1
|
Young-Wolff KC, Kong K, Alexeeff SE, Croen LA, Oberman N, Kirane H, Ansley D, Davignon M, Adams SR, Avalos LA. Prenatal Cannabis Use and Offspring Attention Deficit Hyperactivity Disorder and Disruptive Behavior Disorders: A Retrospective Cohort Study. J Dev Behav Pediatr 2024:00004703-990000000-00212. [PMID: 39400201 DOI: 10.1097/dbp.0000000000001323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 08/20/2024] [Indexed: 10/15/2024]
Abstract
OBJECTIVE To examine whether maternal cannabis use during early pregnancy is associated with offspring attention deficit hyperactivity disorder (ADHD) and disruptive behavior disorders (DBD). METHODS We conducted a population-based retrospective birth cohort study of children (N = 141,570) born between 2011 and 2018 to pregnant individuals (N = 117,130) in Kaiser Permanente Northern California universally screened for any prenatal cannabis use at the entrance to prenatal care (at ∼8-10 wk gestation). Prenatal cannabis use was defined as (1) self-reported use and/or a positive toxicology test, (2) self-reported use, (3) a positive toxicology test, and (4) self-reported use frequency. Cox proportional hazards regression models adjusting for maternal characteristics (sociodemographics, other substance use and substance use disorders, prenatal care initiation, comorbidities) examined associations between prenatal cannabis use and offspring ADHD and DBD diagnosed by age 11 years. RESULTS The sample of pregnant individuals was 27.2% Asian/Pacific Islander, 5.7% Black, 24.5% Hispanic, and 38.8% non-Hispanic White, with a mean (SD) age of 30.9 (5.2) years; 4.6% screened positive for any cannabis use (0.4% daily, 0.5% weekly, 1.1% monthly or less, 2.7% unknown frequency); 3.92% had a positive toxicology test and 1.8% self-reported use; 7.7% of offspring had ADHD and 6.8% had DBD. Maternal prenatal cannabis use was not associated with ADHD (adjusted hazard ratio [aHR]: 0.84, 95% CI, 0.70-1.01), and there was an inverse association with DBD (aHR: 0.83, 95% CI, 0.71-0.97), which remained when cannabis was defined by toxicology testing but not by self-report. Frequency of use was not associated with outcomes. CONCLUSION Maternal prenatal cannabis use was not associated with an increased risk of offspring ADHD or DBD.
Collapse
Affiliation(s)
- Kelly C Young-Wolff
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA
| | - Kevin Kong
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA
| | - Stacey E Alexeeff
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA
| | - Lisa A Croen
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA
| | - Nina Oberman
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA
| | | | - Deborah Ansley
- The Permanente Medical Group, Regional Offices, Oakland, CA
| | | | - Sara R Adams
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA
| | - Lyndsay A Avalos
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA
| |
Collapse
|
2
|
Avalos LA, Oberman N, Alexeeff SE, Croen LA, Davignon MN, Adams SR, Ansley D, Chambers CD, Steuerle K, Young-Wolff KC. Early Maternal Prenatal Cannabis Use and Child Developmental Delays. JAMA Netw Open 2024; 7:e2440295. [PMID: 39422907 DOI: 10.1001/jamanetworkopen.2024.40295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Importance Maternal prenatal cannabis use is associated with adverse neonatal health effects, yet little is known about its association with child developmental outcomes. Objective To evaluate associations between maternal prenatal cannabis use in early pregnancy and child early developmental delays. Design, Setting, and Participants This cohort study included 119 976 children born to 106 240 unique individuals between January 2015 and December 2019 and followed up to aged 5.5 years or younger (through December 31, 2021) at Kaiser Permanente Northern California. Individuals were screened for prenatal cannabis use via self-report and urine toxicology at entrance into prenatal care (approximately 8- to 10-weeks' gestation). Data were analyzed from February 2023 to March 2024. Exposure Maternal prenatal cannabis use defined as any use (self-reported or by urine toxicology testing) and use frequency. Main Outcomes Early developmental delays (speech and language disorders, motor delays, global delays) in children up to age 5.5 years defined by International Statistical Classification of Diseases and Related Health Problems, Ninth Revision and Tenth Revision diagnoses codes ascertained from electronic health records. Results In this cohort of 119 976 pregnancies among 106 240 unique pregnant individuals, there were 29 543 Hispanic pregnancies (24.6%), 6567 non-Hispanic Black pregnancies (5.5%), 46 823 non-Hispanic White pregnancies (39.0%), 12 837 pregnancies (10.7%) to individuals aged 24 years or younger, and 10 365 pregnancies (8.6%) to individuals insured by Medicaid. Maternal prenatal cannabis use was documented for 6778 pregnancies (5.6%). Daily maternal prenatal cannabis use was reported for 618 pregnancies (0.5%), weekly for 722 pregnancies (0.6%), and monthly or less for 1617 pregnancies (1.3%). No association was observed between maternal prenatal cannabis use and child speech and language disorders (HR, 0.93; 95% CI, 0.84-1.03), global developmental delays (HR, 1.04; 95% CI, 0.68-1.59), or motor delays (HR, 0.86; 95% CI, 0.69-1.06). No association was detected between the frequency of maternal prenatal cannabis use and child early developmental delays. Conclusions and Relevance In this cohort study, maternal prenatal cannabis use was not associated with an increased risk of child early developmental delays. Future research is needed to assess different patterns of cannabis use throughout pregnancy. Given the association between maternal prenatal cannabis use and other adverse outcomes, pregnant individuals should be educated on those risks.
Collapse
Affiliation(s)
- Lyndsay A Avalos
- Division of Research, Kaiser Permanente Northern California, Pleasanton
- Bernard J. Tyson Kaiser Permanente School of Medicine, Pasadena, California
| | - Nina Oberman
- Division of Research, Kaiser Permanente Northern California, Pleasanton
| | - Stacey E Alexeeff
- Division of Research, Kaiser Permanente Northern California, Pleasanton
| | - Lisa A Croen
- Division of Research, Kaiser Permanente Northern California, Pleasanton
- Bernard J. Tyson Kaiser Permanente School of Medicine, Pasadena, California
| | - Meghan N Davignon
- Pediatric Developmental Disabilities, Pediatric Subspecialties, Kaiser Permanente Roseville Medical Center, Roseville, California
| | - Sara R Adams
- Division of Research, Kaiser Permanente Northern California, Pleasanton
| | - Deborah Ansley
- Regional Offices, Kaiser Permanente Northern California, Oakland
| | - Christina D Chambers
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla
| | - Kristin Steuerle
- Kaiser Permanente Santa Rosa Medical Center, Santa Rosa, California
| | - Kelly C Young-Wolff
- Division of Research, Kaiser Permanente Northern California, Pleasanton
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
| |
Collapse
|
3
|
Camacho JA, Welch B, Ferguson M, Sepehr E, Vaught C, Zhao Y, Fitzpatrick S, Yourick J, Sprando RL, Hunt PR. Assessment of the effects of cannabidiol and a CBD-rich hemp extract in Caenorhabditis elegans. FRONTIERS IN TOXICOLOGY 2024; 6:1469341. [PMID: 39420966 PMCID: PMC11484448 DOI: 10.3389/ftox.2024.1469341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024] Open
Abstract
Consumer use of cannabidiol (CBD) is growing, but there are still data gaps regarding its possible adverse effects on reproduction and development. Multiple pathways and signaling cascades involved in organismal development and neuronal function, including endocannabinoid synthesis and signaling systems, are well conserved across phyla, suggesting that Caenorhabditis elegans can model the in vivo effects of exogenous cannabinoids. The effects in C. elegans on oxidative stress response (OxStrR), developmental timing, juvenile and adult spontaneous locomotor activity, reproductive output, and organismal CBD concentrations were assessed after exposure to purified CBD or a hemp extract suspended in 0.5% sesame oil emulsions. In C. elegans, this emulsion vehicle is equivalent to a high-fat diet (HFD). As in mammals, HFD was associated with oxidative-stress-related gene expression in C. elegans adults. CBD reduced HFD-induced OxStrR in transgenic adults and counteracted the hypoactivity observed in HFD-exposed wild-type adults. In C. elegans exposed to CBD from the onset of feeding, delays in later milestone acquisition were irreversible, while later juvenile locomotor activity effects were reversible after the removal of CBD exposure. CBD-induced reductions in mean juvenile population body size were cumulative when chronic exposures were initiated at parental reproductive maturity. Purified CBD was slightly more toxic than matched concentrations of CBD in hemp extract for all tested endpoints, and both were more toxic to juveniles than to adults. Dosimetry indicated that all adverse effect levels observed in C. elegans far exceeded recommended CBD dosages for humans.
Collapse
Affiliation(s)
- Jessica A. Camacho
- Division of Food Contact Substances, Office of Food Additive Safety, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, College Park, MD, United States
| | - Bonnie Welch
- Division of Virulence Assessment, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, College Park, MD, United States
| | - Martine Ferguson
- Biostatistics and Bioinformatics Staff, Office of Analytics and Outreach, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, College Park, MD, United States
| | - Estatira Sepehr
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, Laurel, MD, United States
| | - Cory Vaught
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, Laurel, MD, United States
| | - Yang Zhao
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, Laurel, MD, United States
| | - Suzanne Fitzpatrick
- Office of the Center Director, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, College Park, MD, United States
| | - Jeffrey Yourick
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, Laurel, MD, United States
| | - Robert L. Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, Laurel, MD, United States
| | - Piper Reid Hunt
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, Laurel, MD, United States
| |
Collapse
|
4
|
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
|
5
|
Navarro D, Gasparyan A, Navarrete F, Manzanares J. Fetal Cannabinoid Syndrome: Behavioral and Brain Alterations of the Offspring Exposed to Dronabinol during Gestation and Lactation. Int J Mol Sci 2024; 25:7453. [PMID: 39000559 PMCID: PMC11242182 DOI: 10.3390/ijms25137453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
This study establishes a fetal cannabinoid syndrome model to evaluate the effects of high doses of dronabinol (synthetic THC) during pregnancy and lactation on behavioral and brain changes in male and female progeny and their susceptibility to alcohol consumption. Female C57BL/6J mice received dronabinol (10 mg/kg/12 h, p.o.) from gestational day 5 to postnatal day 21. On the weaning day, the offspring were separated by sex, and on postnatal day 60, behavioral and neurobiological changes were analyzed. Mice exposed to dronabinol exhibited increased anxiogenic and depressive-like behaviors and cognitive impairment. These behaviors were associated with neurodevelopment-related gene and protein expression changes, establishing, for the first time, an association among behavioral changes, cognitive impairment, and neurobiological alterations. Exposure to dronabinol during pregnancy and lactation disrupted the reward system, leading to increased motivation to consume alcohol in the offspring. All these modifications exhibited sex-dependent patterns. These findings reveal the pronounced adverse effects on fetal neurodevelopment resulting from cannabis use during pregnancy and lactation and strongly suggest the need to prevent mothers who use cannabis in this period from the severe and permanent side effects on behavior and brain development that may occur in their children.
Collapse
Affiliation(s)
- Daniela Navarro
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
6
|
Carlson E, Teboul E, Canale C, Coleman H, Angeliu C, Garbarini K, Markowski VP. Perinatal Tetrahydrocannabinol Compromises Maternal Care and Increases Litter Attrition in the Long-Evans Rat. TOXICS 2024; 12:311. [PMID: 38787090 PMCID: PMC11126083 DOI: 10.3390/toxics12050311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
The marijuana legalization trend in the U.S. will likely lead to increased use by younger adults during gestation and postpartum. The current study examined the hypothesis that delta-9-tetrahydrocannabinol (THC) would disrupt voluntary maternal care behaviors and negatively impact offspring development. Rat dams were gavaged with 0, 2, 5, or 10 mg/kg THC from the 1st day of gestation through the 21st postnatal day. Somatic growth and developmental milestones were measured in the offspring, and maternal pup retrieval tests were conducted on postnatal days 1, 3, and 5. THC did not affect body growth but produced transient delays in the righting reflex and eye opening in offspring. However, there was significant pup mortality due to impaired maternal care. Dams in all THC groups took significantly longer to retrieve their pups to the nest and often failed to retrieve any pups. Serum levels of THC and metabolites measured at this time were comparable to those in breastfeeding women who are chronic users. Benchmark doses associated with a 10% reduction of pup retrieval or increased pup mortality were 0.383 (BMDL 0.228) and 0.794 (BMDL 0.442) mg/kg THC, respectively. The current findings indicate that maternal care is an important and heretofore overlooked index of THC behavioral toxicity and should be included in future assessments of THC's health risks.
