- Dept. of Biotechnology and Life Sciences, University of Insubria, Busto Arsizio (VA), Italy; Zardi Gori Foundation, Milan, Italy.
| |
Collapse
|
24
|
Laviolette SR. Cannabinoid regulation of opiate motivational processing in the mesolimbic system: the integrative roles of amygdala, prefrontal cortical and ventral hippocampal input pathways. Curr Opin Behav Sci 2017. [DOI: 10.1016/j.cobeha.2016.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
25
|
Maternal marijuana use has independent effects on risk for spontaneous preterm birth but not other common late pregnancy complications. Reprod Toxicol 2016; 62:77-86. [DOI: 10.1016/j.reprotox.2016.04.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 04/08/2016] [Accepted: 04/21/2016] [Indexed: 11/21/2022]
|
26
|
Chromothripsis and epigenomics complete causality criteria for cannabis- and addiction-connected carcinogenicity, congenital toxicity and heritable genotoxicity. Mutat Res 2016; 789:15-25. [PMID: 27208973 DOI: 10.1016/j.mrfmmm.2016.05.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/17/2016] [Accepted: 05/01/2016] [Indexed: 12/30/2022]
Abstract
The recent demonstration that massive scale chromosomal shattering or pulverization can occur abruptly due to errors induced by interference with the microtubule machinery of the mitotic spindle followed by haphazard chromosomal annealing, together with sophisticated insights from epigenetics, provide profound mechanistic insights into some of the most perplexing classical observations of addiction medicine, including cancerogenesis, the younger and aggressive onset of addiction-related carcinogenesis, the heritability of addictive neurocircuitry and cancers, and foetal malformations. Tetrahydrocannabinol (THC) and other addictive agents have been shown to inhibit tubulin polymerization which perturbs the formation and function of the microtubules of the mitotic spindle. This disruption of the mitotic machinery perturbs proper chromosomal segregation during anaphase and causes micronucleus formation which is the primary locus and cause of the chromosomal pulverization of chromothripsis and downstream genotoxic events including oncogene induction and tumour suppressor silencing. Moreover the complementation of multiple positive cannabis-cancer epidemiological studies, and replicated dose-response relationships with established mechanisms fulfils causal criteria. This information is also consistent with data showing acceleration of the aging process by drugs of addiction including alcohol, tobacco, cannabis, stimulants and opioids. THC shows a non-linear sigmoidal dose-response relationship in multiple pertinent in vitro and preclinical genotoxicity assays, and in this respect is similar to the serious major human mutagen thalidomide. Rising community exposure, tissue storage of cannabinoids, and increasingly potent phytocannabinoid sources, suggests that the threshold mutagenic dose for cancerogenesis will increasingly be crossed beyond the developing world, and raise transgenerational transmission of teratogenicity as an increasing concern.
Collapse
|
27
|
Alpár A, Di Marzo V, Harkany T. At the Tip of an Iceberg: Prenatal Marijuana and Its Possible Relation to Neuropsychiatric Outcome in the Offspring. Biol Psychiatry 2016; 79:e33-45. [PMID: 26549491 DOI: 10.1016/j.biopsych.2015.09.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 08/26/2015] [Accepted: 09/14/2015] [Indexed: 12/11/2022]
Abstract
Endocannabinoids regulate brain development via modulating neural proliferation, migration, and the differentiation of lineage-committed cells. In the fetal nervous system, (endo)cannabinoid-sensing receptors and the enzymatic machinery of endocannabinoid metabolism exhibit a cellular distribution map different from that in the adult, implying distinct functions. Notably, cannabinoid receptors serve as molecular targets for the psychotropic plant-derived cannabis constituent Δ(9)-tetrahydrocannainol, as well as synthetic derivatives (designer drugs). Over 180 million people use cannabis for recreational or medical purposes globally. Recreational cannabis is recognized as a niche drug for adolescents and young adults. This review combines data from human and experimental studies to show that long-term and heavy cannabis use during pregnancy can impair brain maturation and predispose the offspring to neurodevelopmental disorders. By discussing the mechanisms of cannabinoid receptor-mediated signaling events at critical stages of fetal brain development, we organize histopathologic, biochemical, molecular, and behavioral findings into a logical hypothesis predicting neuronal vulnerability to and attenuated adaptation toward environmental challenges (stress, drug exposure, medication) in children affected by in utero cannabinoid exposure. Conversely, we suggest that endocannabinoid signaling can be an appealing druggable target to dampen neuronal activity if pre-existing pathologies associate with circuit hyperexcitability. Yet, we warn that the lack of critical data from longitudinal follow-up studies precludes valid conclusions on possible delayed and adverse side effects. Overall, our conclusion weighs in on the ongoing public debate on cannabis legalization, particularly in medical contexts.
