Hormonal regulation of delta opioid receptor immunoreactivity in interneurons and pyramidal cells in the rat hippocampus

https://doi.org/10.1016/j.nlm.2011.01.002Get rights and content

Abstract

Clinical and preclinical studies indicate that women and men differ in relapse vulnerability to drug-seeking behavior during abstinence periods. As relapse is frequently triggered by exposure of the recovered addict to objects previously associated with drug use and the formation of these associations requires memory systems engaged by the hippocampal formation (HF), studies exploring ovarian hormone modulation of hippocampal function are warranted. Previous studies revealed that ovarian steroids alter endogenous opioid peptide levels and trafficking of mu opioid receptors in the HF, suggesting cooperative interaction between opioids and estrogens in modulating hippocampal excitability. However, whether ovarian steroids affect the levels or trafficking of delta opioid receptors (DORs) in the HF is unknown. Here, hippocampal sections of adult male and normal cycling female Sprague-Dawley rats were processed for quantitative immunoperoxidase light microscopy and dual label fluorescence or immunoelectron microscopy using antisera directed against the DOR and neuropeptide Y (NPY). Consistent with previous studies in males, DOR-immunoreactivity (-ir) localized to select interneurons and principal cells in the female HF. In comparison to males, females, regardless of estrous cycle phase, show reduced DOR-ir in the granule cell layer of the dentate gyrus and proestrus (high estrogen) females, in particular, display reduced DOR-ir in the CA1 pyramidal cell layer. Ultrastructural analysis of DOR-labeled profiles in CA1 revealed that while females generally show fewer DORs in the distal apical dendrites of pyramidal cells, proestrus females, in particular, exhibit DOR internalization and trafficking towards the soma. Dual label studies revealed that DORs are found in NPY-labeled interneurons in the hilus, CA3, and CA1. While DOR colocalization frequency in NPY-labeled neuron somata was similar between animals in the hilus, proestrus females had fewer NPY-labeled neurons that co-labeled with DOR in stratum oriens of CA1 and CA3 when compared to males. Ultrastructural analysis of NPY-labeled axon terminals within stratum radiatum of CA1 revealed that NPY-labeled axon terminals contain DORs that are frequently found at or near the plasma membrane. As no differences were noted by sex or estrous cycle phase, DOR activation on NPY-labeled axon terminals would inhibit GABA release probability equally in males and females. Taken together, these findings suggest that ovarian steroids can impact hippocampal function through direct effects on DOR levels and trafficking in principal cells and broad indirect effects through reductions in DOR-ir in NPY-labeled interneurons, particularly in CA1.

Research highlights

► Ovarian hormones modulate DOR levels and trafficking in hippocampal principal cells. ► DOR levels are reduced in granule cells and CA1 pyramidal cell dendrites of females. ► Proestrus females display selective internalization and trafficking of DORs in CA1. ► Proestrus females show fewer NPY-labeled interneurons with DORs in CA1 and CA3.

Introduction

Despite lower rates of use and abuse, clinical evidence suggests that women may be more susceptible to several aspects of addiction than men. For example, women report shorter drug-free periods and higher levels of craving and dysphoria during drug withdrawal (Elman et al., 2001, Griffin et al., 1989, Kosten et al., 1996). Women are also more likely to experience craving following exposure to drug-related cues (Robbins, Ehrman, Childress, & O’Brien, 1999) and relapse to drug-seeking behavior (Elman et al., 2001, McKay et al., 1996, Rubin et al., 1996). Animal models that mimic different phases of the addiction process have been useful in determining whether a biological basis exists for observed sex differences in abuse vulnerability, as these studies control for sociocultural and other factors that may occlude sex differences in clinical studies of drug abuse. In studies of drug relapse or reinstatement, female rats show more extinction responding on the drug-associated lever after drug removal and greater reinstatement after a priming injection than males. Additionally, reinstatement occurs after a lower priming dose in females than in males (Comer et al., 1996, Klein et al., 1997, Lynch and Carroll, 2000). Recent studies using intact and ovariectomized (OVX) rats replaced with estradiol propose that ovarian hormones, particularly estrogen, contribute to the aforementioned sex differences during reinstatement (Fuchs et al., 2005, Kippin et al., 2005, Larson et al., 2005). Taken together, clinical and preclinical studies indicate that women and men differ in relapse vulnerability during abstinence periods. Relapse is frequently triggered by exposure of the recovered addict to objects previously associated with drug use (White, 1996). As the formation of these associations requires the episodic and declarative memory systems engaged by the hippocampal formation (HF) (Hyman and Malenka, 2001, Nestler, 2002, Nestler, 2001, Holden, 2001), studies exploring ovarian hormone modulation of hippocampal function are warranted.