Collapse
Affiliation(s)
- Emma Carlson
- Department of Psychology, State University of New York at Geneseo, One College Circle, Geneseo, NY 14454, USA; (E.C.); (C.C.); (H.C.); (C.A.); (K.G.)
| | - Eric Teboul
- Departments of Neurosurgery and Neuroscience, Brown University & Rhode Island Hospital, Providence, RI 02912, USA;
| | - Charlene Canale
- Department of Psychology, State University of New York at Geneseo, One College Circle, Geneseo, NY 14454, USA; (E.C.); (C.C.); (H.C.); (C.A.); (K.G.)
| | - Harper Coleman
- Department of Psychology, State University of New York at Geneseo, One College Circle, Geneseo, NY 14454, USA; (E.C.); (C.C.); (H.C.); (C.A.); (K.G.)
| | - Christina Angeliu
- Department of Psychology, State University of New York at Geneseo, One College Circle, Geneseo, NY 14454, USA; (E.C.); (C.C.); (H.C.); (C.A.); (K.G.)
| | - Karissa Garbarini
- Department of Psychology, State University of New York at Geneseo, One College Circle, Geneseo, NY 14454, USA; (E.C.); (C.C.); (H.C.); (C.A.); (K.G.)
| | - Vincent P. Markowski
- Department of Psychology, State University of New York at Geneseo, One College Circle, Geneseo, NY 14454, USA; (E.C.); (C.C.); (H.C.); (C.A.); (K.G.)
| |
Collapse
|
7
|
Xue T, Ma RH, Xu C, Sun B, Yan DF, Liu XM, Gao D, Li ZH, Gao Y, Wang CZ. The endocannabinoid system is involved in the anxiety-like behavior induced by dual-frequency 2.65/0.8 GHz electromagnetic radiation in mice. Front Mol Neurosci 2024; 17:1366855. [PMID: 38685914 PMCID: PMC11057378 DOI: 10.3389/fnmol.2024.1366855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/27/2024] [Indexed: 05/02/2024] Open
Abstract
As wireless communication devices gain popularity, concerns about the potential risks of environmental exposure to complex frequency electromagnetic radiation (EMR) on mental health have become a public health issue. Historically, EMR research has predominantly focused on single- frequency electromagnetic waves, neglecting the study of multi-frequency electromagnetic waves, which more accurately represent everyday life. To address these concerns, our study compared the emotional effects of single-frequency and dual-frequency EMR while exploring potential molecular mechanisms and intervention targets. Our results revealed that single-frequency EMR at 2.65 or 0.8 GHz did not induce anxiety-like behavior in mice. However, exposure to dual-frequency EMR at 2.65/0.8 GHz significantly led to anxiety-like behavior in mice. Further analysis of mouse sera revealed substantial increases in corticosterone and corticotrophin releasing hormone levels following exposure to 2.65/0.8 GHz EMR. Transcriptome sequencing indicated a significant decrease in the expression of Cnr1, encoding cannabinoid receptor 1 Type (CB1R), in the cerebral. This finding was consistently verified through western blot analysis, revealing a substantial reduction in CB1R content. Additionally, a significant decrease in the endocannabinoid 2-arachidonoylglycerol was observed in the cerebral cortex. Remarkably, administering the cannabinoid receptor agonist Win55-212-2 significantly alleviated the anxiety-like behavior, and the cannabinoid receptor antagonist AM251 effectively counteracted the anti-anxiety effects of Win55-212-2. In summary, our research confirmed that dual-frequency EMR is more likely to induce anxiety-like behavior in mice than single-frequency EMR, with implications for the hypothalamic-pituitary-adrenal axis and the endocannabinoid system. Furthermore, our findings suggest that Win55-212-2 may represent a novel avenue for researching and developing anti-EMR drugs.
Collapse
Affiliation(s)
- Teng Xue
- Laboratory of Bioelectromagnetics, Beijing Institute of Radiation and Medicine, Beijing, China
- School of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Rui-Han Ma
- Laboratory of Bioelectromagnetics, Beijing Institute of Radiation and Medicine, Beijing, China
- School of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Chou Xu
- Department of Critical Care Medicine, The 983rd Hospital of the Joint Logistics Support Force of PLA, Tianjin, China
- Chinese PLA General Hospital, Beijing, China
| | - Bin Sun
- Laboratory of Bioelectromagnetics, Beijing Institute of Radiation and Medicine, Beijing, China
| | - Dong-Fei Yan
- Laboratory of Bioelectromagnetics, Beijing Institute of Radiation and Medicine, Beijing, China
| | - Xiao-Man Liu
- Laboratory of Bioelectromagnetics, Beijing Institute of Radiation and Medicine, Beijing, China
| | - Dawen Gao
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Zhi-Hui Li
- Laboratory of Bioelectromagnetics, Beijing Institute of Radiation and Medicine, Beijing, China
| | - Yan Gao
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Chang-Zhen Wang
- Laboratory of Bioelectromagnetics, Beijing Institute of Radiation and Medicine, Beijing, China
| |
Collapse
|
8
|
Sandini TM, Onofrychuk TJ, Roebuck AJ, Hammond SA, Udenze D, Hayat S, Herdzik MA, McElroy DL, Orvold SN, Greba Q, Laprairie RB, Howland JG. Repeated Exposure to High-THC Cannabis Smoke during Gestation Alters Sex Ratio, Behavior, and Amygdala Gene Expression of Sprague Dawley Rat Offspring. eNeuro 2023; 10:ENEURO.0100-23.2023. [PMID: 37957008 PMCID: PMC10687874 DOI: 10.1523/eneuro.0100-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/21/2023] Open
Abstract
Because of the legalization of Cannabis in many jurisdictions and the trend of increasing Δ9-tetrahydrocannabinol (THC) content in Cannabis products, an urgent need exists to understand the impact of Cannabis use during pregnancy on fetal neurodevelopment and behavior. To this end, we exposed female Sprague Dawley rats to Cannabis smoke daily from gestational day 6 to 20 or room air. Maternal reproductive parameters, offspring behavior, and gene expression in the offspring amygdala were assessed. Body temperature was decreased in dams following smoke exposure and more fecal boli were observed in the chambers before and after smoke exposure in dams exposed to smoke. Maternal weight gain, food intake, gestational length, litter number, and litter weight were not altered by exposure to Cannabis smoke. A significant increase in the male-to-female ratio was noted in the Cannabis-exposed litters. In adulthood, male and female Cannabis smoke-exposed offspring explored the inner zone of an open field significantly less than control offspring. Gestational Cannabis smoke exposure did not affect behavior on the elevated plus maze test or social interaction test in the offspring. Cannabis offspring were better at visual pairwise discrimination and reversal learning tasks conducted in touchscreen-equipped operant conditioning chambers. Analysis of gene expression in the adult amygdala using RNA sequencing revealed subtle changes in genes related to development, cellular function, and nervous system disease in a subset of the male offspring. These results demonstrate that repeated exposure to high-THC Cannabis smoke during gestation alters maternal physiological parameters, sex ratio, and anxiety-like behaviors in the adulthood offspring.
Collapse
Affiliation(s)
- Thaisa M Sandini
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Timothy J Onofrychuk
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Andrew J Roebuck
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
- School of Liberal Arts, Yukon University, Whitehorse, Yukon Territory Y1A 5K4, Canada
| | - S Austin Hammond
- Global Institute for Food Security, Saskatoon, Saskatchewan S7N 4L8, Canada
| | - Daniel Udenze
- Next Generation Sequencing Facility, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Shahina Hayat
- Deparment of Oncology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Melissa A Herdzik
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Dan L McElroy
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Spencer N Orvold
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Quentin Greba
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Robert B Laprairie
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - John G Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| |
Collapse
|
9
|
Lin A, Dent GL, Davies S, Dominguez ZM, Cioffredi LA, McLemore GL, Maxwell JR. Prenatal cannabinoid exposure: why expecting individuals should take a pregnancy pause from using cannabinoid products. Front Pediatr 2023; 11:1278227. [PMID: 37886232 PMCID: PMC10598870 DOI: 10.3389/fped.2023.1278227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023] Open
Abstract
Cannabinoid use in all populations is increasing as legalization across the United States continues. Concerningly, there is a lack of caution provided by medical providers to pregnant individuals as to the impact the use of cannabinoids could have on the developing fetus. Research continues in both the preclinical and clinical areas, and is severely needed, as the potency of delta-9-tetrahydrocannabinol (THC), the primary psychoactive component of cannabis, has increased dramatically since the initial studies were completed. Thus far, clinical studies raise compelling evidence for short term memory deficits, impulse control issues, and attention deficiencies following prenatal cannabinoid exposure (PCE). These changes may be mediated through epigenetic modifications that not only impact the current offspring but could carry forward to future generations. While additional studies are needed, a pregnancy pause from cannabinoid products should be strongly recommended by providers to ensure the optimal health and well-being of our future generations.
Collapse
Affiliation(s)
- Alexis Lin
- Gustavus Adolphus College, Saint Peter, MN, United States
| | - Gelonia L. Dent
- Department of Mathematics, Medgar Evers College, CUNY, Brooklyn, NY, United States
| | - Suzy Davies
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, United States
| | - Zarena M. Dominguez
- Department of Pediatrics, University of New Mexico, Albuquerque, NM, United States
| | | | | | - Jessie R. Maxwell
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, United States
- Department of Pediatrics, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
10
|
Sarikahya MH, Cousineau SL, De Felice M, Szkudlarek HJ, Wong KKW, DeVuono MV, Lee K, Rodríguez-Ruiz M, Gummerson D, Proud E, Ng THJ, Hudson R, Jung T, Hardy DB, Yeung KKC, Schmid S, Rushlow W, Laviolette SR. Prenatal THC exposure induces long-term, sex-dependent cognitive dysfunction associated with lipidomic and neuronal pathology in the prefrontal cortex-hippocampal network. Mol Psychiatry 2023; 28:4234-4250. [PMID: 37525013 DOI: 10.1038/s41380-023-02190-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 08/02/2023]
Abstract
With increasing maternal cannabis use, there is a need to investigate the lasting impact of prenatal exposure to Δ9-tetrahydrocannabinol (THC), the main psychotropic compound in cannabis, on cognitive/memory function. The endocannabinoid system (ECS), which relies on polyunsaturated fatty acids (PUFAs) to function, plays a crucial role in regulating prefrontal cortical (PFC) and hippocampal network-dependent behaviors essential for cognition and memory. Using a rodent model of prenatal cannabis exposure (PCE), we report that male and female offspring display long-term deficits in various cognitive domains. However, these phenotypes were associated with highly divergent, sex-dependent mechanisms. Electrophysiological recordings revealed hyperactive PFC pyramidal neuron activity in both males and females, but hypoactivity in the ventral hippocampus (vHIPP) in males, and hyperactivity in females. Further, cortical oscillatory activity states of theta, alpha, delta, beta, and gamma bandwidths were strongly sex divergent. Moreover, protein expression analyses at postnatal day (PD)21 and PD120 revealed primarily PD120 disturbances in dopamine D1R/D2 receptors, NMDA receptor 2B, synaptophysin, gephyrin, GAD67, and PPARα selectively in the PFC and vHIPP, in both regions in males, but only the vHIPP in females. Lastly, using matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS), we identified region-, age-, and sex-specific deficiencies in specific neural PUFAs, namely docosahexaenoic acid (DHA) and arachidonic acid (ARA), and related metabolites, in the PFC and hippocampus (ventral/dorsal subiculum, and CA1 regions). This study highlights several novel, long-term and sex-specific consequences of PCE on PFC-hippocampal circuit dysfunction and the potential role of specific PUFA signaling abnormalities underlying these pathological outcomes.
Collapse
Affiliation(s)
- Mohammed H Sarikahya
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Samantha L Cousineau
- Departments of Chemistry and Biochemistry, Western University, London, Ontario, N6A 3K7, Canada
| | - Marta De Felice
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Hanna J Szkudlarek
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Karen K W Wong
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Marieka V DeVuono
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Kendrick Lee
- Departments of Physiology and Pharmacology and Obstetrics and Gynaecology, Western University, London, Ontario, N6A 5C1, Canada
- Children's Health Research Institute, St. Josephs Health Care,, London, Ontario, N6C 2R5, Canada
| | - Mar Rodríguez-Ruiz
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Dana Gummerson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Emma Proud
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Tsun Hay Jason Ng
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Roger Hudson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Tony Jung
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
| | - Daniel B Hardy
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
- Departments of Physiology and Pharmacology and Obstetrics and Gynaecology, Western University, London, Ontario, N6A 5C1, Canada
- Children's Health Research Institute, St. Josephs Health Care,, London, Ontario, N6C 2R5, Canada
| | - Ken K-C Yeung
- Departments of Chemistry and Biochemistry, Western University, London, Ontario, N6A 3K7, Canada
| | - Susanne Schmid
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
- Department of Psychology, Western University, London, Ontario, N6A 3K7, Canada
| | - Walter Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada
- Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, N6C 2R5, Canada
- Department of Psychiatry, Western University, London, Ontario, N6A 3K7, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada.
- Department of Anatomy and Cell Biology, Western University, London, Ontario, N6A 3K7, Canada.
- Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, N6C 2R5, Canada.
- Department of Psychiatry, Western University, London, Ontario, N6A 3K7, Canada.
| |
Collapse
|
11
|
Emerging Roles of Endocannabinoids as Key Lipid Mediators for a Successful Pregnancy. Int J Mol Sci 2023; 24:ijms24065220. [PMID: 36982295 PMCID: PMC10048990 DOI: 10.3390/ijms24065220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
In recent years, Cannabis use/misuse for treating pregnancy-related symptoms and other chronic conditions has increased among pregnant women, favored by decriminalization and/or legalization of its recreational uses in addition to its easy accessibility. However, there is evidence that prenatal Cannabis exposure might have adverse consequences on pregnancy progression and a deleterious impact on proper neurodevelopmental trajectories in the offspring. Maternal Cannabis use could interfere with the complex and finely controlled role performed by the endocannabinoid system in reproductive physiology, impairing multiple gestational processes from blastocyst implantation to parturition, with long-lasting intergenerational effects. In this review, we discuss current clinical and preclinical evidence regarding the role of endocannabinoids in development, function, and immunity of the maternal–fetal interface, focusing on the impact of Cannabis constituents on each of these gestational processes. We also discuss the intrinsic limitations of the available studies and the future perspectives in this challenging research field.
