Persistent inhibitory circuit defects and disrupted social behaviour following in utero exogenous cannabinoid exposure

G A Vargish, K A Pelkey, X Yuan, R Chittajallu, D Collins, C Fang, C J McBain, G A Vargish, K A Pelkey, X Yuan, R Chittajallu, D Collins, C Fang, C J McBain

Abstract

Placental transfer of Δ9-tetrahydrocannabinol (THC) during pregnancy has the potential to interfere with endogenous cannabinoid (CB) regulation of fetal nervous system development in utero. Here we examined the effect of maternal CB intake on mouse hippocampal interneurons largely focusing on cholecystokinin-expressing interneurons (CCK-INTs), a prominent CB subtype-1 receptor (CB1R) expressing neuronal population throughout development. Maternal treatment with THC or the synthetic CB1R agonist WIN55,212-2 (WIN) produced a significant loss of CCK-INTs in the offspring. Further, residual CCK-INTs in animals prenatally treated with WIN displayed decreased dendritic complexity. Consistent with these anatomical deficits, pups born to CB-treated dams exhibited compromised CCK-INT-mediated feedforward and feedback inhibition. Moreover, pups exposed to WIN in utero lacked constitutive CB1R-mediated suppression of inhibition from residual CCK-INTs and displayed altered social behavior. Our findings add to a growing list of potential cell/circuit underpinnings that may underlie cognitive impairments in offspring of mothers that abuse marijuana during pregnancy.

Conflict of interest statement

The authors have no conflicts of interest.