Hippocampal output is regulated through a series of principal cell and inhibitory interneuron connections that are susceptible to modulation by endogenous opioids and exogenous opiates. Ovarian steroid hormones have been shown to influence levels of the endogenous hippocampal opioid peptides, enkephalin and dynorphin, which directly modulate hippocampal excitability (Roman et al., 2006, Drake et al., 2007, Torres-Reveron et al., 2008, Torres-Reveron et al., 2009a, Williams et al., 2010). For example, leu-enkephalin levels are increased in sub-regions of the dentate gyrus (DG) and CA3 of young adult females when estrogen levels are relatively high (Torres-Reveron et al., 2008). Dynorphin levels are similarly increased in the DG and select CA3 lamina 24 h following estrogen exposure (Torres-Reveron et al., 2009a). In addition, ovarian steroid hormones modulate mu opioid receptor (MOR) trafficking in hippocampal interneurons. Specifically, elevated levels of estrogens, either in proestrus females or after 72 h of pulsatile estradiol replacement in OVX females, increased the availability of MORs on the plasma membrane of subpopulations of GABAergic basket cells in the hilar region of the DG (Torres-Reveron et al., 2009b). Such altered trafficking of MORs could potentially alter the disinhibitory effects of endogenous or exogenous opiates, given that MOR activation in the DG produces excitation by inhibiting GABAergic transmission (Bramham and Sarvey, 1996, Drake et al., 2007, Morris and Johnston, 1995, Xie and Lewis, 1995a). Activation of the delta opioid receptor (DOR) at the circuit level also affects excitatory transmission and the induction of synaptic plasticity in principal cells of the DG (Bausch and Chavkin, 1997, Bramham and Sarvey, 1996, Piguet and North, 1993) and CA1 (Bao et al., 2007) through both direct effects on principal cells and indirect effects via inhibition of interneurons. In addition, recent reports suggest that opioid peptides acting primarily on DORs play an important role in mediating cue- and context-induced drug-seeking behavior (Marinelli et al., 2009). Thus, ovarian steroid modulation of DOR immunoreactivity (-ir) levels and trafficking in hippocampal neurons merits further investigation.

Prior studies in male rats demonstrated that the highest DOR mRNA levels were found in GABAergic neurons in the principal cell layers, stratum oriens, and dentate hilus (Stumm, Zhou, Schulz, & Hollt, 2004). DOR-ir, moreover, was commonly found in somatostatin (SOM)/neuropeptide Y (NPY) containing GABAergic interneurons in hippocampal lamina (Commons and Milner, 1996, Commons and Milner, 1997). Moreover, in CA1 specifically, DORs were found also on pyramidal cell dendrites and somata and could therefore directly modulate pyramidal cell activity (Commons & Milner, 1997). The present study sought to extend these observations, where applicable, to the female hippocampus. Thus, single and dual immunolabeling approaches were used to assess ovarian hormone influences on DOR-ir and distribution within hippocampal principal cells and interneurons. Quantitative densitometric immunocytochemistry and immunoelectron microscopy were employed to measure altered DOR-ir and trafficking in principal cells of normal cycling proestrus (high estrogen) and diestrus (low estrogen) female rats in comparison to male rats. Dual label immunofluorescence and immunoelectron microscopy was used to examine DOR localization to NPY-labeled interneurons and axon terminals in select hippocampal lamina of males and normal cycling females. The current study focused on findings in the dorsal hippocampus, where estrogen-induced morphological changes have been consistently reported (Cooke & Woolley, 2005).

Section snippets

Animals and estrous cycle determination

Adult male (275–325 g; approximately 60 days old) and female (225–250 g; approximately 60 days old) Sprague-Dawley rats from Charles River Laboratories (Wilmington, MA) were pair-housed with ad libitum access to food and water and with 12:12 light/dark cycles (lights on 0600–1800). All procedures were approved by the Weill Cornell Medical College Institutional Animal Care and Use Committee and were in accordance with the National Institutes of Health guidelines. Female rats were allowed to

Ovarian hormones influence DOR-ir in the DG and CA1

In the dentate gyrus, consistent with previously reported DOR-labeling patterns (Commons & Milner, 1996), proestrus and diestrus females and males display diffuse DOR-ir in the granule cell layer as well as more intensely labeled hilar neurons varying in size and shape (Fig. 1A). To determine whether sex or hormonal status influenced DOR-labeling in the DG, we compared DOR-ir levels in granule cells and numbers of DOR-labeled neurons in the hilus between groups. Quantitative densitometric

Discussion

This study is the first to demonstrate that ovarian hormones present in normal cycling females modulate DOR levels and trafficking in hippocampal principal cells. Specifically, in comparison to males: (1) DOR levels are reduced in granule cell somata and distal dendrites of CA1 pyramidal cells of females, regardless of estrous cycle phase, and (2) proestrus (high estrogen) females, in particular, display trafficking of DORs away from the plasma membrane and into the cytoplasm of distal

Acknowledgments

This work was supported by National Institutes of Health grants DA08259, HL18974, HL096571, DA007274, DA028072, Minority Supplement to DA08259, NIH-MSTP grant GM07739, the American Psychological Association Diversity Program in Neuroscience, and the UNCF-Merck Science Initiative. We appreciate the insightful comments and expertise provided by Dr. Bruce McEwen. We are also thankful for the technical assistance of Dr. Diane Lane, Dr. Yuko Hara, Ms. Katherine Mitterling and Ms. Louisa Thompson.

