Elsevier

Neuropharmacology

Volume 196, 15 September 2021, 108702
Neuropharmacology

Lateral hypothalamus-projecting noradrenergic locus coeruleus pathway modulates binge-like ethanol drinking in male and female TH-ires-cre mice

https://doi.org/10.1016/j.neuropharm.2021.108702Get rights and content

Highlights

  • The NE reuptake inhibitor, reboxetine, blunts binge-like ethanol intake, suggesting a potential therapeutic target.

  • Chemogenetic activation of the locus coeruleus blunts binge-like ethanol intake.

  • Chemogenetic activation of a norepinephrine circuit from the LC to lateral hypothalamus (LH) blunts binge-like ethanol intake.

  • Pharmacological manipulation of adrenergic receptor in the LH selectively blunts binge-like ethanol intake.

Abstract

A growing body of literature implicates noradrenergic (NE) signaling in the modulation of ethanol consumption. However, relatively few studies have detailed specific brain pathways that mediate NE-associated binge-like ethanol consumption. To begin to fill this gap in the literature, male and female C57BL6/J and TH-ires-cre mice underwent pharmacological and chemogenetic testing, respectively, in combination with “drinking in the dark” procedures to model binge-like consumption of ethanol or sucrose solutions. First, we showed that intraperitoneal administration of the NE reuptake inhibitor, reboxetine, blunted binge-like ethanol intake in C57BL6/J mice. Chemogenetic activation of locus coeruleus (LC) tyrosine hydroxylase (TH)-expressing neurons blunted binge-like ethanol intake regardless of sex. Chemogenetic activation of LC projections to the lateral hypothalamus (LH), a region implicated in ethanol consumption, blunted binge-like ethanol drinking without altering sucrose intake in ethanol-experienced or ethanol-naïve mice. In C57BL/6 J mice, LH-targeted microinfusion of an α1-adrenergic receptor (AR) agonist blunted binge-like ethanol intake across both sexes, while LH infusion of a β-AR agonist blunted binge-like ethanol intake in females exclusively. Finally, in mice with high baseline ethanol intake both an α1- AR agonist and an α-2 AR antagonist blunted binge-like ethanol intake. The present results provide novel evidence that increased NE tone in a circuit arising from the LC and projecting to the LH reduces binge-like ethanol drinking in mice, and may represent a novel approach to treating binge or heavy drinking prior to the development of dependence.

This article is part of the special Issue on “Neurocircuitry Modulating Drug and Alcohol Abuse”.

Introduction

Alcohol abuse poses significant health risks and economic costs to individuals around the world (Lim et al., 2012; Rehm et al., 2009). Repeated bouts of binge drinking, defined as a pattern of drinking yielding blood ethanol concentrations (BECs) achieving the U.S. legal limit of 80 mg/dl within a short (2-h) time window (NIAAA, 2004), are thought to contribute to the development of later-life alcohol dependence (Hingson et al., 2006). Recent estimates indicate that nearly 90% of U.S. adults that drink excessively consume alcohol in the form of a binge (Esser et al., 2014). Thus, understanding the neurochemical signaling systems that modulates binge-like ethanol consumption is critical for identification of potential therapies for reducing this dangerous pattern of behavior. To this end, the 4-day “drinking-in-the-dark” (DID) paradigm and its variants are among the more popular paradigms for modeling voluntary binge-like ethanol intake in mice (Rhodes et al., 2005; Thiele and Navarro, 2014) and rats (Bell et al., 2011; Holgate et al., 2017), promoting ingestion of large quantities of ethanol and reliably generating BECs exceeding 80 mg/dl. Using DID procedures, researchers have begun to examine roles for neurochemical signaling systems and circuitry that modulate binge-like ethanol intake (Sprow and Thiele, 2012). Interestingly, there is evidence that overlapping neurochemical pathways in the brain modulate both alcohol use disorders and eating disorders (Thiele et al., 2003), suggesting that at least in some circumstances common therapeutic strategies may be identified.