Figures

Figure 1. Loss of CCK-INTs following in…
Figure 1. Loss of CCK-INTs following in utero THC or WIN exposure
A) Representative images of CCK, PV, SOM, CR and VIP labeling in hippocampal sections from mice born to VH or WIN treated mothers. B) Summary plot of the density of labeled cells for each marker in paired VH/WIN treated litters. 11 mice from 11 litters/treatment were examined for CCK and PV, 10 mice from 10 litters/treatment for SOM, and 12 mice from 12 litters/treatment for CR and VIP (*p=5×10−6). C) Summary plot of the density of labeled cells for CCK, PV and SOM in paired VH/THC treated litters. Three mice from three litters/treatment were examined for each marker (*p=0.001). D) Representative images (left) and summary plot (right) of GFP+ cell labeling in 5HT3AR-GFP and Nkx2.1-Cre:RCE-GFP mice born to VH or WIN treated mothers. Dashed lines indicate the region of interest for cell counts. For 5HT3AR-GFP 7 mice from seven WIN or VH treated litters were examined (*p=3.9×10−9) and for Nkx2.1-Cre:RCE-GFP mice 3 mice from 3 litters/treatment were analyzed. E) Representative images (left) and group data counts (right) for CCK-INT bouton stains with CB1R after prenatal treatment with VH or WIN. 4 mice from 4 litters/treatment were counted (*p=0.02). F) Drawings of reconstructed CCK-BCs (left) and SCAs (right) recorded VH (top) and WIN (bottom) treated animals. Dendrites are black for both treatment groups while axons are colored red and blue for VH and WIN treated animals, respectively. Dashed lines indicate the borders of SP. G) Quantification of terminal density along the axon of CCK-BCs and SCAs in VH and WIN treated animals (CCK-BCs: VH: n=5, WIN=4; SCAs: n=4/treatment). H) Quantification of total dendrite length for CCK-BS and SCAs in VH and WIN treated animals (*p=0.03). I) Sholl analyses for dendrites of CCK-BCs (left) and SCAs (right) in VH or WIN treated mice (*p=0.03, for both). Values are plotted as mean±SEM.
Figure 2. WIN exposure does not affect…
Figure 2. WIN exposure does not affect PC morphology or physiology
A) Images of L1NCAM (red) and Hoechst (blue) staining in both VH (top) and WIN (bottom) treated animals. Images show normal patterning of projections in both the corpus collosum (left) and fimbria (right). B) Left panel shows sample images of Hoechst stain in CA1 of VH (top) and WIN (bottom) treated animals. Right panel shows summarized data of PC density in CA1 of VH and WIN treated animals (VH: n=4 mice/4 litters, WIN: n=4 mice/4 litters). C) Left panel shows drawings of reconstructed PCs in VH (left) and WIN (right) treated mice. Right panel shows summarized plot of PC dendritic length in VH and WIN treated animals (VH: n=6 cells/3 mice/2 litters, WIN: n=6 cells/3 mice/2 litters). D) Continuous traces (left) and representative waveform (middle) of sEPSCs recorded in CA1 PCs of VH (top) and WIN (bottom) treated mice. Right panel shows summarized data for sEPSC amplitude and frequency in VH and WIN mice (VH: n=9 cells/2 mice/2 litters, WIN: n=9 mice/2 mice/2 litters). E) Left panel shows representative traces of field EPSPs recorded in CA1 stratum radiatum of VH (top) and WIN (bottom) animals following stimulation of the Schaffer collateral pathway at increasing intensity. Left panel shows summarized plot of input/out dynamics in VH and WIN mice depicting field EPSP amplitude at various stimulation intensities (VH: n=9 slices/3 mice/2 litters, WIN: n=9 slices/3 mice/2 litters).
Figure 3. Deficient CCK-INT mediated inhibitory drive…
Figure 3. Deficient CCK-INT mediated inhibitory drive in prenatally WIN treated mice