Collapse
|
12
|
Blayac L, Ponte C, Lavaud M, Micallef J, Lapeyre-Mestre M. Increase of cannabis and cocaine use by pregnant women in France from 2005 to 2018: Insights of the annual cross sectional OPPIDUM survey. Therapie 2023; 78:201-211. [PMID: 36283856 DOI: 10.1016/j.therap.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/21/2022] [Indexed: 11/06/2022]
Abstract
CONTEXT Substance use disorders during pregnancy are associated with antenatal, neonatal and long-term adverse drug reactions in children. The aim of this study was to describe, using data from OPPIDUM survey, the evolution of illicit substance use or diverted prescription drug use among pregnant women visiting French addiction care centres from 2005 to 2018. MATERIAL AND METHOD The current study focused on women of childbearing age (15-44years old) identified as pregnant, and included in the OPPIDUM survey from 2005 to 2018. OPPIDUM is a French nationwide survey repeated each year since 1995, which anonymously collects information on drug abuse and dependence observed in patients recruited in addiction care centres. Patterns of illicit substance use, psychoactive medications, and opioid maintenance treatment (OMT) were described over time (by period of 2 years) and changes between 2005-2006 and 2017-2018 were investigated. RESULTS From 2005 to 2018, 784 pregnant women were included in the OPPIDUM survey (median age 28, interquartile range (IQR): 25-33). The proportion of women using exclusively cannabis (1.2% vs. 17.1%, P=0.0004) and cocaine (including crack) use (4.7% vs. 14.3%, P=0.0384) significantly increased over the period. Considering the first substance reported as leading to dependence, heroin significantly decreased (78.8% vs. 50.0%, P=0.0002) whereas cannabis significantly increased (5.9% vs. 25.7%, P=0.0005). CONCLUSION This study highlighted an important change of patterns of drug use by French pregnant women over a 14-year-period. Health professionals must be aware of these changes to adapt prevention and care among women of childbearing age and pregnant women.
Collapse
Affiliation(s)
- Léna Blayac
- Service de pharmacologie médicale et clinique, centre d'addictovigilance (CEIP-A), CHU de Toulouse - UFR santé, université de Toulouse, 31000 Toulouse, France.
| | - Camille Ponte
- Service de pharmacologie médicale et clinique, centre d'addictovigilance (CEIP-A), CHU de Toulouse - UFR santé, université de Toulouse, 31000 Toulouse, France
| | - Monique Lavaud
- École de sages-femmes, université de Toulouse, 31059 Toulouse, France
| | - Joëlle Micallef
- UMR 1106, Inserm, service de pharmacologie clinique, centre d'addictovigilance, AP-HM, Aix-Marseille université, 13005 Marseille, France
| | - Maryse Lapeyre-Mestre
- Service de pharmacologie médicale et clinique, centre d'addictovigilance (CEIP-A), CHU de Toulouse - UFR santé, université de Toulouse, 31000 Toulouse, France
| |
Collapse
|
13
|
Shah S, Schwenk ES, Sondekoppam RV, Clarke H, Zakowski M, Rzasa-Lynn RS, Yeung B, Nicholson K, Schwartz G, Hooten WM, Wallace M, Viscusi ER, Narouze S. ASRA Pain Medicine consensus guidelines on the management of the perioperative patient on cannabis and cannabinoids. Reg Anesth Pain Med 2023; 48:97-117. [PMID: 36596580 DOI: 10.1136/rapm-2022-104013] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/08/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND The past two decades have seen an increase in cannabis use due to both regulatory changes and an interest in potential therapeutic effects of the substance, yet many aspects of the substance and their health implications remain controversial or unclear. METHODS In November 2020, the American Society of Regional Anesthesia and Pain Medicine charged the Cannabis Working Group to develop guidelines for the perioperative use of cannabis. The Perioperative Use of Cannabis and Cannabinoids Guidelines Committee was charged with drafting responses to the nine key questions using a modified Delphi method with the overall goal of producing a document focused on the safe management of surgical patients using cannabinoids. A consensus recommendation required ≥75% agreement. RESULTS Nine questions were selected, with 100% consensus achieved on third-round voting. Topics addressed included perioperative screening, postponement of elective surgery, concomitant use of opioid and cannabis perioperatively, implications for parturients, adjustment in anesthetic and analgesics intraoperatively, postoperative monitoring, cannabis use disorder, and postoperative concerns. Surgical patients using cannabinoids are at potential increased risk for negative perioperative outcomes. CONCLUSIONS Specific clinical recommendations for perioperative management of cannabis and cannabinoids were successfully created.
Collapse
Affiliation(s)
- Shalini Shah
- Dept of Anesthesiology & Perioperative Care, UC Irvine Health, Orange, California, USA
| | - Eric S Schwenk
- Anesthesiology and Perioperative Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Hance Clarke
- Anesthesiology and Pain Medicine, Univ Toronto, Toronto, Ontario, Canada
| | - Mark Zakowski
- Anesthesiology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | - Brent Yeung
- Anesthesiology and Perioperative Care, University of California Irvine, Irvine, California, USA
| | | | - Gary Schwartz
- AABP Integrative Pain Care, Melville, New York, USA.,Anesthesiology, Maimonides Medical Center, Brooklyn, New York, USA
| | | | - Mark Wallace
- Anesthesiology, Division of Pain Medicine, University of California San Diego, La Jolla, California, USA
| | - Eugene R Viscusi
- Anesthesiology and Perioperative Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Samer Narouze
- Center for Pain Medicine, Western Reserve Hospital, Cuyahoga Falls, Ohio, USA
| |
Collapse
|
14
|
Lamanna-Rama N, MacDowell KS, López G, Leza JC, Desco M, Ambrosio E, Soto-Montenegro ML. Neuroimaging revealed long-lasting glucose metabolism changes to morphine withdrawal in rats pretreated with the cannabinoid agonist CP-55,940 during periadolescence. Eur Neuropsychopharmacol 2023; 69:60-76. [PMID: 36780817 DOI: 10.1016/j.euroneuro.2023.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/28/2022] [Accepted: 01/22/2023] [Indexed: 02/13/2023]
Abstract
This study evaluates the long-term effects of a six and 14-week morphine withdrawal in rats pretreated with a cannabinoid agonist (CP-55,940, CP) during periadolescence. Wistar rats (33 males; 32 females) were treated with CP or its vehicle (VH) from postnatal day (PND) 28-38. At PND100, rats performed morphine self-administration (MSA, 15d/12 h/session). Eight groups were defined according to pretreatment (CP), treatment (morphine), and sex. Three [18F]FDG-PET brain images were acquired: after MSA, and after six and 14 weeks of withdrawal. PET data were analyzed with SPM12. Endocannabinoid (EC) markers were evaluated in frozen brain tissue at endpoint. Females showed a higher mean number of self-injections than males. A main Sex effect on global brain metabolism was found. FDG uptake in males was discrete, whereas females showed greater brain metabolism changes mainly in areas of the limbic system after morphine treatment. Moreover, the morphine-induced metabolic pattern in females was exacerbated when CP was previously present. In addition, the CP-Saline male group showed reduced CB1R, MAGL expression, and NAPE/FAAH ratio compared to the control group, and morphine was able to reverse CB1R and MAGL expression almost to control levels. In conclusion, females showed greater and longer-lasting metabolic changes after morphine withdrawal than males, indicating a higher vulnerability and a different sensitivity to morphine in subjects pre-exposed to CP. In contrast, males primarily showed changes in EC markers. Together, our results suggest that CP pre-exposure contributes to the modulation of brain metabolism and EC systems in a sex-dependent manner.
Collapse
Affiliation(s)
- N Lamanna-Rama
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain
| | - K S MacDowell
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Departamento de Farmacología & Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Imas12, IUIN, Spain
| | - G López
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Psicobiología, Facultad de Psicología, National University for Distance Learning (UNED), Madrid, Spain; Faculty of Health Science, Universidad Internacional de La Rioja (UNIR), Spain
| | - J C Leza
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Departamento de Farmacología & Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Imas12, IUIN, Spain
| | - M Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| | - E Ambrosio
- Departamento de Psicobiología, Facultad de Psicología, National University for Distance Learning (UNED), Madrid, Spain.
| | - M L Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Grupo de Fisiopatología y Farmacología del Sistema Digestivo de la Universidad Rey Juan Carlos (NEUGUT), Madrid, España.
| |
Collapse
|
15
|
Guma E, Cupo L, Ma W, Gallino D, Moquin L, Gratton A, Devenyi GA, Chakravarty MM. Investigating the "two-hit hypothesis": Effects of prenatal maternal immune activation and adolescent cannabis use on neurodevelopment in mice. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110642. [PMID: 36150422 DOI: 10.1016/j.pnpbp.2022.110642] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 12/09/2022]
Abstract
Prenatal exposure to maternal immune activation (MIA) and chronic adolescent cannabis use are both risk factors for neuropsychiatric disorders. However, exposure to a single risk factor may not result in major mental illness, indicating that multiple exposures may be required for illness onset. Here, we examine whether combined exposure to prenatal MIA and adolescent delta-9-tetrahydrocannabinol (THC), the main psychoactive component of cannabis, lead to enduring neuroanatomical and behavioural changes in adulthood. Mice were prenatally exposed to viral mimetic, poly I:C (5 mg/kg), or vehicle at gestational day (GD) 9, and postnatally exposed to chronic THC (5 mg/kg, intraperitoneal) or vehicle during adolescence (postnatal day [PND]28-45). Longitudinal magnetic resonance imaging (MRI) was performed pre-treatment, PND 25, post-treatment, PND 50, and in adulthood, PND85, followed by behavioural tests for anxiety-like, social, and sensorimotor gating. Post-mortem assessment of cannabinoid (CB)1 and 2 receptor expressing cells was performed in altered regions identified by MRI (anterior cingulate and somatosensory cortices, striatum, and hippocampus). Subtle deviations in neurodevelopmental trajectory and subthreshold anxiety-like behaviours were observed in mice exposed to both risk factors. Sex-dependent effects were observed in patterns of shared brain-behaviour covariation, indicative of potential sex differences in response to MIA and THC. Density of CB1 and CB2 receptor positive cells was significantly decreased in all mice exposed to MIA, THC, or both. These findings suggest that there may be a cumulative effect of risk factor exposure on gross neuroanatomical development, and that the endocannabinoid system may be sensitive to both prenatal MIA, adolescent THC, or the combination.
Collapse
Affiliation(s)
- Elisa Guma
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Developmental Neurogenomics Unit, Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, USA.
| | - Lani Cupo
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Weiya Ma
- Douglas Mental Health University Institute, McGill University, Montréal, Québec, Canada
| | - Daniel Gallino
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Luc Moquin
- Douglas Mental Health University Institute, McGill University, Montréal, Québec, Canada
| | - Alain Gratton
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, McGill University, Montréal, Québec, Canada
| | - Gabriel A Devenyi
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, McGill University, Montréal, Québec, Canada
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, McGill University, Montréal, Québec, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
16
|
Iyer P, Niknam Y, Campbell M, Moran F, Kaufman F, Kim A, Sandy M, Zeise L. Animal evidence considered in determination of cannabis smoke and
Δ
9
‐tetrahydrocannabinol (
Δ
9
‐THC
) as causing reproductive toxicity (developmental endpoint); part
II
. Neurodevelopmental effects. Birth Defects Res 2022; 114:1155-1168. [DOI: 10.1002/bdr2.2084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Poorni Iyer
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Yassaman Niknam
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Marlissa Campbell
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Francisco Moran
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Farla Kaufman
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Allegra Kim
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Martha Sandy
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| | - Lauren Zeise
- Office of Environmental Health Hazard Assessment (OEHHA) Sacramento California USA
| |
Collapse
|
17
|
Ramírez S, Miguez G, Quezada-Scholz VE, Pardo L, Alfaro F, Varas FI, Laborda MA. Behavioral effects on the offspring of rodent mothers exposed to Tetrahydrocannabinol (THC): A meta-analysis. Front Psychol 2022; 13:934600. [PMID: 36092118 PMCID: PMC9462465 DOI: 10.3389/fpsyg.2022.934600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/08/2022] [Indexed: 11/17/2022] Open
Abstract
Pre and perinatal administration of Tetrahydrocannabinol (THC) in rodents and their offspring has many effects that have been studied using different methods that have not been integrated using quantitative methods. The effect of THC administration on behavior can be better understood by meta-analytic techniques. We examined whether there is an overall effect on the behavior of the offspring when THC is administered to mothers. Eligibility criteria included experiments using an experimental design with a control group without THC, in which THC is administered to mothers during pregnancy and lactation in rodents, and in which at least one type of behavioral (locomotor, emotional or cognitive) measurement in the offspring was implemented. Cohen’s d was obtained for each study, then each individual study was weighted, and moderator analysis was performed. Analysis was performed using fixed and random effect models, and the heterogeneity was assessed by calculating Qb, I2 and the prediction interval. Furthermore, 3 sub-meta-analyses were carried out according to the type of behavior. The general analysis determined a low weighted effect size of THC on the behavior of the offspring, moderated by type of rat strain. The sub-meta-analyses showed a medium effect for cognitive effects of THC in the offspring, and a low effect on locomotor activity and emotional behavior. In addition, publication bias was not detected. More research is needed to contribute to the understanding of the effect of THC exposure on offspring.