Collapse
Affiliation(s)
- Alán Alpár
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, Budapest, Hungary; Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Instituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Pozzuoli, Naples, Italy
| | - Tibor Harkany
- Division of Molecular Neurosciences, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
28
|
Grewen K, Salzwedel AP, Gao W. Functional Connectivity Disruption in Neonates with Prenatal Marijuana Exposure. Front Hum Neurosci 2015; 9:601. [PMID: 26582983 PMCID: PMC4631947 DOI: 10.3389/fnhum.2015.00601] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/18/2015] [Indexed: 01/17/2023] Open
Abstract
Prenatal marijuana exposure (PME) is linked to neurobehavioral and cognitive impairments; however, findings in childhood and adolescence are inconsistent. Type-1 cannabinoid receptors (CB1R) modulate fetal neurodevelopment, mediating PME effects on growth of functional circuitry sub-serving behaviors critical for academic and social success. The purpose of this study was to investigate the effects of prenatal marijuana on development of early brain functional circuitry prior to prolonged postnatal environmental influences. We measured resting state functional connectivity during unsedated sleep in infants at 2–6 weeks (+MJ: 20 with PME in combination with nicotine, alcohol, opiates, and/or selective serotonin reuptake inhibitors; −MJ: 23 exposed to the same other drugs without marijuana, CTR: 20 drug-free controls). Connectivity of subcortical seed regions with high fetal CB1R expression was examined. Marijuana-specific differences were observed in insula and three striatal connections: anterior insula–cerebellum, right caudate–cerebellum, right caudate–right fusiform gyrus/inferior occipital, left caudate–cerebellum. +MJ neonates had hypo-connectivity in all clusters compared with −MJ and CTR groups. Altered striatal connectivity to areas involved in visual spatial and motor learning, attention, and in fine-tuning of motor outputs involved in movement and language production may contribute to neurobehavioral deficits reported in this at-risk group. Disrupted anterior insula connectivity may contribute to altered integration of interoceptive signals with salience estimates, motivation, decision-making, and later drug use. Compared with CTRs, both +MJ and −MJ groups demonstrated hyper-connectivity of left amygdala seed with orbital frontal cortex and hypo-connectivity of posterior thalamus seed with hippocampus, suggesting vulnerability to multiple drugs in these circuits.
Collapse
Affiliation(s)
- Karen Grewen
- Department of Psychiatry, Neurobiology, and Psychology, University of North Carolina Chapel Hill , Chapel Hill, NC , USA
| | - Andrew P Salzwedel
- Department of Radiology, Biomedical Research Imaging Center, University of North Carolina Chapel Hill , Chapel Hill, NC , USA ; Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute, Cedars-Sinai Medical Cente , Los Angeles, CA , USA
| | - Wei Gao
- Department of Radiology, Biomedical Research Imaging Center, University of North Carolina Chapel Hill , Chapel Hill, NC , USA ; Department of Biomedical Sciences and Imaging, Biomedical Imaging Research Institute, Cedars-Sinai Medical Cente , Los Angeles, CA , USA
| |
Collapse
|
29
|
Fetal Alcohol Spectrum Disorder: Potential Role of Endocannabinoids Signaling. Brain Sci 2015; 5:456-93. [PMID: 26529026 PMCID: PMC4701023 DOI: 10.3390/brainsci5040456] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/19/2015] [Accepted: 10/19/2015] [Indexed: 12/15/2022] Open
Abstract
One of the unique features of prenatal alcohol exposure in humans is impaired cognitive and behavioral function resulting from damage to the central nervous system (CNS), which leads to a spectrum of impairments referred to as fetal alcohol spectrum disorder (FASD). Human FASD phenotypes can be reproduced in the rodent CNS following prenatal ethanol exposure. Several mechanisms are expected to contribute to the detrimental effects of prenatal alcohol exposure on the developing fetus, particularly in the developing CNS. These mechanisms may act simultaneously or consecutively and differ among a variety of cell types at specific developmental stages in particular brain regions. Studies have identified numerous potential mechanisms through which alcohol can act on the fetus. Among these mechanisms are increased oxidative stress, mitochondrial damage, interference with the activity of growth factors, glia cells, cell adhesion molecules, gene expression during CNS development and impaired function of signaling molecules involved in neuronal communication and circuit formation. These alcohol-induced deficits result in long-lasting abnormalities in neuronal plasticity and learning and memory and can explain many of the neurobehavioral abnormalities found in FASD. In this review, the author discusses the mechanisms that are associated with FASD and provides a current status on the endocannabinoid system in the development of FASD.
Collapse
|
30
|
Vaiserman A. Epidemiologic evidence for association between adverse environmental exposures in early life and epigenetic variation: a potential link to disease susceptibility? Clin Epigenetics 2015; 7:96. [PMID: 26366233 PMCID: PMC4567831 DOI: 10.1186/s13148-015-0130-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/01/2015] [Indexed: 12/21/2022] Open
Abstract
A growing body of evidence suggests that the risk of development and progression of a variety of human chronic diseases depends on epigenetic modifications triggered by environmental cues during early life sensitive stages. Exposures to environmental factors such as adverse nutritional, psychological, and social conditions, as well as pollutants and substance abuse in early life, have been shown to be important determinants of epigenetic programming of chronic pathological conditions in human populations. Over the past years, it has become increasingly clear due to the epigenome-wide association studies (EWASs) that early life adverse environmental events may trigger widespread and persistent alterations in transcriptional profiling. Several candidate genes have been identified underlying these associations. In this context, DNA methylation is the most intensively studied epigenetic phenomenon. In this review, the clinical and epidemiological evidence for the role of epigenetic factors in mediating the link between early life experiences and long-term health outcomes are summarized.