Norepinephrine (NE) is centrally synthesized in brainstem nuclei, with the majority of its synthesis occurring via the locus coeruleus (LC) (Sawchenko and Swanson, 1982), and has long been implicated in ethanol ingestion (Arango et al., 1994; Gilpin and Koob, 2010; Lu et al., 1997; O'Neil et al., 2013; Rasmussen et al., 2014a; Rasmussen et al., 2009; Rasmussen et al., 2014b; Simpson et al., 2009; Verplaetse et al., 2012) as well as feeding behavior (Bello et al., 2019). In fact, the NE/dopamine (DA) reuptake inhibitor bupropion (BUP), when combined with the non-selective opioid antagonist, naltrexone (NAL), has been used successfully to treat binge eating in human (Halseth et al., 2018), and we have recently shown that BUP alone and in combination with NAL blunts binge-like ethanol intake in mice (Navarro et al., 2019). Further, in a preliminary open-label study we recently reported that BUP + NAL therapy blunts the frequency of binge ethanol drinking in humans (Walter et al., 2020). These observations suggest that blunted NE tone contributes to binge behavior, and increasing NE tone is protective. However, there is a gap in the literature on the specific NE neurocircuitry that modulates binge ethanol drinking.

Insight into the NE mechanisms involved in modulating binge behavior comes from studies that have used c-Fos immunoreactivity (IR) as a marker of neuronal activation. It has been shown that the LC is activated following voluntary binge-like ethanol consumption (Burnham and Thiele, 2017). Interestingly, i. p. injection of ethanol into rats bred for high versus low ethanol consumption revealed reduced ethanol-induced c-Fos IR in the LC among rats bred for high levels of intake (Thiele et al., 1997). More recently, we showed that an inbred line of mice that was selectively bred to achieve binge-like blood ethanol levels (the iHDID-1 line) failed to exhibit ethanol-induced c-Fos expression in the LC at doses that induced c-Fos expression in the control HS/Npt line (Robinson et al., 2020). These observations suggest that LC activity is triggered by ethanol, and the observation that this activity is blunted in high drinking lines of rats and mice supports a role for ethanol-induced LC activity as a protective mechanism to limit ethanol intake.

In rodents, the lateral hypothalamus (LH) has been implicated in modulation of many behaviors including ethanol consumption (Chen et al., 2013, 2014; Navarro et al., 2016; Sprow et al., 2016; Wayner et al., 1971) and seeking (Marchant et al., 2009, 2014). Though brainstem NE nuclei innervate numerous regions (Robertson et al., 2016), the LH connects reciprocally with the LC (Jones and Moore, 1977; Papp and Palkovits, 2014) and contains rich populations of α-1, α-2, and β-adrenergic receptors (ARs) (Leibowitz et al., 1982). Thus, the LC may signal through the LH to regulate binge-like ethanol consumption. Experiments herein elucidated the role of the LC in binge-like ethanol intake. First, we show that peripheral administration of a NE reuptake inhibitor (NRI) significantly blunted binge-like ethanol consumption without altering overall motor behavior or anxiety-like behavior, consistent with our previous studies using BUP. Using excitatory Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), we demonstrated that general activation the LC blunted binge-like ethanol and sucrose consumption. Specific activation of TH-expressing LC neuron projecting to the LH similarly reduced binge-like ethanol consumption without altering sucrose drinking in ethanol-experienced or ethanol-naïve mice. Finally, we show that LH-directed adrenergic receptor (AR) manipulations reduced ethanol consumption that were compound-specific, and in some cases sex-specific and dependent on baseline levels of ethanol intake. The present results provide novel evidence that increased NE tone in a circuit arising from the LC and projecting to the LH reduces binge-like ethanol drinking, and may represent a novel strategy for treating binge drinking disorders prior to the development of ethanol dependence.

Section snippets

Animals

Male and female TH-ires-Cre mice (Savitt et al., 2005), bred in house and backcrossed on a C57BL6/J strain, were utilized for DREADD manipulations and tracing studies. Male and female C57BL6/J mice (stock # 000664, Jackson Laboratory), 6–8 weeks old upon arrival were utilized for pharmacological manipulations. All mice were individually housed at least 1 week prior to testing onset with ad libitum access to Prolab® RMH 3000 (Purina labDiet®; St. Louis, MO) and water except where noted. The

Experiment 1: effects of systemic administration of a peripherally bioavailable NRI on binge-like ethanol intake

Data from this experiment are presented in Fig. 1. A repeated-measures ANOVA (sex x treatment) failed to reveal a main effect of sex or sex by treatment interaction, so data from males and females were collapsed. Paired t-test for total ethanol intake revealed a significant reduction in intake following 10 mg/kg reboxetine pretreatment (Fig. 1A) [t-test: t (20) = 4.199, p < 0.001]. 10 mg/kg reboxetine similarly blunted BECs (Fig. 1B) [t-test: t (20) = 3.473, p = 0.002]. Fig. 1C shows a

