Availability of Central α4β2* Nicotinic Acetylcholine Receptors in Human Obesity

Purpose: Obesity is thought to arise, in part, from deficits in the inhibitory control over appetitive behavior. Such motivational processes are regulated by neuromodulators, specifically acetylcholine (ACh), via α4β2* nicotinic ACh receptors (nAChR). These nAChR are highly enriched in the thalamus and contribute to the thalamic gating of cortico-striatal signaling, but also act on the mesoaccumbal reward system. The changes in α4β2* nAChR availability, however, have not been demonstrated in human obesity thus far. The aim of our study was, thus, to investigate whether there is altered brain α4β2* nAChR availability in individuals with obesity compared to normal-weight healthy controls. Methods: We studied 15 non-smoking individuals with obesity (body mass index, BMI: 37.8 ± 3.1 kg/m2; age: 39 ± 14 years, 9 females) and 16 normal-weight controls (non-smokers, BMI: 21.9 ± 1.7 kg/m2; age: 28 ± 7 years, 13 females) by using PET and the α4β2* nAChR selective (−)-[18F]flubatine, which was applied within a bolus-infusion protocol (294 ± 16 MBq). Volume-of-interest (VOI) analysis was performed in order to calculate the regional total distribution volume (VT). Results: No overall significant difference in VT between the individuals with obesity and the normal-weight volunteers was found, while the VT in the nucleus basalis of Meynert tended to be lower in the individuals with obesity (10.1 ± 2.1 versus 11.9 ± 2.2; p = 0.10), and the VT in the thalamus showed a tendency towards higher values in the individuals with obesity (26.5 ± 2.5 versus 25.9 ± 4.2; p = 0.09). Conclusion: While these first data do not show greater brain α4β2* nAChR availability in human obesity overall, the findings of potentially aberrant α4β2* nAChR availability in the key brain regions that regulate feeding behavior merit further exploration.

[1]  Lieneke K. Janssen,et al.  Molecular Imaging of Central Dopamine in Obesity: A Qualitative Review across Substrates and Radiotracers , 2022, Brain sciences.

[2]  Sabine Kastner,et al.  Thalamic functions in distributed cognitive control , 2017, Nature Neuroscience.

[3]  K. Lutfy,et al.  Effects of nicotine on homeostatic and hedonic components of food intake. , 2017, The Journal of endocrinology.

[4]  Ralf D. Wimmer,et al.  Thalamic amplification of cortical connectivity sustains attentional control , 2017, Nature.

[5]  Laszlo Zaborszky,et al.  The Input-Output Relationship of the Cholinergic Basal Forebrain. , 2017, Cell reports.

[6]  R. E. Carson,et al.  Imaging of cerebral α4β2* nicotinic acetylcholine receptors with (−)-[18F]Flubatine PET: Implementation of bolus plus constant infusion and sensitivity to acetylcholine in human brain , 2016, NeuroImage.

[7]  Alexander M. Herman,et al.  A cholinergic basal forebrain feeding circuit modulates appetite suppression , 2016, Nature.

[8]  S. Ostlund,et al.  Nucleus Accumbens Acetylcholine Receptors Modulate Dopamine and Motivation , 2016, Neuropsychopharmacology.

[9]  Gudrun Wagenknecht,et al.  First-in-human PET quantification study of cerebral α4β2* nicotinic acetylcholine receptors using the novel specific radioligand (−)-[18F]Flubatine , 2015, NeuroImage.

[10]  T. Brandon,et al.  Smoking as alternative to eating among restrained eaters: effect of food prime on young adult female smokers. , 2014, Health psychology : official journal of the Division of Health Psychology, American Psychological Association.

[11]  M. Smolka,et al.  Nicotine administration in healthy non‐smokers reduces appetite but does not alter plasma ghrelin , 2014, Human psychopharmacology.