Collapse
|
31
|
Higuera-Matas A, Ucha M, Ambrosio E. Long-term consequences of perinatal and adolescent cannabinoid exposure on neural and psychological processes. Neurosci Biobehav Rev 2015; 55:119-46. [PMID: 25960036 DOI: 10.1016/j.neubiorev.2015.04.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/30/2015] [Accepted: 04/29/2015] [Indexed: 10/23/2022]
Abstract
Marihuana is the most widely consumed illicit drug, even among adolescents and pregnant women. Given the critical developmental processes that occur in the adolescent and fetal nervous system, marihuana consumption during these stages may have permanent consequences on several brain functions in later adult life. Here, we review what is currently known about the long-term consequences of perinatal and adolescent cannabinoid exposure. The most consistent findings point to long-term impairments in cognitive function that are associated with structural alterations and disturbed synaptic plasticity. In addition, several neurochemical modifications are also evident after prenatal or adolescent cannabinoid exposure, especially in the endocannabinoid, glutamatergic, dopaminergic and opioidergic systems. Important sexual dimorphisms are also evident in terms of the long-lasting effects of cannabinoid consumption during pregnancy and adolescence, and cannabinoids possibly have a protective effect in adolescents who have suffered traumatic life challenges, such as maternal separation or intense stress. Finally, we suggest some future research directions that may encourage further advances in this exciting field.
Collapse
Affiliation(s)
- Alejandro Higuera-Matas
- Department of Psychobiology, School of Psychology, National University of Distance Learning (UNED), C/ Juan del Rosal 10, 28040 Madrid, Spain.
| | - Marcos Ucha
- Department of Psychobiology, School of Psychology, National University of Distance Learning (UNED), C/ Juan del Rosal 10, 28040 Madrid, Spain
| | - Emilio Ambrosio
- Department of Psychobiology, School of Psychology, National University of Distance Learning (UNED), C/ Juan del Rosal 10, 28040 Madrid, Spain
| |
Collapse
|
32
|
Maccarrone M, Guzman M, Mackie K, Doherty P, Harkany T. Programming of neural cells by (endo)cannabinoids: from physiological rules to emerging therapies. Nat Rev Neurosci 2014; 15:786-801. [PMID: 25409697 PMCID: PMC4765324 DOI: 10.1038/nrn3846] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Among the many signalling lipids, endocannabinoids are increasingly recognized for their important roles in neuronal and glial development. Recent experimental evidence suggests that, during neuronal differentiation, endocannabinoid signalling undergoes a fundamental switch from the prenatal determination of cell fate to the homeostatic regulation of synaptic neurotransmission and bioenergetics in the mature nervous system. These studies also offer novel insights into neuropsychiatric disease mechanisms and contribute to the public debate about the benefits and the risks of cannabis use during pregnancy and in adolescence.
Collapse
Affiliation(s)
- Mauro Maccarrone
- School of Medicine and Center of Integrated Research, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, I-00128 Rome, Italy
- European Center for Brain Research/Santa Lucia Foundation, Via del Fosso di Fiorano 65, I-00143 Rome, Italy
| | - Manuel Guzman
- Department of Biochemistry and Molecular Biology I and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Complutense University, E-28040 Madrid, Spain
| | - Ken Mackie
- Department of Psychological & Brain Sciences, Indiana University, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
| | - Patrick Doherty
- Wolfson Centre for Age-Related Diseases, King's College London SE1 1UL, United Kingdom
| | - Tibor Harkany
- Division of Molecular Neuroscience, Department of Medical Biochemistry & Biophysics, Scheeles väg 1:A1, Karolinska Institutet, SE-17177 Stockholm Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria
| |
Collapse
|
33
|
Carr RL, Graves CA, Mangum LC, Nail CA, Ross MK. Low level chlorpyrifos exposure increases anandamide accumulation in juvenile rat brain in the absence of brain cholinesterase inhibition. Neurotoxicology 2013; 43:82-89. [PMID: 24373905 DOI: 10.1016/j.neuro.2013.12.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 12/15/2022]
Abstract
The prevailing dogma is that chlorpyrifos (CPF) mediates its toxicity through inhibition of cholinesterase (ChE). However, in recent years, the toxicological effects of developmental CPF exposure have been attributed to an unknown non-cholinergic mechanism of action. We hypothesize that the endocannabinoid system may be an important target because of its vital role in nervous system development. We have previously reported that repeated exposure to CPF results in greater inhibition of fatty acid amide hydrolase (FAAH), the enzyme that metabolizes the endocannabinoid anandamide (AEA), than inhibition of either forebrain ChE or monoacylglycerol lipase (MAGL), the enzyme that metabolizes the endocannabinoid 2-arachidonylglycerol (2-AG). This exposure resulted in the accumulation of 2-AG and AEA in the forebrain of juvenile rats; however, even at the lowest dosage level used (1.0mg/kg), forebrain ChE inhibition was still present. Thus, it is not clear if FAAH activity would be inhibited at dosage levels that do not inhibit ChE. To determine this, 10 day old rat pups were exposed daily for 7 days to either corn oil or 0.5mg/kg CPF by oral gavage. At 4 and 12h post-exposure on the last day of administration, the activities of serum ChE and carboxylesterase (CES) and forebrain ChE, MAGL, and FAAH were determined as well as the forebrain AEA and 2-AG levels. Significant inhibition of serum ChE and CES was present at both 4 and 12h. There was no significant inhibition of the activities of forebrain ChE or MAGL and no significant change in the amount of 2-AG at either time point. On the other hand, while no statistically significant effects were observed at 4h, FAAH activity was significantly inhibited at 12h resulting in a significant accumulation of AEA. Although it is not clear if this level of accumulation impacts brain maturation, this study demonstrates that developmental CPF exposure at a level that does not inhibit brain ChE can alter components of endocannabinoid signaling.
