1
|
Laule C, Sayar-Atasoy N, Aklan I, Kim H, Ates T, Davis D, Atasoy D. Stress integration by an ascending adrenergic-melanocortin circuit. Neuropsychopharmacology 2024; 49:1361-1372. [PMID: 38326456 PMCID: PMC11251172 DOI: 10.1038/s41386-024-01810-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
Stress is thought to be an important contributing factor for eating disorders; however, neural substrates underlying the complex relationship between stress and appetite are not fully understood. Using in vivo recordings from awake behaving mice, we show that various acute stressors activate catecholaminergic nucleus tractus solitarius (NTSTH) projections in the paraventricular hypothalamus (PVH). Remarkably, the resulting adrenergic tone inhibits MC4R-expressing neurons (PVHMC4R), which are known for their role in feeding suppression. We found that PVHMC4R silencing encodes negative valence in sated mice and is required for avoidance induced by visceral malaise. Collectively, these findings establish PVHMC4R neurons as an effector of stress-activated brainstem adrenergic input in addition to the well-established hypothalamic-pituitary-adrenal axis. Convergent modulation of stress and feeding by PVHMC4R neurons implicates NTSTH → PVHMC4R input in stress-associated appetite disorders.
Collapse
Affiliation(s)
- Connor Laule
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nilufer Sayar-Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Iltan Aklan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hyojin Kim
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Tayfun Ates
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Debbie Davis
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
2
|
Tackett WR, Yalakala J, Hambuchen MD. Co-administration of naloxone and dexmedetomidine to simultaneously reverse acute effects of fentanyl and methamphetamine in rats. Drug Alcohol Depend 2024; 259:111301. [PMID: 38640863 DOI: 10.1016/j.drugalcdep.2024.111301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND The incidence of combination methamphetamine (METH)-opioid overdose has substantially increased in recent years. While agitation is uncommon after the naloxone (NLX) reversal of opioids, it is a major clinical concern in acute METH intoxication and can be physiologically antagonized by opioid-induced sedation. This study aimed to perform initial preclinical analysis of the safety and efficacy of dexmedetomidine (DEXMED) co-administered with NLX to attenuate METH-induced locomotor activity, as a rat model of agitation, after the reversal of fentanyl (FENT)-induced sedation. METHODS Male Sprague Dawley rats were administered subcutaneous (SC) 0.1mg/kg FENT ± 1mg/kg METH. Fifteen min later, SC 0.1mg/kg NLX ± an increasing (0, 0.032, 0.056, and 0.1mg/kg) DEXMED dose was administered prior to the measurement of locomotor activity. After a washout period, the FENT ± METH and NLX ± DEXMED administration with the highest dose of DEXMED was administered for measurement of blood oxygen saturation and heart rate. RESULTS After the NLX reversal of FENT-induced sedation, adjunct DEXMED substantially and significantly reduced METH-induced locomotor activity (p<0.05) at all doses tested. While the addition of DEXMED did not significantly reduce blood oxygenation in METH treated rats, it did so in the absence of METH. Also, DEXMED significantly reduced heart rate compared to non-DEXMED treated groups and resulted in further significant reductions in the animals not exposed to METH (p<0.05). CONCLUSIONS These data provide preclinical evidence that DEXMED may be a safe and effective chemical restraint for METH-induced agitation after NLX opioid reversal.
Collapse
Affiliation(s)
- Wesley R Tackett
- Department of Pharmaceutical Science, Marshall University School of Pharmacy, Stephen J. Kopp Hall 353, 1 John Marshall Drive, Huntington, WV 25755, USA
| | - Jyostna Yalakala
- Department of Pharmaceutical Science, Marshall University School of Pharmacy, Stephen J. Kopp Hall 353, 1 John Marshall Drive, Huntington, WV 25755, USA
| | - Michael D Hambuchen
- Department of Pharmaceutical Science, Marshall University School of Pharmacy, Stephen J. Kopp Hall 353, 1 John Marshall Drive, Huntington, WV 25755, USA.
| |
Collapse
|
3
|
Sayar-Atasoy N, Laule C, Aklan I, Kim H, Yavuz Y, Ates T, Coban I, Koksalar-Alkan F, Rysted J, Davis D, Singh U, Alp MI, Yilmaz B, Cui H, Atasoy D. Adrenergic modulation of melanocortin pathway by hunger signals. Nat Commun 2023; 14:6602. [PMID: 37857606 PMCID: PMC10587058 DOI: 10.1038/s41467-023-42362-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023] Open
Abstract
Norepinephrine (NE) is a well-known appetite regulator, and the nor/adrenergic system is targeted by several anti-obesity drugs. To better understand the circuitry underlying adrenergic appetite control, here we investigated the paraventricular hypothalamic nucleus (PVN), a key brain region that integrates energy signals and receives dense nor/adrenergic input, using a mouse model. We found that PVN NE level increases with signals of energy deficit and decreases with food access. This pattern is recapitulated by the innervating catecholaminergic axon terminals originating from NTSTH-neurons. Optogenetic activation of rostral-NTSTH → PVN projection elicited strong motivation to eat comparable to overnight fasting whereas its inhibition attenuated both fasting-induced & hypoglycemic feeding. We found that NTSTH-axons functionally targeted PVNMC4R-neurons by predominantly inhibiting them, in part, through α1-AR mediated potentiation of GABA release from ARCAgRP presynaptic terminals. Furthermore, glucoprivation suppressed PVNMC4R activity, which was required for hypoglycemic feeding response. These results define an ascending nor/adrenergic circuit, NTSTH → PVNMC4R, that conveys peripheral hunger signals to melanocortin pathway.
Collapse
Affiliation(s)
- Nilufer Sayar-Atasoy
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Connor Laule
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Iltan Aklan
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hyojin Kim
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yavuz Yavuz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Tayfun Ates
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ilknur Coban
- Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | | | - Jacob Rysted
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Debbie Davis
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Uday Singh
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Muhammed Ikbal Alp
- Department of Physiology, School of Medicine, Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Bayram Yilmaz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Huxing Cui
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Deniz Atasoy
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
4
|
Wartko PD, Weiss NS, Enquobahrie DA, Chan KCG, Stephenson-Famy A, Mueller BA, Dublin S. Maternal Gestational Weight Gain in Relation to Antidepressant Continuation in Pregnancy. Am J Perinatol 2021; 38:1442-1452. [PMID: 32604448 PMCID: PMC8487259 DOI: 10.1055/s-0040-1713652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Both excessive and inadequate gestational weight gain (GWG) are associated with adverse health outcomes for the woman and her child. Antidepressant use in pregnancy could affect GWG, based on evidence in nonpregnant women that some antidepressants may cause weight gain and others weight loss. Previous studies of antidepressant use and GWG were small with limited ability to account for confounding, including by maternal mental health status and severity. We assessed the association of antidepressant continuation in pregnancy with GWG among women using antidepressants before pregnancy. STUDY DESIGN Our retrospective cohort study included singleton livebirths from 2001 to 2014 within Kaiser Permanente Washington, an integrated health care system. Data were obtained from electronic health records and linked Washington State birth records. Among women with ≥1 antidepressant fill within 6 months before pregnancy, women who filled an antidepressant during pregnancy were considered "continuers;" women without a fill were "discontinuers." We calculated mean differences in GWG and relative risks (RR) of inadequate and excessive weight gain based on Institute of Medicine guidelines. Using inverse probability of treatment weighting with generalized estimating equations, we addressed differences in maternal characteristics, including mental health conditions. RESULTS Among the 2,887 births, 1,689 (59%) were to women who continued antidepressants in pregnancy and 1,198 (42%) were to discontinuers. After accounting for confounding, continuers had similar weight gain to those who discontinued (mean difference: 1.3 lbs, 95% confidence interval [CI]: -0.1 to 2.8 lbs) and similar risks of inadequate and excessive GWG (RR: 0.95, 95% CI: 0.80-1.14 and RR: 1.06, 95% CI: 0.98-1.14, respectively). Findings were comparable for specific antidepressants and trimesters of exposure. CONCLUSION We did not find evidence that continuation of antidepressants in pregnancy led to differences in GWG. KEY POINTS · Antidepressant use is associated with weight change in nonpregnant populations.. · Prior evidence on whether antidepressant use in pregnancy affects gestational weight gain is sparse.. · We accounted for confounding by characteristics such as mental health conditions and their severity.. · We found no association between pregnancy antidepressant continuation and gestational weight gain..
Collapse
Affiliation(s)
- Paige D Wartko
- Department of Epidemiology, University of Washington,
Seattle, Washington.,Kaiser Permanente Washington Health Research Institute,
Seattle, Washington
| | - Noel S Weiss
- Department of Epidemiology, University of Washington,
Seattle, Washington.,Public Health Sciences Division, Fred Hutchinson Cancer
Research Center, Seattle, WA, USA
| | | | - KC Gary Chan
- Department of Biostatistics, University of Washington,
Seattle, Washington
| | | | - Beth A Mueller
- Department of Epidemiology, University of Washington,
Seattle, Washington.,Public Health Sciences Division, Fred Hutchinson Cancer
Research Center, Seattle, WA, USA
| | - Sascha Dublin
- Department of Epidemiology, University of Washington,
Seattle, Washington.,Kaiser Permanente Washington Health Research Institute,
Seattle, Washington
| |
Collapse
|
5
|
Pruccoli J, Parmeggiani A, Cordelli DM, Lanari M. The Role of the Noradrenergic System in Eating Disorders: A Systematic Review. Int J Mol Sci 2021; 22:11086. [PMID: 34681746 PMCID: PMC8537146 DOI: 10.3390/ijms222011086] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 01/30/2023] Open
Abstract
Noradrenaline (NE) is a catecholamine acting as both a neurotransmitter and a hormone, with relevant effects in modulating feeding behavior and satiety. Several studies have assessed the relationship between the noradrenergic system and Eating Disorders (EDs). This systematic review aims to report the existing literature on the role of the noradrenergic system in the development and treatment of EDs. A total of 35 studies were included. Preclinical studies demonstrated an involvement of the noradrenergic pathways in binge-like behaviors. Genetic studies on polymorphisms in genes coding for NE transporters and regulating enzymes have shown conflicting evidence. Clinical studies have reported non-unanimous evidence for the existence of absolute alterations in plasma NE values in patients with Anorexia Nervosa (AN) and Bulimia Nervosa (BN). Pharmacological studies have documented the efficacy of noradrenaline-modulating therapies in the treatment of BN and Binge Eating Disorder (BED). Insufficient evidence was found concerning the noradrenergic-mediated genetics of BED and BN, and psychopharmacological treatments targeting the noradrenergic system in AN. According to these data, further studies are required to expand the existing knowledge on the noradrenergic system as a potential target for treatments of EDs.
Collapse
Affiliation(s)
- Jacopo Pruccoli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Centro Regionale per i Disturbi della Nutrizione e dell’Alimentazione in età Evolutiva, U.O. Neuropsichiatria dell’età Pediatrica, 40138 Bologna, Italy; (A.P.); (D.M.C.)
- Dipartimento di Scienze Mediche e Chirurgiche (DIMEC), Università di Bologna, 40138 Bologna, Italy;
| | - Antonia Parmeggiani
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Centro Regionale per i Disturbi della Nutrizione e dell’Alimentazione in età Evolutiva, U.O. Neuropsichiatria dell’età Pediatrica, 40138 Bologna, Italy; (A.P.); (D.M.C.)