A) At left are continuous traces of representative CA1 PC recordings showing sIPSCs before and after DSI induction (+70mV, truncated) in VH or WIN treated mice. Middle panel shows averaged sIPSC waveforms for events obtained for 5s before and after DSI induction. Group data plotted at right highlights changes in amplitude, frequency and charge of sIPSCs following DSI induction expressed as percentage of pre-DSI levels (VH: n=8 cells/3 animals/2 litters, WIN: n=11 cells/3 animals/2 litters; *Amplitude p=0.03, *Charge: p=0.03. B) Schematic of feedback inhibition evoked by alvear stimulation (left) and averaged CA1 IPSCS (right) from representative recordings illustrating sensitivity to both GABA (Bicuculline, 10μM) and glutamate (DNQX, 10μM/dl-AP5, 50μM) antagonism confirming their disynaptic nature. C) At left are averaged disynaptically evoked IPSC waveforms from representative recordings before and after acute WIN (1μM) application in VH and WIN treated mice. Group data plots to the right show acute WIN effects on IPSC amplitudes for each recording. D) Summarized data shown as % of control amplitudes obtained prior to acute WIN application (right; VH: n=20 cells/11 animals/5 litters, WIN: n=16 cells/10 animals/6 litters; * p=0.01). E) Representative example traces (left) and group data summary (right) illustrating the effect of acute WIN application on 5 pulse trains of disynaptically evoked IPSCs in VH and WIN treated mice (VH: n=20 cells/11 animals/5 litters, WIN: n=16 cells/10 animals/6 litters; p=0.01, 0.02, 0.10, 0.03, and 0.05 for pulses 1–5 respectively). F) Schematic of feedforward inhibition evoked by Schaffer collateral stimulation (left). At middle are averaged feedforward evoked waveforms showing the inward monosynaptic excitatory current and outward disynaptic inhibitory current. Right panel shows summarized plot of the % of disynaptic inhibition that is sensitive to DSI in both VH and WIN treated mice (VH: n=5 cells/3 animals/2 litters, WIN: n=6 cells/3 animals/3 litters; p=0.03).
Figure 4. Loss of tonic eCB mediated…
Figure 4. Loss of tonic eCB mediated inhibition of CCK-BC release in WIN treated mice
A) Schematic diagram (left), averaged IPSC traces from representative recordings (middle), and group data summary plots (right) illustrating the effects of the CB1R antagonist AM251 (10 μM) on basal CCK-BC to CA1 PC transmission in VH and WIN treated mice (VH: n=10 cells/4 animals/1 litter, WIN: n=9 cells/3 animals/2 litters; * p=0.01). B) Effect of AM251 on trains of monosynaptically evoked CCK-BC to PC IPSCs in VH and WIN treated mice. Shown are averaged IPSC trains from representative recordings (left) and group data summaries for changes in total charge during the train (middle, *p=0.03) or amplitudes of each event during the trains (right, p=0.01, 0.11, 0.06, 0.13, and 0.03 for pulses 1–5 respectively) expressed as %s of control responses obtained before AM251 (VH: n=10 cells/4 animals/1 litters, WIN: n=9 cells/3 animals/2 litters,). C) Schematic diagram of CCK-INT to PC paired recording (left), representative traces of unitary events before and after DSI induction (middle) and group data summary of DSI amplitude changes expressed as % inhibition of pre-DSI levels (right) for connected pairs in VH and WIN treated mice (VH: n=6 pairs/4 animals/2 litters), WIN: n=6 pairs/4 animals/2 litters). D) Single trial traces from representative CCK-INT to PC pairs (left) and group data summary (right) illustrating the degree of asynchronous release from CCK-INTs during train activity in VH and WIN treated mice. Asynchronicity was measured as the charge transfer during a 300ms window centered on the peak of the last AP in the train (boxed region; VH: n=9 pairs/6 animals/2 litters, WIN: n=8 pairs/6 animals/2 litters).
Figure 5. Reduced social interaction in WIN…
Figure 5. Reduced social interaction in WIN treated mice
A) Schematic diagram of three-chamber social interaction test. B) Group data summarizing the number entries mice from each treatment group made into chambers containing the novel mouse or object. C) Group data showing the total time mice from each treatment group spent interacting with the novel mouse and novel object (*p=0.01). D–E) Elevated plus maze group data summarizing the number of entries (D) and total time spent (E) in the open and closed arms of the maze for VH and WIN treated mice. F) Summary of the number of head dips mice from each treatment made during elevated plus maze testing. 11 mice from 3 litters were evaluated per treatment group for each behavioral test.