Collapse
Affiliation(s)
- Russell L Carr
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA.
| | - Casey A Graves
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA
| | - Lee C Mangum
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA
| | - Carole A Nail
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA
| | - Matthew K Ross
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA
| |
Collapse
|
34
|
Hurd YL, Michaelides M, Miller ML, Jutras-Aswad D. Trajectory of adolescent cannabis use on addiction vulnerability. Neuropharmacology 2013; 76 Pt B:416-24. [PMID: 23954491 DOI: 10.1016/j.neuropharm.2013.07.028] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 07/16/2013] [Accepted: 07/19/2013] [Indexed: 12/22/2022]
Abstract
The adolescent brain is a period of dynamic development making it vulnerable to environmental factors such as drug exposure. Of the illicit drugs, cannabis is most used by teenagers since it is perceived by many to be of little harm. This perception has led to a growing number of states approving its legalization and increased accessibility. Most of the debates and ensuing policies regarding cannabis were done without consideration of its impact on one of the most vulnerable population, namely teens, or without consideration of scientific data. We provide an overview of the endocannabinoid system in relation to adolescent cannabis exposure and provide insights regarding factors such as genetics and behavioral traits that confer risk for subsequent addiction. While it is clear that more systematic scientific studies are needed to understand the long-term impact of adolescent cannabis exposure on brain and behavior, the current evidence suggests that it has a far-reaching influence on adult addictive behaviors particularly for certain subsets of vulnerable individuals. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Yasmin L Hurd
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; James J. Peters Veterans Administration, Bronx, NY, USA.
| | | | | | | |
Collapse
|
35
|
Reiss D, Leve LD, Neiderhiser JM. How genes and the social environment moderate each other. Am J Public Health 2013; 103 Suppl 1:S111-21. [PMID: 23927504 DOI: 10.2105/ajph.2013.301408] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Recent research has suggested that the social environment can moderate the expression of genetic influences on health and that genetic influences can shape an individual's sensitivity to the social environment. Evidence supports 4 major mechanisms: genes can influence an individual's response to environmental stress, genes may enhance an individual's sensitivity to both favorable and adverse environments, inherited characteristics may better fit with some environments than with others, and inherited capabilities may only become manifest in challenging or responsive environments. Further progress depends on better recognition of patterns of gene-environment interaction, improved methods of assessing the environment and its impact on genetic mechanisms, the use of appropriately designed laboratory studies, identification of heritable differences in an individual before environmental moderation occurs, and clarification of the timing of the impact of social and genetic moderation.
Collapse
Affiliation(s)
- David Reiss
- David Reiss is with the Child Study Center, Yale School of Medicine, New Haven, CT. Leslie D. Leve is with the University of Oregon, Department of Counseling Psychology and Human Services, and the Oregon Social Learning Center, Eugene. Jenae M. Neiderhiser is with the Department of Psychology, The Pennsylvania State University, University Park
| | | | | |
Collapse
|
36
|
Anandamide-CB1 receptor signaling contributes to postnatal ethanol-induced neonatal neurodegeneration, adult synaptic, and memory deficits. J Neurosci 2013; 33:6350-66. [PMID: 23575834 DOI: 10.1523/jneurosci.3786-12.2013] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The transient exposure of immature rodents to ethanol during postnatal day 7 (P7), which is comparable with the third trimester in human pregnancy, induces synaptic dysfunctions. However, the molecular mechanisms underlying these dysfunctions are still poorly understood. Although the endocannabinoid system has been shown to be an important modulator of ethanol sensitivity in adult mice, its potential role in synaptic dysfunctions in mice exposed to ethanol during early brain development is not examined. In this study, we investigated the potential role of endocannabinoids and the cannabinoid receptor type 1 (CB1R) in neonatal neurodegeneration and adult synaptic dysfunctions in mice exposed to ethanol at P7. Ethanol treatment at P7, which induces neurodegeneration, increased anandamide (AEA) but not 2-arachidonylglycerol biosynthesis and CB1R protein expression in the hippocampus and cortex, two brain areas that are important for memory formation and storage, respectively. N-Arachidonoyl phosphatidylethanolamine-phospholipase D (NAPE-PLD), glycerophosphodiesterase (GDE1), and CB1R protein expression were enhanced by transcriptional activation of the genes encoding NAPE-PLD, GDE1, and CB1R proteins, respectively. In addition, ethanol inhibited ERK1/2 and AKT phosphorylation. The blockade of CB1Rs before ethanol treatment at P7 relieved ERK1/2 but not AKT phosphorylation and prevented neurodegeneration. CB1R knock-out mice exhibited no ethanol-induced neurodegeneration and inhibition of ERK1/2 phosphorylation. The protective effects of CB1R blockade through pharmacological or genetic deletion resulted in normal adult synaptic plasticity and novel object recognition memory in mice exposed to ethanol at P7. The AEA/CB1R/pERK1/2 signaling pathway may be directly responsible for the synaptic and memory deficits associated with fetal alcohol spectrum disorders.