[12]  M. Mesulam,et al.  Cholinergic circuitry of the human nucleus basalis and its fate in Alzheimer's disease , 2013, The Journal of comparative neurology.

[13]  Gudrun Wagenknecht,et al.  Fully automated radiosynthesis of both enantiomers of [18F]Flubatine under GMP conditions for human application. , 2013, Applied radiation and isotopes : including data, instrumentation and methods for use in agriculture, industry and medicine.

[14]  Sepideh Sadaghiani,et al.  High density of nicotinic receptors in the cingulo-insular network , 2013, NeuroImage.

[15]  T. Robinson,et al.  Cholinergic Control over Attention in Rats Prone to Attribute Incentive Salience to Reward Cues , 2013, The Journal of Neuroscience.

[16]  M. Picciotto,et al.  Nicotinic regulation of energy homeostasis. , 2012, Nicotine & tobacco research : official journal of the Society for Research on Nicotine and Tobacco.

[17]  M. Baxter,et al.  Cholinergic modulation of a specific memory function of prefrontal cortex , 2011, Nature Neuroscience.

[18]  R. Dolan,et al.  Cholinergic modulation of cognition: Insights from human pharmacological functional neuroimaging , 2011, Progress in neurobiology.

[19]  R. Seeley,et al.  Neuroscience: Weight loss through smoking , 2011, Nature.

[20]  Yan Rao,et al.  Nicotine Decreases Food Intake Through Activation of POMC Neurons , 2011, Science.

[21]  A. N. van den Pol,et al.  Nicotine excites hypothalamic arcuate anorexigenic proopiomelanocortin neurons and orexigenic neuropeptide Y neurons: similarities and differences. , 2011, Journal of neurophysiology.

[22]  Ciprian Catana,et al.  Toward Implementing an MRI-Based PET Attenuation-Correction Method for Neurologic Studies on the MR-PET Brain Prototype , 2010, The Journal of Nuclear Medicine.

[23]  D. James Surmeier,et al.  Thalamic Gating of Corticostriatal Signaling by Cholinergic Interneurons , 2010, Neuron.

[24]  L. Carrozzi,et al.  Weight gain after smoking cessation. , 2009, Monaldi archives for chest disease = Archivio Monaldi per le malattie del torace.

[25]  D. Bessesen Update on obesity. , 2008, The Journal of clinical endocrinology and metabolism.

[26]  Marina R. Picciotto,et al.  It is not “either/or”: Activation and desensitization of nicotinic acetylcholine receptors both contribute to behaviors related to nicotine addiction and mood , 2008, Progress in Neurobiology.

[27]  A. Lajtha,et al.  Food Reward-Induced Neurotransmitter Changes in Cognitive Brain Regions , 2007, Neurochemical Research.

[28]  D. Bertrand,et al.  Nicotinic acetylcholine receptors and nicotinic cholinergic mechanisms of the central nervous system. , 2007, Annual review of pharmacology and toxicology.

[29]  J. Verbalis,et al.  CNS Site of Action and Brainstem Circuitry Responsible for the Intravenous Effects of Nicotine on Gastric Tone , 2002, The Journal of Neuroscience.

[30]  D. Bertrand,et al.  Chronic Exposure to Nicotine Upregulates the Human α4β2 Nicotinic Acetylcholine Receptor Function , 2001, The Journal of Neuroscience.

[31]  William D. Dupont,et al.  Power and sample size calculations. A review and computer program. , 1990, Controlled clinical trials.

[32]  M. McNamee,et al.  Desensitization of the nicotinic acetylcholine receptor: Molecular mechanisms and effect of modulators , 1989, Cellular and Molecular Neurobiology.

[33]  M. Picciotto,et al.  Molecular mechanisms underlying behaviors related to nicotine addiction. , 2013, Cold Spring Harbor perspectives in medicine.

[34]  A. Stunkard,et al.  The three-factor eating questionnaire to measure dietary restraint, disinhibition and hunger. , 1985, Journal of psychosomatic research.