- Dipartimento di Scienze Mediche e Chirurgiche (DIMEC), Università di Bologna, 40138 Bologna, Italy;
| | - Duccio Maria Cordelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Centro Regionale per i Disturbi della Nutrizione e dell’Alimentazione in età Evolutiva, U.O. Neuropsichiatria dell’età Pediatrica, 40138 Bologna, Italy; (A.P.); (D.M.C.)
- Dipartimento di Scienze Mediche e Chirurgiche (DIMEC), Università di Bologna, 40138 Bologna, Italy;
| | - Marcello Lanari
- Dipartimento di Scienze Mediche e Chirurgiche (DIMEC), Università di Bologna, 40138 Bologna, Italy;
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico di S. Orsola, U.O. Pediatria d’urgenza, Pronto Soccorso Pediatrico e OBI, 40138 Bologna, Italy
| |
Collapse
|
6
|
Cerda IH, Fitzgerald PJ. An electrophysiological model of major depression: Relevance to clinical subtyping and pharmacological management. Psychiatry Res 2021; 303:114054. [PMID: 34153629 DOI: 10.1016/j.psychres.2021.114054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 06/02/2021] [Indexed: 11/24/2022]
Abstract
We present a neurochemical model of unipolar major depressive disorder that makes predictions for optimizing pharmacological treatment of this debilitating neuropsychiatric disorder. We suggest that there are two principal electrophysiological subtypes of depression, with the more common one involving a high excitatory/inhibitory (E/I) electrophysiological ratio, and a less common low E/I subtype. The high E/I subtype is paradoxically a variant of previous conceptions of atypical depression, whereas the low E/I subtype is a variant of melancholic depression. We focus on the ratio of norepinephrine (NE) to serotonin (5HT) as primary determinants of E/I ratio, which have opposing effects on mood regulation. We suggest that high NE/5HT (or E/I) ratio depressions should be treated with pharmacological agents that boost 5HT (such as SSRIs) and/or drugs that reduce noradrenergic transmission (such as clonidine, guanfacine, propranolol, prazosin). In contrast, low NE/5HT (or E/I) depressions should be treated with agents that boost NE (such as most tricyclics) and/or drugs that reduce serotonergic transmission. Our model predicts that the rapidly acting antidepressant ketamine (and possibly scopolamine), which has an acutely excitatory electrophysiological profile that may be followed by sustained increased inhibition, should improve the high NE/5HT subtype and worsen the low subtype.
Collapse
Affiliation(s)
- Ivo H Cerda
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109, USA.
| | - Paul J Fitzgerald
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109, USA.
| |
Collapse
|
7
|
Apóstol Del Rosal GD, Limón ID, Martínez I, Patricio-Martínez A. The Chronic Oral Administration of Clobenzorex or Amphetamine Decreases Motor Behavior and Induces Glial Activation in the Striatum Without Dopaminergic Degeneration. Neurotox Res 2021; 39:1405-1417. [PMID: 34279823 DOI: 10.1007/s12640-021-00395-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/22/2021] [Accepted: 07/12/2021] [Indexed: 01/12/2023]
Abstract
Described as amphetamine-like due to their structural and stimulant similarities, clobenzorex is one of the five most-commonly used drugs in Mexico for the treatment of obesity. Various studies have shown that amphetamines induce dopaminergic neurotoxicity and neuroinflammation in the striatum, symptoms which are associated with motor damage. For this reason, the present study aimed to evaluate the effect of chronic clobenzorex administration on motor behaviors, TH immunoreactivity, gliosis, and the neurodegenerative process in the striatum and substantia nigra pars compacta (SNpc). The present research was conducted on three experimental groups of male Wistar rats: the vehicle group, the amphetamine group (2 mg/kg), and the clobenzorex group (30 mg/kg). All groups were subject to oral administration every 24 h for 31 days. Motor activity and motor coordination were evaluated in the open field test and the beam walking test, respectively. The animals were euthanized after the last day of treatment to enable the extraction of their brains for the evaluation of tyrosine hydroxylase (TH) levels, the immunoreactivity of the glial cells, and the neurodegeneration of both the striatum and SNpc via amino-cupric-silver stain. The results obtained show that amphetamine and clobenzorex administration decrease motor activity and motor coordination in the beam walking test and cause increased gliosis in the striatum, while no significant changes were observed in terms of immunoreactivity to TH and neurodegeneration in both the striatum and SNpc. These results suggest that the chronic administration of clobenzorex may decrease motor function in a manner similar to amphetamine, via the neuroadaptive and non-neurotoxic changes caused to the striatum under this administration scheme.
Collapse
Affiliation(s)
- Grego David Apóstol Del Rosal
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Isabel Martínez
- Laboratorio de Neuroquímica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico.
- Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Blvd. Valsequillo y Av. San Claudio Edificio BIO-1 C.U. Col. Jardines de San Manuel A.P, 72570, Puebla, Mexico.
| |
Collapse
|
8
|
Deng Y, Deng G, Grobe JL, Cui H. Hypothalamic GPCR Signaling Pathways in Cardiometabolic Control. Front Physiol 2021; 12:691226. [PMID: 34262481 PMCID: PMC8274634 DOI: 10.3389/fphys.2021.691226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/26/2021] [Indexed: 01/22/2023] Open
Abstract
Obesity is commonly associated with sympathetic overdrive, which is one of the major risk factors for the development of cardiovascular diseases, such as hypertension and heart failure. Over the past few decades, there has been a growing understanding of molecular mechanisms underlying obesity development with central origin; however, the relative contribution of these molecular changes to the regulation of cardiovascular function remains vague. A variety of G-protein coupled receptors (GPCRs) and their downstream signaling pathways activated in distinct hypothalamic neurons by different metabolic hormones, neuropeptides and monoamine neurotransmitters are crucial not only for the regulation of appetite and metabolic homeostasis but also for the sympathetic control of cardiovascular function. In this review, we will highlight the main GPCRs and associated hypothalamic nuclei that are important for both metabolic homeostasis and cardiovascular function. The potential downstream molecular mediators of these GPCRs will also be discussed.
Collapse
Affiliation(s)
- Yue Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- FOE Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- Obesity Research and Educational Initiative, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|
9
|
Yang B, Sanches-Padilla J, Kondapalli J, Morison SL, Delpire E, Awatramani R, Surmeier DJ. Locus coeruleus anchors a trisynaptic circuit controlling fear-induced suppression of feeding. Neuron 2021; 109:823-838.e6. [PMID: 33476548 PMCID: PMC9272546 DOI: 10.1016/j.neuron.2020.12.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/17/2020] [Accepted: 12/28/2020] [Indexed: 12/19/2022]
Abstract
The circuit mechanisms underlying fear-induced suppression of feeding are poorly understood. To help fill this gap, mice were fear conditioned, and the resulting changes in synaptic connectivity among the locus coeruleus (LC), the parabrachial nucleus (PBN), and the central nucleus of amygdala (CeA)-all of which are implicated in fear and feeding-were studied. LC neurons co-released noradrenaline and glutamate to excite PBN neurons and suppress feeding. LC neurons also suppressed inhibitory input to PBN neurons by inducing heterosynaptic, endocannabinoid-dependent, long-term depression of CeA synapses. Blocking or knocking down endocannabinoid receptors in CeA neurons prevented fear-induced depression of CeA synaptic transmission and fear-induced suppression of feeding. Altogether, these studies demonstrate that LC neurons play a pivotal role in modulating the circuitry that underlies fear-induced suppression of feeding, pointing to new ways of alleviating stress-induced eating disorders.
Collapse
Affiliation(s)
- Ben Yang
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Javier Sanches-Padilla
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jyothisri Kondapalli
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sage L Morison
- Department of Neurology and Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Rajeshwar Awatramani
- Department of Neurology and Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
10
|
Morales I, Berridge KC. 'Liking' and 'wanting' in eating and food reward: Brain mechanisms and clinical implications. Physiol Behav 2020; 227:113152. [PMID: 32846152 PMCID: PMC7655589 DOI: 10.1016/j.physbeh.2020.113152] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 08/17/2020] [Accepted: 08/21/2020] [Indexed: 01/02/2023]
Abstract
It is becoming clearer how neurobiological mechanisms generate 'liking' and 'wanting' components of food reward. Mesocorticolimbic mechanisms that enhance 'liking' include brain hedonic hotspots, which are specialized subregions that are uniquely able to causally amplify the hedonic impact of palatable tastes. Hedonic hotspots are found in nucleus accumbens medial shell, ventral pallidum, orbitofrontal cortex, insula cortex, and brainstem. In turn, a much larger mesocorticolimbic circuitry generates 'wanting' or incentive motivation to obtain and consume food rewards. Hedonic and motivational circuitry interact together and with hypothalamic homeostatic circuitry, allowing relevant physiological hunger and satiety states to modulate 'liking' and 'wanting' for food rewards. In some conditions such as drug addiction, 'wanting' is known to dramatically detach from 'liking' for the same reward, and this may also occur in over-eating disorders. Via incentive sensitization, 'wanting' selectively becomes higher, especially when triggered by reward cues when encountered in vulnerable states of stress, etc. Emerging evidence suggests that some cases of obesity and binge eating disorders may reflect an incentive-sensitization brain signature of cue hyper-reactivity, causing excessive 'wanting' to eat. Future findings on the neurobiological bases of 'liking' and 'wanting' can continue to improve understanding of both normal food reward and causes of clinical eating disorders.
Collapse
Affiliation(s)
- Ileana Morales
- Department of Psychology, University of Michigan, Ann Arbor, Michigan 48109-1043, United States.
| | - Kent C Berridge
- Department of Psychology, University of Michigan, Ann Arbor, Michigan 48109-1043, United States
| |
Collapse
|
11
|
Tsofliou F, Pitsiladis YP, Lara J, Hadjicharalambous M, Macdonald IA, Wallace MA, Lean MEJ. The effects of moderate alterations in adrenergic activity on acute appetite regulation in obese women: A randomised crossover trial. Nutr Health 2020; 26:311-322. [PMID: 32729763 PMCID: PMC7534026 DOI: 10.1177/0260106020942117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Previous evidence has demonstrated that serum leptin is correlated with appetite in combination with, but not without, modest exercise. AIM The present experiments investigated the effects of exogenous adrenaline and α/β adrenoceptor blockade in combination with moderate exercise on serum leptin concentrations, appetite/satiety sensations and subsequent food intake in obese women. METHODS A total of 10 obese women ((mean ± SEM), age: 50 (1.9) years, body mass index 36 (4.1) kg/m2, waist 104.8 (4.1) cm) participated in two separate, double-blind randomised experimental trials. Experiment 1: moderate exercise after α/β adrenergic blocker (labetalol, 100 mg orally) versus moderate exercise plus placebo; experiment 2: adrenaline infusion for 20 minutes versus saline infusion. Appetite/satiety and biochemistry were measured at baseline, pre- and immediately post-intervention, then 1 hour post-intervention (i.e., before dinner). Food intake was assessed via ad libitum buffet-style dinner. RESULTS No differences were found in appetite/satiety, subsequent food intake or serum leptin in any of the studies (experiment 1 or experiment 2). In experiment 1, blood glucose was higher (p < 0.01) and plasma free fatty acids lower (p = 0.04) versus placebo. In experiment 2, plasma free fatty acids (p < 0.05) increased after adrenaline versus saline infusion. CONCLUSIONS Neither inhibition of exercise-induced adrenergic activity by combined α/β adrenergic blockade nor moderate increases in adrenergic activity induced by intravenous adrenaline infusion affected acute appetite regulation.