Collapse
|
37
|
Long-term health consequences of early-life exposure to substance abuse: an epigenetic perspective. J Dev Orig Health Dis 2013; 4:269-79. [DOI: 10.1017/s2040174413000123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A growing body of evidence highlights the importance of the nutritional or other environmental stimuli during critical periods of development in the long-term programming of organ systems and homeostatic pathways of the organism. The adverse influences early in development and particularly during intrauterine life have been shown to programme the risks for adverse health outcomes in adult life. The mechanisms underlying developmental programming remain still unclear. However, increasing evidence has been accumulated indicating the important role of epigenetic regulation including DNA methylation, histone modifications and non-coding RNAs in the developmental programming of late-onset pathologies, including cancer, neurodegenerative diseases, and type 2 diabetes. The maternal substance abuse during pregnancy, including smoking, drinking and psychoactive drug intake, is one of the important factors determining the process of developmental programming in modern human beings. The impact of prenatal drug/substance exposure on infant and early childhood development is currently in the main focus. The long-term programming effects of such exposures on aging and associated pathologies, however, have been reported only rarely. The purpose of this review is to provide a summary of recent research findings which indicate that maternal substance abuse during pregnancy and/or neonatal period can programme not only a child's health status, but also can cause long-term or even life-long health outcomes via mechanisms of epigenetic memory.
Collapse
|
38
|
Opioid system and Alzheimer's disease. Neuromolecular Med 2012; 14:91-111. [PMID: 22527793 DOI: 10.1007/s12017-012-8180-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 04/04/2012] [Indexed: 12/15/2022]
Abstract
The opioid system may be involved in the pathogenesis of AD, including cognitive impairment, hyperphosphorylated tau, Aβ production, and neuroinflammation. Opioid receptors influence the regulation of neurotransmitters such as acetylcholine, norepinephrine, GABA, glutamate, and serotonin which have been implicated in the pathogenesis of AD. Opioid system has a close relation with Aβ generation since dysfunction of opioid receptors retards the endocytosis and degradation of BACE1 and γ-secretase and upregulates BACE1 and γ-secretase, and subsequently, the production of Aβ. Conversely, activation of opioid receptors increases the endocytosis of BACE1 and γ-secretase and downregulates BACE1 and γ-secretase, limiting the production of Aβ. The dysfunction of opioid system (opioid receptors and opioid peptides) may contribute to hyperphosphorylation of tau and neuroinflammation, and accounts for the degeneration of cholinergic neurons and cognitive impairment. Thus, the opioid system is potentially related to AD pathology and may be a very attractive drug target for novel pharmacotherapies of AD.
Collapse
|
39
|
Morris CV, DiNieri JA, Szutorisz H, Hurd YL. Molecular mechanisms of maternal cannabis and cigarette use on human neurodevelopment. Eur J Neurosci 2011; 34:1574-83. [PMID: 22103415 PMCID: PMC3226730 DOI: 10.1111/j.1460-9568.2011.07884.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Prenatal development is highly sensitive to maternal drug use due to the vulnerability for disruption of the fetal brain with its ongoing neurodevelopment, resulting in lifelong consequences that can enhance risk for psychiatric disorders. Cannabis and cigarettes are the most commonly used illicit and licit substances, respectively, among pregnant women. Although the behavioral consequences of prenatal cannabis and cigarette exposure have been well-documented in epidemiological and clinical studies, only recently have investigations into the molecular mechanisms associated with the developmental impact of early drug exposure been addressed. This article reviews the literature relevant to long-term gene expression disturbances in the human fetal brain in relation to maternal cannabis and cigarette use. To provide translational insights, we discuss animal models in which protracted molecular consequences of prenatal cannabis and cigarette exposure can be better explored and which enable future evaluation of epigenetic pathways, such as DNA methylation and histone modification, that could potentially maintain abnormal gene regulation and related behavioral disturbances. Altogether, this information may help to address the current gaps of knowledge regarding the impact of early drug exposure that set in motion lifelong molecular disturbances that underlie vulnerability to psychiatric disorders.
Collapse
Affiliation(s)
- Claudia V Morris
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | |
Collapse
|
40
|
DiNieri JA, Wang X, Szutorisz H, Spano SM, Kaur J, Casaccia P, Dow-Edwards D, Hurd YL. Maternal cannabis use alters ventral striatal dopamine D2 gene regulation in the offspring. Biol Psychiatry 2011; 70:763-769. [PMID: 21820648 PMCID: PMC3186868 DOI: 10.1016/j.biopsych.2011.06.027] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 06/27/2011] [Accepted: 06/29/2011] [Indexed: 10/17/2022]
Abstract
BACKGROUND Prenatal cannabis exposure has been linked to addiction vulnerability, but the neurobiology underlying this risk is unknown. METHODS Striatal dopamine and opioid-related genes were studied in human fetal subjects exposed to cannabis (as well as cigarettes and alcohol). Cannabis-related gene disturbances observed in the human fetus were subsequently characterized with an animal model of prenatal Δ-9-tetrahydrocannabinol (THC) (.15 mg/kg) exposure. RESULTS Prenatal cannabis exposure decreased dopamine receptor D2 (DRD2) messenger RNA expression in the human ventral striatum (nucleus accumbens [NAc]), a key brain reward region. No significant alterations were observed for the other genes in cannabis-exposed subjects. Maternal cigarette use was associated with reduced NAc prodynorphin messenger RNA expression, and alcohol exposure induced broad alterations primarily in the dorsal striatum of most genes. To explore the mechanisms underlying the cannabis-associated disturbances, we exposed pregnant rats to THC and examined the epigenetic regulation of the NAc Drd2 gene in their offspring at postnatal day 2, comparable to the human fetal period studied, and in adulthood. Chromatin immunoprecipitation of the adult NAc revealed increased 2meH3K9 repressive mark and decreased 3meH3K4 and RNA polymerase II at the Drd2 gene locus in the THC-exposed offspring. Decreased Drd2 expression was accompanied by reduced dopamine D2 receptor (D(2)R) binding sites and increased sensitivity to opiate reward in adulthood. CONCLUSIONS These data suggest that maternal cannabis use alters developmental regulation of mesolimbic D(2)R in offspring through epigenetic mechanisms that regulate histone lysine methylation, and the ensuing reduction of D(2)R might contribute to addiction vulnerability later in life.