Discussion

This series of studies sought to uncover the roles of the LC and its projection to the LH in modulation of voluntary binge-like ethanol consumption. First, we first showed that systemic administration of the NRI, reboxetine, blunted binge-like ethanol intake in both sexes without altering locomotor or anxiety-like behavior. These findings are consistent with our previous observation that the NE and dopamine re-uptake inhibitor, BUP, significantly blunts binge-like ethanol drinking mice (Navarro

Conclusions

Though well-established as playing a critical role in depression and anxiety disorders (Montoya et al., 2016), a growing body of literature is unpacking a role for central NE signaling in the modulation of drug and alcohol intake. Our results indicate that elevated NE signaling in the NE LC→LH circuit plays protective roles against binge-like ethanol intake. We recently reported that the dopamine/norepinephrine reuptake inhibitor, bupropion, blunts binge-like ethanol intake in mice (Navarro et

Declaration of competing interest

The authors declare no competing financial interests. Dr. Thiele owns shares of Glauser Life Sciences, a copy the aims to develop therapeutics for mental health disorders. The work that is presented in this paper is not directly related to the scientific aims of Glauser Life Sciences.

Acknowledgments

We thank Rhiannon Thomas, Timothy Gilliam, Sophie Bendrath, and Sonia Sabater for their expert assistance with the present projects. This research was funded by NIH grants AA013573, AA022048, & AA025809.

References (99)

  • A. Guillamon et al.

    Effects of sex steroids on the development of the locus coeruleus in the rat

    Brain Res.

    (1988)
  • J.H. Jennings et al.

    Visualizing hypothalamic network dynamics for appetitive and consummatory behaviors

    Cell

    (2015)
  • L. Kochenborger et al.

    Modulation of fear/anxiety responses, but not food intake, following alpha-adrenoceptor agonist microinjections in the nucleus accumbens shell of free-feeding rats

    Neuropharmacology

    (2012)
  • G.F. Koob et al.

    Drug addiction, dysregulation of reward, and allostasis

    Neuropsychopharmacology

    (2001)
  • S.F. Leibowitz et al.

    Distribution of alpha-adrenergic, beta-adrenergic and dopaminergic receptors in discrete hypothalamic areas of rat

    Brain Res.

    (1982)
  • S.S. Lim et al.

    A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010

    Lancet

    (2012)
  • L.S. Lubbers et al.

    Estrogen receptor (ER) subtype agonists alter monoamine levels in the female rat brain

    J. Steroid Biochem. Mol. Biol.

    (2010)
  • J.M. Luque et al.

    Sexual dimorphism of the dopamine-beta-hydroxylase-immunoreactive neurons in the rat locus ceruleus

    Brain Res Dev Brain Res

    (1992)
  • S.S. Mansur et al.

    Changes in food intake and anxiety-like behaviors after clonidine injected into the median raphe nucleus

    Behav. Brain Res.

    (2010)
  • S.S. Mansur et al.

    Phenylephrine into the median raphe nucleus evokes an anxiolytic-like effect in free-feeding rats but does not alter food intake in free feeding rats

    Behav. Brain Res.

    (2011)
  • C.Y. Ostock et al.

    Modulation of L-DOPA's antiparkinsonian and dyskinetic effects by alpha2-noradrenergic receptors within the locus coeruleus

    Neuropharmacology

    (2015)
  • H. Pinos et al.

    The development of sex differences in the locus coeruleus of the rat

    Brain Res. Bull.

    (2001)
  • D.D. Rasmussen et al.

    The alpha2-adrenergic receptor agonist, clonidine, reduces alcohol drinking in alcohol-preferring (P) rats

    Alcohol

    (2014)
  • D.E. Redmond et al.

    Hyperphagia and hyperdipsia after locus coeruleus lesions in the stumptailed monkey

    Life Sci.

    (1977)
  • J. Rehm et al.

    Global burden of disease and injury and economic cost attributable to alcohol use and alcohol-use disorders

    Lancet

    (2009)
  • J.S. Rhodes et al.

    Evaluation of a simple model of ethanol drinking to intoxication in C57BL/6J mice

    Physiol. Behav.