References (105)

  • E.B. Larson et al.

    Effect of short- vs. long-term estrogen on reinstatement of cocaine-seeking behavior in female rats

    Pharmacology Biochemistry and Behavior

    (2005)
  • B.J. Morris et al.

    A role for hippocampal opioids in long-term functional plasticity

    Trends in Neurosciences

    (1995)
  • N.H. Nakamura et al.

    Changes in interneuronal phenotypes regulated by estradiol in the adult rat hippocampus: A potential role for neuropeptide Y

    Neuroscience

    (2005)
  • E.J. Nestler

    Common molecular and cellular substrates of addiction and memory

    Neurobiology of Learning and Memory

    (2002)
  • A.I. Persson et al.

    Comparison of immunoblotted delta opioid receptor proteins expressed in the adult rat brain and their regulation by growth hormone

    Neuroscience Research

    (2005)
  • S.J. Robbins et al.

    Comparing levels of cocaine cue reactivity in male and female outpatients

    Drug And Alcohol Dependence.

    (1999)
  • E. Roman et al.

    Variations in opioid peptide levels during the estrous cycle in Sprague-Dawley rats

    Neuropeptides

    (2006)
  • L.C. Saland et al.

    Chronic ethanol modulates delta and mu-opioid receptor expression in rat CNS: Immunohistochemical analysis with quantitiative confocal microscopy

    Neuroscience Letters

    (2005)
  • T.S. Shippenberg et al.

    Delta-opioid receptor antagonists prevent sensitization to the conditioned rewarding effects of morphine

    Biological Psychiatry

    (2009)
  • A. Torres-Reveron et al.

    Ovarian steroids modulate leu-enkephalin levels and target leu-enkephalinergic profiles in the female hippocampal mossy fiber pathway

    Brain Research

    (2008)
  • A. Torres-Reveron et al.

    Hippocampal dynorphin immunoreactivity increases in response to gonadal steroids and is positioned for direct modulation by ovarian steroid receptors

    Neuroscience

    (2009)
  • A. Torres-Reveron et al.

    Ovarian steroids alter mu opioid receptor trafficking in hippocampal parvalbumin GABAergic interneurons

    Experimental Neurology

    (2009)
  • P.I. Tsao et al.

    Type-specific sorting of G protein-coupled receptors after endocytosis

    Journal of Biological Chemistry

    (2000)
  • I. Vathy et al.

    Prenatal exposure to morphine differentially alters gonadal hormone regulation of delta-opioid receptor binding in male and female rats

    Brain Research Bulletin

    (2000)
  • A.B. Ali et al.

    Facilitating pyramid to horizontal oriens-alveus interneurone inputs: dual intracellular recordings in slices of rat hippocampus

    Journal of Physiology

    (1998)
  • G. Bao et al.

    Morphine and heroin differentially modulate in vivo hippocampal LTP in opiate-dependent rat

    Neuropsychopharmacology

    (2007)
  • S.B. Bausch et al.

    Changes in hippocampal circuitry after pilocarpine-induced seizures as revealed by opioid receptor distribution and activation

    Journal of Neuroscience

    (1997)
  • A. Belanger et al.

    Ovarian progestins, androgens and estrogen throughout the 4-day estrous cycle in the rat

    Biology of Reproduction

    (1981)
  • T.V. Bliss et al.

    A synaptic model of memory: Long-term potentiation in the hippocampus

    Nature

    (1993)
  • C.R. Bramham et al.

    Endogenous activation of mu and delta-1 opioid receptors is required for long-term potentiation induction in the lateral perforant path: Dependence on GABAergic inhibition

    Journal of Neuroscience

    (1996)
  • C.M. Cahill et al.

    Prolonged morphine treatment targets delta opioid receptors to neuronal plasma membranes and enhances delta-mediated antinociception

    Journal of Neuroscience

    (2001)
  • V.I. Chefer et al.

    Augmentation of morphine-induced sensitization but reduction in morphine tolerance and reward in delta-opioid receptor knockout mice

    Neuropsychopharmacology

    (2009)
  • P.Y. Cheng et al.