Collapse
Affiliation(s)
- Jennifer A DiNieri
- Departments of Psychiatry, Neuroscience, Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, United States
| | - Xinyu Wang
- Section of Psychiatry, Department of Clinical Neuroscience, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Henrietta Szutorisz
- Departments of Psychiatry, Neuroscience, Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, United States
| | - Sabrina M Spano
- Section of Psychiatry, Department of Clinical Neuroscience, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Jasbir Kaur
- Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, New York, 10029, United States
| | - Patrizia Casaccia
- Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, New York, 10029, United States
| | - Diana Dow-Edwards
- Department of Physiology and Pharmacology, State University of New York, Downstate Medical Center, Box 29, 450 Clarkson Avenue, Brooklyn, NY 11203, United States
| | - Yasmin L Hurd
- Departments of Psychiatry, Neuroscience, Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York.
| |
Collapse
|
41
|
Wu CS, Jew CP, Lu HC. Lasting impacts of prenatal cannabis exposure and the role of endogenous cannabinoids in the developing brain. FUTURE NEUROLOGY 2011; 6:459-480. [PMID: 22229018 PMCID: PMC3252200 DOI: 10.2217/fnl.11.27] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cannabis is the most commonly used illicit substance among pregnant women. Human epidemiological and animal studies have found that prenatal cannabis exposure influences brain development and can have long-lasting impacts on cognitive functions. Exploration of the therapeutic potential of cannabis-based medicines and synthetic cannabinoid compounds has given us much insight into the physiological roles of endogenous ligands (endocannabinoids) and their receptors. In this article, we examine human longitudinal cohort studies that document the long-term influence of prenatal exposure to cannabis, followed by an overview of the molecular composition of the endocannabinoid system and the temporal and spatial changes in their expression during brain development. How endocannabinoid signaling modulates fundamental developmental processes such as cell proliferation, neurogenesis, migration and axonal pathfinding are also summarized.
Collapse
Affiliation(s)
- Chia-Shan Wu
- The Cain Foundation Laboratories, Jan & Dan Duncan Neurological Research Institute at Texas Children's Hospital, 1250 Moursund St Suite 1225, Houston, TX 77030, USA
| | - Christopher P Jew
- The Cain Foundation Laboratories, Jan & Dan Duncan Neurological Research Institute at Texas Children's Hospital, 1250 Moursund St Suite 1225, Houston, TX 77030, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hui-Chen Lu
- The Cain Foundation Laboratories, Jan & Dan Duncan Neurological Research Institute at Texas Children's Hospital, 1250 Moursund St Suite 1225, Houston, TX 77030, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
42
|
Carr RL, Borazjani A, Ross MK. Effect of developmental chlorpyrifos exposure, on endocannabinoid metabolizing enzymes, in the brain of juvenile rats. Toxicol Sci 2011; 122:112-20. [PMID: 21507991 DOI: 10.1093/toxsci/kfr081] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The endogenous cannabinoids 2-arachidonoylglycerol (2-AG) and arachidonoyl ethanolamide (AEA or anandamide) play vital roles during nervous system development including regulating axonal guidance and synaptogenesis. The enzymatic degradation of 2-AG and AEA is highly susceptible to inhibition by organophosphate compounds in vitro. Furthermore, acute in vivo exposure of adult animals to the agricultural insecticide chlorpyrifos (CPS) caused moderate inhibition of both 2-AG and AEA hydrolysis. However, the effects of repeated exposure to lower levels of CPS, especially during development, on endocannabinoid metabolism in the brain is not known. To examine this, rat pups were orally exposed daily from postnatal days 10-16 to either 1.0, 2.5, or 5.0 mg/kg CPS. Body weight gain was reduced by 5.0 mg/kg on all days of treatment whereas 2.5 mg/kg reduced the weight gain only on the last two days of treatment. At 4-h postexposure on day 16, forebrain cholinesterase (ChE) activity and hydrolysis of 2-AG and AEA were inhibited in a dose-related manner, and the extent of inhibition from highest to lowest level was AEA hydrolysis > ChE activity > 2-AG hydrolysis. The extent of inhibition of AEA hydrolysis was approximately twice than that of ChE activity with AEA hydrolysis being virtually eliminated by 2.5 and 5.0 mg/kg and 1.0 mg/kg causing 40% inhibition. The sensitivity of AEA hydrolysis, compared with canonical targets such as ChE activity, suggests a potential alternative developmental target for CPS. Inhibition of AEA hydrolysis could result in accumulation of endocannabinoids, which could alter normal endocannabinoid transmission during brain maturation.