References

    1. National Survey on Drug Use and Health (U.S.), United States. Substance Abuse and Mental Health Services Administration. Office of Applied Studies. National Survey on Drug Use and Health series. Dept. of Health and Human Services, Substance Abuse and Mental Health Services Administration, Office of Applied Studies; Rockville, MD: 2003. Results from the ... National Survey on Drug Use and Health : national findings; p. v.
    1. Grotenhermen F. Pharmacokinetics and pharmacodynamics of cannabinoids. Clinical pharmacokinetics. 2003;42(4):327–360.
    1. Pijlman FT, Rigter SM, Hoek J, Goldschmidt HM, Niesink RJ. Strong increase in total delta-THC in cannabis preparations sold in Dutch coffee shops. Addict Biol. 2005;10(2):171–180.
    1. Mehmedic Z, Chandra S, Slade D, Denham H, Foster S, Patel AS, et al. Potency trends of Delta9-THC and other cannabinoids in confiscated cannabis preparations from 1993 to 2008. Journal of forensic sciences. 2010;55(5):1209–1217.
    1. Hayatbakhsh MR, Flenady VJ, Gibbons KS, Kingsbury AM, Hurrion E, Mamun AA, et al. Birth outcomes associated with cannabis use before and during pregnancy. Pediatric research. 2012;71(2):215–219.
    1. Goldschmidt L, Day NL, Richardson GA. Effects of prenatal marijuana exposure on child behavior problems at age 10. Neurotoxicol Teratol. 2000;22(3):325–336.
    1. Goldschmidt L, Richardson GA, Willford JA, Severtson SG, Day NL. School achievement in 14-year-old youths prenatally exposed to marijuana. Neurotoxicol Teratol. 2012;34(1):161–167.
    1. Leech SL, Larkby CA, Day R, Day NL. Predictors and correlates of high levels of depression and anxiety symptoms among children at age 10. Journal of the American Academy of Child and Adolescent Psychiatry. 2006;45(2):223–230.
    1. Noland JS, Singer LT, Short EJ, Minnes S, Arendt RE, Kirchner HL, et al. Prenatal drug exposure and selective attention in preschoolers. Neurotoxicol Teratol. 2005;27(3):429–438.
    1. Howlett AC. The cannabinoid receptors. Prostaglandins & other lipid mediators. 2002;68–69:619–631.
    1. Berghuis P, Dobszay MB, Wang X, Spano S, Ledda F, Sousa KM, et al. Endocannabinoids regulate interneuron migration and morphogenesis by transactivating the TrkB receptor. Proc Natl Acad Sci U S A. 2005;102(52):19115–19120.
    1. Berghuis P, Rajnicek AM, Morozov YM, Ross RA, Mulder J, Urban GM, et al. Hardwiring the brain: endocannabinoids shape neuronal connectivity. Science. 2007;316(5828):1212–1216.
    1. Mulder J, Aguado T, Keimpema E, Barabas K, Ballester Rosado CJ, Nguyen L, et al. Endocannabinoid signaling controls pyramidal cell specification and long-range axon patterning. Proc Natl Acad Sci U S A. 2008;105(25):8760–8765.
    1. Diaz-Alonso J, Aguado T, Wu CS, Palazuelos J, Hofmann C, Garcez P, et al. The CB(1) cannabinoid receptor drives corticospinal motor neuron differentiation through the Ctip2/Satb2 transcriptional regulation axis. J Neurosci. 2012;32(47):16651–16665.
    1. Tortoriello G, Morris CV, Alpar A, Fuzik J, Shirran SL, Calvigioni D, et al. Miswiring the brain: Delta9-tetrahydrocannabinol disrupts cortical development by inducing an SCG10/stathmin-2 degradation pathway. The EMBO journal. 2014;33(7):668–685.
    1. Alpar A, Tortoriello G, Calvigioni D, Niphakis MJ, Milenkovic I, Bakker J, et al. Endocannabinoids modulate cortical development by configuring Slit2/Robo1 signalling. Nat Commun. 2014;5:4421.
    1. Wu CS, Zhu J, Wager-Miller J, Wang S, O’Leary D, Monory K, et al. Requirement of cannabinoid CB(1) receptors in cortical pyramidal neurons for appropriate development of corticothalamic and thalamocortical projections. Eur J Neurosci. 2010;32(5):693–706.
    1. de Salas-Quiroga A, Diaz-Alonso J, Garcia-Rincon D, Remmers F, Vega D, Gomez-Canas M, et al. Prenatal exposure to cannabinoids evokes long-lasting functional alterations by targeting CB1 receptors on developing cortical neurons. Proc Natl Acad Sci U S A. 2015
    1. Morozov YM, Freund TF. Post-natal development of type 1 cannabinoid receptor immunoreactivity in the rat hippocampus. Eur J Neurosci. 2003;18(5):1213–1222.
    1. Morozov YM, Torii M, Rakic P. Origin, early commitment, migratory routes, and destination of cannabinoid type 1 receptor-containing interneurons. Cereb Cortex. 2009;19(Suppl 1):i78–89.