Collapse
Affiliation(s)
- Fotini Tsofliou
- Human Nutrition, School of Medicine, University of Glasgow, United Kingdom
- Department of Rehabilitation and Sport Sciences, Faculty of Health and Social Sciences, Bournemouth University, United Kingdom
| | - Yannis P Pitsiladis
- College of Medical Veterinary and Life Science, Institute of Cardiovascular & Medical Sciences, University of Glasgow, United Kingdom
- Centre for Sport and Exercise Science and Medicine, University of Brighton, United Kingdom
| | - Jose Lara
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, United Kingdom
| | - Marios Hadjicharalambous
- College of Medical Veterinary and Life Science, Institute of Cardiovascular & Medical Sciences, University of Glasgow, United Kingdom
- Human Performance Laboratory, Department of Life & Health Sciences, School of Sciences and Engineering, University of Nicosia, Cyprus
| | - Ian A Macdonald
- School of Life Sciences, University of Nottingham Medical School, Queen’s Medical Centre, United Kingdom
| | - Mike A Wallace
- University Department of Pathological Biochemistry, Glasgow Royal Infirmary, United Kingdom
| | - Mike E J Lean
- Human Nutrition, School of Medicine, University of Glasgow, United Kingdom
| |
Collapse
|
12
|
Hicks C, Sabino V, Cottone P. The Alpha-1 Adrenergic Receptor Antagonist Prazosin Reduces Binge-Like Eating in Rats. Nutrients 2020; 12:nu12061569. [PMID: 32481494 PMCID: PMC7352795 DOI: 10.3390/nu12061569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/26/2020] [Indexed: 01/19/2023] Open
Abstract
Background: Binge-eating disorder is a pervasive addiction-like disorder that is defined by excessive and uncontrollable consumption of food within brief periods of time. The aim of the current study was to examine the role of the brain noradrenergic system in binge-like eating through the use of the alpha-1 adrenergic receptor antagonist prazosin. Methods: For this purpose, we employed a limited access model whereby male Wistar rats were allowed to nosepoke for either chow (Chow rats) or a sugary, highly palatable food (Palatable rats) for 1 h/day. The effects of prazosin (0, 0.5, 1 and 2 mg/kg, i.p.) were tested in a fixed ratio 1 (FR1) and progressive ratio (PR) schedule of reinforcement. Results: The results show that prazosin preferentially reduced the responses for palatable food in a FR1 reinforcement schedule; when tested in a PR schedule of reinforcement, prazosin increased breakpoint in both Chow and Palatable rats, but more potently and more efficaciously in the latter. Our results suggest that prazosin treatment preferentially increased the motivational properties of the palatable diet. Conclusions: The current findings provide the characterization of the effects of prazosin on binge-like eating and offer support to the existing literature showing the important role of the noradrenergic system in addiction-like behavior.
Collapse
|
13
|
Neuroprotective and Neuromodulatory Effects Induced by Cannabidiol and Cannabigerol in Rat Hypo-E22 cells and Isolated Hypothalamus. Antioxidants (Basel) 2020; 9:antiox9010071. [PMID: 31941059 PMCID: PMC7022242 DOI: 10.3390/antiox9010071] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/23/2019] [Accepted: 01/10/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cannabidiol (CBD) and cannabigerol (CBG) are non-psychotropic terpenophenols isolated from Cannabis sativa, which, besides their anti-inflammatory/antioxidant effects, are able to inhibit, the first, and to stimulate, the second, the appetite although there are no studies elucidating their role in the hypothalamic appetite-regulating network. Consequently, the aim of the present research is to investigate the role of CBD and CBG in regulating hypothalamic neuromodulators. Comparative evaluations between oxidative stress and food intake-modulating mediators were also performed. METHODS Rat hypothalamic Hypo-E22 cells and isolated tissues were exposed to either CBD or CBG, and the gene expressions of neuropeptide (NP)Y, pro-opiomelanocortin (POMC) and fatty acid amide hydrolase were assessed. In parallel, the influence of CBD on the synthesis and release of dopamine (DA), norepinephrine (NE), and serotonin (5-HT) was evaluated. The 3-hydroxykinurenine/kinurenic acid (3-HK/KA) ratio was also determined. RESULTS Both CBD and CBG inhibited NPY and POMC gene expression and decreased the 3-HK/KA ratio in the hypothalamus. The same compounds also reduced hypothalamic NE synthesis and DA release, whereas the sole CBD inhibited 5-HT synthesis. CONCLUSION The CBD modulates hypothalamic neuromodulators consistently with its anorexigenic role, whereas the CBG effect on the same mediators suggests alternative mechanisms, possibly involving peripheral pathways.
Collapse
|
14
|
McConn BR, Siegel PB, Cline MA, Gilbert ER. Anorexigenic effects of mesotocin in chicks are genetic background-dependent and are associated with changes in the paraventricular nucleus and lateral hypothalamus. Comp Biochem Physiol A Mol Integr Physiol 2019; 232:79-90. [DOI: 10.1016/j.cbpa.2019.03.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/08/2019] [Accepted: 03/12/2019] [Indexed: 11/15/2022]
|
15
|
Genetic and Neurobiological Analyses of the Noradrenergic-like System in Vulnerability to Sugar Overconsumption Using a Drosophila Model. Sci Rep 2017; 7:17642. [PMID: 29247240 PMCID: PMC5732301 DOI: 10.1038/s41598-017-17760-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 11/30/2017] [Indexed: 12/22/2022] Open
Abstract
Regular overconsumption of sugar is associated with obesity and type-2 diabetes, but how genetic factors contribute to variable sugar preferences and intake levels remains mostly unclear. Here we provide evidence for the usefulness of a Drosophila larva model to investigate genetic influence on vulnerability to sugar overconsumption. Using genetic and RNA interference approaches, we show that the activity of the Oamb gene, which encodes a receptor for octopamine (OA, the invertebrate homologue of norepinephrine), plays a major role in controlled sugar consumption. Furthermore, Oamb appears to suppress sugar food intake in fed larvae in an acute manner, and neurons expressing this Oamb receptor do not overlap with neurons expressing Octβ3R, another OA receptor previously implicated in hunger-driven exuberant sugar intake. Together, these results suggest that two separate sub-circuits, defined by Oamb and Octβ3R respectively, co-regulate sugar consumption according to changes in energy needs. We propose that the noradrenergic-like system defines an ancient regulatory mechanism for prevention of sugar overload.
Collapse
|
16
|
Recinella L, Leone S, Ferrante C, Chiavaroli A, Shohreh R, Di Nisio C, Vacca M, Orlando G, Salvatori R, Brunetti L. Effects of growth hormone-releasing hormone gene targeted ablation on ghrelin-induced feeding. Growth Horm IGF Res 2017; 37:40-46. [PMID: 29121520 DOI: 10.1016/j.ghir.2017.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/30/2022]
Abstract
Impairment of growth hormone (GH) signaling has been associated with increased feeding and adiposity. The gastric hormone ghrelin, in addition to its GH-secretagogue effects, stimulates food intake after both central and peripheral administration. In the present study we further investigated the feeding regulatory role of the ghrelin-GH axis in a mouse model of isolated GH deficiency due to targeted ablation of the GH-releasing hormone (GHRH) gene [GHRH knockout (GHRHKO)]. We evaluated the effects of intracerebroventricular ghrelin administration on feeding behavior, related hypothalamic neuropeptides and neurotransmitters, and serum ghrelin levels in mice homozygous for GHRHKO allele (-/-) and heterozygous (+/-) control animals. Vehicle-treated GHRHKO mice showed increased food intake compared to heterozygotes, associated with increased circulating ghrelin levels. Moreover, -/- mice showed elevated hypothalamic levels of neuropeptide Y (NPY), agouti-related peptide (AgRP) mRNAs and norepinephrine (NE) and decreased corticotropin-releasing hormone (CRH) mRNA levels. Ghrelin treatment significantly augmented food intake in both genotypes, but the relative increase compared to vehicle-treated animals was higher in -/- than +/- mice. In the hypothalamus, ghrelin increased AgRP and decreased CRH gene expression only in heterozygous mice, while it induced a significant reduction in proopiomelanocortin (POMC) mRNA levels in -/- mice. Ghrelin treatment also decreased hypothalamic serotonin (5-hydroxytriptamine, 5-HT) and dopamine (DA) levels in both genotypes. Additionally, we observed increased DA metabolism induced by ghrelin in both genotypes. In conclusion, dysregulation of the ghrelin-GHRH-GH axis in GHRHKO mice could lead to increased feeding secondary to elevated circulating levels of ghrelin, and the obesogenic phenotype is likely mediated by elevated NPY and AgRP, and decreased CRH gene expression in the hypothalamus.
Collapse
Affiliation(s)
- Lucia Recinella
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy
| | - Sheila Leone
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy
| | | | | | - Rugia Shohreh
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy
| | - Chiara Di Nisio
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy
| | - Michele Vacca
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy
| | - Giustino Orlando
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy.
| | - Roberto Salvatori
- Division of Endocrinology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luigi Brunetti
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy
| |
Collapse
|
17
|
Mollica A, Pelliccia S, Famiglini V, Stefanucci A, Macedonio G, Chiavaroli A, Orlando G, Brunetti L, Ferrante C, Pieretti S, Novellino E, Benyhe S, Zador F, Erdei A, Szucs E, Samavati R, Dvrorasko S, Tomboly C, Ragno R, Patsilinakos A, Silvestri R. Exploring the first Rimonabant analog-opioid peptide hybrid compound, as bivalent ligand for CB1 and opioid receptors. J Enzyme Inhib Med Chem 2017; 32:444-451. [PMID: 28097916 PMCID: PMC6009935 DOI: 10.1080/14756366.2016.1260565] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Cannabinoid (CB) and opioid systems are both involved in analgesia, food intake, mood and behavior. Due to the co-localization of µ-opioid (MOR) and CB1 receptors in various regions of the central nervous system (CNS) and their ability to form heterodimers, bivalent ligands targeting to both these systems may be good candidates to investigate the existence of possible cross-talking or synergistic effects, also at sub-effective doses. In this work, we selected from a small series of new Rimonabant analogs one CB1R reverse agonist to be conjugated to the opioid fragment Tyr-D-Ala-Gly-Phe-NH2. The bivalent compound (9) has been used for in vitro binding assays, for in vivo antinociception models and in vitro hypothalamic perfusion test, to evaluate the neurotransmitters release.