Collapse
Affiliation(s)
- Russell L Carr
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762-6100, USA.
| | | | | |
Collapse
|
43
|
Khurshid N, Hameed LS, Mohanasundaram S, Iyengar S. Opioid modulation of cell proliferation in the ventricular zone of adult zebra finches (Taenopygia guttata). FASEB J 2010; 24:3681-95. [PMID: 20495180 DOI: 10.1096/fj.09-146746] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Besides modulating pain, stress, physiological functions, motivation, and reward, the opioid system has been implicated in developmental and adult mammalian neurogenesis and gliogenesis. In adult male songbirds including zebra finches, neurons generated from the ventricular zone (VZ) of the lateral ventricles are incorporated throughout the telencephalon, including the song control nuclei, HVC, and area X. Although the endogenous opioid met-enkephalin is present in neurons adjacent to the VZ and is upregulated in song control regions during singing, it is not known whether the opioid system can modulate adult neurogenesis/gliogenesis in zebra finches. We used quantitative RT-PCR and in situ hybridization to demonstrate that μ- and δ-opioid receptors are expressed by the VZ of adult male zebra finches. Treating cultured VZ cells from male birds with the opioid antagonist naloxone led to an increase in cell proliferation measured by 5-bromo-2-deoxyuridine incorporation, whereas administering met-enkephalin had the opposite effect, compared with saline-treated cultures. Systemically administering naloxone (2.5 mg/kg body wt) to adult male zebra finches for 4 d also led to a significant increase in cell proliferation in the ventral VZ of these birds, compared with saline-treated controls. Our results show that cell proliferation is augmented by naloxone in the VZ adjacent to the anterior commissure, suggesting that the endogenous opioids modulate adult neurogenesis/gliogenesis by inhibiting cell proliferation in songbirds.
Collapse
Affiliation(s)
- Nazia Khurshid
- Division of Systems Neuroscience, National Brain Research Centre, Manesar, India
| | | | | | | |
Collapse
|
44
|
Expression of δ- and μ-opioid receptors in the ventricular and subventricular zones of the developing human neocortex. Neurosci Res 2008; 61:257-70. [DOI: 10.1016/j.neures.2008.03.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 03/14/2008] [Accepted: 03/17/2008] [Indexed: 11/24/2022]
|
45
|
López-Moreno JA, González-Cuevas G, Moreno G, Navarro M. The pharmacology of the endocannabinoid system: functional and structural interactions with other neurotransmitter systems and their repercussions in behavioral addiction. Addict Biol 2008; 13:160-87. [PMID: 18422831 DOI: 10.1111/j.1369-1600.2008.00105.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Addiction is a chronic, recurring and complex disorder. It is characterized by anomalous behaviors that are linked to permanent or long-lasting neurobiological alterations. Furthermore, the endocannabinoid system has a crucial role in mediating neurotransmitter release as one of the main neuromodulators of the mammalian central nervous system. The purpose of the present review is to instruct readers about the functional and structural interactions between the endocannabinoid system and the main neurotransmitter systems of the central nervous system in the context of drug addiction. With this aim, we have systematically reviewed the main findings of most of the existing literature that explores cross-talk in the five brain areas that are most traditionally implicated in addiction: amygdala, prefrontal cortex, nucleus accumbens, hippocampus and ventral tegmental area (VTA). The neurotransmission systems influenced by the pharmacology of the endocannabinoid system in these brain areas, which are reviewed here, are gamma-aminobutyric acid (GABA), glutamate, the main biogenic amines (dopamine, noradrenaline and serotonin), acetylcholine and opioids. We show that all of these neurotransmitter systems can be modulated differentially in each brain area by the activation or deactivation of cannabinoid CB1 brain receptors. Specifically, most of the studies relate to the hippocampus and nucleus accumbens. Moreover, the neurotransmitter with the fewest number of related studies is acetylcholine (excepting in the hippocampus), whereas there is a large number that evaluates GABA, glutamate and dopamine. Finally, we propose a possible interpretation of the role of the endocannabinoid system in the phenomenon of addiction.
Collapse
Affiliation(s)
- José Antonio López-Moreno
- Department of Psychobiology, Faculty of Psychology, Campus de Somosaguas, Complutense University of Madrid, Spain.
