Glucagon-like peptide-1 receptor regulation of basal dopamine transporter activity is species-dependent

https://doi.org/10.1016/j.neuint.2020.104772Get rights and content

Highlights

  • GLP-1 (7–36)-amide increased DA uptake, DAT expression and DA clearance in rats.

  • No effect of exenatide on DA uptake and no change in DA uptake in GLP-1R KO in mice.

  • No acute effect of exenatide on DAT availability in striatum in healthy individuals.

Abstract

Introduction

A solid body of preclinical evidence shows that glucagon-like peptide-1 receptor (GLP-1R) agonists attenuate the effects of substance use disorder related behaviors. The mechanisms underlying these effects remain elusive. In the present study, we hypothesized that GLP-1R activation modulates dopaminetransporter (DAT) and thus dopamine (DA) homeostasis in striatum. This was evaluated in three different experiments: two preclinical and one clinical.

Methods

Rat striatal DA uptake, DA clearance and DAT cell surface expression was assessed following GLP-1 (7-36)-amide exposure in vitro. DA uptake in mice was assesed ex vivo following systemic treatment with the GLP-1R agonist exenatide. In addition, DA uptake was measured in GLP-1R knockout mice and compared with DA-uptake in wild type mice. In healthy humans, changes in DAT availability was assessed during infusion of exenatide measured by single-photon emission computed tomography imaging.

Results

In rats, GLP-1 (7-36)-amide increased DA uptake, DA clearance and DAT cell surface expression in striatum. In mice, exenatide did not change striatal DA uptake. In GLP-1R knockout mice, DA uptake was similar to what was measured in wildtype mice. In humans, systemic infusion of exenatide did not result in acute changes in striatal DAT availability.

Conclusions

The GLP-1R agonist-induced modulation of striatal DAT activity in vitro in rats could not be replicated ex vivo in mice and in vivo in humans. Therefore, the underlying mechanisms of action for the GLP-1R agonists-induced efficacy in varios addiction-like behavioural models still remain.

Introduction

Natural reward and drug reward converge on a common neural pathway, the mesolimbic dopamine system. Since drugs activate the same reward system that underlies food reward (Volkow et al., 2013) it is perhaps no surprise that appetite-regulating peptides, besides governing energy homeostasis, also target brain areas associated with reward and addiction (Engel and Jerlhag, 2014). Glucagon-like peptide-1 (GLP-1) is a peptide that acts both as an incretin hormone that regulates blood sugar and as a neuropeptide in the brain regulating satiety (Holst, 2013). Its anorexic and glucoregulatory effects are well-established and GLP-1 receptor (GLP-1R) agonists (GLP-1RA) are approved for clinical use in type 2 diabetes and obesity (Isaacs et al., 2016). Recent years of research have broadened the understanding of the GLP-1 system especially by linking it to the rewarding properties of food (Dickson et al., 2012; Skibicka, 2013; Sisley et al., 2014). In the brain, GLP-1Rs are found in areas associated with reward, reinforcement and addiction including the ventral tegmental area (VTA) and striatum (Göke et al., 1995; Merchenthaler et al., 1999; Alhadeff et al., 2012). The VTA and its dopaminergic projections to the nucleus accumbens (NAc) orchestrate motivated behavior to obtain natural reward (such as food and sex) but are prone to be hijacked by artificial reward substances (such as drugs of abuse and alcohol) ultimately leading to addiction (Koob, 1992). Importantly, many appetite-regulating hormones including GLP-1 exert a direct influence on the VTA and NAc (Fulton, 2010). Taken together these data have led to the hypothesis that the central GLP-1R system modulates the reward system. Indeed, systemic or central administration of GLP-1RAs in rodents and non-human primates attenuate addiction-related effects of alcohol, central stimulants and nicotine (Graham et al., 2013; Egecioglu et al., 2013; Sørensen et al., 2015; Reddy et al., 2016; Fortin and Roitman, 2017; Thomsen et al., 2017, 2019; Brunchmann et al., 2019, Tuesta et al., 2017). In addition, gene variants of GLP-1R in humans have been associated with the prevalence of alcohol use disorder (AUD) (Suchankova et al., 2015).