Collapse
Affiliation(s)
- Adriano Mollica
- a Dipartimento di Farmacia , Università di Chieti-Pescara "G. d'Annunzio" , Chieti , Italy
| | - Sveva Pelliccia
- b Dipartimento di Chimica e Tecnologie del Farmaco , Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza Università di Roma , Roma , Italy
| | - Valeria Famiglini
- b Dipartimento di Chimica e Tecnologie del Farmaco , Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza Università di Roma , Roma , Italy
| | - Azzurra Stefanucci
- a Dipartimento di Farmacia , Università di Chieti-Pescara "G. d'Annunzio" , Chieti , Italy
| | - Giorgia Macedonio
- a Dipartimento di Farmacia , Università di Chieti-Pescara "G. d'Annunzio" , Chieti , Italy
| | - Annalisa Chiavaroli
- a Dipartimento di Farmacia , Università di Chieti-Pescara "G. d'Annunzio" , Chieti , Italy
| | - Giustino Orlando
- a Dipartimento di Farmacia , Università di Chieti-Pescara "G. d'Annunzio" , Chieti , Italy
| | - Luigi Brunetti
- a Dipartimento di Farmacia , Università di Chieti-Pescara "G. d'Annunzio" , Chieti , Italy
| | - Claudio Ferrante
- a Dipartimento di Farmacia , Università di Chieti-Pescara "G. d'Annunzio" , Chieti , Italy
| | - Stefano Pieretti
- c Dipartimento del Farmaco , Istituto Superiore di Sanità , Rome , Italy
| | - Ettore Novellino
- d Dipartimento di Farmacia , Università di Napoli "Federico II" , Naples , Italy
| | - Sandor Benyhe
- e Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences , Szeged , Hungary
| | - Ferenc Zador
- e Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences , Szeged , Hungary
| | - Anna Erdei
- e Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences , Szeged , Hungary
| | - Edina Szucs
- e Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences , Szeged , Hungary
| | - Reza Samavati
- e Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences , Szeged , Hungary
| | - Szalbolch Dvrorasko
- e Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences , Szeged , Hungary
| | - Csaba Tomboly
- e Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences , Szeged , Hungary
| | - Rino Ragno
- f Dipartimento di Chimica e Tecnologie del Farmaco , Rome Center for Molecular Design, Sapienza Università di Roma , Roma , Italy.,g Alchemical Dynamics s.r.l , Roma , Italy
| | - Alexandros Patsilinakos
- f Dipartimento di Chimica e Tecnologie del Farmaco , Rome Center for Molecular Design, Sapienza Università di Roma , Roma , Italy.,g Alchemical Dynamics s.r.l , Roma , Italy
| | - Romano Silvestri
- b Dipartimento di Chimica e Tecnologie del Farmaco , Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza Università di Roma , Roma , Italy
| |
Collapse
|
18
|
Interaction Between Nociceptin/Orphanin FQ and Adrenergic System on Food Intake in Neonatal Chicken. Int J Pept Res Ther 2016. [DOI: 10.1007/s10989-016-9548-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
19
|
Abstract
Weight gain is a side-effect commonly associated with drugs used for headache prophylaxis. Weight gain can adversely affect patient health, exacerbate comorbid metabolic disorders and encourage noncompliance. Few studies have been conducted specifically on the effect of headache medications on weight, and it is important for physicians to have accurate information about weight-gain side-effects when identifying appropriate pharmacological regimens. This review discusses the potential effects on weight of the more common headache medications.
Collapse
Affiliation(s)
- W B Young
- Jefferson Headache Center, Department of Neurology, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA.
| | | |
Collapse
|
20
|
Brunetti L, Di Nisio C, Orlando G, Ferrante C, Vacca M. The Regulation of Feeding: A Cross Talk between Peripheral and Central Signalling. Int J Immunopathol Pharmacol 2016; 18:201-12. [PMID: 15888244 DOI: 10.1177/039463200501800203] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Feeding and energy expenditures are modulated by the interplay of hormones and neurotransmitters in the central nervous system (CNS), where the hypothalamus plays a pivotal role in the transduction of peripheral afferents into satiety and feeding signals. Aminergic neurotransmitters such as dopamine (DA), norepinephrine (NE) and serotonin (5-hydroxytryptamine, 5-HT) are historically considered to play a key role, but a number of peptides are involved in finely tuning feeding regulation. This review summarizes the current understanding of the CNS mechanisms of orexigenic peptides, such as neuropeptide Y, orexins, and ghrelin, as well as anorectic peptides, such as leptin, neurotensin (NT), cocaine- and amphetamine regulated transcript (CART) peptide, thyrotropin-releasing hormone (TRH), corticotropin-releasing hormone (CRH), urocortin, amylin.
Collapse
Affiliation(s)
- L Brunetti
- Department of Drug Sciences, G. D'Annunzio University, Chieti, Italy
| | | | | | | | | |
Collapse
|
21
|
Brierley DI, Samuels J, Duncan M, Whalley BJ, Williams CM. Cannabigerol is a novel, well-tolerated appetite stimulant in pre-satiated rats. Psychopharmacology (Berl) 2016; 233:3603-13. [PMID: 27503475 PMCID: PMC5021742 DOI: 10.1007/s00213-016-4397-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/19/2016] [Indexed: 11/26/2022]
Abstract
RATIONALE The appetite-stimulating properties of cannabis are well documented and have been predominantly attributed to the hyperphagic activity of the psychoactive phytocannabinoid, ∆(9)-tetrahydrocannabinol (∆(9)-THC). However, we have previously shown that a cannabis extract devoid of ∆(9)-THC still stimulates appetite, indicating that other phytocannabinoids also elicit hyperphagia. One possible candidate is the non-psychoactive phytocannabinoid cannabigerol (CBG), which has affinity for several molecular targets with known involvement in the regulation of feeding behaviour. OBJECTIVES The objective of the study was to assess the effects of CBG on food intake and feeding pattern microstructure. METHODS Male Lister hooded rats were administered CBG (30-120 mg/kg, per ora (p.o.)) or placebo and assessed in open field, static beam and grip strength tests to determine a neuromotor tolerability profile for this cannabinoid. Subsequently, CBG (at 30-240 mg/kg, p.o.) or placebo was administered to a further group of pre-satiated rats, and hourly intake and meal pattern data were recorded over 2 h. RESULTS CBG produced no adverse effects on any parameter in the neuromotor tolerability test battery. In the feeding assay, 120-240 mg/kg CBG more than doubled total food intake and increased the number of meals consumed, and at 240 mg/kg reduced latency to feed. However, the sizes or durations of individual meals were not significantly increased. CONCLUSIONS Here, we demonstrate for the first time that CBG elicits hyperphagia, by reducing latency to feed and increasing meal frequency, without producing negative neuromotor side effects. Investigation of the therapeutic potential of CBG for conditions such as cachexia and other disorders of eating and body weight regulation is thus warranted.
Collapse
Affiliation(s)
- Daniel I Brierley
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
- School of Chemistry, Food and Pharmacy, University of Reading, Reading, UK
| | - James Samuels
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| | | | - Benjamin J Whalley
- School of Chemistry, Food and Pharmacy, University of Reading, Reading, UK
| | - Claire M Williams
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK.
| |
Collapse
|
22
|
|
23
|
Eriksson A, Williams MJ, Voisin S, Hansson I, Krishnan A, Philippot G, Yamskova O, Herisson FM, Dnyansagar R, Moschonis G, Manios Y, Chrousos GP, Olszewski PK, Frediksson R, Schiöth HB. Implication of coronin 7 in body weight regulation in humans, mice and flies. BMC Neurosci 2015; 16:13. [PMID: 25887538 PMCID: PMC4364644 DOI: 10.1186/s12868-015-0151-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/25/2015] [Indexed: 12/22/2022] Open
Abstract
Background Obesity is a growing global concern with strong associations with cardiovascular disease, cancer and type-2 diabetes. Although various genome-wide association studies have identified more than 40 genes associated with obesity, these genes cannot fully explain the heritability of obesity, suggesting there may be other contributing factors, including epigenetic effects. Results We performed genome wide DNA methylation profiling comparing normal-weight and obese 9–13 year old children to investigate possible epigenetic changes correlated with obesity. Of note, obese children had significantly lower methylation levels at a CpG site located near coronin 7 (CORO7), which encodes a tryptophan-aspartic acid dipeptide (WD)-repeat containing protein most likely involved in Golgi complex morphology and function. Anatomical profiling of coronin 7 (Coro7) mRNA expression in mice revealed that it is highly expressed in appetite and energy balance regulating regions, including the hypothalamus, striatum and locus coeruleus, the main noradrenergic brain site. Interestingly, we found that food deprivation in mice downregulates hypothalamic Coro7 mRNA levels, and injecting ethanol, an appetite stimulant, increased the number of Coro7 expressing cells in the locus coeruleus. Finally, by employing the genetically-tractable Drosophila melanogaster model we were able to demonstrate an evolutionarily conserved metabolic function for the CORO7 homologue pod1. Knocking down the pod1 in the Drosophila adult nervous system increased their resistance to starvation. Furthermore, feeding flies a high-calorie diet significantly increased pod1 expression. Conclusion We conclude that coronin 7 is involved in the regulation of energy homeostasis and this role stems, to some degree, from the effect on feeding for calories and reward.
Collapse
Affiliation(s)
- Anders Eriksson
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Husargatan 3, Box 593, Uppsala, 75 124, Sweden.
| | - Michael J Williams
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Husargatan 3, Box 593, Uppsala, 75 124, Sweden.
| | - Sarah Voisin
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Husargatan 3, Box 593, Uppsala, 75 124, Sweden.
| | - Ida Hansson
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Husargatan 3, Box 593, Uppsala, 75 124, Sweden.
| | - Arunkumar Krishnan
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Husargatan 3, Box 593, Uppsala, 75 124, Sweden.
| | - Gaetan Philippot
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Husargatan 3, Box 593, Uppsala, 75 124, Sweden.
| | - Olga Yamskova
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Husargatan 3, Box 593, Uppsala, 75 124, Sweden.
| | - Florence M Herisson
- Department of Biological Sciences, University of Waikato, Hamilton, New Zealand.
| | - Rohit Dnyansagar
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Husargatan 3, Box 593, Uppsala, 75 124, Sweden.
| | - George Moschonis
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece.
| | - Yannis Manios
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece.
| | - George P Chrousos
- First Department of Pediatrics, Aghia Sophia Children's Hospital, Athens University Medical School, Athens, Greece.
| | - Pawel K Olszewski
- Department of Biological Sciences, University of Waikato, Hamilton, New Zealand.
| | - Robert Frediksson
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Husargatan 3, Box 593, Uppsala, 75 124, Sweden.
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Husargatan 3, Box 593, Uppsala, 75 124, Sweden.
| |
Collapse
|
24
|
Ho JM, Anekonda VT, Thompson BW, Zhu M, Curry RW, Hwang BH, Morton GJ, Schwartz MW, Baskin DG, Appleyard SM, Blevins JE. Hindbrain oxytocin receptors contribute to the effects of circulating oxytocin on food intake in male rats. Endocrinology 2014; 155:2845-57. [PMID: 24877632 PMCID: PMC4098005 DOI: 10.1210/en.2014-1148] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oxytocin (OT)-elicited hypophagia has been linked to neural activity in the nucleus of the solitary tract (NTS). Because plasma OT levels increase after a meal, we hypothesized that circulating OT acts at both peripheral and hindbrain OT receptors (OTRs) to limit food intake. To initially determine whether circulating OT inhibits food intake by acting at hindbrain OTRs, we pretreated rats with an OTR antagonist administered into the fourth ventricle (4V) followed by either central or systemic OT administration. Administration of the OTR antagonist into the 4V blocked anorexia induced by either 4V or i.p. injection of OT. However, blockade of peripheral OTRs also weakened the anorectic response to ip OT. Our data suggest a predominant role for hindbrain OTRs in the hypophagic response to peripheral OT administration. To elucidate central mechanisms of OT hypophagia, we tested whether OT activates NTS catecholaminergic neurons. OT (ip) increased the number of NTS cells expressing c-Fos, of which 10%-15% were catecholaminergic. Furthermore, electrophysiological studies in mice revealed that OT stimulated 47% (8 of 17) of NTS catecholamine neurons through a presynaptic mechanism. However, OT-elicited hypophagia did not appear to require activation of α1-adrenoceptors, and blockade of glucagon-like peptide-1 receptors similarly did not attenuate anorexia induced by OT. These findings demonstrate that OT elicits satiety through both central and peripheral OTRs and that although catecholamine neurons are a downstream target of OT signaling in the NTS, the hypophagic effect is mediated independently of α1-adrenoceptor signaling.