Collapse
Affiliation(s)
- Simón Ramírez
- Department of Psychology, Universidad de Chile, Santiago, Chile
| | - Gonzalo Miguez
- Department of Psychology, Universidad de Chile, Santiago, Chile
| | | | - Luis Pardo
- Department of Psychology, Universidad de Chile, Santiago, Chile
| | - Felipe Alfaro
- Department of Social Sciences and Humanities, Universidad de Aysén, Coyhaique, Chile
| | - Felipe I. Varas
- Department of Psychology, Universidad de Chile, Santiago, Chile
| | - Mario A. Laborda
- Department of Psychology, Universidad de Chile, Santiago, Chile
- *Correspondence: Mario A. Laborda,
| |
Collapse
|
18
|
Martínez-Peña AA, Lee K, Pereira M, Ayyash A, Petrik JJ, Hardy DB, Holloway AC. Prenatal Exposure to Delta-9-tetrahydrocannabinol (THC) Alters the Expression of miR-122-5p and Its Target Igf1r in the Adult Rat Ovary. Int J Mol Sci 2022; 23:ijms23148000. [PMID: 35887347 PMCID: PMC9323798 DOI: 10.3390/ijms23148000] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 02/06/2023] Open
Abstract
As cannabis use during pregnancy increases, it is important to understand its effects on the developing fetus. Particularly, the long-term effects of its psychoactive component, delta-9-tetrahydrocannabinol (THC), on the offspring’s reproductive health are not fully understood. This study examined the impact of gestational THC exposure on the miRNA profile in adult rat ovaries and the possible consequences on ovarian health. Prenatal THC exposure resulted in the differential expression of 12 out of 420 evaluated miRNAs. From the differentially expressed miRNAs, miR-122-5p, which is highly conserved among species, was the only upregulated target and had the greatest fold change. The upregulation of miR-122-5p and the downregulation of its target insulin-like growth factor 1 receptor (Igf1r) were confirmed by RT-qPCR. Prenatally THC-exposed ovaries had decreased IGF-1R-positive follicular cells and increased follicular apoptosis. Furthermore, THC decreased Igf1r expression in ovarian explants and granulosa cells after 48 h. As decreased IGF-1R has been associated with diminished ovarian health and fertility, we propose that these THC-induced changes may partially explain the altered ovarian follicle dynamics observed in THC-exposed offspring. Taken together, our data suggests that prenatal THC exposure may impact key pathways in the developing ovary, which could lead to subfertility or premature reproductive senescence.
Collapse
Affiliation(s)
- Annia A. Martínez-Peña
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada; (A.A.M.-P.); (A.A.)
| | - Kendrick Lee
- The Children’s Health Research Institute, The Lawson Health Research Institute, Departments of Obstetrics and Gynecology and Physiology and Pharmacology, Western University, London, ON N6A 3K7, Canada; (K.L.); (D.B.H.)
| | - Madison Pereira
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (M.P.); (J.J.P.)
| | - Ahmed Ayyash
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada; (A.A.M.-P.); (A.A.)
| | - James J. Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (M.P.); (J.J.P.)
| | - Daniel B. Hardy
- The Children’s Health Research Institute, The Lawson Health Research Institute, Departments of Obstetrics and Gynecology and Physiology and Pharmacology, Western University, London, ON N6A 3K7, Canada; (K.L.); (D.B.H.)
| | - Alison C. Holloway
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4K1, Canada; (A.A.M.-P.); (A.A.)
- Correspondence: ; Tel.: +1-(905)-525-9140 (ext. 22130)
| |
Collapse
|
19
|
Maciel IDS, de Abreu GH, Johnson CT, Bonday R, Bradshaw HB, Mackie K, Lu HC. Perinatal CBD or THC Exposure Results in Lasting Resistance to Fluoxetine in the Forced Swim Test: Reversal by Fatty Acid Amide Hydrolase Inhibition. Cannabis Cannabinoid Res 2022; 7:318-327. [PMID: 34182795 PMCID: PMC9225394 DOI: 10.1089/can.2021.0015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Introduction: There is widespread acceptance of cannabis for medical or recreational use across the society, including pregnant women. Concerningly, numerous studies find that the developing central nervous system (CNS) is vulnerable to the detrimental effects of Δ9-tetrahydrocannabinol (THC). In contrast, almost nothing on the consequences of perinatal cannabidiol (CBD) exposure. In this study, we used mice to investigate the adult impact of perinatal cannabinoid exposure (PCE) with THC, CBD, or a 1:1 ratio of THC and CBD on behaviors. Furthermore, the lasting impact of PCE on fluoxetine sensitivity in the forced swim test (FST) was evaluated to probe neurochemical pathways interacting with the endocannabinoid system (ECS). Methods: Pregnant CD1 dams were injected subcutaneously daily with vehicle, 3 mg/kg THC, 3 mg/kg CBD, or 3 mg/kg THC +3 mg/kg CBD from gestational day 5 to postnatal day 10. Mass spectroscopic (MS) analyses were conducted to measure the THC and CBD brain levels in dams and their embryonic progenies. PCE adults were subjected to a battery of behavioral tests: open field arena, sucrose preference test, marble burying test, nestlet shredding test, and FST. Results: MS analysis found substantial levels of THC and CBD in embryonic brains. Our behavioral testing found that PCE females receiving THC or CBD buried significantly more marbles than control mice. Interestingly, PCE males receiving CBD or THC+CBD had significantly increased sucrose preference. While PCE with THC or CBD did not affect FST immobility, PCE with THC or CBD prevented fluoxetine from decreasing immobility in both males and females. Excitingly, fatty acid amide hydrolase (FAAH) inhibition with a dose of URB597 that was behaviorally inactive in the FST rescued fluoxetine efficacy in PCE mice of both sexes. Conclusions: Our data suggest that PCE with either THC, CBD, or THC+CBD alters repetitive and hedonic behaviors in a phytocannabinoid and sex-dependent manner. In addition, PCE with THC or CBD prevents fluoxetine from enhancing coping behavior. The restoration of fluoxetine responsiveness in THC or CBD PCE adults by inhibition of FAAH suggests that PCE causes a lasting reduction of the ECS and that enhancement of anandamide signaling represents a potential treatment for behavioral deficits following PCE.
Collapse
Affiliation(s)
- Izaque de Sousa Maciel
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA
| | - Gabriel H.D. de Abreu
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA.,Program in Neuroscience, Indiana University, Bloomington, Indiana, USA
| | - Claire T. Johnson
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA.,Program in Neuroscience, Indiana University, Bloomington, Indiana, USA
| | - Rida Bonday
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA
| | - Heather B. Bradshaw
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA.,Program in Neuroscience, Indiana University, Bloomington, Indiana, USA
| | - Ken Mackie
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA.,Program in Neuroscience, Indiana University, Bloomington, Indiana, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA.,Program in Neuroscience, Indiana University, Bloomington, Indiana, USA.,Address correspondence to: Hui-Chen Lu, PhD, The Linda and Jack Gill Center for Biomolecular Science, Indiana University, 702 N Walnut Grove Ave, IN 47405, USA,
| |
Collapse
|
20
|
Hussain S, Breit KR, Thomas JD. The effects of prenatal nicotine and THC E-cigarette exposure on motor development in rats. Psychopharmacology (Berl) 2022; 239:1579-1591. [PMID: 35338387 DOI: 10.1007/s00213-022-06095-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/13/2022] [Indexed: 10/18/2022]
Abstract
RATIONALE In the USA, nicotine and cannabis are the most common licit and illicit drugs used among pregnant women. Importantly, nicotine and cannabis are now being combined for consumption via e-cigarettes, an increasingly popular delivery device. Both nicotine and tetrahydrocannabinol (THC), the primary psychoactive component of cannabis, cross the placenta barrier. However, the consequences of prenatal cannabis use are not well understood, and less is known about potential combination effects when consumed with nicotine, especially via e-cigarettes. OBJECTIVE The present study used a rodent model to examine how prenatal e-cigarette exposure to nicotine, THC, and the combination impacts motor development among offspring. METHODS Pregnant Sprague-Dawley rats were exposed to nicotine (36 mg/mL), THC (100 mg/mL), the combination, or vehicle via e-cigarette inhalation from gestational days (GD) 5-20. One sex pair per litter was tested on an early sensorimotor development task (postnatal days [PD] 12-20) and a parallel bar motor coordination task (PD 30-32). RESULTS Combined prenatal exposure to nicotine and THC delayed sensorimotor development, even though neither drug produced impairments on their own. In contrast, prenatal exposure to either nicotine or THC impaired motor coordination, whereas combined exposure exacerbated these effects, particularly among females. CONCLUSIONS These data illustrate that prenatal exposure to either nicotine or THC may alter motor development, and that the combination may produce more severe effects. These findings have important implications for pregnant women as we better understand the teratogenic effects of these drugs consumed via e-cigarettes.
Collapse
Affiliation(s)
- S Hussain
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, CA, San Diego, USA
| | - K R Breit
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, CA, San Diego, USA.,Department of Psychology, West Chester University of Pennsylvania, West Chester, PA, USA
| | - J D Thomas
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, CA, San Diego, USA.
| |
Collapse
|
21
|
Ikeda AS, Knopik VS, Bidwell LC, Parade SH, Goodman SH, Emory EK, Palmer RHC. A Review of Associations between Externalizing Behaviors and Prenatal Cannabis Exposure: Limitations & Future Directions. TOXICS 2022; 10:toxics10010017. [PMID: 35051059 PMCID: PMC8779620 DOI: 10.3390/toxics10010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/13/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023]
Abstract
In utero cannabis exposure can disrupt fetal development and increase risk for various behavioral disruptions, including hyperactivity, inattention, delinquent behaviors, and later substance abuse, among others. This review summarizes the findings from contemporary investigations linking prenatal cannabis exposure to the development of psychopathology and identifies the limitations within the literature, which constrain our interpretations and generalizability. These limitations include a lack of genetic/familial control for confounding and limited data examining real world products, the full range of cannabinoids, and motives for use specifically in pregnant women. Taken together, our review reveals the need to continue to improve upon study designs in order to allow researchers to accurately draw conclusions about the development of behavioral consequences of prenatal cannabis exposure. Findings from such studies would inform policy and practices regarding cannabis use during pregnancy and move the field toward developing a comprehensive teratogenic profile of cannabis similar to what is characterized in the prenatal alcohol and tobacco literature.
Collapse
Affiliation(s)
- Ami S. Ikeda
- Behavioral Genetics of Addiction Laboratory, Emory University, Atlanta, GA 30322, USA
- Department of Psychology, Emory University, Atlanta, GA 30322, USA; (S.H.G.); (E.K.E.)
- Correspondence: (A.S.I.); (R.H.C.P.)
| | - Valerie S. Knopik
- Department of Human Development and Family Studies, College of Health and Human Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - L. Cinnamon Bidwell
- Institute of Cognitive Science, University of Colorado Boulder, Boulder, CO 80309, USA;
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Stephanie H. Parade
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI 02903, USA;
- Bradley/Hasbro Children’s Research Center, E.P. Bradley Hospital, East Providence, RI 02915, USA
| | - Sherryl H. Goodman
- Department of Psychology, Emory University, Atlanta, GA 30322, USA; (S.H.G.); (E.K.E.)
| | - Eugene K. Emory
- Department of Psychology, Emory University, Atlanta, GA 30322, USA; (S.H.G.); (E.K.E.)
| | - Rohan H. C. Palmer
- Behavioral Genetics of Addiction Laboratory, Emory University, Atlanta, GA 30322, USA
- Department of Psychology, Emory University, Atlanta, GA 30322, USA; (S.H.G.); (E.K.E.)
- Correspondence: (A.S.I.); (R.H.C.P.)
| |
Collapse
|
22
|
Conrad SE, Davis D, Vilcek N, Thompson JB, Guarino S, Papini S, Papini MR. Frustrative nonreward and cannabinoid receptors: Chronic (but not acute) WIN 55,212-2 treatment increased resistance to change in two reward downshift tasks. Pharmacol Biochem Behav 2022; 213:173320. [PMID: 34990705 DOI: 10.1016/j.pbb.2021.173320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 10/19/2022]
Abstract
Assessing the role of cannabinoid (CB) receptors in behavior is relevant given the trend toward the legalization of medicinal and recreational marijuana. The present research aims at bridging a gap in our understanding of CB-receptor function in animal models of frustrative nonreward. These experiments were designed to (1) determine the effects of chronic administration of the nonselective CB1-receptor agonist WIN 55,212-2 (WIN) on reward downshift in rats and (2) determine whether the effects of chronic WIN were reducible to acute effects. In Experiment 1, chronic WIN (7 daily injections, 10 mg/kg, ip) accelerated the recovery of consummatory behavior after a 32-to-4% sucrose downshift relative to vehicle controls. In addition, chronic WIN eliminated the preference for an unshifted lever when the other lever was subject to a 12-to-2 pellet downshift in free-choice trials, but only in animals with previous experience with a sucrose downshift. In Experiment 2, acute WIN (1 mg/kg, ip) reduced consummatory behavior, but did not affect recovery from a 32-to-4% sucrose downshift. The antagonist SR 141716A (3 mg/kg, ip) also failed to interfere with recovery after the sucrose downshift. In Experiment 3, acute WIN administration (1 mg/kg, ip) did not affect free-choice behavior after a pellet downshift, although it reduced lever pressing and increased magazine entries relative to vehicle controls. The effects of chronic WIN on frustrative nonreward were not reducible to acute effects of the drug. Chronic WIN treatment in rats, like chronic marijuana use in humans, seems to increase resistance to the effects of frustrative nonreward.
