Cannabinoid exposure and altered DNA methylation in rat and human sperm

Susan K Murphy, Nilda Itchon-Ramos, Zachary Visco, Zhiqing Huang, Carole Grenier, Rose Schrott, Kelly Acharya, Marie-Helene Boudreau, Thomas M Price, Douglas J Raburn, David L Corcoran, Joseph E Lucas, John T Mitchell, F Joseph McClernon, Marty Cauley, Brandon J Hall, Edward D Levin, Scott H Kollins, Susan K Murphy, Nilda Itchon-Ramos, Zachary Visco, Zhiqing Huang, Carole Grenier, Rose Schrott, Kelly Acharya, Marie-Helene Boudreau, Thomas M Price, Douglas J Raburn, David L Corcoran, Joseph E Lucas, John T Mitchell, F Joseph McClernon, Marty Cauley, Brandon J Hall, Edward D Levin, Scott H Kollins

Abstract

Little is known about the reproductive effects of paternal cannabis exposure. We evaluated associations between cannabis or tetrahydrocannabinol (THC) exposure and altered DNA methylation in sperm from humans and rats, respectively. DNA methylation, measured by reduced representation bisulfite sequencing, differed in the sperm of human users from non-users by at least 10% at 3,979 CpG sites. Pathway analyses indicated Hippo Signaling and Pathways in Cancer as enriched with altered genes (Bonferroni p < 0.02). These same two pathways were also enriched with genes having altered methylation in sperm from THC-exposed versus vehicle-exposed rats (p < 0.01). Data validity is supported by significant correlations between THC exposure levels in humans and methylation for 177 genes, and substantial overlap in THC target genes in rat sperm (this study) and genes previously reported as having altered methylation in the brain of rat offspring born to parents both exposed to THC during adolescence. In humans, cannabis use was also associated with significantly lower sperm concentration. Findings point to possible pre-conception paternal reproductive risks associated with cannabis use.

Keywords: Cannabis; DNA methylation; epigenetic reprogramming; human; rat; sperm; tetrahydrocannabinol.

Figures

Figure 1.
Figure 1.
Study procedures for screening and consent. Twenty-four men, including 12 cannabis users and 12 non-users, were enrolled and participated in the study.
Figure 2.
Figure 2.
Cannabis use is associated with DNA methylation in sperm. (a) Heat map showing DNA methylation relative to the median for 708 CpG sites (rows) associated with 46 genes with differential methylation between cannabis users (left columns) and non-user controls (right columns). Each column represents one participant, while each row represents one CpG site. Row clustering was unsupervised. Methylation levels are median-centered. (b) Top, bisulfite pyrosequencing data compared to RRBS data from users to non-users for a CpG within the intragenic repeated sequence of AHRR by linear regression. Bottom, bisulfite pyrosequencing data for one of the few genes, PRDM16, showing increased methylation in the user group. Each data point is the average of replicate measures. The mean is shown by the center dashed bar with error bars representing standard deviation. Unpaired t test, one-tailed, F test to compare variances (p = 0.44). D’Agostino & Pearson normality test showed data was normally distributed in each group. (c) Top graph: bisulfite pyrosequencing data for COL18A1 showing all eight CpG sites analyzed together discriminate users from non-users. Each data point is the average of replicate measures. The mean is shown by the center dashed bar with error bars representing standard deviation. Unpaired t test, one-tailed, F test to compare variances (p = 0.27). D’Agostino & Pearson normality test showed data was normally distributed in each group. Bottom five graphs: Comparison of methylation values from RRBS versus bisulfite pyrosequencing for the five CpG sites identified as differentially methylated by RRBS, analyzed using linear regression. (d) Top graph: bisulfite pyrosequencing data for PTPRN2 showing all seven CpG sites analyzed together discriminate users from non-users. Each data point is the average of replicate measures. The mean is shown by the center bar with error bars representing standard deviation. Unpaired t test with Welch’s correction, one-tailed, F test to compare variances (p = 0.03). D’Agostino & Pearson normality test showed data was normally distributed in each group. Bottom five graphs: Comparison of methylation values from RRBS versus bisulfite pyrosequencing for the five CpG sites identified as differentially methylated by RRBS, analyzed by linear regression. (e) Correlations between urinary THC concentration in the human cannabis user group and sperm DNA methylation levels for CpGs identified as differentially methylated in PTGIR (top) and CSNK1E (bottom), analyzed by linear regression. Panels B-D: Users, open circles; non-users, closed circles.