Collapse
Affiliation(s)
- Jacqueline M Ho
- Research and Development Service (J.M.H., V.T.A., B.W.T., R.W.C., B.H.H., D.G.B., J.E.B.), Veterans Affairs Puget Sound Health Care System, Seattle, Washington 98108; Department of Medicine (J.M.H., G.J.M., M.W.S., D.G.B., J.E.B.), Division of Metabolism, Endocrinology, and Nutrition, and Diabetes and Obesity Center of Excellence (G.J.M., M.W.S.), Department of Medicine, University of Washington, Seattle, Washington 98195; Program in Neuroscience (M.Z., S.M.A.), Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington 99164
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Catoira NP, Viale L, Di Girolamo G, Gonzalez C. New centrally acting agents for appetite control: from biological mechanisms to clinical efficacy. Curr Med Res Opin 2014; 30:961-9. [PMID: 24432840 DOI: 10.1185/03007995.2014.884494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Obesity is one of the major problems of health policy in different countries. Pharmacological attempts have been made to help affected people without a definitive solution. Some agents--either with peripheral or central effect--are available in the market. On July 2012, the FDA approved two novel preparations for obese patients: (1) topiramate-phentermine--the first one an anticonvulsant and the second one a sympathomimetic amine--and (2) lorcaserin, a 5-HT2CR agonist. Both preparations emerged as new options for weight management. SCOPE Based on the complex biology of eating behavior, in this review we discuss the features, mechanisms of action, pharmacokinetics, advantages and possible disadvantages of these new agents. CONCLUSION With differences in efficacy (higher for the topiramate-phentermine combination), both preparations are active in reducing appetite and body weight, as well as in improving comorbidities. Additional information will be collected from Phase IV surveillance. Focus on cardiovascular, neuropsychiatric (for both introductions) and embrio-fetal safety (especially for topiramate) is expected.
Collapse
Affiliation(s)
- Natalia Paola Catoira
- Hospital Enrique Tornú, Department of Pharmacology, Universidad de Buenos Aires, Buenos Aires , Argentina
| | | | | | | |
Collapse
|
26
|
Brunetti L, Chiavaroli A, Cocco A, Ferrante C, Ferrucci A, Luisi G, Orlando G, Pinnen F, Vacca M. Synthesis and neuromodulatory effects of TRH-related peptides: inhibitory activity on catecholamine release in vitro. Pharmacol Rep 2013; 65:823-35. [PMID: 24145076 DOI: 10.1016/s1734-1140(13)71063-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 02/16/2013] [Indexed: 11/27/2022]
Abstract
BACKGROUND A detailed comprehension of central mechanisms underlying feeding behavior holds considerable promise for the treatment of alimentary disorders. METHODS In order to elucidate the tight interrelationships occurring at the hypothalamic neuronal endings between aminergic neurotransmitters and co-localized appetite modulators, we initially studied the effects of two anorexigenic peptides structurally related to thyrotropin-releasing hormone (TRH, 1), namely cyclo(His-Pro) (CHP, 2) and pGlu-His-Gly-OH (3), on [(3)H]-norepinephrine and [(3)H]-dopamine release from perfused rat hypothalamic synaptosomes. Furthermore, a number of TRH and CHP analogues were synthesized and tested for their ability to influence neurotransmitter release in the selected neuronal model. RESULTS Peptide 3 showed only a slight inhibitory activity on norepinephrine release, whereas no effect was observed for compound 2. TRH analogue 8, metabolically stabilized by the replacement of pyroglutamate with the pyrohomocysteic acid (pHcs), was found to be inactive. Conversely, a significant inhibitory effect on dopamine and norepinephrine release was observed for the CHP-related diketopiperazines cyclo(Leu-Pro) (11) and cyclo(His-Gly) (14). CONCLUSIONS These results suggest a potential role for cyclo-dipeptides 11 and 14 in the hypothalamic modulation of appetite suppressant circuitry.
Collapse
Affiliation(s)
- Luigi Brunetti
- Department of Pharmacy, G. d'Annunzio University, via dei Vestini 31, 66013 Chieti, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Brunetti L, Ferrante C, Orlando G, Recinella L, Leone S, Chiavaroli A, Di Nisio C, Shohreh R, Manippa F, Ricciuti A, Mollica A, Vacca M. Orexigenic effects of endomorphin-2 (EM-2) related to decreased CRH gene expression and increased dopamine and norepinephrine activity in the hypothalamus. Peptides 2013; 48:83-8. [PMID: 23916912 DOI: 10.1016/j.peptides.2013.07.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 07/25/2013] [Accepted: 07/25/2013] [Indexed: 10/26/2022]
Abstract
Endomorphin-1 (EM-1) and endomorphin-2 (EM-2) are opioid peptides which are selective partial agonists of μ-opioid receptor. We studied the effects of EM-2 injected into the arcuate nucleus (ARC) of the hypothalamus on feeding behavior and gene expression of orexigenic [agouti-related peptide (AgRP), neuropeptide Y (NPY) and orexin-A] and anorexigenic [cocaine and amphetamine-regulated transcript (CART), corticotrophin releasing hormone (CRH) and proopiomelanocortin (POMC)] peptides in male Wistar rats fed a standard laboratory diet. Furthermore, we evaluated the effects of EM-2 on dopamine (DA), norepinephrine (NE) and serotonin (5-hydroxytryptamine, 5-HT) steady state concentrations, in the hypothalamus. 64 rats (16 for each group of treatment) were injected into the ARC, at 9.00 am, with either vehicle or EM-2 (0.50-0.75 μmol/kg) or EM-2 (0.50 μmol/kg) plus β-funaltrexamine (0.20 μmol/kg). Food intake was recorded through 24h following injection, and hypothalamic DA, NE, 5-HT levels and neuropeptide gene expression were evaluated 24h after EM-2 administration. Compared to vehicle, EM-2 significantly increased food intake, throughout 24h post-injection. Furthermore, EM-2 treatment led to a significant increase of DA and NE concentrations and a decrease of CRH mRNA levels. On the other hand, β-funaltrexamine administration reverted both feeding stimulatory and neuromodulatory effects induced by EM-2. We can conclude that the orexigenic effect of μ-opioid receptor activation by EM-2 could be related to both inhibition of CRH and stimulation of dopamine and norepinephrine levels, in the hypothalamus.
Collapse
Affiliation(s)
- Luigi Brunetti
- Department of Pharmacy, "G. d'Annunzio" University, via dei Vestini 31, 66013 Chieti, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Brunetti L, Orlando G, Ferrante C, Recinella L, Leone S, Chiavaroli A, Di Nisio C, Shohreh R, Manippa F, Ricciuti A, Vacca M. Orexigenic effects of omentin-1 related to decreased CART and CRH gene expression and increased norepinephrine synthesis and release in the hypothalamus. Peptides 2013; 44:66-74. [PMID: 23538212 DOI: 10.1016/j.peptides.2013.03.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/01/2013] [Accepted: 03/01/2013] [Indexed: 12/21/2022]
Abstract
Omentin-1, a visceral fat depot-specific secretory protein, is inversely correlated with obesity and insulin resistance. We investigated, in rats, the effects of chronic omentin-1 administration (8 μg/kg, intraperitoneally, once daily for 14-days) on feeding behavior and related hypothalamic peptides and neurotransmitters. Food intake and body weight were recorded daily throughout the study. We found a significantly increased food intake compared to controls, but only in days 10-14, while body weight significantly increased since day 12 (P<0.05). Compared with vehicle, omentin-1 treatment led to a significant reduction in both cocaine and amphetamine-regulated transcript (CART) (P<0.05) and corticotrophin releasing hormone (CRH) (P<0.05) gene expression, while pro-opiomelanocortin (POMC), agouti-related peptide (AgRP), neuropeptide Y (NPY) and orexin-A gene expression were not modified with respect to vehicle-treated rats. We also found an increase in hypothalamic levodopa (l-dopa) (P<0.05) and norepinephrine (NE) (P<0.01) synthesis, without any effect on dopamine (DA) and serotonin (5-hydroxytryptamine, 5-HT) metabolism. Furthermore, in hypothalamic synaptosomes, omentin-1 (10-100 ng/ml) stimulated basal NE release (ANOVA, P<0.0001; post hoc, P<0.001 vs. vehicle), in a dose-dependent manner, leaving unaffected both basal and depolarization-induced DA and 5-HT release. Finally, when synaptosomes were co-perfused with leptin and omentin-1, we observed that leptin was able to reverse omentin-1-induced stimulation of NE. In conclusion, the orexigenic effects of omentin-1 could be related, at least in part, to decreased CART and CRH gene expression and increased NE synthesis and release in the hypothalamus.
Collapse
Affiliation(s)
- Luigi Brunetti
- Department of Pharmacy, "G. d'Annunzio" University, via dei Vestini 31, 66013 Chieti, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ogawa N, Ito M, Yamaguchi H, Shiuchi T, Okamoto S, Wakitani K, Minokoshi Y, Nakazato M. Intestinal fatty acid infusion modulates food preference as well as calorie intake via the vagal nerve and midbrain-hypothalamic neural pathways in rats. Metabolism 2012; 61:1312-20. [PMID: 22445513 DOI: 10.1016/j.metabol.2012.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 02/04/2012] [Accepted: 02/23/2012] [Indexed: 01/19/2023]
Abstract
The intestine plays important roles in the regulation of feeding behavior by sensing macronutrients. Intestinal fatty acids strongly suppress food intake, but little is known about whether intestinal fatty acids affect food preference. We investigated the effects of jejunal fatty acids infusion on food preference by conducting two-diet choice experiments in rats fed a high-fat diet (HFD) and a high-carbohydrate diet (HCD). Jejunal linoleic acid (18:2) infusion reduced HFD intake dose-dependently, while HCD intake increased with the middle dose of the infusion we examined (100 μL/h) and reduced to the control level with the higher doses (150 and 200 μL/h). α-Linolenic acid (18:3), but not caprylic acid (8:0), altered the food preference and total calorie intake in the same manner as linoleic acid. Linoleic acid infusion dose-dependently increased plasma glucagon-like peptide-1, peptide YY and cholecystokinin levels, but not ghrelin levels. Subdiaphragmatic vagotomy or midbrain transection prevented the change in food preference and total calorie intake by linoleic acid infusion. Jejunal linoleic acid infusion increased norepinephrine turnover in the paraventricular hypothalamic nucleus, while intracerebroventricular injection of idazoxan, an α2-adrenergic receptor (AR) antagonist, suppressed the increased HCD intake, but did not affect the decreased HFD intake. These findings indicated that intestinal long-chain fatty acids modulated food preference as well as total calorie intake via the vagal nerve and midbrain-hypothalamic neural pathways. The effects of the α2-AR antagonist in the brain suggested that the brain distinctly controlled HCD and HFD intake in response to jejunal linoleic acid infusion.