Collapse
Affiliation(s)
- Shannon E Conrad
- Department of Psychology, Texas Christian University, Fort Worth, TX 76129, USA
| | - Delaney Davis
- Department of Psychology, Texas Christian University, Fort Worth, TX 76129, USA
| | - Natalia Vilcek
- Department of Psychology, Texas Christian University, Fort Worth, TX 76129, USA
| | - Joanna B Thompson
- Department of Psychology, Texas Christian University, Fort Worth, TX 76129, USA
| | - Sara Guarino
- Department of Psychology, Texas Christian University, Fort Worth, TX 76129, USA
| | - Santiago Papini
- Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA
| | - Mauricio R Papini
- Department of Psychology, Texas Christian University, Fort Worth, TX 76129, USA.
| |
Collapse
|
23
|
Sasso EB, Bolshakova M, Bogumil D, Johnson B, Komatsu E, Sternberg J, Cortessis V, Mullin P. Marijuana use and perinatal outcomes in obstetric patients at a safety net hospital. Eur J Obstet Gynecol Reprod Biol 2021; 266:36-41. [PMID: 34562806 DOI: 10.1016/j.ejogrb.2021.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/31/2021] [Accepted: 09/13/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To characterize the association between antepartum marijuana exposure and maternal and neonatal outcomes at our institution. STUDY DESIGN Retrospective chart review identified an obstetric cohort of singleton gestations. Women with self-reported marijuana use were compared with non-users. Demographic characteristics, risk factors, and maternal-fetal outcomes were evaluated. Associations between outcomes and marijuana use were assessed with regression analysis. RESULTS Of 2792 deliveries, 5.4% reported marijuana use. Compared to non-users, marijuana users entered prenatal care later, were younger, non-Hispanic, and used other illicit substances. Marijuana users had a higher rate of cesarean delivery (p = 0.01). After adjusting for confounders, marijuana use remained associated with 4.1-fold risk of delivering a small for gestational age (SGA) infant and 2.89-fold risk of neonatal oxygen use. CONCLUSION At a safety net hospital, antepartum marijuana use is significantly associated with cesarean delivery, SGA and supplemental oxygen use at birth. Healthcare disparities associated with marijuana use make this a population of critical interest.
Collapse
Affiliation(s)
- Elizabeth B Sasso
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Maria Bolshakova
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David Bogumil
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brittney Johnson
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Emi Komatsu
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jennifer Sternberg
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Victoria Cortessis
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Patrick Mullin
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
24
|
The Impact of Early Life Exposure to Cannabis: The Role of the Endocannabinoid System. Int J Mol Sci 2021; 22:ijms22168576. [PMID: 34445282 PMCID: PMC8395329 DOI: 10.3390/ijms22168576] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 01/14/2023] Open
Abstract
Cannabis use during pregnancy has continued to rise, particularly in developed countries, as a result of the trend towards legalization and lack of consistent, evidence-based knowledge on the matter. While there is conflicting data regarding whether cannabis use during pregnancy leads to adverse outcomes such as stillbirth, preterm birth, low birthweight, or increased admission to neonatal intensive care units, investigations into long-term effects on the offspring’s health are limited. Historically, studies have focused on the neurobehavioral effects of prenatal cannabis exposure on the offspring. The effects of cannabis on other physiological aspects of the developing fetus have received less attention. Importantly, our knowledge about cannabinoid signaling in the placenta is also limited. The endocannabinoid system (ECS) is present at early stages of development and represents a potential target for exogenous cannabinoids in utero. The ECS is expressed in a broad range of tissues and influences a spectrum of cellular functions. The aim of this review is to explore the current evidence surrounding the effects of prenatal exposure to cannabinoids and the role of the ECS in the placenta and the developing fetus.
Collapse
|
25
|
Silva MH. Chlorpyrifos and Δ 9 Tetrahydrocannabinol exposure and effects on parameters associated with the endocannabinoid system and risk factors for obesity. Curr Res Toxicol 2021; 2:296-308. [PMID: 34467221 PMCID: PMC8384771 DOI: 10.1016/j.crtox.2021.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 12/15/2022] Open
Abstract
Marilyn Silva. Retired from a career in toxicology and risk assessment. Increased childhood and adult obesity are associated with chlorpyrifos (CPF), an organophosphate pesticide. Cannabis (Δ9Tetrahydrocannabinol: Δ9THC) use has increased globally with legalization. CPF applications on cannabis crops lacks federally regulated tolerances and may pose health risks through exposure during development and in adulthood. Both CPF and Δ9THC affect the endocannabinoid system (eCBS), a regulator of appetite, energy balance, and gut microbiota, which, if disrupted, increases risk for obesity and related diseases. CPF inhibits eCB metabolism and Δ9THC is a partial agonist/antagonist at the cannabinoid receptor (CB1R). Effects of each on obesogenic parameters were examined via literature search. Male rodents with CPF exposure showed increased body weights, dysbiosis, inflammation and oxidative stress, potentially associated with increased eCBs acting through the gut-microbiota-adipose-brain regulatory loop. Δ9THC generally decreased body weights via partial agonism at the CB1R, lowering levels of eCBs. Dysbiosis and/or oxidative stress associated inflammation occurred with CPF, but these parameters were not tested with Δ9THC. Database deficiencies included limited endpoints to compare between chemicals/age-groups, inter-study variables (dose ranges, dosing vehicle, rodent strain, treatment duration, etc.). CPF and Δ9THC were not tested together, but human co-chemical effects would depend on exposure ratio, subject age, exposure duration, and health status, among others. An overriding concern is that both chemicals are well-documented developmental neurotoxins in addition to their low dose effects on energy balance. A co-exposure risk assessment is warranted with increased use and lack of federal CPF regulation on cannabis.
Collapse
Affiliation(s)
- Marilyn H. Silva
- Retired from a career in toxicology and risk assessment 2437, Evenstar Lane, Davis, CA 95616, United States
| |
Collapse
|
26
|
Akhtar A, Pilkhwal Sah S. Advances in the pharmacotherapeutic management of post-traumatic stress disorder. Expert Opin Pharmacother 2021; 22:1919-1930. [PMID: 34124975 DOI: 10.1080/14656566.2021.1935871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Post-traumatic stress disorder (PTSD), a mental disorder, is associated with anxiety, depression, and social awkwardness resulting from past traumatic episodes like natural disasters, accidents, terrorist attacks, war, rape, and sexual violence. It affects primarily the amygdala, cortex, and hippocampus where neurochemical changes result in altered behavior. PTSD patients display impaired fear extinction, and past events keep haunting them. The topic presents relevant sections like PTSD pharmacotherapy, associated challenges, and the novel targets and drugs for future research and therapy.Areas covered: The authors discuss the current pharmacotherapy like SSRIs, NDRIs, SNRIs, anticonvulsants, antidepressants, and benzodiazepines, used to attenuate the associated symptoms. However, the primary focus being the novel and potential targets which can be explored better to understand possible future research and advanced therapy in PTSD. For the same, an account of both preclinical and clinical studies has been covered.Expert opinion: Excessive adverse effects, limited efficacy, and lower patient compliance are some of the major challenges with conventional drugs. Moreover, they correct only fewer symptoms without halting the disease progression. Several agents are investigated in different preclinical and clinical phases, which can potentially overcome the pitfalls and limitations associated with conventional therapies.
Collapse
Affiliation(s)
- Ansab Akhtar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Sangeeta Pilkhwal Sah
- Pharmacology Division, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
27
|
Pinky PD, Majrashi M, Fujihashi A, Bloemer J, Govindarajulu M, Ramesh S, Reed MN, Moore T, Suppiramaniam V, Dhanasekaran M. Effects of prenatal synthetic cannabinoid exposure on the cerebellum of adolescent rat offspring. Heliyon 2021; 7:e06730. [PMID: 33912711 PMCID: PMC8066425 DOI: 10.1016/j.heliyon.2021.e06730] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/19/2020] [Accepted: 04/01/2021] [Indexed: 11/25/2022] Open
Abstract
Cannabis is the most commonly used illicit drug worldwide. Recently, cannabis use among young pregnant women has greatly increased. However, prenatal cannabinoid exposure leads to long-lasting cognitive, motor, and behavioral deficits in the offspring and alterations in neural circuitry through various mechanisms. Although these effects have been studied in the hippocampus, the effects of prenatal cannabinoid exposure on the cerebellum are not well elucidated. The cerebellum plays an important role in balance and motor control, as well as cognitive functions such as attention, language, and procedural memories. The aim of this study was to investigate the effects of prenatal cannabinoid exposure on the cerebellum of adolescent offspring. Pregnant rats were treated with synthetic cannabinoid agonist WIN55,212-2, and the offspring were evaluated for various cerebellar markers of oxidative stress, mitochondrial function, and apoptosis. Additionally, signaling proteins associated with glutamate dependent synaptic plasticity were examined. Administration of WIN55,212-2 during pregnancy altered markers of oxidative stress by significantly reducing oxidative stress and nitrite content. Mitochondrial Complex I and Complex IV activities were also enhanced following prenatal cannabinoid exposure. With regard to apoptosis, pP38 levels were significantly increased, and proapoptotic factor caspase-3 activity, pERK, and pJNK levels were significantly decreased. CB1R and GluA1 levels remained unchanged; however, GluN2A was significantly reduced. There was a significant decrease in MAO activity although tyrosine hydroxylase activity was unaltered. Our study indicates that the effects of prenatal cannabinoid exposure on the cerebellum are unique compared to other brain regions by enhancing mitochondrial function and promoting neuronal survival. Further studies are required to evaluate the mechanisms by which prenatal cannabinoid exposure alters cerebellar processes and the impact of these alterations on behavior.
Collapse
Affiliation(s)
- Priyanka D. Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
- Center for Neuroscience Initiatives, Auburn University, Auburn, AL, USA
| | - Mohammed Majrashi
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Ayaka Fujihashi
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
- Department of Biomedical and Pharmaceutical Sciences, Touro College of Pharmacy, New York, NY, USA
| | - Manoj Govindarajulu
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
- Center for Neuroscience Initiatives, Auburn University, Auburn, AL, USA
| | - Sindhu Ramesh
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
- Center for Neuroscience Initiatives, Auburn University, Auburn, AL, USA
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
- Center for Neuroscience Initiatives, Auburn University, Auburn, AL, USA
| | - Timothy Moore
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
- Center for Neuroscience Initiatives, Auburn University, Auburn, AL, USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
- Center for Neuroscience Initiatives, Auburn University, Auburn, AL, USA
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
- Center for Neuroscience Initiatives, Auburn University, Auburn, AL, USA
| |
Collapse
|
28
|
Walker OS, Gurm H, Sharma R, Verma N, May LL, Raha S. Delta-9-tetrahydrocannabinol inhibits invasion of HTR8/SVneo human extravillous trophoblast cells and negatively impacts mitochondrial function. Sci Rep 2021; 11:4029. [PMID: 33597628 PMCID: PMC7889882 DOI: 10.1038/s41598-021-83563-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 01/29/2021] [Indexed: 01/31/2023] Open
Abstract
Prenatal cannabis use is a significant problem and poses important health risks for the developing fetus. The molecular mechanisms underlying these changes are not fully elucidated but are thought to be attributed to delta-9-tetrahydrocannabinol (THC), the main bioactive constituent of cannabis. It has been reported that THC may target the mitochondria in several tissue types, including placental tissue and trophoblast cell lines, and alter their function. In the present study, in response to 48-h THC treatment of the human extravillous trophoblast cell line HTR8/SVneo, we demonstrate that cell proliferation and invasion are significantly reduced. We further demonstrate THC-treatment elevated levels of cellular reactive oxygen species and markers of lipid damage. This was accompanied by evidence of increased mitochondrial fission. We also observed increased expression of cellular stress markers, HSP70 and HSP60, following exposure to THC. These effects were coincident with reduced mitochondrial respiratory function and a decrease in mitochondrial membrane potential. Taken together, our results suggest that THC can induce mitochondrial dysfunction and reduce trophoblast invasion; outcomes that have been previously linked to poor placentation. We also demonstrate that these changes in HTR8/SVneo biology may be variably mediated by cannabinoid receptors CB1 and CB2.
Collapse
Affiliation(s)
- O’Llenecia S. Walker
- grid.25073.330000 0004 1936 8227Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| | - Harmeet Gurm
- grid.25073.330000 0004 1936 8227Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| | - Reeti Sharma
- grid.25073.330000 0004 1936 8227 Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| | - Navkiran Verma
- grid.25073.330000 0004 1936 8227 Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| | - Linda L. May
- grid.25073.330000 0004 1936 8227 Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| | - Sandeep Raha
- grid.25073.330000 0004 1936 8227Graduate Program in Medical Sciences, Department of Pediatrics, McMaster University, HSC 4H7, Hamilton, ON L8S 4K1 Canada
| |
Collapse
|
29
|
Breijyeh Z, Jubeh B, Bufo SA, Karaman R, Scrano L. Cannabis: A Toxin-Producing Plant with Potential Therapeutic Uses. Toxins (Basel) 2021; 13:117. [PMID: 33562446 PMCID: PMC7915118 DOI: 10.3390/toxins13020117] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023] Open
Abstract
For thousands of years, Cannabis sativa has been utilized as a medicine and for recreational and spiritual purposes. Phytocannabinoids are a family of compounds that are found in the cannabis plant, which is known for its psychotogenic and euphoric effects; the main psychotropic constituent of cannabis is Δ9-tetrahydrocannabinol (Δ9-THC). The pharmacological effects of cannabinoids are a result of interactions between those compounds and cannabinoid receptors, CB1 and CB2, located in many parts of the human body. Cannabis is used as a therapeutic agent for treating pain and emesis. Some cannabinoids are clinically applied for treating chronic pain, particularly cancer and multiple sclerosis-associated pain, for appetite stimulation and anti-emesis in HIV/AIDS and cancer patients, and for spasticity treatment in multiple sclerosis and epilepsy patients. Medical cannabis varies from recreational cannabis in the chemical content of THC and cannabidiol (CBD), modes of administration, and safety. Despite the therapeutic effects of cannabis, exposure to high concentrations of THC, the main compound that is responsible for most of the intoxicating effects experienced by users, could lead to psychological events and adverse effects that affect almost all body systems, such as neurological (dizziness, drowsiness, seizures, coma, and others), ophthalmological (mydriasis and conjunctival hyperemia), cardiovascular (tachycardia and arterial hypertension), and gastrointestinal (nausea, vomiting, and thirst), mainly associated with recreational use. Cannabis toxicity in children is more concerning and can cause serious adverse effects such as acute neurological symptoms (stupor), lethargy, seizures, and even coma. More countries are legalizing the commercial production and sale of cannabis for medicinal use, and some for recreational use as well. Liberalization of cannabis laws has led to increased incidence of toxicity, hyperemesis syndrome, lung disease cardiovascular disease, reduced fertility, tolerance, and dependence with chronic prolonged use. This review focuses on the potential therapeutic effects of cannabis and cannabinoids, as well as the acute and chronic toxic effects of cannabis use on various body systems.