References

    1. Gunn JK, Rosales CB, Center KE, et al. Prenatal exposure to cannabis and maternal and child health outcomes: a systematic review and meta-analysis. BMJ Open. 2016;6:e009986.
    1. Funkhouser M. Governing the states and localities: state Marijuana laws in 2017 map. Governing News. Available from: Dr. Mark Funkhouser, 2017.
    1. Center for Behavioral Health Statistics and Quality (CBHSQ) SAaMHSAS U.S. Department of Health and Human Services (HHS), and RTI international. results from the 2015 national survey on drug use and health: detailed tables In: Types of illicit drug use in lifetime PY, and past month among persons aged 18 to 25: percentages, 2014 and 2015. 2017.
    1. Volkow ND, Swanson JM, Evins AE, et al. Effects of cannabis use on human behavior, including cognition, motivation, and psychosis: a review. JAMA Psychiatry. 2016;73:292–297.
    1. Szutorisz H, Hurd YL.. High times for cannabis: epigenetic imprint and its legacy on brain and behavior. Neurosci Biobehav Rev. 2018;85:93–101.
    1. Pitsilis G, Spyridakos D, Nomikos GG, et al. Adolescent female cannabinoid exposure diminishes the reward-facilitating effects of Delta9-tetrahydrocannabinol and d-amphetamine in the adult male offspring. Front Pharmacol. 2017;8:225.
    1. Byrnes JJ, Johnson NL, Schenk ME, et al. Cannabinoid exposure in adolescent female rats induces transgenerational effects on morphine conditioned place preference in male offspring. J Psychopharmacol. 2012;26:1348–1354.
    1. Soubry A, Guo L, Huang Z, et al. Obesity-related DNA methylation at imprinted genes in human sperm: results from the TIEGER study. Clin Epigenetics. 2016;8:51.
    1. Ariel M, Cedar H, McCarrey J.. Developmental changes in methylation of spermatogenesis-specific genes include reprogramming in the epididymis. Nat Genet. 1994;7:59–63.
    1. Donkin I, Versteyhe S, Ingerslev LR, et al. Obesity and bariatric surgery drive epigenetic variation of spermatozoa in humans. Cell Metab. 2016;23:369–378.
    1. Meissner A, Gnirke A, Bell GW, et al. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res. 2005;33:5868–5877.
    1. Lu AT, Hannon E, Levine ME, et al. Genetic architecture of epigenetic and neuronal ageing rates in human brain regions. Nat Commun. 2017;8:15353.
    1. Huen K, Solomon O, Kogut K, et al. PON1 DNA methylation and neurobehavior in Mexican-American children with prenatal organophosphate exposure. Environ Int. 2018;121:31–40.
    1. Non AL, Binder AM, Kubzansky LD, et al. Genome-wide DNA methylation in neonates exposed to maternal depression, anxiety, or SSRI medication during pregnancy. Epigenetics. 2014;9:964–972.
    1. Joubert BR, Haberg SE, Nilsen RM, et al. 450K epigenome-wide scan identifies differential DNA methylation in newborns related to maternal smoking during pregnancy. Environ Health Perspect. 2012;120:1425–1431.
    1. Laqqan M, Tierling S, Alkhaled Y, et al. Spermatozoa from males with reduced fecundity exhibit differential DNA methylation patterns. Andrology. 2017;5:971–978.
    1. Yang WS, Stockwell BR. Inhibition of casein kinase 1-epsilon induces cancer-cell-selective, PERIOD2-dependent growth arrest. Genome Biol. 2008;9:R92.
    1. Bryant CD, Parker CC, Zhou L, et al. Csnk1e is a genetic regulator of sensitivity to psychostimulants and opioids. Neuropsychopharmacology. 2012;37:1026–1035.
    1. Kent WJ, Sugnet CW, Furey TS, et al. The human genome browser at UCSC. Genome Res. 2002;12:996–1006.
    1. Watson CT, Szutorisz H, Garg P, et al. Genome-wide DNA methylation profiling reveals epigenetic changes in the rat nucleus accumbens associated with cross-generational effects of adolescent THC exposure. Neuropsychopharmacology. 2015;40:2993–3005.
    1. Tang WW, Dietmann S, Irie N, et al. A unique gene regulatory network resets the human germline epigenome for development. Cell. 2015;161:1453–1467.
    1. Gundersen TD, Jorgensen N, Andersson AM, et al. Association between use of Marijuana and male reproductive hormones and semen quality: A study among 1,215 healthy young men. Am J Epidemiol. 2015;182:473–481.
    1. Hembree WC 3rd, Nahas GG, Zeidenberg P, et al. Changes in human spermatozoa associated with high dose marihuana smoking. Adv Biosci. 1978;22–23:429–439.