Collapse
Affiliation(s)
- Nobuya Ogawa
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Pharmacological inhibition of monoamine reuptake transporters has been known for many years as an effective therapy to reduce food intake and body weight in obese subjects. However, most of the marketed drugs failed after a distinct period in clinical use and had to be withdrawn because of serious adverse effects resulting in a negative benefit-risk profile. The most common side effects for this drug class included increases in systemic or pulmonary blood pressure and/or heart rate, cardiac valvulopathies, higher cardiovascular event rates, psychiatric disorders, or high abuse potential. The recent withdrawal of sibutramine as result of its adverse actions on the cardiovascular system highlighted again the problems with this drug class in antiobesity therapy. Recent developments to combine reuptake inhibitors with other drug classes, for example, opioid antagonists seem to be a promising approach to improve the benefit-risk profile of these compounds.This chapter will discuss the history of this drug class in appetite control, its mechanism of action, and the clinical effects of selected drugs from this class.
Collapse
Affiliation(s)
- Ulrich Kintscher
- Charité - Universitätsmedizin, Institute of Pharmacology, Berlin, Germany.
| |
Collapse
|
31
|
Kanzler SA, Januario AC, Paschoalini MA. Involvement of β3-adrenergic receptors in the control of food intake in rats. Braz J Med Biol Res 2011; 44:1141-7. [PMID: 21971689 DOI: 10.1590/s0100-879x2011007500127] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 09/09/2011] [Indexed: 11/22/2022] Open
Abstract
This study examined the food intake changes evoked by intracerebroventricular (icv) injection of a selective agonist (BRL37344, 2 and 20 nmol) or antagonist (SR59230A, 10 and 50 nmol) of β3-adrenergic receptors in 24-h fasted rats (adult male Wistar rats, 200-350 g, N = 6/treatment). The animals were also pretreated with saline icv (SAL) or SR59230A (50 nmol) followed by BRL37344 (20 nmol) or SAL in order to determine the selectivity of the effects evoked by BRL37344 on food intake or the selectivity of the effects evoked by SR59230A on risk assessment (RA) behavior. The highest dose of BRL37344 (N = 7) decreased food intake 1 h after the treatment (6.4 ± 0.5 g in SAL-treated vs 4.2 ± 0.8 g in drug-treated rats). While both doses of SR59230A failed to affect food intake (5.1 ± 1.1 g for 10 nmol and 6.0 ± 1.8 g for 50 nmol), this treatment reduced the RA frequency (number/30 min) (4 ± 2 for SAL-treated vs 1 ± 1 for 10 nmol and 0.5 ± 1 for 50 nmol SR59230A-treated rats), an ethological parameter related to anxiety. While pretreatment with SR59230A (7.0 ± 0.5 g) abolished the hypophagia induced by BRL37344 (3.6 ± 0.9 g), BRL37344 suppressed the reduction in RA frequency caused by SR59230A. These results show that the hypophagia caused by BRL37344 is selectively mediated by β3-adrenergic receptors within the central nervous system. Moreover, they suggest the involvement of these receptors in the control of anxiety.
Collapse
Affiliation(s)
- S A Kanzler
- Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | | |
Collapse
|
32
|
Taksande BG, Kotagale NR, Nakhate KT, Mali PD, Kokare DM, Hirani K, Subhedar NK, Chopde CT, Ugale RR. Agmatine in the hypothalamic paraventricular nucleus stimulates feeding in rats: involvement of neuropeptide Y. Br J Pharmacol 2011; 164:704-18. [PMID: 21564088 PMCID: PMC3188911 DOI: 10.1111/j.1476-5381.2011.01484.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 04/02/2011] [Accepted: 05/02/2011] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Agmatine, a multifaceted neurotransmitter, is abundantly expressed in the hypothalamic paraventricular nucleus (PVN). Our aim was to assess (i) the effect of agmatine on feeding behaviour and (ii) its association, if any, with neuropeptide Y (NPY). EXPERIMENTAL APPROACH Satiated rats fitted with intra-PVN cannulae were administered agmatine, alone or jointly with (i) α₂-adrenoceptor agonist, clonidine, or antagonist, yohimbine; (ii) NPY, NPY Y₁ receptor agonist, [Leu³¹, Pro³⁴]-NPY, or antagonist, BIBP3226; or (iii) yohimbine and NPY. Cumulative food intake was monitored at different post-injection time points. Furthermore, the expression of hypothalamic NPY following i.p. treatment with agmatine, alone or in combination with yohimbine (i.p.), was evaluated by immunocytochemistry. KEY RESULTS Agmatine robustly increased feeding in a dose-dependent manner. While pretreatment with clonidine augmented, yohimbine attenuated the orexigenic response to agmatine. Similarly, NPY and [Leu³¹, Pro³⁴]-NPY potentiated the agmatine-induced hyperphagia, whereas BIBP3226 inhibited it. Moreover, yohimbine attenuated the synergistic orexigenic effect induced by the combination of NPY and agmatine. Agmatine increased NPY immunoreactivity in the PVN fibres and in the cells of the hypothalamic arcuate nucleus (ARC) and this effect was prevented by pretreatment with yohimbine. NPY immunoreactivity in the fibres of the ARC, dorsomedial, ventromedial and lateral nuclei of the hypothalamus was not affected by any of the above treatments. CONCLUSIONS AND IMPLICATIONS The orexigenic effect of agmatine is coupled to increased NPY activity mediated by stimulation of α₂-adrenoceptors within the PVN. This signifies the importance of agmatine or α₂-adrenoceptor modulators in the development of novel therapeutic agents to treat feeding-related disorders.
Collapse
Affiliation(s)
- BG Taksande
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of PharmacyNew Kamptee, Nagpur, Maharashtra, India
| | - NR Kotagale
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of PharmacyNew Kamptee, Nagpur, Maharashtra, India
| | - KT Nakhate
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University CampusNagpur, Maharashtra, India
| | - PD Mali
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of PharmacyNew Kamptee, Nagpur, Maharashtra, India
| | - DM Kokare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University CampusNagpur, Maharashtra, India
| | - K Hirani
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of MedicineMiami, FL, USA
| | - NK Subhedar
- Indian Institute of Science Education and Research (IISER)Sutarwadi, Pashan, Pune, India
| | - CT Chopde
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of PharmacyNew Kamptee, Nagpur, Maharashtra, India
| | - RR Ugale
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of PharmacyNew Kamptee, Nagpur, Maharashtra, India
| |
Collapse
|
33
|
Kochenborger L, Zanatta D, Berretta LM, Lopes APF, Wunderlich BL, Januário AC, Neto JM, Terenzi MG, Paschoalini MA, Faria MS. Modulation of fear/anxiety responses, but not food intake, following α-adrenoceptor agonist microinjections in the nucleus accumbens shell of free-feeding rats. Neuropharmacology 2011; 62:427-35. [PMID: 21871465 DOI: 10.1016/j.neuropharm.2011.08.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 08/10/2011] [Accepted: 08/11/2011] [Indexed: 10/17/2022]
Abstract
This study investigated the effect of α-adrenoceptor agonists microinjected into the shell region of the accumbens nucleus (AcbSh) on feeding and anxiety-related behaviors in free-feeding rats. Male Wistar rats with a chronically implanted cannula into the AcbSh were unilaterally microinjected with either clonidine (CLON, α(2)-adrenoceptor agonist) or phenylephrine (PHEN, α(1)-adrenoceptor agonist) at the doses of 6 and 20 nmol and submitted to the elevated plus-maze (EPM), a pre-clinical test of anxiety. Immediately after the EPM test, the animals underwent food intake evaluation for 30 min. The data showed that rats microinjected with CLON (20 nmol/0.2 μl) into the AcbSh exhibited increased %Open arm time, which is compatible with an anxiolytic-like effect. The CLON-induced anxiolysis was corroborated by increased head-dipping and decreased stretched-attend posture, two ethologically derived behaviors which are fear/anxiety-motivated. The animal's locomotor activity was not changed by 20 nmol CLON microinjection into the AcbSh. However, neither dose of PHEN microinjected into the AcbSh was able to alter either the spatial-temporal or ethological variables representative of fear/anxiety and locomotion. Food intake was not altered by any dose of CLON and PHEN microinjected into the AcbSh, but the 20 nmol CLON microinjection induced increased motor activity in the feeding test. The data suggests that noradrenergic projections to the AcbSh may underlie fear/anxiety modulation through α(2)-adrenoceptor in the AcbSh, while feeding behavior was unaffected by noradrenergic modulation in the AcbSh of free-feeding rats. This article is part of a Special Issue entitled 'Anxiety and Depression'.
Collapse
Affiliation(s)
- Larissa Kochenborger
- Department of Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), 88040-900 Florianópolis, SC, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Smith KS, Berridge KC, Aldridge JW. Disentangling pleasure from incentive salience and learning signals in brain reward circuitry. Proc Natl Acad Sci U S A 2011; 108:E255-64. [PMID: 21670308 PMCID: PMC3131314 DOI: 10.1073/pnas.1101920108] [Citation(s) in RCA: 281] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Multiple signals for reward-hedonic impact, motivation, and learned associative prediction-are funneled through brain mesocorticolimbic circuits involving the nucleus accumbens and ventral pallidum. Here, we show how the hedonic "liking" and motivation "wanting" signals for a sweet reward are distinctly modulated and tracked in this circuit separately from signals for Pavlovian predictions (learning). Animals first learned to associate a fixed sequence of Pavlovian cues with sucrose reward. Subsequent intraaccumbens microinjections of an opioid-stimulating drug increased the hedonic liking impact of sucrose in behavior and firing signals of ventral pallidum neurons, and likewise, they increased incentive salience signals in firing to the reward-proximal incentive cue (but did not alter firing signals to the learned prediction value of a reward-distal cue). Microinjection of a dopamine-stimulating drug instead enhanced only the motivation component but did not alter hedonic impact or learned prediction signals. Different dedicated neuronal subpopulations in the ventral pallidum tracked signal enhancements for hedonic impact vs. incentive salience, and a faster firing pattern also distinguished incentive signals from slower hedonic signals, even for a third overlapping population. These results reveal separate neural representations of wanting, liking, and prediction components of the same reward within the nucleus accumbens to ventral pallidum segment of mesocorticolimbic circuitry.
Collapse
Affiliation(s)
- Kyle S Smith
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | |
Collapse
|
35
|
The tempted brain eats: pleasure and desire circuits in obesity and eating disorders. Brain Res 2010; 1350:43-64. [PMID: 20388498 DOI: 10.1016/j.brainres.2010.04.003] [Citation(s) in RCA: 542] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 03/23/2010] [Accepted: 04/02/2010] [Indexed: 01/12/2023]
Abstract
What we eat, when and how much, all are influenced by brain reward mechanisms that generate "liking" and "wanting" for foods. As a corollary, dysfunction in reward circuits might contribute to the recent rise of obesity and eating disorders. Here we assess brain mechanisms known to generate "liking" and "wanting" for foods and evaluate their interaction with regulatory mechanisms of hunger and satiety, relevant to clinical issues. "Liking" mechanisms include hedonic circuits that connect together cubic-millimeter hotspots in forebrain limbic structures such as nucleus accumbens and ventral pallidum (where opioid/endocannabinoid/orexin signals can amplify sensory pleasure). "Wanting" mechanisms include larger opioid networks in nucleus accumbens, striatum, and amygdala that extend beyond the hedonic hotspots, as well as mesolimbic dopamine systems, and corticolimbic glutamate signals that interact with those systems. We focus on ways in which these brain reward circuits might participate in obesity or in eating disorders.