    (2005)
  • J.A. Rinker et al.

    Extended amygdala to ventral tegmental area corticotropin-releasing factor circuit controls binge ethanol intake

    Biol. Psychiatry

    (2017)
  • S.D. Robertson et al.

    Uncovering diversity in the development of central noradrenergic neurons and their efferents

    Brain Res.

    (2016)
  • M.A. Roni et al.

    Effects of lobeline and reboxetine, fluoxetine, or bupropion combination on depression-like behaviors in mice

    Pharmacol. Biochem. Behav.

    (2015)
  • B.J. Sahakian et al.

    Changes in body weight and food-related behaviour induced by destruction of the ventral or dorsal noradrenergic bundle in the rat

    Neuroscience

    (1983)
  • P.E. Sawchenko et al.

    The organization of noradrenergic pathways from the brainstem to the paraventricular and supraoptic nuclei in the rat

    Brain Res.

    (1982)
  • D.A. Schreihofer et al.

    Cholecystokinin induces Fos expression in catecholaminergic neurons of the macaque monkey caudal medulla

    Brain Res.

    (1997)
  • N.R. Sciolino et al.

    Recombinase-dependent mouse lines for chemogenetic activation of genetically defined cell types

    Cell Rep.

    (2016)
  • P. Simon et al.

    Thigmotaxis as an index of anxiety in mice. Influence of dopaminergic transmissions

    Behav. Brain Res.

    (1994)
  • G.M. Sprow et al.

    The neurobiology of binge-like ethanol drinking: evidence from rodent models

    Physiol. Behav.

    (2012)
  • T.E. Thiele et al.

    Drinking in the dark" (DID) procedures: a model of binge-like ethanol drinking in non-dependent mice

    Alcohol

    (2014)
  • T.E. Thiele et al.

    Alcoholism and obesity: overlapping neuropeptide pathways?

    Neuropeptides

    (2003)
  • T.E. Thiele et al.

    Ethanol-induced c-Fos expression in rat lines selected for low and high alcohol consumption

    Brain Res.

    (1997)
  • N. Uemura et al.

    Induction of Fos-like immunoreactivity in the lower brainstem and the spinal cord of the rat by intraperitoneal administration of an endogenous satiety substance, 2-buten-4-olide

    Neurosci. Lett.

    (1997)
  • E. Vardy et al.

    A new DREADD facilitates the multiplexed chemogenetic interrogation of behavior

    Neuron

    (2015)
  • I. Vathy et al.

    Ovarian steroids and hypothalamic norepinephrine release: studies using in vivo brain microdialysis

    Life Sci.

    (1988)
  • M. Wang et al.

    Alpha2A-adrenoceptors strengthen working memory networks by inhibiting cAMP-HCN channel signaling in prefrontal cortex

    Cell

    (2007)
  • M.J. Wayner et al.

    Ethanol drinking elicited during electrical stimulation of the lateral hypothalamus

    Physiol. Behav.

    (1971)
  • C.H. West et al.

    Locus coeruleus neuronal activity determines proclivity to consume alcohol in a selectively-bred line of rats that readily consumes alcohol

    Alcohol

    (2015)
  • G.K. Aghajanian et al.

    Alpha 2-adrenoceptor-mediated hyperpolarization of locus coeruleus neurons: intracellular studies in vivo

    Science

    (1982)
  • L. Albrechet-Souza et al.

    Corticotropin releasing factor binding protein and CRF2 receptors in the ventral tegmental area: modulation of ethanol binge drinking in C57bl/6J mice

    Alcohol Clin. Exp. Res.

    (2015)
  • N.T. Bello et al.

    Reduced sensory-evoked locus coeruleus-norepinephrine neural activity in female rats with a history of dietary-induced binge eating

    Front. Psychol.

    (2019)
  • E.A. Carbone et al.

    An open-label trial on the efficacy and tolerability of naltrexone/bupropion SR for treating altered eating behaviours and weight loss in binge eating disorder

    Eat. Weight Disord.

    (2020)
  • Y.W. Chen et al.

    Differential role of D1 and D2 receptors in the perifornical lateral hypothalamus in controlling ethanol drinking and food intake: possible interaction with local orexin neurons

    Alcohol Clin. Exp. Res.

    (2014)
  • Cited by (0)

    View full text