Collapse
Affiliation(s)
- Zeinab Breijyeh
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem Abu Dis P144, Palestine; (Z.B.); (B.J.)
| | - Buthaina Jubeh
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem Abu Dis P144, Palestine; (Z.B.); (B.J.)
| | - Sabino A. Bufo
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
- Department of Geography, Environmental Management & Energy Studies, University of Johannesburg, Johannesburg 2092, South Africa
| | - Rafik Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem Abu Dis P144, Palestine; (Z.B.); (B.J.)
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
| | - Laura Scrano
- Department of European Cultures (DICEM), University of Basilicata, 75100 Matera, Italy;
| |
Collapse
|
30
|
Odom GC, Cottler LB, Striley CW, Lopez-Quintero C. Perceived Risk of Weekly Cannabis Use, Past 30-Day Cannabis Use, and Frequency of Cannabis Use Among Pregnant Women in the United States. Int J Womens Health 2020; 12:1075-1088. [PMID: 33235517 PMCID: PMC7678496 DOI: 10.2147/ijwh.s266540] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/12/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND While accumulated evidence has shown that the prevalence of cannabis use among pregnant women in the US has increased in recent years, little is known about the specific subpopulations affected. The aim of this study was to estimate the prevalence and correlates of the perceived risk of weekly cannabis use, past 30-day cannabis use, and frequency of past 30-day cannabis use among US pregnant women. METHODS We analyzed data from 2,247 pregnant women 14 to 44 years of age surveyed in the 2015 to 2017 cross-sectional National Survey on Drug Use and Health. Analyses account for the sampling design. Primary outcomes included perceived risk of weekly cannabis use, past 30-day cannabis use, and frequency of cannabis use. We conducted multivariable logistic and negative binomial regression models to assess the associations between the primary outcomes and multiple correlates. RESULTS Among US pregnant women, 21.6% (95% CI=19.4, 23.8) did not perceive any risk associated with weekly cannabis use, 5.3% (95% CI=4.2, 6.5) used cannabis in the past 30 days, and among past-month users, the average number of days of use was 15.6 (95% CI=13.5, 17.7). Pregnant women living below the poverty line were both more likely to perceive no risk of weekly cannabis use (aOR=1.8; 95% CI=1.3, 2.5) and use cannabis more often in the past 30 days (aOR=2.9; 95% CI=1.5, 5.7) than pregnant women within an income bracket of more than two times the federal poverty threshold. Age, race, trimester of pregnancy, co-use of tobacco and/or alcohol were also associated with these outcomes. CONCLUSION Younger age, living in poverty, early trimester of pregnancy, and co-use of tobacco and/or alcohol increased the odds of cannabis use among pregnant women. As cannabis legalization spreads and cannabis use is increasingly perceived as safe, there is a growing need for research to determine the reasons why women in the identified at-risk subgroups are using cannabis during pregnancy.
Collapse
Affiliation(s)
- Gage C Odom
- Department of Epidemiology, University of Florida, Gainesville, FL32611, USA
| | - Linda B Cottler
- Department of Epidemiology, University of Florida, Gainesville, FL32611, USA
| | - Catherine W Striley
- Department of Epidemiology, University of Florida, Gainesville, FL32611, USA
| | | |
Collapse
|
31
|
Perinatal THC exposure via lactation induces lasting alterations to social behavior and prefrontal cortex function in rats at adulthood. Neuropsychopharmacology 2020; 45:1826-1833. [PMID: 32428929 PMCID: PMC7608083 DOI: 10.1038/s41386-020-0716-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/16/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022]
Abstract
Cannabis is the world's most widely abused illicit drug and consumption amongst women during and surrounding the period of pregnancy is increasing. Previously, we have shown that cannabinoid exposure via lactation during the early postnatal period disrupts early developmental trajectories of prefrontal cortex maturation and induces behavioral abnormalities during the first weeks of life in male and female rat progeny. Here, we investigated the lasting consequences of this postnatal cannabinoid exposure on synaptic and behavioral parameters in the adult offspring of ∆9-tetrahydrocannabinol (THC)-treated dams. At adulthood, these perinatally THC-exposed rats exhibits deficits in social discrimination accompanied by an overall augmentation of social exploratory behavior. These behavioral alterations were further correlated with multiple abnormalities in synaptic plasticity in the prefrontal cortex, including lost endocannabinoid-mediated long-term depression (LTD), lost long-term potentiation and augmented mGlu2/3-LTD. Finally, basic parameters of intrinsic excitability at prefrontal cortex pyramidal neurons were similarly altered by the perinatal THC exposure. Thus, perinatal THC exposure via lactation induces lasting deficits in behavior and synaptic function which persist into adulthood life in male and female progeny.
Collapse
|
32
|
Weimar HV, Wright HR, Warrick CR, Brown AM, Lugo JM, Freels TG, McLaughlin RJ. Long-term effects of maternal cannabis vapor exposure on emotional reactivity, social behavior, and behavioral flexibility in offspring. Neuropharmacology 2020; 179:108288. [PMID: 32860776 DOI: 10.1016/j.neuropharm.2020.108288] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/11/2020] [Accepted: 08/24/2020] [Indexed: 01/21/2023]
Abstract
The use of cannabis during pregnancy is a growing public health concern. As more countries implement legislation permitting recreational cannabis use, there is an urgent need to better understand its impact on fetal neurodevelopment and its long-term effects in exposed offspring. Studies examining effects of prenatal cannabis exposure typically employ injections of synthetic cannabinoids or isolated cannabis constituents that may not accurately model cannabis use in human populations. To address this limitation, we developed a novel e-cigarette technology-based system to deliver vaporized cannabis extracts to pregnant Long Evans rats. We used this model to determine effects of prenatal cannabis exposure on emotional, social, and cognitive endpoints of male and female offspring during early development and into adulthood. Dams were exposed to cannabis vapor (CANTHC: 400 mg/ml), vehicle vapor (VEH), or no vapor (AIR) twice daily during mating and gestation. Offspring exposed to CANTHC and VEH showed reduced weight gain relative to AIR offspring prior to weaning. CANTHC offspring made more isolation-induced ultrasonic vocalizations (USVs) on postnatal day 6 (P6) relative to VEH-exposed offspring, which is indicative of increased emotional reactivity. Male CANTHC offspring engaged in fewer social investigation behaviors than VEH-exposed male offspring during a social play test on P26. In adulthood, CANTHC-exposed offspring spent less time exploring the open arms of the elevated plus maze and exhibited dose-dependent deficits in behavioral flexibility in an attentional set-shifting task relative to AIR controls. These data collectively indicate that prenatal cannabis exposure may cause enduring effects on the behavioral profile of offspring.
Collapse
Affiliation(s)
- Halle V Weimar
- Departments of Integrative Physiology and Neuroscience, Washington State University, WA, USA
| | - Hayden R Wright
- Departments of Integrative Physiology and Neuroscience, Washington State University, WA, USA
| | - Collin R Warrick
- Departments of Integrative Physiology and Neuroscience, Washington State University, WA, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Amanda M Brown
- Departments of Integrative Physiology and Neuroscience, Washington State University, WA, USA
| | - Janelle M Lugo
- Departments of Integrative Physiology and Neuroscience, Washington State University, WA, USA
| | - Timothy G Freels
- Departments of Integrative Physiology and Neuroscience, Washington State University, WA, USA
| | - Ryan J McLaughlin
- Departments of Integrative Physiology and Neuroscience, Washington State University, WA, USA; Department of Psychology, Washington State University, WA, USA.
| |
Collapse
|
33
|
Barnes JL, Mohr C, Ritchey CR, Erikson CM, Shiina H, Rossi DJ. Developmentally Transient CB1Rs on Cerebellar Afferents Suppress Afferent Input, Downstream Synaptic Excitation, and Signaling to Migrating Neurons. J Neurosci 2020; 40:6133-6145. [PMID: 32631938 PMCID: PMC7406284 DOI: 10.1523/jneurosci.1931-19.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 06/15/2020] [Accepted: 06/20/2020] [Indexed: 01/13/2023] Open
Abstract
The endocannabinoid system plays important roles in brain development, but mechanistic studies have focused on neuronal differentiation, migration, and synaptogenesis, with less attention to transcellular interactions that coordinate neurodevelopmental processes across developing neural networks. We determined that, in the developing rodent cerebellar cortex (of both sexes), there is a transient window when the dominant brain cannabinoid receptor, CB1R, is expressed on afferent terminals instead of output neuron Purkinje cell synapses that dominate the adult cerebellum. Activation of these afferent CB1Rs suppresses synaptic transmission onto developing granule cells, and consequently also suppresses excitation of downstream neurons in the developing cortical network, including nonsynaptic, migrating neurons. Application of a CB1R antagonist during afferent stimulation trains and depolarizing voltage steps caused a significant, sustained potentiation of synaptic amplitude. Our data demonstrate that transiently expressed afferent CB1Rs regulate afferent synaptic strength during synaptogenesis, which enables coordinated dampening of transcortical developmental signals.SIGNIFICANCE STATEMENT The endogenous cannabinoid system plays diverse roles in brain development, which, combined with the rapidly changing legal and medical status of cannabis-related compounds, makes understanding how exogenous cannabinoids affect brain development an important biomedical objective. The cerebellum is a key brain region in a variety of neurodevelopmental disorders, and the adult cerebellum has one of the highest expression levels of CB1R, but little is known about CB1R in the developing cerebellum. Here we report a developmentally distinct expression and function of CB1R in the cerebellum, in which endogenous or exogenous activation of CB1Rs modifies afferent synaptic strength and coordinated downstream network signaling. These findings have implications for recreational and medical use of exogenous cannabinoids by pregnant and breastfeeding women.
Collapse
MESH Headings
- Action Potentials
- Animals
- Cannabinoid Receptor Antagonists/pharmacology
- Cell Movement
- Excitatory Postsynaptic Potentials
- Female
- Male
- Mice
- Mice, Inbred C57BL
- Neurogenesis
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- Neurons, Afferent/physiology
- Purkinje Cells/drug effects
- Purkinje Cells/metabolism
- Purkinje Cells/physiology
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Synapses/metabolism
- Synapses/physiology
Collapse
Affiliation(s)
- Jesse L Barnes
- Washington State University, Integrative Physiology and Neuroscience, Pullman, Washington 99164
| | - Claudia Mohr
- Washington State University, Integrative Physiology and Neuroscience, Pullman, Washington 99164
| | - Caitlin R Ritchey
- Washington State University, Integrative Physiology and Neuroscience, Pullman, Washington 99164
| | - Chloe M Erikson
- Washington State University, Integrative Physiology and Neuroscience, Pullman, Washington 99164
| | - Hiroko Shiina
- Washington State University, Integrative Physiology and Neuroscience, Pullman, Washington 99164
| | - David J Rossi
- Washington State University, Integrative Physiology and Neuroscience, Pullman, Washington 99164
| |
Collapse
|
34
|
Mustafa HJ, Cross SN, Jacobs KM, Tessier KM, Tofte AN, McCarter AR, Narasimhan SL. Preterm Birth of Infants Prenatally Diagnosed with Congenital Heart Disease, Characteristics, Associations, and Outcomes. Pediatr Cardiol 2020; 41:972-978. [PMID: 32356015 PMCID: PMC7394484 DOI: 10.1007/s00246-020-02345-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/08/2020] [Indexed: 11/26/2022]
Abstract
There are limited data on the relation between congenital heart disease (CHD) and preterm birth (PTB). We aimed to estimate the risk of PTB in newborns with CHD, to study associations and risk factors (modifiable and non-modifiable) as well as investigate postnatal outcomes. This was a retrospective cohort study of 336 pregnancies diagnosed with CHD between 2011 and 2016. Groups consisted of those delivered at or after 37 weeks, and those who delivered prior to 37 weeks. Collected data included maternal and fetal characteristics as well postnatal outcomes. Complete data were obtained from 237 singleton pregnancies. The overall proportion of PTB was 23.2% for all CHD, of which 38.2% were spontaneous PTB which was almost unchanged after excluding extracardiac anomalies and pathogenic chromosomal abnormalities. Significant non-modifiable risk factors were pregnancy-related HTN disorders (P < 0.001), fetal growth restriction (P = 0.01), and pathogenic chromosomal abnormalities (P = 0.046). Significant PTB modifiable risk factors included prenatal marijuana use (P = 0.01). Pregnancies delivered at 37-38 weeks had significantly more newborns with birthweight < 2500 g (P < 0.001), required more pre-operative NICU support including intubation (P = 0.049), vasopressors (P = 0.04), prostaglandins (P = 0.003), antibiotics (P = 0.01), and had longer hospital stay (P = 0.001) than those delivered at ≥ 39 weeks. Prenatally diagnosed pregnancies with CHD had higher PTB rate compared to the general population, with spontaneous PTB comprising 38.2% of these preterm deliveries. Most PTB risk factors were non-modifiable, however, significant modifiable factors included marijuana use in pregnancy. Outcomes were favorable in neonates delivered at or beyond 39 weeks.