Collapse
|
36
|
Baghbanzadeh A, Hajinezhad MR, Shohreh B, Maleklou R. Intralateral hypothalamic area injection of isoproterenol and propranolol affects food and water intake in broilers. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2010; 196:221-6. [DOI: 10.1007/s00359-010-0507-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 01/25/2010] [Accepted: 01/27/2010] [Indexed: 10/19/2022]
|
37
|
Vidarsdottir S, Roelfsema F, Streefland T, Holst JJ, Rehfeld JF, Pijl H. Short-term treatment with olanzapine does not modulate gut hormone secretion: olanzapine disintegrating versus standard tablets. Eur J Endocrinol 2010; 162:75-83. [PMID: 19779025 DOI: 10.1530/eje-09-0433] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Treatment with olanzapine (atypical antipsychotic drug) is frequently associated with various metabolic anomalies, including obesity, dyslipidemia, and diabetes mellitus. Recent data suggest that olanzapine orally disintegrating tablets (ODT), which dissolve instantaneously in the mouth, might cause less weight gain than olanzapine standard oral tablets (OST). DESIGN AND METHODS Ten healthy men received olanzapine ODT (10 mg o.d., 8 days), olanzapine OST (10 mg o.d., 8 days), or no intervention in a randomized crossover design. At breakfast and dinner, blood samples were taken for measurement of pancreatic polypeptide, peptide YY, glucagon-like peptide-1, total glucagon, total ghrelin, and cholecystokinin (CCK) concentrations. RESULTS With the exception of pre- and postprandial concentration of ghrelin at dinner and preprandial CCK concentrations at breakfast, which were all slightly increased (respectively P=0.048, P=0.034 and P=0.042), olanzapine did not affect gut hormone concentrations. Thus, olanzapine ODT and OST had similar effects on gut hormone secretion. CONCLUSION Short-term treatment with olanzapine does not have major impact on the plasma concentration of gut hormones we measured in healthy men. Moreover, despite pharmacological difference, gut hormone concentrations are similar during treatment with olanzapine ODT and OST. The capacity of olanzapine to induce weight gain and diabetes is unlikely to be caused by modulation of the secretion of gut hormones measured here. We cannot exclude the possibility that olanzapine's impact on other gut hormones, to impair insulin sensitivity and stimulate weight gain, exists.
Collapse
Affiliation(s)
- Solrun Vidarsdottir
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
It is now axiomatic that neurons in the hypothalamic arcuate nucleus have a primary role in responding to changes in circulating levels of leptin and transmitting signals to downstream circuits that influence eating and energy expenditure. Signals generated from the gastrointestinal tract during meals reach the brainstem, via the vagus nerve and other routes, and impinge on neural circuits that influence the timing and size of meals and amount of food consumed. One of the mechanisms by which leptin exerts its anorexic effects is by increasing the effectiveness of intestinal signals that cause satiation during a meal. It is clear that the effects of gut satiation signals such as CCK can be amplified by leptin acting in the CNS, and in the arcuate nucleus in particular. The present article describes the state of our knowledge about specific neural circuits between the hypothalamus and brainstem that play a role in the interaction of leptin and meal-control signals to control food intake.
Collapse
|
39
|
Morris HV, Nilsson S, Dixon CI, Stephens DN, Clifton PG. Alpha1- and alpha2-containing GABAA receptor modulation is not necessary for benzodiazepine-induced hyperphagia. Appetite 2009; 52:675-683. [PMID: 19501766 DOI: 10.1016/j.appet.2009.03.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 02/23/2009] [Accepted: 03/12/2009] [Indexed: 10/21/2022]
Abstract
Benzodiazepines increase food intake, an effect attributed to their ability to enhance palatability. We investigated which GABA(A) receptor subtypes may be involved in mediating benzodiazepine-induced hyperphagia. The role of the alpha2 subtype was investigated by observing the effects of midazolam, on the behavioural satiety sequence in mice with targeted deletion of the alpha2 gene (alpha2 knockout). Midazolam (0.125, 0.25 and 0.5mg/kg) increased food intake and the amount of time spent feeding in alpha2 knockout mice, suggesting that BZ-induced hyperphagia does not involve alpha2-containing GABA(A) receptors. We further investigated the roles of alpha1- and alpha3-containing GABA(A) receptors in mediating BZ-induced hyperphagia. We treated alpha2(H101R) mice, in which alpha2-containing receptors are rendered benzodiazepine insensitive, with L-838417, a compound which acts as a partial agonist at alpha2-, alpha3- and alpha5-receptors but is inactive at alpha1-containing receptors. L-838417 (10 and 30 mg/kg) increased food intake and the time spent feeding in both wildtype and alpha2(H101R) mice, demonstrating that benzodiazepine-induced hyperphagia does not require alpha1- and alpha2-containing GABA(A) receptors. These observations, together with evidence against the involvement of alpha5-containing GABA(A) receptors, suggest that alpha3-containing receptors mediate BZ-induced hyperphagia in the mouse.
Collapse
Affiliation(s)
- H V Morris
- Department of Psychology, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - S Nilsson
- Department of Psychology, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - C I Dixon
- Department of Psychology, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - D N Stephens
- Department of Psychology, University of Sussex, Falmer, Brighton, BN1 9QG, UK
| | - P G Clifton
- Department of Psychology, University of Sussex, Falmer, Brighton, BN1 9QG, UK.
| |
Collapse
|
40
|
Brunetti L, Orlando G, Recinella L, Leone S, Ferrante C, Chiavaroli A, Lazzarin F, Vacca M. Glucagon-like peptide 1 (7-36) amide (GLP-1) and exendin-4 stimulate serotonin release in rat hypothalamus. Peptides 2008; 29:1377-81. [PMID: 18502539 DOI: 10.1016/j.peptides.2008.04.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 04/07/2008] [Accepted: 04/08/2008] [Indexed: 11/15/2022]
Abstract
Glucagon-like peptide 1 (7-36) amide (GLP-1) and exendin-4 are gastrointestinal hormones as well as neuropeptides involved in glucose homeostasis and feeding regulation, both peripherally and at the central nervous system (CNS), acting through the same GLP-1 receptor. Aminergic neurotransmitters play a role in the modulation of feeding in the hypothalamus and we have previously found that peripheral hormones and neuropeptides, which are known to modulate feeding in the central nervous system, are able to modify catecholamine and serotonin release in the hypothalamus. In the present paper we have evaluated the effects of GLP-1 and exendin-4 on dopamine, norepinephrine, and serotonin release from rat hypothalamic synaptosomes, in vitro. We found that glucagon-like peptide 1 (7-36) amide and exendin-4 did not modify either basal or depolarization-induced dopamine and norepinephrine release; on the other hand glucagon-like peptide 1 (7-36) amide and exendin-4 stimulated serotonin release, in a dose dependent manner. We can conclude that the central anorectic effects of GLP-1 agonists could be partially mediated by increased serotonin release in the hypothalamus, leaving the catecholamine release unaffected.
Collapse
Affiliation(s)
- Luigi Brunetti
- Department of Drug Sciences, G. d'Annunzio University, Chieti, Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Scopinho AA, Resstel LBM, Corrêa FMA. alpha(1)-Adrenoceptors in the lateral septal area modulate food intake behaviour in rats. Br J Pharmacol 2008; 155:752-6. [PMID: 18641672 DOI: 10.1038/bjp.2008.295] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND PURPOSE Control of food intake is a complex behaviour which involves many interconnected brain structures. The present work assessed if the noradrenergic system in the lateral septum (LS) was involved in the feeding behaviour of rats. EXPERIMENTAL APPROACH In the first protocol, the food intake of rats was measured. Then non-food-deprived animals received either 100 nL of 21 nmol of noradrenaline or vehicle unilaterally in the LS 10 min after local 10 nmol of WB4101, an alpha(1)-adrenoceptor antagonist, or vehicle. In the second protocol, different doses of WB4101 (1, 10 or 20 nmol in 100 nL) were microinjected bilaterally into the LS of rats, deprived of food for 18 h and food intake was compared to that of satiated animals. KEY RESULTS One-sided microinjection of noradrenaline into the LS of normal-fed rats evoked food intake, compared with vehicle-injected control animals, which was significantly reduced by alpha(1)-adrenoceptor antagonism. In a further investigation, food intake was significantly higher in food-deprived animals, compared to satiated controls. Pretreatment of the LS with WB4101 reduced food intake in only food-deprived animals in a dose-related manner, suggesting that the LS noradrenergic system was involved in the control of food intake. CONCLUSION AND IMPLICATIONS Activation by local microinjection of noradrenaline of alpha(1)-adrenoceptors in the LS evoked food intake behaviour in rats. In addition, blockade of the LS alpha(1)-adrenoceptors inhibited food intake in food-deprived animals, suggesting that the LS noradrenergic system modulated food intake behaviour and satiation.
Collapse
Affiliation(s)
- A A Scopinho
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | | | | |
Collapse
|
42
|
Hoebel BG, Wellman PJ. Overview of methodological approaches to the study of ingestive behavior. ACTA ACUST UNITED AC 2008; Chapter 8:Unit 8.6A. [PMID: 18428544 DOI: 10.1002/0471142301.ns0806as03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Some examples of procedures used by feeding researchers are discussed in this unit and include ablation of neural function; inhibition of behaviors by selective neurotoxins and antisense oligonucleotides; staining of sensory and motor mechanisms; electrical stimulation of the brain; local injection and microdialysis of nutrients, neurotransmitters, and drugs; autoradiography and in situ hybridization of neurotransmitters with their receptors; electrophysiological techniques for multi- and single-unit recording of cells in the hypothalamus; and gene technology using inbred strains of genetically obese mice.
Collapse
Affiliation(s)
- B G Hoebel
- Princeton University, Princeton, New Jersey, USA
| | | |
Collapse
|
43
|
Wellman PJ, McMahon LR. Basic measures of food intake. ACTA ACUST UNITED AC 2008; Chapter 8:Unit 8.6B. [PMID: 18428545 DOI: 10.1002/0471142301.ns0806bs03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This unit describes a method for measuring the consumption of a pellet diet during a 30-min testing session conducted during the late portion of the day. The procedure generates reliable and stable baseline measures of feeding, and is appropriate for assessing experimental manipulations that either enhance (e.g., injecting a peptide into the brain) or suppress feeding (e.g., systemic injections of amphetamine).
Collapse
Affiliation(s)
- P J Wellman
- Texas A&M University, College Station, Texas, USA
| | | |
Collapse
|
44
|
Abstract
A cohesive picture has emerged over the last decade regarding the pathophysiology and treatment of bipolar disorder, a serious mental disorder that cycles between mania and depression. Mania is associated with overactive PKC intracellular signaling, and recent genome-wide association studies of bipolar disorder have implicated an enzyme that reduces the activation of PKC. Overactive PKC signaling in the prefrontal cortex may explain many of the symptoms of mania. Functional imaging studies have shown reduced activity in the right prefrontal cortex during the manic state. Dysfunction of the right prefrontal cortex is known to lead to a disinhibited profile, including poor impulse control, risk-taking, distractibility, poor sustained attention and delusions, which resemble the symptoms of mania. Structural imaging studies have further shown a loss of prefrontal volume in untreated patients with bipolar disorder. This loss of function and gray matter in the prefrontal cortex may arise from abnormal signaling cascades, notably PKC signaling. Studies in animals have shown that elevated PKC activity markedly and rapidly impairs the functioning of the prefrontal cortex, providing a link to the the loss of prefrontal regulation of thought and emotion during the manic state. Sustained elevation in PKC signaling may also lead to loss of gray matter in the prefrontal cortex, which can be protected by PKC inhibitors. Importantly, the current mainstays in the treatment of mania, lithium (a monovalent cation) and valproate (a small fatty acid), indirectly inhibit PKC. Proof-of-principle studies with the antiestrogenic PKC inhibitor tamoxifen have shown rapid antimanic effects. Recent evidence suggests that lithium may also protect prefrontal gray matter in patients. These data indicate that new, direct PKC inhibitors are needed as potential new treatments for bipolar disorder.