    1. Pacey AA, Povey AC, Clyma JA, et al. Participating centres of chaps UK. Modifiable and non-modifiable risk factors for poor sperm morphology. Hum Reprod. 2014;29:1629–1636.
    1. Johnston L, O’Malley P, Miech R, et al. Monitoring the future national results on drug use: 1975–2013: Overview, key findings on adolescent drug use.: Ann Arbor: Institute for Social Research. University of Michigan; 2014.
    1. ElSohly MA, Mehmedic Z, Foster S, et al. Changes in Cannabis potency over the last 2 decades (1995–2014): analysis of current data in the United States. Biol Psychiatry. 2016;79:613–619.
    1. Hackett JA, Sengupta R, Zylicz JJ, et al. Germline DNA demethylation dynamics and imprint erasure through 5-hydroxymethylcytosine. Science. 2013;339:448–452.
    1. Abbasi J. The paternal epigenome makes its mark. JAMA. 2017;317:2049–2051.
    1. Day J, Savani S, Krempley BD, et al. Influence of paternal preconception exposures on their offspring: through epigenetics to phenotype. Am J Stem Cells. 2016;5:11–18.
    1. Soubry A, Murphy SK, Wang F, et al. Newborns of obese parents have altered DNA methylation patterns at imprinted genes. Int J Obes (Lond). 2015;39:650–657.
    1. Shnorhavorian M, Schwartz SM, Stansfeld B, et al. Differential DNA methylation regions in adult human sperm following adolescent chemotherapy: potential for epigenetic inheritance. PLoS One. 2017;12:e0170085.
    1. Laqqan M, Tierling S, Alkhaled Y, et al. Aberrant DNA methylation patterns of human spermatozoa in current smoker males. Reprod Toxicol. 2017;71:126–133.
    1. Alkhaled Y, Laqqan M, Tierling S, et al. Impact of cigarette-smoking on sperm DNA methylation and its effect on sperm parameters. Andrologia. 2018;50:e12950.
    1. Jenkins TG, James ER, Alonso DF, et al. Cigarette smoking significantly alters sperm DNA methylation patterns. Andrology. 2017;5:1089–1099.
    1. Adams IB, Martin BR. Cannabis: pharmacology and toxicology in animals and humans. Addiction. 1996;91:1585–1614.
    1. Grotenhermen F, Russo E, eds. Cannabis and Cannabinoids: pharmacology, toxicology and therapeutic potential. New York: The Hawthorne Press, Inc; 2002.
    1. Scallet AC. Neurotoxicology of cannabis and THC: a review of chronic exposure studies in animals. Pharmacol Biochem Behav. 1991;40:671–676.
    1. Marques CJ, Joao Pinho M, Carvalho F, et al. DNA methylation imprinting marks and DNA methyltransferase expression in human spermatogenic cell stages. Epigenetics. 2011;6:1354–1361.
    1. Sheehan DV, Lecrubier Y, Harnett-Sheehan K, et al. Reliability and validity of the M.I.N.I. international neuropsychiatric interview (M.I.N.I.): according to the SCID-P. Eur Psychiatry. 1997;12:232–241.
    1. Lecrubier Y, Sheehan D, Weiller E, et al. The M.I.N.I. International Neuropsychiatric Interview (M.I.N.I.) A short diagnostic structured interview: reliability and validity according to the CIDI. Eur Psychiatry. 1997;12:224–231.
    1. Cooper TG, Noonan E, von Eckardstein S, et al. World Health Organization reference values for human semen characteristics. Hum Reprod Update. 2010;16:231–245.
    1. Irimia C, Polis IY, Stouffer D, et al. Persistent effects of chronic Delta9-THC exposure on motor impulsivity in rats. Psychopharmacology (Berl). 2015;232:3033–3043.
    1. Harte LC, Dow-Edwards D. Sexually dimorphic alterations in locomotion and reversal learning after adolescent tetrahydrocannabinol exposure in the rat. Neurotoxicol Teratol. 2010;32:515–524.
    1. Rubino T, Realini N, Braida D, et al. Changes in hippocampal morphology and neuroplasticity induced by adolescent THC treatment are associated with cognitive impairment in adulthood. Hippocampus. 2009;19:763–772.
    1. Krueger F, Andrews SR. Bismark: a flexible aligner and methylation caller for bisulfite-seq applications. Bioinformatics. 2011;27:1571–1572.
    1. Murphy SK, Huang Z, Hoyo C. Differentially methylated regions of imprinted genes in prenatal, perinatal and postnatal human tissues. PLoS One. 2012;7:e40924.
    1. Bassil CF, Huang Z, Murphy SK. Bisulfite pyrosequencing. Methods Mol Biol. 2013;1049:95–107.
    1. Glusman G, Caballero J, Mauldin DE, et al. Kaviar: an accessible system for testing SNV novelty. Bioinformatics. 2011;27:3216–3217.
    1. Huang Da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4:44–57.

Source: PubMed

3
Abonnere