    1. Antypa M, Faux C, Eichele G, Parnavelas JG, Andrews WD. Differential gene expression in migratory streams of cortical interneurons. Eur J Neurosci. 2011;34(10):1584–1594.
    1. Saez TM, Aronne MP, Caltana L, Brusco AH. Prenatal exposure to the CB1 and CB2 cannabinoid receptor agonist WIN 55,212-2 alters migration of early-born glutamatergic neurons and GABAergic interneurons in the rat cerebral cortex. Journal of neurochemistry. 2014;129(4):637–648.
    1. Morozov YM, Freund TF. Postnatal development and migration of cholecystokinin-immunoreactive interneurons in rat hippocampus. Neuroscience. 2003;120(4):923–939.
    1. Nagode DA, Tang AH, Yang K, Alger BE. Optogenetic identification of an intrinsic cholinergically driven inhibitory oscillator sensitive to cannabinoids and opioids in hippocampal CA1. J Physiol. 2014;592(Pt 1):103–123.
    1. Sauer JF, Struber M, Bartos M. Impaired fast-spiking interneuron function in a genetic mouse model of depression. eLife. 2015;4
    1. Yang M, Silverman JL, Crawley JN. Automated three-chambered social approach task for mice. In: Crawley Jacqueline N, et al., editors. Current protocols in neuroscience / editorial board. Unit 8. Chapter 8. 2011. p. 26.
    1. Mereu G, Fa M, Ferraro L, Cagiano R, Antonelli T, Tattoli M, et al. Prenatal exposure to a cannabinoid agonist produces memory deficits linked to dysfunction in hippocampal long-term potentiation and glutamate release. Proc Natl Acad Sci U S A. 2003;100(8):4915–4920.
    1. Mato S, Chevaleyre V, Robbe D, Pazos A, Castillo PE, Manzoni OJ. A single in-vivo exposure to delta 9THC blocks endocannabinoid-mediated synaptic plasticity. Nat Neurosci. 2004;7(6):585–586.
    1. Chittajallu R, Craig MT, McFarland A, Yuan X, Gerfen S, Tricoire L, et al. Dual origins of functionally distinct O-LM interneurons revealed by differential 5-HT(3A)R expression. Nat Neurosci. 2013;16(11):1598–1607.
    1. Tricoire L, Pelkey KA, Erkkila BE, Jeffries BW, Yuan X, McBain CJ. A blueprint for the spatiotemporal origins of mouse hippocampal interneuron diversity. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2011;31(30):10948–10970.
    1. Xu Q, Tam M, Anderson SA. Fate mapping Nkx2. 1-lineage cells in the mouse telencephalon. The Journal of comparative neurology. 2008;506(1):16–29.
    1. Lee S, Hjerling-Leffler J, Zagha E, Fishell G, Rudy B. The largest group of superficial neocortical GABAergic interneurons expresses ionotropic serotonin receptors. J Neurosci. 2010;30(50):16796–16808.
    1. Somogyi J, Baude A, Omori Y, Shimizu H, El Mestikawy S, Fukaya M, et al. GABAergic basket cells expressing cholecystokinin contain vesicular glutamate transporter type 3 (VGLUT3) in their synaptic terminals in hippocampus and isocortex of the rat. Eur J Neurosci. 2004;19(3):552–569.
    1. Bezaire MJ, Soltesz I. Quantitative assessment of CA1 local circuits: knowledge base for interneuron-pyramidal cell connectivity. Hippocampus. 2013;23(9):751–785.
    1. Hill EL, Gallopin T, Ferezou I, Cauli B, Rossier J, Schweitzer P, et al. Functional CB1 receptors are broadly expressed in neocortical GABAergic and glutamatergic neurons. J Neurophysiol. 2007;97(4):2580–2589.
    1. Donato F, Rompani SB, Caroni P. Parvalbumin-expressing basket-cell network plasticity induced by experience regulates adult learning. Nature. 2013;504(7479):272–276.
    1. Cope DW, Maccaferri G, Marton LF, Roberts JD, Cobden PM, Somogyi P. Cholecystokinin-immunopositive basket and Schaffer collateral-associated interneurones target different domains of pyramidal cells in the CA1 area of the rat hippocampus. Neuroscience. 2002;109(1):63–80.
    1. Kawamura Y, Fukaya M, Maejima T, Yoshida T, Miura E, Watanabe M, et al. The CB1 cannabinoid receptor is the major cannabinoid receptor at excitatory presynaptic sites in the hippocampus and cerebellum. J Neurosci. 2006;26(11):2991–3001.
    1. Pitler TA, Alger BE. Depolarization-induced suppression of GABAergic inhibition in rat hippocampal pyramidal cells: G protein involvement in a presynaptic mechanism. Neuron. 1994;13(6):1447–1455.
    1. Wilson RI, Nicoll RA. Endogenous cannabinoids mediate retrograde signalling at hippocampal synapses. Nature. 2001;410(6828):588–592.
    1. Glickfeld LL, Scanziani M. Distinct timing in the activity of cannabinoid-sensitive and cannabinoid-insensitive basket cells. Nature Neuroscience. 2006;9(6):807–815.