Collapse
Affiliation(s)
- AFT Arnsten
- Yale University School of Medicine, Department of Neurobiology, 333 Cedar Street, New Haven, CT 06510, USA
| | - HK Manji
- National Institute of Mental Health, National Institutes of Health, Building 35, 1C912, Bethesda, MD 20892, USA
| |
Collapse
|
45
|
Wellman PJ, Ho DH, Nation JR. Differential impact of cocaine on meal patterns in female and male rats. Life Sci 2007; 82:359-66. [PMID: 18207198 DOI: 10.1016/j.lfs.2007.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Revised: 11/01/2007] [Accepted: 11/09/2007] [Indexed: 10/22/2022]
Abstract
Female rats, relative to males, exhibit greater behavioral activation to cocaine and other psychostimulants, but the effect of sex and the estrous cycle in modulating the hypophagic action of cocaine has not been evaluated. Meal patterns were recorded in automated food hoppers during the first 3 h of the dark phase in adult female and male rats after administration of ascending cocaine doses (0, 7.5, and 15 mg/kg cocaine, i.p.) on successive trials. Cocaine produced a greater suppression of feeding as well as a reduction in meal number over a 3 h test period in female rats during estrus, relative to that noted during diestrus. In contrast, during the 180 min test period, male rats showed minimal hypophagic responses to 7.5 or 15 mg/kg cocaine. These results extend the range of behavioral perturbations induced by cocaine that are modulated by sex and by the estrous cycle and are consistent with the notion that estradiol may modulate the neurochemical actions of cocaine.
Collapse
Affiliation(s)
- Paul J Wellman
- Cellular and Behavioral Neuroscience Program, Department of Psychology, Texas A&M University, College Station, TX 77843-4235, USA.
| | | | | |
Collapse
|
46
|
Garina DV, Kuz'mina VV, Gerasimov YV. The effect of epinephrine on feeding and motion patterns in goldfish Carassius auratus (L.). Comp Biochem Physiol A Mol Integr Physiol 2007; 148:544-9. [PMID: 17698382 DOI: 10.1016/j.cbpa.2007.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Revised: 07/02/2007] [Accepted: 07/07/2007] [Indexed: 10/23/2022]
Abstract
The effect of intraperitoneal injections of epinephrine (0.14 and 0.70 mg/kg) on some characteristics of feeding activity (ration and total time of feeding) as well as on motion patterns (time of swimming in group and separately) in juvenile goldfish has been investigated. Two-phase (short-term decrease in the first phase, increase in the second one) feeding response under both doses of epinephrine has been revealed. More pronounced effect of epinephrine at the dose of 0.14 mg/kg on the ration and time of feeding (comparing to the dose of 0.70 mg/kg and Ringer injection) was observed in the second phase. Furthermore, significant decrease of time of "separated" swimming in the first phase under both doses of the hormone is revealed. The hyperglycemic response induced by the injections of epinephrine, with synchronous reduction of the concentration of glycogen in hepatopancreas allows to suggest that glycogen-phosphorylase activating mechanism was underlying the "first-phase" change of feeding reactions of goldfish.
Collapse
Affiliation(s)
- D V Garina
- Institute for Biology of Inland Waters RAS, Borok, Nekouz, Yaroslavl region, Russia.
| | | | | |
Collapse
|
47
|
Clifford PS, Davis KW, Elliott AE, Wellman PJ. Effects of ICV administration of the alpha1A-adrenoceptor antagonist 5-methylurapidil on concurrent measures of eating and locomotion after cocaine in the rat. Life Sci 2007; 81:1059-65. [PMID: 17881010 PMCID: PMC2097960 DOI: 10.1016/j.lfs.2007.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 07/25/2007] [Accepted: 08/03/2007] [Indexed: 10/22/2022]
Abstract
Psychostimulants including amphetamine and cocaine induce locomotion and stereotypy and suppress eating. Although the capacity of cocaine to alter locomotion is usually viewed as related to dopamine neurotransmission, recent studies suggest that norepinephrine, acting through alpha1-adrenergic receptors (alpha1-ARs) can facilitate cocaine-stimulated locomotion. Of the three alpha1-AR subtypes (alpha(1A), alpha(1B), and alpha(1D)) identified to date, inactivation of the alpha(1B)-AR subtype diminishes cocaine-stimulated locomotion, whereas the impact of inactivation of the alpha(1A)-AR subtype on either eating or locomotion is unknown. In the present study, we assessed the relative impact of ICV administration of the alpha(1B)-AR antagonist 5-methylurapidil (5-MU) on cocaine-stimulated hyperlocomotion and hypophagia, using a concurrent method [Wellman, P.J., Ho, D.H., Davis, K.W., 2005. Concurrent measures of feeding and locomotion in rats. Physiology of Behavior 84 (5), 769-774.]. Rats were infused ICV with one of 3 doses of 5-MU (0, 3, or 30 nmol) and then injected (i.p.) with 0, 2.5, 5.0, 10.0, or 20.0 mg/kg cocaine HCl on each of five tests. Rats always received the same 5-MU dose, but a different cocaine dose on each trial. Feeding and locomotion were assessed concurrently during a 45-min postinjection period. Significant suppression of eating was noted at 2.5 mg/kg cocaine, a dose that does not alter forward locomotion in the rat. Administration of 5-MU did not alter locomotion in rats treated with saline, but did significantly increase baseline food intake. Neither cocaine-induced hypophagia nor hyperlocomotion was altered by ICV administration of 5-MU. These results suggest that the capacity of alpha1-AR agonists (e.g. phenylpropanolamine) to suppress eating may be related to activation of the alpha(1A)-AR subtype, whereas cocaine does not act through the alpha(1A)-AR subtype to suppress eating nor does this subtype modulate cocaine-induced hyperlocomotion.
Collapse
Affiliation(s)
- P Shane Clifford
- Behavioral Neuroscience Program, Department of Psychology, Texas A&M University, College Station, TX 77843-4235, USA
| | | | | | | |
Collapse
|
48
|
Kramer PR, Guan G, Wellman PJ, Bellinger LL. Nicotine's attenuation of body weight involves the perifornical hypothalamus. Life Sci 2007; 81:500-8. [PMID: 17655879 PMCID: PMC2725327 DOI: 10.1016/j.lfs.2007.06.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Revised: 05/31/2007] [Accepted: 06/14/2007] [Indexed: 11/28/2022]
Abstract
Previously we showed that intermittent administration of nicotine (NIC) in the dark phase decreased food intake and body weight and this could be blocked when the NIC receptor antagonist mecamylamine was infused into the fourth ventricle. Catecholaminergic neurons adjacent to the fourth ventricle contain NIC receptors and directly innervate the perifornical hypothalamus (PFH) which has been shown to be involved in regulation of feeding. This study explored whether NIC regulates feeding behavior by modulating catecholaminergic input to the PFH. Epinephrine and norepinephrine neuronal input was ablated within the PFH by infusion of 6-hydroxydopamine hydrobromide (6-OHDA), while bupropion was infused to protect dopaminergic neurons. After recovery of body weights to pre-surgery levels, food intake, meal size, meal number and body weight were measured after intermittent NIC injections. The results showed the PFH lesioned animals did not exhibit the typical prolonged drop in food intake, meal size and body weight normally associated with NIC administration. High performance liquid chromatography analyses demonstrated that compared to control rats, 6-OHDA administration significantly reduced PFH norepinephrine and epinephrine levels, but not dopamine levels. These results are consistent with NIC reducing food intake in part by acting through catecholaminergic neurons within or extending through the PFH.
Collapse
Affiliation(s)
- Phillip R Kramer
- Department of Biomedical Sciences, Baylor College of Dentistry, Texas A&M University System Health Science Center, 3302 Gaston Ave. Dallas, TX 75246, USA.
| | | | | | | |
Collapse
|
49
|
Date Y, Shimbara T, Koda S, Toshinai K, Ida T, Murakami N, Miyazato M, Kokame K, Ishizuka Y, Ishida Y, Kageyama H, Shioda S, Kangawa K, Nakazato M. Peripheral ghrelin transmits orexigenic signals through the noradrenergic pathway from the hindbrain to the hypothalamus. Cell Metab 2006; 4:323-31. [PMID: 17011505 DOI: 10.1016/j.cmet.2006.09.004] [Citation(s) in RCA: 175] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2005] [Revised: 07/19/2006] [Accepted: 09/13/2006] [Indexed: 02/07/2023]
Abstract
Ghrelin, a gastrointestinal peptide, stimulates feeding when administered peripherally. Blockade of the vagal afferent pathway abolishes ghrelin-induced feeding, indicating that the vagal afferent pathway may be a route conveying orexigenic ghrelin signals to the brain. Here, we demonstrate that peripheral ghrelin signaling, which travels to the nucleus tractus solitarius (NTS) at least in part via the vagus nerve, increases noradrenaline (NA) in the arcuate nucleus of the hypothalamus, thereby stimulating feeding at least partially through alpha-1 and beta-2 noradrenergic receptors. In addition, bilateral midbrain transections rostral to the NTS, or toxin-induced loss of neurons in the hindbrain that express dopamine beta hydroxylase (an NA synthetic enzyme), abolished ghrelin-induced feeding. These findings provide new evidence that the noradrenergic system is necessary in the central control of feeding behavior by peripherally administered ghrelin.
Collapse
Affiliation(s)
- Yukari Date
- Third Department of Internal Medicine, Miyazaki Medical College, University of Miyazaki, Miyazaki 889-1692, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Park YM, Chung YC, Lee SH, Lee KJ, Kim H, Byun YC, Lim SW, Paik JW, Lee HJ. Weight gain associated with the alpha2a-adrenergic receptor -1,291 C/G polymorphism and olanzapine treatment. Am J Med Genet B Neuropsychiatr Genet 2006; 141B:394-7. [PMID: 16583406 DOI: 10.1002/ajmg.b.30311] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Weight gain can be an adverse effect of antipsychotics and is an important factor for long-term health and treatment compliance. Many reports have shown that the alpha(2)-adrenergic receptor may be related to eating behaviors or lipolytic activities, both associated with body weight change. We hypothesized that there might be a relationship between the alpha(2a)-adrenergic receptor -1,291 C/G polymorphism and olanzapine-induced weight gain. A group of 62 Korean schizophrenic patients participated in a study; weight and height measurements were obtained prior to starting olanzapine and measured again after long-term treatment. Genotyping for the -1291 C/G polymorphism was performed on all participants. Body weight changes from baseline to endpoint were significantly associated with genotypes (P = 0.028). The frequency of the G allele was significantly higher in subjects who had severe weight gain (defined as a more than 10% weight gain from baseline) compared to subjects who did not have extreme weight gain (less than 10% weight gain from baseline) (X(2) = 6.120, P = 0.013; OR = 2.58, 95% CI = 1.21-5.51). Therefore, the findings from this study support a relationship between the -1291 C/G polymorphism of the alpha(2a)-adrenergic receptor and weight gain in Korean schizophrenic patients receiving olanzapine treatment.
Collapse
Affiliation(s)
- Young-Min Park
- Department of Psychiatry, Inje University College of Medicine, Goyang, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|