Collapse
Affiliation(s)
- Hiba J Mustafa
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Women's Health, University of Minnesota, Minneapolis, MN, USA.
| | - Sarah N Cross
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Women's Health, University of Minnesota, Minneapolis, MN, USA
| | - Katherine M Jacobs
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Women's Health, University of Minnesota, Minneapolis, MN, USA
| | - Katelyn M Tessier
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Shanti L Narasimhan
- Department of Pediatrics Cardiology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
35
|
Brancato A, Castelli V, Lavanco G, Marino RAM, Cannizzaro C. In utero Δ9-tetrahydrocannabinol exposure confers vulnerability towards cognitive impairments and alcohol drinking in the adolescent offspring: Is there a role for neuropeptide Y? J Psychopharmacol 2020; 34:663-679. [PMID: 32338122 DOI: 10.1177/0269881120916135] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cannabinoid consumption during pregnancy has been increasing on the wave of the broad-based legalisation of cannabis in Western countries, raising concern about the putative detrimental outcomes on foetal neurodevelopment. Indeed, since the endocannabinoid system regulates synaptic plasticity, emotional and cognitive processes from early stages of life interfering with it and other excitability endogenous modulators, such as neuropeptide Y (NPY), might contribute to the occurrence of a vulnerable phenotype later in life. AIMS This research investigated whether in utero exposure to Δ9-tetrahydrocannabinol (THC) may induce deficits in emotional/cognitive processes and alcohol vulnerability in adolescent offspring. NPY and excitatory postsynaptic density (PSD) machinery were measured as markers of neurobiological vulnerability. METHODS Following in utero THC exposure (2 mg/kg delivered subcutaneously), preadolescent male rat offspring were assessed for: behavioural reactivity in the open field test, neutral declarative memory and aversive limbic memory in the Novel Object and Emotional Object Recognition tests, immunofluorescence for NPY neurons and the PSD proteins Homer-1, 1b/c and 2 in the prefrontal cortex, amygdala and nucleus accumbens at adolescence (cohort 1); and instrumental learning, alcohol taking, relapse and conflict behaviour in the operant chamber throughout adolescence until early adulthood (cohort 2). RESULTS In utero THC-exposed adolescent rats showed: (a) increased locomotor activity; (b) no alteration in neutral declarative memory; (c) impaired aversive limbic memory; (d) decreased NPY-positive neurons in limbic regions; (e) region-specific variations in Homer-1, 1b/c and 2 immunoreactivity; (f) decreased instrumental learning and increased alcohol drinking, relapse and conflict behaviour in the operant chamber. CONCLUSION Gestational THC impaired the formation of memory traces when integration between environmental encoding and emotional/motivational processing was required and promoted the development of alcohol-addictive behaviours. The abnormalities in NPY signalling and PSD make-up may represent the common neurobiological background, suggesting new targets for future research.
Collapse
Affiliation(s)
- Anna Brancato
- Department of Health Promotion, Mother-Child Care, Internal Medicine and Medical Specialties of Excellence 'G. D'Alessandro', University of Palermo, Palermo, Italy
| | - Valentina Castelli
- Department of Health Promotion, Mother-Child Care, Internal Medicine and Medical Specialties of Excellence 'G. D'Alessandro', University of Palermo, Palermo, Italy.,Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Gianluca Lavanco
- INSERM U1215, NeuroCentre Magendie, Bordeaux, France.,University of Bordeaux, Bordeaux, France.,Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Rosa Anna Maria Marino
- Department of Anatomy and Neurobiology, School of Medicine, University of Maryland, Baltimore, USA
| | - Carla Cannizzaro
- Department of Health Promotion, Mother-Child Care, Internal Medicine and Medical Specialties of Excellence 'G. D'Alessandro', University of Palermo, Palermo, Italy
| |
Collapse
|
36
|
Sex differences in the interactive effects of early life stress and the endocannabinoid system. Neurotoxicol Teratol 2020; 80:106893. [PMID: 32437941 DOI: 10.1016/j.ntt.2020.106893] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/15/2022]
Abstract
Sex differences in both the endocannabinoid system and stress responses have been established for decades. While there is ample evidence that the sexes respond differently to stress and that the endocannabinoid system is involved in this response, what is less clear is whether the endocannabinoid system mediates this response to stress differently in both sexes. Also, do the sexes respond similarly to exogenous cannabinoids (CBs) following stress? Can the administration of exogenous CBs normalize the effects of stress and if so, does this happen similarly in male and female subjects? This review will attempt to delineate the stress induced neurochemical alterations in the endocannabinoid system and the resulting behavioral changes across periods of development: prenatal, early neonatal or adolescent in males and females. Within this frame work, we will then examine the neurochemical and behavioral effects of exogenous CBs and illustrate that the response to CBs is determined by the stress history of the animal. The theoretical framework for this endeavor relates to the established effects of adverse childhood experiences (ACE) in increasing substance abuse, depression and anxiety and the possibility that individuals with high ACE scores may consume cannabinoids to "self-medicate". Overall, we see that while there are instances where exogenous cannabinoids "normalize" the adverse effects produced by early stress, this normalization does not occur in all animal models with any sort of consistency. The most compelling report where CB administration appears to normalize behaviors altered by early stress, shows minimal differences between the sexes (Alteba et al., 2016). This is in stark contrast to the majority of studies on early stress and the endocannabinoid system where both sexes are included and show quite divergent, in fact opposite, effects in males and females. Frequently there is a disconnect between neurochemical changes and behavioral changes and often, exogenous CBs have greater effects in stressed animals compared to non-stressed controls. This report as well as others reviewed here do support the concept that the effects of exogenous CBs are different in individuals experiencing early stress and that these differences are not equal in males and females. However, due to the wide variety of stressors used and the range of ages when the stress is applied, additional careful studies are warranted to fully understand the interactive effects of stress and the endocannabinoid system in males and females. In general, the findings do not support the statement that CB self-administration is an effective treatment for the adverse behavioral effects of early maltreatment in either males or females. Certainly this review should draw the attention of clinicians working with children, adolescents and adults exposed to early trauma and provide some perspective on the dysregulation of the endocannabinoid system in the response to trauma, the complex actions of exogenous CBs based on stress history and the unique effects of these factors in men and women.
Collapse
|
37
|
Scheyer AF, Borsoi M, Wager-Miller J, Pelissier-Alicot AL, Murphy MN, Mackie K, Manzoni OJJ. Cannabinoid Exposure via Lactation in Rats Disrupts Perinatal Programming of the Gamma-Aminobutyric Acid Trajectory and Select Early-Life Behaviors. Biol Psychiatry 2020; 87:666-677. [PMID: 31653479 PMCID: PMC7056509 DOI: 10.1016/j.biopsych.2019.08.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/26/2019] [Accepted: 08/26/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Cannabis usage is increasing with its widespread legalization. Cannabis use by mothers during lactation transfers active cannabinoids to the developing offspring during this critical period and alters postnatal neurodevelopment. A key neurodevelopmental landmark is the excitatory to inhibitory gamma-aminobutyric acid (GABA) switch caused by reciprocal changes in expression ratios of the K+/Cl- transporters potassium-chloride cotransporter 2 (KCC2) and sodium-potassium-chloride transporter (NKCC1). METHODS Rat dams were treated with Δ9-tetrahydrocannabinol or a synthetic cannabinoid during the first 10 days of postnatal development, and experiments were then conducted in the offspring exposed to these drugs via lactation. The network influence of GABA transmission was analyzed using cell-attached recordings. KCC2 and NKCC1 levels were determined using Western blot and quantitative polymerase chain reaction analyses. Ultrasonic vocalization and homing behavioral experiments were carried out at relevant time points. RESULTS Treating rat dams with cannabinoids during early lactation retards transcriptional upregulation and expression of KCC2, thereby delaying the GABA switch in pups of both sexes. This perturbed trajectory was corrected by the NKCC1 antagonist bumetanide and accompanied by alterations in ultrasonic vocalization without changes in homing behavior. Neurobehavioral deficits were prevented by CB1 receptor antagonism during maternal exposure, showing that the CB1 receptor underlies the cannabinoid-induced alterations. CONCLUSIONS These results reveal how perinatal cannabinoid exposure retards an early milestone of development, delaying the trajectory of GABA's polarity transition and altering early-life communication.
Collapse
Affiliation(s)
- Andrew F Scheyer
- Institut de neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale U1249, Marseille, France; Aix-Marseille University, Marseille, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, Institut National de la Santé et de la Recherche Médicale-Aix-Marseille University/Indiana University
| | - Milene Borsoi
- Institut de neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale U1249, Marseille, France; Aix-Marseille University, Marseille, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, Institut National de la Santé et de la Recherche Médicale-Aix-Marseille University/Indiana University
| | - Jim Wager-Miller
- Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, Institut National de la Santé et de la Recherche Médicale-Aix-Marseille University/Indiana University; Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana; Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | - Anne-Laure Pelissier-Alicot
- Institut de neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale U1249, Marseille, France; Aix-Marseille University, Marseille, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, Institut National de la Santé et de la Recherche Médicale-Aix-Marseille University/Indiana University; Service de Psychiatrie, CHU Conception, Assistance Publique - Hôpitaux de Marseille, Marseille, France; Service de Médecine Légale, CHU Timone-Adultes, Assistance Publique - Hôpitaux de Marseille, Marseille, France
| | - Michelle N Murphy
- Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, Institut National de la Santé et de la Recherche Médicale-Aix-Marseille University/Indiana University; Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana; Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | - Ken Mackie
- Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, Institut National de la Santé et de la Recherche Médicale-Aix-Marseille University/Indiana University; Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana; Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana.
| | - Olivier J J Manzoni
- Institut de neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale U1249, Marseille, France; Aix-Marseille University, Marseille, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, Institut National de la Santé et de la Recherche Médicale-Aix-Marseille University/Indiana University.
| |
Collapse
|
38
|
Carr RL, Alugubelly N, de Leon K, Loyant L, Mohammed AN, Patterson ME, Ross MK, Rowbotham NE. Inhibition of fatty acid amide hydrolase by chlorpyrifos in juvenile rats results in altered exploratory and social behavior as adolescents. Neurotoxicology 2020; 77:127-136. [PMID: 31931040 DOI: 10.1016/j.neuro.2020.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/31/2019] [Accepted: 01/07/2020] [Indexed: 12/26/2022]
Abstract
The organophosphorus insecticide chlorpyrifos (CPF) is suspected to cause developmental neurotoxicity in children leading to long term effects. Developmental exposure of rat pups to CPF at low levels disrupts degradation of the brain endocannabinoids through the inhibition of fatty acid amide hydrolase (FAAH) and decreases the reactivity of juvenile rats in an emergence test. In this study, we further investigated the effects of developmental CPF exposure on behavior but also included exposure to PF-04457845, a specific inhibitor of FAAH, for comparison of behavior altered by FAAH inhibition with behavior altered by CPF. Ten day old rat pups were exposed orally either to 0.5, 0.75, or 1.0 mg/kg CPF or 0.02 mg/kg PF-04457845 daily for 7 days. In an open field (day 23), the high CPF and PF-04457845 groups exhibited increased motor activity but no differences in the time spent in the field's center. In an elevated plus maze (day 29), all treatment groups had increased open arm activity but ethological behaviors associated with anxiety were not altered. Behaviors in the maze associated with increased general activity and exploratory drive were increased. Social interactions (day 36) were measured and all treatment groups exhibited increased levels of play behavior. The similarities in behavior between PF-04457845 and CPF suggest that enhanced endocannabinoid signaling during the exposure period plays a role in the persistent alteration of behavior observed following developmental CPF exposure.
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, USA.
| | - Navatha Alugubelly
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Kathryne de Leon
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Louise Loyant
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Afzaal N Mohammed
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - M Elizabeth Patterson
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Matthew K Ross
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Nicole E Rowbotham
- Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| |
Collapse
|
39
|
Franks AL, Berry KJ, DeFranco DB. Prenatal drug exposure and neurodevelopmental programming of glucocorticoid signalling. J Neuroendocrinol 2020; 32:e12786. [PMID: 31469457 PMCID: PMC6982551 DOI: 10.1111/jne.12786] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/25/2019] [Accepted: 08/27/2019] [Indexed: 12/21/2022]
Abstract
Prenatal neurodevelopment is dependent on precise functioning of multiple signalling pathways in the brain, including those mobilised by glucocorticoids (GC) and endocannabinoids (eCBs). Prenatal exposure to drugs of abuse, including opioids, alcohol, cocaine and cannabis, has been shown to not only impact GC signalling, but also alter functioning of the hypothalamic-pituitary-adrenal (HPA) axis. Such exposures can have long-lasting neurobehavioural consequences, including alterations in the stress response in the offspring. Furthermore, cannabis contains cannabinoids that signal via the eCB pathway, which is linked to some components of GC signalling in the adult brain. Given that GCs are frequently used in pregnancy to prevent complications of prematurity, and also that rates of cannabis use in pregnancy are increasing, the likelihood of foetal co-exposure to these compounds is high and may have additional implications for long-term neurodevelopment. Here, we present a discussion of GC signalling and the HPA axis, as well as the effects of prenatal drug exposure on these pathways and the stress response, and we explore the interactions between GC and EC signalling in the developing brain and potential for neurodevelopmental consequences.