Although GLP-1R activation attenuates the rewarding properties of food, drugs and alcohol, the underlying mechanisms remain largely unknown. We recently suggested a link between GLP-1R activation and the dopamine transporter (DAT) that regulates dopamine (DA) homeostasis in the lateral septum (LS) of the brain (Reddy et al., 2016), and elsewhere. GLP-1Rs are highly expressed in LS, which is also associated with reward (OLDS and MILNER, 1954). Brain DA plays a pivotal role in drug addiction and since plasma membrane DAT is essential for terminating DA neurotransmission, regulation of DAT may or may not underlie the observed effects of GLP-1R on reward. Since the striatum serves as a central interface in the reward system (Yager et al., 2015) we wanted to investigate if the GLP-1R system modulates DAT and consequently, levels of synaptic DA.

Here we report data using native GLP-1 (rodents) and GLP-1RA (humans) as tools in three different studies; two studies in rodents and one study in healthy humans. Our hypothesis was supported by the present rat striatal data, but not by data from mice or humans.

Section snippets

Animals and striatal slice preparation

Experimentally naïve male Sprague-Dawley rats (275–300g Charles River) were used. Corticostriatal hemislices (300 μm; hereafter “striatal slices”) were prepared with a vibratome (Leica VT1000S) in an ice cold oxygenated (95% O2/5% CO2) sucrose cutting solution consisting of (in mM): 210 sucrose, 20 NaCl, 2.5 KCl, 1 MgCl2, 1.2 NaH2PO4, 10 glucose, 26 NaHCO3. Slices were then transferred to oxygenated artificial cerebrospinal fluid (aCSF) at 28 °C for a minimum of 1 h. The aCSF consisted of (in

Data analysis

In Experiment 1, DA uptake data were analyzed by one-way ANOVA followed by Bonferroni-Holm post-hoc test. DAT data were analyzed by one tailed paired t-test and DA clearance data were analyzed by Student's t-test. In Experiment 2, DA uptake data were analyzed by two-way ANOVA with exenatide treatment or genotype as between-subjects factor and DA concentration as repeated-measures factor. In Experiment 3, data were analyzed by one-way ANOVA with BPP as repeated-measures factor. All data are

Experiment 1 – GLP-1 (7–36)-amide increased DA uptake, DAT expression and DA clearance in rats

Incubating slices with 100 nM GLP-1 (7–36)-amide significantly increased DA uptake relative to aCSF control (Fig. 1A). Cell surface DAT expression, labeled by biotinylation, was also significantly increased in GLP-1 (7–36)-amide -treated slices compared to vehicle (Fig. 1B). Using high-speed chronoamperometry (HSCA), we measured the effect of locally microinjected GLP-1 (7–36)-amide in vivo on DA clearance in the striatum. GLP-1(7–36)-amide produced a transient increase in the rate of DA

Preclinical results

A regulatory role of GLP-1Rs for reward modulating DAT and DA homeostasis seems likely since it has been shown that GLP-1Rs are widely expressed in the mesolimbic dopamine system including striatum (Merchenthaler et al., 1999) and several preclinical studies have demonstrated an attenuating effect on feeding as well as alcohol- and drug-related behavior (Engel and Jerlhag, 2014; Skibicka, 2013). We have previously demonstrated that the GLP-1RA exenatide attenuates cocaine-induced DA release in

Conclusion

A solid body of preclinical evidence shows that GLP-1R agonists attenuate the effects of substance use disorder-related behaviors. The mechanisms underlying these effects remain elusive. In the present study, we hypothesized that GLP-1R activation modulates DAT and thus DA homeostasis in striatum. In rats and using GLP-1 (7–36)-amide, we found support for this hypothesis corresponding with our previous findings in the LS (Reddy et al., 2016). However, we could not replicate the present rat

Author contribution statement

Conceptualization-MEJ, UG, AG, TV, GM, AFJ

Methodology-All authors

Validation, Formal analysis, Investigation-All authors

Resources, Data Curation-All authors

Writing - Original Draft-MEJ

Writing - Review & Editing-All authors

Supervision-AG, UG, TV, GM, AFJ

Project administration-AG, UG, TV, GM, AFJ

Funding acquisition-AG, UG, TV, GM, AFJ

Funding

Funding was provided by The Research Foundation, Mental Health Services, Capital Region of Denmark, The Research Foundation, Capital Region of Denmark and by the National Institutes of Health [NIH DA043960]. MT was supported by NIH grant AA025071. LCD was supported by NIH grant MH64489.

Acknowledgements

We thank the staff, especially Svitlana Olsen and Peter Jensen for excellent technical support.

References (58)

  • A.L. Alhadeff et al.