    1. Basu J, Srinivas KV, Cheung SK, Taniguchi H, Huang ZJ, Siegelbaum SA. A cortico-hippocampal learning rule shapes inhibitory microcircuit activity to enhance hippocampal information flow. Neuron. 2013;79(6):1208–1221.
    1. Neu A, Foldy C, Soltesz I. Postsynaptic origin of CB1-dependent tonic inhibition of GABA release at cholecystokinin-positive basket cell to pyramidal cell synapses in the CA1 region of the rat hippocampus. J Physiol. 2007;578(Pt 1):233–247.
    1. Kim J, Alger BE. Reduction in endocannabinoid tone is a homeostatic mechanism for specific inhibitory synapses. Nat Neurosci. 2010;13(5):592–600.
    1. Hefft S, Jonas P. Asynchronous GABA release generates long-lasting inhibition at a hippocampal interneuron-principal neuron synapse. Nature Neuroscience. 2005;8(10):1319–1328.
    1. Foldy C, Malenka RC, Sudhof TC. Autism-associated neuroligin-3 mutations commonly disrupt tonic endocannabinoid signaling. Neuron. 2013;78(3):498–509.
    1. Trezza V, Campolongo P, Cassano T, Macheda T, Dipasquale P, Carratu MR, et al. Effects of perinatal exposure to delta-9-tetrahydrocannabinol on the emotional reactivity of the offspring: a longitudinal behavioral study in Wistar rats. Psychopharmacology (Berl) 2008;198(4):529–537.
    1. Omiya Y, Uchigashima M, Konno K, Yamasaki M, Miyazaki T, Yoshida T, et al. VGluT3-expressing CCK-positive basket cells construct invaginating synapses enriched with endocannabinoid signaling proteins in particular cortical and cortex-like amygdaloid regions of mouse brains. J Neurosci. 2015;35(10):4215–4228.
    1. Aguado T, Palazuelos J, Monory K, Stella N, Cravatt B, Lutz B, et al. The endocannabinoid system promotes astroglial differentiation by acting on neural progenitor cells. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2006;26(5):1551–1561.
    1. Keimpema E, Mackie K, Harkany T. Molecular model of cannabis sensitivity in developing neuronal circuits. Trends Pharmacol Sci. 2011;32(9):551–561.
    1. Shabani M, Hosseinmardi N, Haghani M, Shaibani V, Janahmadi M. Maternal exposure to the CB1 cannabinoid agonist WIN 55212-2 produces robust changes in motor function and intrinsic electrophysiological properties of cerebellar Purkinje neurons in rat offspring. Neuroscience. 2011;172:139–152.
    1. Campolongo P, Trezza V, Cassano T, Gaetani S, Morgese MG, Ubaldi M, et al. Perinatal exposure to delta-9-tetrahydrocannabinol causes enduring cognitive deficits associated with alteration of cortical gene expression and neurotransmission in rats. Addict Biol. 2007;12(3–4):485–495.
    1. Spano MS, Ellgren M, Wang X, Hurd YL. Prenatal cannabis exposure increases heroin seeking with allostatic changes in limbic enkephalin systems in adulthood. Biol Psychiatry. 2007;61(4):554–563.
    1. Maccarrone M, Guzman M, Mackie K, Doherty P, Harkany T. Programming of neural cells by (endo)cannabinoids: from physiological rules to emerging therapies. Nat Rev Neurosci. 2014;15(12):786–801.
    1. Kehrer C, Maziashvili N, Dugladze T, Gloveli T. Altered Excitatory-Inhibitory Balance in the NMDA-Hypofunction Model of Schizophrenia. Frontiers in molecular neuroscience. 2008;1:6.
    1. Calfa G, Li W, Rutherford JM, Pozzo-Miller L. Excitation/Inhibition imbalance and impaired synaptic inhibition in hippocampal area CA3 of Mecp2 knockout mice. Hippocampus. 2014
    1. Yizhar O, Fenno LE, Prigge M, Schneider F, Davidson TJ, O’Shea DJ, et al. Neocortical excitation/inhibition balance in information processing and social dysfunction. Nature. 2011;477(7363):171–178.
    1. Ziburkus J, Cressman JR, Schiff SJ. Seizures as imbalanced up states: excitatory and inhibitory conductances during seizure-like events. J Neurophysiol. 2013;109(5):1296–1306.
    1. Cass DK, Flores-Barrera E, Thomases DR, Vital WF, Caballero A, Tseng KY. CB1 cannabinoid receptor stimulation during adolescence impairs the maturation of GABA function in the adult rat prefrontal cortex. Molecular psychiatry. 2014;19(5):536–543.
    1. Lee SH, Soltesz I. Requirement for CB1 but not GABAB receptors in the cholecystokinin mediated inhibition of GABA release from cholecystokinin expressing basket cells. J Physiol. 2011;589(Pt 4):891–902.
    1. Etherton M, Foldy C, Sharma M, Tabuchi K, Liu X, Shamloo M, et al. Autism-linked neuroligin-3 R451C mutation differentially alters hippocampal and cortical synaptic function. Proc Natl Acad Sci U S A. 2011;108(33):13764–13769.

Source: PubMed

3
Se inscrever