Collapse
Affiliation(s)
- Alexis L Franks
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kimberly J Berry
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Donald B DeFranco
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology and Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
40
|
Hemingway SJA, Davies JK, Jirikowic T, Olson EM. What proportion of the brain structural and functional abnormalities observed among children with fetal alcohol spectrum disorder is explained by their prenatal alcohol exposure and their other prenatal and postnatal risks? ADVANCES IN PEDIATRIC RESEARCH 2020; 7:41. [PMID: 33335991 PMCID: PMC7744001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Individuals with prenatal alcohol exposure (PAE) often present with a myriad of other prenatal (e.g. exposure to tobacco and other illicit drugs, poor prenatal care) and postnatal risk factors (e.g. multiple home placements, physical/sexual abuse, low socio-economic status)-all of which are likely contributing to their adverse outcomes. METHODS A comprehensive neuropsychological battery, coupled with magnetic resonance imaging, was administered to children with fetal alcohol spectrum disorders (FASD) in 2009. Study participants diagnosed with FASD by the University of Washington using the FASD 4-Digit Code were compared to typically-developing peers with no PAE. Data from this MRI study were used to explore the proportion of variance in brain structural and functional abnormalities explained by PAE and 14 other prenatal and postnatal risk factors. RESULTS PAE was the dominant risk factor explaining the largest proportion of variance in regional brain size (total brain, frontal lobe, caudate, hippocampus and corpus callosum) and brain function (intellect, achievement, memory, language, executive-function, motor, adaptation, behavior-attention and mental health symptoms). Other prenatal and postnatal risk factors were 3 to 7-fold more prevalent than in the general population. Individually, each risk factor explained a statistically significant, but smaller proportion of variance in brain outcome compared to PAE. In combination, the proportion of variance explained by the presence of multiple prenatal and postnatal risks rivaled that of PAE. CONCLUSION A better understanding of the impact other prenatal and postnatal risk factors have on the neurodevelopmental outcomes of individuals with FASD can inform more effective prevention and intervention strategies.
Collapse
|
41
|
Alshaarawy O, Anthony JC. Cannabis use among women of reproductive age in the United States: 2002-2017. Addict Behav 2019; 99:106082. [PMID: 31421581 PMCID: PMC6791768 DOI: 10.1016/j.addbeh.2019.106082] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND In this study of cannabis use in large nationally representative samples of United States (US) women aged 12-44 years, we evaluate variation by pregnancy month and by trimester. We also evaluate cannabis dependence, which might explain why some women continue using cannabis during pregnancy. METHODS Large nationally representative samples drawn for the US National Surveys on Drug Use and Health included 12-44-year-old women asked about pregnancy month, cannabis use, and cannabis dependence (n = 381,199). For this research, we produced month-specific estimates across four-time intervals (2002-2005, 2006-2009, 2010-2013, 2014-2017). RESULTS Overall from 2002 to 2017, estimates for non-pregnant women and for pregnant women in Trimester 1 indicate 7%-8% had used cannabis at least once in the 30 days prior to assessment. For pregnancy Month 1, the corresponding estimate is 11%, double Month 3 estimate of 5%. This degree of month-to-month variation is not seen for pregnant women in Trimesters 2 and 3, for whom estimates are 3% and 2%, respectively. Among women using cannabis during pregnancy, an estimated 19% have cannabis dependence, versus an expected value of 13% among non-pregnant women (p < .05). CONCLUSION Evidence of a possibly ameliorative pregnancy-associated reduction of cannabis use prevalence was seen by Month 3 during pregnancy. Cannabis dependence may help account for cannabis use early during pregnancy. Identification and outreach to reproductive age women with cannabis dependence might decrease prenatal cannabis exposure.
Collapse
Affiliation(s)
- Omayma Alshaarawy
- Department of Family Medicine, Michigan State University, East Lansing, MI, USA.
| | - James C Anthony
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
42
|
Philippot G, Forsberg E, Tahan C, Viberg H, Fredriksson R. A Single δ 9-Tetrahydrocannabinol (THC) Dose During Brain Development Affects Markers of Neurotrophy, Oxidative Stress, and Apoptosis. Front Pharmacol 2019; 10:1156. [PMID: 31636565 PMCID: PMC6787269 DOI: 10.3389/fphar.2019.01156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/09/2019] [Indexed: 11/13/2022] Open
Abstract
δ9-tetrahydrocannabinol (THC) is one of the most used drugs during pregnancy and lactation and efficiently crosses the placental and blood–brain barriers. Despite the recent legalization initiatives worldwide, the adverse outcome pathway (AOP) of THC following exposure during brain development is incompletely understood. We have previously reported that a single injection of THC on postnatal day (PND) 10 altered adult spontaneous behavior and habituation rates in adult mice. Similar behavioral alterations have been reported following PND 10 exposure to the commonly used over-the-counter analgesic acetaminophen (AAP; also known as paracetamol); as both THC and AAP interact with the endocannabinoid system, we hypothesize that this system might be involved in the AOP of both these pharmaceuticals/drugs. Here, we report that a single THC dose on PND 10 decreased transcript levels of tropomyosin receptor kinase b (Trkb) 24 h after exposure in both the frontal and parietal cortex, and in the hippocampus in mice. An increase in the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1) ratio were also found in both the parietal cortex and hippocampus following neonatal exposure to THC. In addition, THC exposure increased transcript levels of cannabinoid receptor type 1 (Cb1r) in the parietal cortex and increased the apoptosis regulator BAX in the frontal cortex. This study is important for mainly 3 reasons: 1) we are starting to get information on the developmental neurotoxic AOP of PND 10 exposure to THC, where we suggest that transcriptional changes of the neurotrophic receptor Trkb are central, 2) our PND 10 exposure model provides information relevant to human exposure and 3) since PND 10 exposure to AAP also decreased Trkb transcript levels, we suggest THC and AAP may share key events in their respective AOP through endocannabinoid-mediated alterations of the brain-derived neurotrophic factor (BDNF)-TRKB signaling pathway.
Collapse
Affiliation(s)
- Gaëtan Philippot
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Erica Forsberg
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| | - Caroline Tahan
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| | - Henrik Viberg
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
43
|
Scheyer AF, Melis M, Trezza V, Manzoni OJJ. Consequences of Perinatal Cannabis Exposure. Trends Neurosci 2019; 42:871-884. [PMID: 31604585 DOI: 10.1016/j.tins.2019.08.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/10/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022]
Abstract
Cannabis exposure during the perinatal period results in varied and significant consequences in affected offspring. The prevalence of detrimental outcomes of perinatal cannabis exposure is likely to increase in tandem with the broadening of legalization and acceptance of the drug. As such, it is crucial to highlight the immediate and protracted consequences of cannabis exposure on pre- and postnatal development. Here, we identify lasting changes in neurons' learning flexibility (synaptic plasticity) and epigenetic misregulation in animal models of perinatal cannabinoid exposure (using synthetic cannabinoids or active components of the cannabis plant), in addition to significant alterations in social behavior and executive functions. These findings are supported by epidemiological data indicating similar behavioral outcomes throughout life in human offspring exposed to cannabis during pregnancy. Further, we indicate important lingering questions regarding accurate modeling of perinatal cannabis exposure as well as the need for sex- and age-dependent outcome measures in future studies.
Collapse
Affiliation(s)
- Andrew F Scheyer
- INMED, INSERM U1249, Marseille, France; Aix-Marseille University, Provence, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Aix-Marseille University, Provence, France/Indiana University, Bloomington, IN, USA
| | - Miriam Melis
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy; National Institute of Neuroscience, Cagliari, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University 'Roma Tre', Rome, Italy
| | - Olivier J J Manzoni
- INMED, INSERM U1249, Marseille, France; Aix-Marseille University, Provence, France; Cannalab, Cannabinoids Neuroscience Research International Associated Laboratory, INSERM-Aix-Marseille University, Provence, France/Indiana University, Bloomington, IN, USA.
| |
Collapse
|
44
|
Ehrenkranz J, Levine MA. Bones and Joints: The Effects of Cannabinoids on the Skeleton. J Clin Endocrinol Metab 2019; 104:4683-4694. [PMID: 31393556 DOI: 10.1210/jc.2019-00665] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/24/2019] [Indexed: 01/12/2023]
Abstract
CONTEXT The endocannabinoid system uses tissue-specific lipid ligands and G protein‒coupled transmembrane receptors to regulate neurologic, metabolic, and immune responses. Recent studies demonstrate that the endocannabinoid system influences bone metabolism. With the increasing use of endocannabinoid mimetics (e.g., tetrahydrocannabinol and cannabidiol), the involvement of endocannabinoids in bone growth and remodeling has become clinically relevant. EVIDENCE ACQUISITION This literature review is based on a search of PubMed and Google Scholar databases as of June 2019 for all English-language publications relating to cannabinoids and bone. We evaluated retrieved articles for relevance, experimental design, data acquisition, statistical analysis, and conclusions. EVIDENCE SYNTHESIS Preclinical studies establish a role for endocannabinoids in bone metabolism. These studies yield complex and often contradictory results attributed to differences in the specific experimental model examined. Studies using human cells or subjects are limited. CONCLUSIONS In vitro and animal models document that endocannabinoids are involved in bone biology. The relevance of these observations to humans is not clear. The increasing long-term use of medical and recreational cannabis underscores the need to better understand the role of endocannabinoids in human bone metabolism. Moreover, it is important to evaluate the role of endocannabinoids as a therapeutic target to prevent and treat disorders associated with bone loss.
Collapse
Affiliation(s)
- Joel Ehrenkranz
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Michael A Levine
- Center for Bone Health and Division of Pediatric Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
45
|
Basavarajappa BS, Joshi V, Shivakumar M, Subbanna S. Distinct functions of endogenous cannabinoid system in alcohol abuse disorders. Br J Pharmacol 2019; 176:3085-3109. [PMID: 31265740 DOI: 10.1111/bph.14780] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022] Open
Abstract
Δ9 -tetrahydrocannabinol, the principal active component in Cannabis sativa extracts such as marijuana, participates in cell signalling by binding to cannabinoid CB1 and CB2 receptors on the cell surface. The CB1 receptors are present in both inhibitory and excitatory presynaptic terminals and the CB2 receptors are found in neuronal subpopulations in addition to microglial cells and astrocytes and are present in both presynaptic and postsynaptic terminals. Subsequent to the discovery of the endocannabinoid (eCB) system, studies have suggested that alcohol alters the eCB system and that this system plays a major role in the motivation to abuse alcohol. Preclinical studies have provided evidence that chronic alcohol consumption modulates eCBs and expression of CB1 receptors in brain addiction circuits. In addition, studies have further established the distinct function of the eCB system in the development of fetal alcohol spectrum disorders. This review provides a recent and comprehensive assessment of the literature related to the function of the eCB system in alcohol abuse disorders.
Collapse
Affiliation(s)
- Balapal S Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,New York State Psychiatric Institute, New York, NY, USA.,Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Vikram Joshi
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Madhu Shivakumar
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Shivakumar Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| |
Collapse
|
46
|
Marijuana Use in Pregnancy: An Updated Look at Marijuana Use and Its Impact on Pregnancy. Clin Obstet Gynecol 2019; 62:185-190. [PMID: 30531376 DOI: 10.1097/grf.0000000000000415] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This article aims to provide an updated look at the use of marijuana in the United States and its impact on pregnancy. First, the prevalence of marijuana use is examined, including use both in and outside of pregnancy. The literature surrounding attitudes and beliefs with regard to use in pregnancy is reviewed. The impact on pregnancy outcomes is reviewed along with the evidence of marijuana's impact on neural development of the fetus. Finally, clinical considerations for providers are discussed.
Collapse
|
47
|
Bond S. Updates from the Literature, March/April 2019. J Midwifery Womens Health 2019; 64:236-240. [DOI: 10.1111/jmwh.12963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 11/27/2022]
|
48
|
Csaba G. Hormonal Imprinting: The First Cellular-level Evidence of Epigenetic Inheritance and its Present State. Curr Genomics 2019; 20:409-418. [PMID: 32476998 PMCID: PMC7235388 DOI: 10.2174/1389202920666191116113524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 09/24/2019] [Accepted: 10/21/2019] [Indexed: 12/28/2022] Open
Abstract
Hormonal imprinting takes place perinatally at the first encounter between the developing hormone receptor and its target hormone. This process is needed for the normal function of the receptor-hormone pair and its effect is life-long. However, in this critical period, when the developmental window is open, related molecules (members of the same hormone family, synthetic hormones and hormone-like molecules, endocrine disruptors) also can be bound by the receptor, causing life-long faulty imprinting. In this case, the receptors’ binding capacity changes and alterations are caused at adult age in the sexual and behavioral sphere, in the brain and bones, inclination to diseases and manifestation of diseases, etc. Hereby, faulty hormonal imprinting is the basis of metabolic and immunological imprinting as well as the developmental origin of health and disease (DOHaD). Although the perinatal period is the most critical for faulty imprinting, there are other critical periods as weaning and adolescence, when the original imprinting can be modified or new imprintings develop. Hormonal imprinting is an epigenetic process, without changing the base sequence of DNA, it is inherited in the cell line of the imprinted cells and also transgenerationally (up to 1000 generations in unicellulars and up to the 3rd generation in mammals are justified). Considering the enormously growing number and amount of faulty imprinters (endocrine disruptors) and the hereditary character of faulty imprinting, this latter is threatening the whole human endocrine system.
Collapse
Affiliation(s)
- György Csaba
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
49
|
Bukiya AN. Physiology of the Endocannabinoid System During Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1162:13-37. [PMID: 31332732 DOI: 10.1007/978-3-030-21737-2_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The endocannabinoid (eCB) system comprises endogenously produced cannabinoids (CBs), enzymes of their production and degradation, and CB-sensing receptors and transporters. The eCB system plays a critical role in virtually all stages of animal development. Studies on eCB system components and their physiological role have gained increasing attention with the rising legalization and medical use of marijuana products. The latter represent exogenous interventions that target the eCB system. This chapter summarizes knowledge in the field of CB contribution to gametogenesis, fertilization, embryo implantation, fetal development, birth, and adolescence-equivalent periods of ontogenesis. The material is complemented by the overview of data from our laboratory documenting the functional presence of the eCB system within cerebral arteries of baboons at different stages of development.
Collapse
Affiliation(s)
- Anna N Bukiya
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
50
|
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
|