    GLP-1 neurons in the nucleus of the solitary tract project directly to the ventral tegmental area and nucleus accumbens to control for food intake

    Endocrinology

    (2012)
  • K.K. Antonsen

    Does glucagon-like peptide-1 (GLP-1) receptor agonist stimulation reduce alcohol intake in patients with alcohol dependence: study protocol of a randomised, double-blinded, placebo-controlled clinical trial

    BMJ Open

    (2018)
  • G.M. Bray

    Exenatide. Am J Health Syst Pharm

    (2006)
  • B.J. Ciliax

    Immunocytochemical localization of the dopamine transporter in human brain

    J. Comp. Neurol.

    (1999)
  • L.C. Daws et al.
  • L.C. Daws et al.

    Neurotransmitter Transporters 53-81

    (2016)
  • M.E. Desimone et al.
  • S.L. Dickson

    The glucagon-like peptide 1 (GLP-1) analogue, exendin-4, decreases the rewarding value of food: a new role for mesolimbic GLP-1 receptors

    J. Neurosci.

    (2012)
  • C.M. Edwards

    Exendin-4 reduces fasting and postprandial glucose and decreases energy intake in healthy volunteers

    Am. J. Physiol. Endocrinol. Metab.

    (2001)
  • E. Egecioglu et al.

    The glucagon-like peptide 1 analogue, exendin-4, attenuates the rewarding properties of psychostimulant drugs in mice

    PloS One

    (2013)
  • J.A. Engel et al.

    Role of appetite-regulating peptides in the pathophysiology of addiction: implications for pharmacotherapy

    CNS Drugs

    (2014)
  • M. Fineman

    Pharmacokinetics and pharmacodynamics of exenatide extended-release after single and multiple dosing

    Clin. Pharmacokinet.

    (2011)
  • M.M. Fletcher et al.

    The complexity of signalling mediated by the glucagon-like peptide-1 receptor

    Biochem. Soc. Trans.

    (2016)
  • R. Göke et al.

    Distribution of GLP-1 binding sites in the rat brain: evidence that exendin-4 is a ligand of brain GLP-1 binding sites

    Eur. J. Neurosci.

    (1995)
  • D.L. Graham et al.

    GLP-1 analog attenuates cocaine reward

    Mol. Psychiatr.

    (2013)
  • A. Hamilton et al.

    Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system

    Neuroreport

    (2009)
  • A.E. Harasta

    Septal glucagon-like peptide 1 receptor expression determines suppression of cocaine-induced behavior

    Neuropsychopharmacology

    (2015)
  • K.M. Heppner

    Expression and distribution of glucagon-like peptide-1 receptor mRNA, protein and binding in the male nonhuman primate (Macaca mulatta) brain

    Endocrinology

    (2015)
  • N.S. Hernandez et al.

    Activation of glucagon-like peptide-1 receptors in the nucleus accumbens attenuates cocaine seeking in rats

    Addiction Biol.

    (2019)
  • Cited by (11)

    • A novel approach to treating opioid use disorders: Dual agonists of glucagon-like peptide-1 receptors and neuropeptide Y<inf>2</inf> receptors

      2021, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      First, there may be species-specific differences in the efficacy of exendin-4 to reduce opioid-mediated behaviors. Central GLP-1R expression is different in mice and rats (Cork et al., 2015b; Graham et al., 2020; Gu et al., 2013; Merchenthaler et al., 1999) and this may affect the ability of GLP-1R agonists to modulate the mesolimbic dopamine system (Fortin and Roitman, 2017; Jensen et al., 2020). In addition, the pharmacokinetic profile of exendin-4 is not as well characterized in mice as in rats (Chen et al., 2013).

    • A novel dual agonist of glucagon-like peptide-1 receptors and neuropeptide Y2 receptors attenuates fentanyl taking and seeking in male rats

      2021, Neuropharmacology
      Citation Excerpt :

      First, there may be species-specific differences in the efficacy of exendin-4. Central GLP-1R expression is different in mice and rats (Cork et al., 2015a; Graham et al., 2020; Gu et al., 2013; Merchenthaler et al., 1999) and this may affect the ability of GLP-1R agonists to suppress drug-induced dopamine release in the striatum (Fortin and Roitman, 2017; Jensen et al., 2020). In addition, the pharmacokinetic profile of exendin-4 is not as well characterized in mice as in rats (Chen et al., 2013).

    View all citing articles on Scopus
    View full text