| | | | | | | |
Collapse
|
46
|
Trezza V, Cuomo V, Vanderschuren LJMJ. Cannabis and the developing brain: insights from behavior. Eur J Pharmacol 2008; 585:441-52. [PMID: 18413273 DOI: 10.1016/j.ejphar.2008.01.058] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2007] [Revised: 12/20/2007] [Accepted: 01/22/2008] [Indexed: 02/05/2023]
Abstract
The isolation and identification, in 1964, of delta-9-tetrahydrocannabinol (THC), the primary psychoactive compound in cannabis, opened the door to a whole new field of medical research. The exploration of the therapeutic potential of THC and other natural and synthetic cannabinoid compounds was paralleled by the discovery of the endocannabinoid system, comprising cannabinoid receptors and their endogenous ligands, which offered exciting new insights into brain function. Besides its well-known involvement in specific brain functions, such as control of movement, memory and emotions, the endocannabinoid system plays an important role in fundamental developmental processes such as cell proliferation, migration and differentiation. For this reason, changes in its activity during stages of high neuronal plasticity, such as the perinatal and the adolescent period, can have long-lasting neurobehavioral consequences. Here, we summarize human and animal studies examining the behavioral and neurobiological effects of in utero and adolescent exposure to cannabis. Since cannabis preparations are widely used and abused by young people, including pregnant women, understanding how cannabinoid compounds affect the developing brain, leading to neurobehavioral alterations or neuropsychiatric disorders later in life, is a serious health issue. In addition, since the endocannabinoid system is emerging as a novel therapeutic target for the treatment of several neuropsychiatric diseases, a detailed investigation of possible adverse effects of cannabinoid compounds on the central nervous system (CNS) of immature individuals is warranted.
Collapse
Affiliation(s)
- Viviana Trezza
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | |
Collapse
|
47
|
Abstract
This paper is the 29th consecutive installment of the annual review of research concerning the endogenous opioid system, now spanning 30 years of research. It summarizes papers published during 2006 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurological disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY 11367, United States.
| |
Collapse
|
48
|
Sheng WS, Hu S, Herr G, Ni HT, Rock RB, Gekker G, Lokensgard JR, Peterson PK. Human Neural Precursor Cells Express Functional κ-Opioid Receptors. J Pharmacol Exp Ther 2007; 322:957-63. [PMID: 17538007 DOI: 10.1124/jpet.107.121988] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neural stem cells (NSCs) play an important role in the developing as well as adult brain. NSCs have been shown to migrate toward sites of injury in the brain and to participate in the process of brain repair. Like NSCs, cultured human neural precursor cells (NPCs) are self-renewing, multipotent cells capable of differentiating into neurons, astrocytes, and oligodendrocytes and of migrating toward chemotactic stimuli. Cellular and environmental factors are important for NPC proliferation and migration. Expression of kappa-opioid receptors (KORs) and mu-opioid receptors (MORs) in murine embryonic stem cells and of MORs and delta-opioid receptors in rodent neuronal precursors, as well as hippocampal progenitors has been reported by other investigators. In this study, we demonstrated robust expression of KORs in highly enriched (>90% nestin-positive) human fetal brain-derived NPCs. We found that KOR ligands, dynorphin(1-17) and trans-3,4-dichloro-N-methyl-N[2-(1-pyrolidinyl)cyclohexyl] benzeneacetamide methanesulfonate (U50,488) but not dynorphin(2-17), stimulated proliferation and migration of NPCs in a concentration-dependent manner. NPC proliferation was maximally stimulated at 10(-14) M dynorphin(1-17) and 10(-12) M U50,488. The KOR selective antagonist, nor-binaltorphimine, partially blocked the migratory and proliferative effects of KOR agonists supporting, at least in part, the involvement of a KOR-related mechanism. As has been described for rodent P19 embryonal carcinoma stem cells, retinoic acid treatment markedly suppressed KOR mRNA expression in human NPCs. Taken together, the results of this study suggest that activation of KORs alters functional properties of NPCs/NSCs that are relevant to human brain development and repair.
Collapse
Affiliation(s)
- Wen S Sheng
- The Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Spano MS, Ellgren M, Wang X, Hurd YL. Prenatal cannabis exposure increases heroin seeking with allostatic changes in limbic enkephalin systems in adulthood. Biol Psychiatry 2007; 61:554-63. [PMID: 16876136 DOI: 10.1016/j.biopsych.2006.03.073] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Revised: 01/19/2006] [Accepted: 03/22/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND Prenatal cannabis exposure is a growing concern with little known about the long-term consequences on behavior and neural systems relevant for reward and emotional processing. METHODS We used an animal model to study the effects of prenatal exposure to Delta(9)-tetrahydrocannabinol (THC) on heroin self-administration behavior and opioid neural systems in adult males (postnatal day 62). Rats were exposed to THC (.15 mg/kg) or vehicle from gestational day 5 to postnatal day 2. RESULTS Both pretreatment groups showed similar heroin intake, but THC-exposed rats exhibited shorter latency to the first active lever press, responded more for low heroin doses, and had higher heroin-seeking during mild stress and drug extinction. THC exposure reduced preproenkephalin (PENK) mRNA expression in the nucleus accumbens during early development, but was elevated in adulthood; no adult striatal changes on preprodynorphin mRNA or PENK in caudate-putamen. PENK mRNA was also increased in the central and medial amygdala in adult THC-exposed animals. THC animals had reduced heroin-induced locomotor activity and nucleus accumbens mu opioid receptor coupling. CONCLUSIONS This study demonstrates enduring effects of prenatal THC exposure into adulthood that is evident on heroin-seeking behavior during extinction and allostatic changes in mesocorticolimbic PENK systems relevant to drug motivation/reward and stress response.
Collapse
Affiliation(s)
- M Sabrina Spano
- Karolinska Institute, Department of Clinical Neuroscience, Psychiatry Section, S-17176 Stockholm, Sweden
| | | | | | | |
Collapse
|