    Ultrastructural immunolabeling shows prominent presynaptic vesicular localization of delta-opioid receptor within both enkephalin- and nonenkephalin-containing axon terminals in the superficial layers of the rat cervical spinal cord

    Journal of Neuroscience

    (1995)
  • S.D. Comer et al.

    Combined effects of buprenorphine and a nondrug alternative reinforcer on i.v. Cocaine self-administration in rats maintained under FR schedules

    Psychopharmacology

    (1996)
  • K.G. Commons et al.

    Ultrastructural heterogeneity of enkephalin-containing terminals in the rat hippocampal formation

    Journal of Comparative Neurology

    (1995)
  • K.G. Commons et al.

    Localization of delta opioid receptor immunoreactivity in interneurons and pyramidal cells in the rat hippocampus

    Journal of Comparative Neurology

    (1997)
  • B.M. Cooke et al.

    Gonadal hormone modulation of dendrites in the mammalian CNS

    Journal of Neurobiology

    (2005)
  • A.D. Corbett et al.

    Selectivities of opioid peptide analogues as agonists and antagonists at the delta-receptor

    British Journal of Pharmacology

    (1984)
  • H.S. Crombag et al.

    Review. Context-induced relapse to drug seeking: A review

    Philosophical Transactions of the Royal Society of London Series B-Biological Sciences

    (2008)
  • C.T. Drake et al.

    Opioid systems in the dentate gyrus

    Progress in Brain Research

    (2007)
  • C.T. Drake et al.

    Mu opioid receptors are in discrete hippocampal interneuron subpopulations

    Hippocampus

    (2002)
  • C.B. Eckersell et al.

    Estrogen-induced alteration of mu-opioid receptor immunoreactivity in the medial preoptic nucleus and medial amygdala

    Journal of Neuroscience

    (1998)
  • I. Elman et al.

    Gender differences in cocaine craving among non-treatment-seeking individuals with cocaine dependence

    American Journal of Drug and Alcohol Abuse

    (2001)
  • T.F. Freund et al.

    Interneurons of the hippocampus

    Hippocampus

    (1996)
  • R.A. Fuchs et al.

    Interactions of the basolateral amygdala with the dorsal hippocampus and dorsomedial prefrontal cortex regulate drug context-induced reinstatement of cocaine-seeking in rats

    European Journal of Neuroscience

    (2007)
  • R.A. Fuchs et al.

    The role of the dorsomedial prefrontal cortex, basolateral amygdala, and dorsal hippocampus in contextual reinstatement of cocaine seeking in rats

    Neuropsychopharmacology

    (2005)
  • M.L. Griffin et al.

    A comparison of male and female cocaine abusers

    Archives of General Psychiatry

    (1989)
  • Y. Hara et al.

    Dopamine D1 receptors have subcellular distributions conducive to interactions with prodynorphin in the rat nucleus accumbens shell

    Synapse

    (2006)
  • K.M. Harris et al.

    Three-dimensional structure of dendritic spines and synapses in rat hippocampus (CA1) at postnatal day 15 and adult ages: Implications for the maturation of synaptic physiology and long-term potentiation

    Journal of Neuroscience

    (1992)
  • C. Holden

    Drug addiction. Zapping memory center triggers drug craving

    Science

    (2001)
  • Cited by (36)

    • Sex and chronic stress alter delta opioid receptor distribution within rat hippocampal CA1 pyramidal cells following behavioral challenges

      2020, Neurobiology of Stress
      Citation Excerpt :

      A person blinded to experimental conditions performed the analyses. Densitometric quantification for DOR-ir within the CA1 region of the hippocampus (Fig. 1A) was done using previously described methods (Pierce et al., 2014; Williams and Milner, 2011; Williams et al., 2011b). Briefly, images of the hippocampus were captured at 4x on a Nikon Eclipse 80i microscope using a Micropublisher 5.0 digital camera (Q-imaging, BC, Canada) and IP Lab software (Scanalytics IPLab, RRID:SCR_002775).

    • Sex Differences in Neuroplasticity- and Stress-Related Gene Expression and Protein Levels in the Rat Hippocampus Following Oxycodone Conditioned Place Preference

      2019, Neuroscience
      Citation Excerpt :

      Increases in both NPY/DOR containing cells, as well as the pool of DORs readily available to be inserted into the plasma membrane (Boudin et al., 1998) following oxycodone CPP, could have functional consequences. Hilar NPY interneurons are known to project to granule cell dendrites where they converge with entorhinal afferents (Milner and Veznedaroglu, 1992; Sik et al., 1995; Williams et al., 2011b). Since NPY release is inhibited by DORs, activation of DORs on NPY-containing interneurons could promote lpp LTP (Sperk et al., 2007).

    View all citing articles on Scopus
    View full text