1
|
Yoshimura M, Flynn BP, Kershaw YM, Zhao Z, Ueta Y, Lightman SL, Conway-Campbell BL. Phase-shifting the circadian glucocorticoid profile induces disordered feeding behaviour by dysregulating hypothalamic neuropeptide gene expression. Commun Biol 2023; 6:998. [PMID: 37775688 PMCID: PMC10541449 DOI: 10.1038/s42003-023-05347-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 09/12/2023] [Indexed: 10/01/2023] Open
Abstract
Here we demonstrate, in rodents, how the timing of feeding behaviour becomes disordered when circulating glucocorticoid rhythms are dissociated from lighting cues; a phenomenon most commonly associated with shift-work and transmeridian travel 'jetlag'. Adrenalectomized rats are infused with physiological patterns of corticosterone modelled on the endogenous adrenal secretory profile, either in-phase or out-of-phase with lighting cues. For the in-phase group, food intake is significantly greater during the rats' active period compared to their inactive period; a feeding pattern similar to adrenal-intact control rats. In contrast, the feeding pattern of the out-of-phase group is significantly dysregulated. Consistent with a direct hypothalamic modulation of feeding behaviour, this altered timing is accompanied by dysregulated timing of anorexigenic and orexigenic neuropeptide gene expression. For Neuropeptide Y (Npy), we report a glucocorticoid-dependent direct transcriptional regulation mechanism mediated by the glucocorticoid receptor (GR). Taken together, our data highlight the adverse behavioural outcomes that can arise when two circadian systems have anti-phasic cues, in this case impacting on the glucocorticoid-regulation of a process as fundamental to health as feeding behaviour. Our findings further highlight the need for development of rational approaches in the prevention of metabolic dysfunction in circadian-disrupting activities such as transmeridian travel and shift-work.
Collapse
Affiliation(s)
- Mitsuhiro Yoshimura
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
- Department of Physiology, University of Occupational and Environmental Health, Japan 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Benjamin P Flynn
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Yvonne M Kershaw
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Zidong Zhao
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Yoichi Ueta
- Department of Physiology, University of Occupational and Environmental Health, Japan 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Stafford L Lightman
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Becky L Conway-Campbell
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| |
Collapse
|
2
|
Monica Shih MC, Huang CCJ, Chu HP, Hsu NC, Chung BC. Embryonic Steroids Control Developmental Programming of Energy Balance. Endocrinology 2021; 162:6380292. [PMID: 34599818 DOI: 10.1210/endocr/bqab196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Indexed: 12/25/2022]
Abstract
Glucose is a major energy source for growth. At birth, neonates must change their energy source from maternal supply to its own glucose production. The mechanism of this transition has not been clearly elucidated. To evaluate the possible roles of steroids in this transition, here we examine the defects associated with energy production of a mouse line that cannot synthesize steroids de novo due to the disruption of its Cyp11a1 (cytochrome P450 family 11 subfamily A member 1) gene. The Cyp11a1 null embryos had insufficient blood insulin and failed to store glycogen in the liver since embryonic day 16.5. Their blood glucose dropped soon after maternal deprivation, and the expression of hepatic gluconeogenic and glycogenic genes were reduced. Insulin was synthesized in the mutant fetal pancreas but failed to be secreted. Maternal glucocorticoid supply rescued the amounts of blood glucose, insulin, and liver glycogen in the fetus but did not restore expression of genes for glycogen synthesis, indicating the requirement of de novo glucocorticoid synthesis for glycogen storage. Thus, our investigation of Cyp11a1 null embryos reveals that the energy homeostasis is established before birth, and fetal steroids are required for the regulation of glycogen synthesis, hepatic gluconeogenesis, and insulin secretion at the fetal stage.
Collapse
Affiliation(s)
- Meng-Chun Monica Shih
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Chen-Che Jeff Huang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Hsueh-Ping Chu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Institute of Molecular and Cell Biology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Nai-Chi Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Bon-Chu Chung
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
3
|
Perry RJ, Resch JM, Douglass AM, Madara JC, Rabin-Court A, Kucukdereli H, Wu C, Song JD, Lowell BB, Shulman GI. Leptin's hunger-suppressing effects are mediated by the hypothalamic-pituitary-adrenocortical axis in rodents. Proc Natl Acad Sci U S A 2019; 116:13670-13679. [PMID: 31213533 PMCID: PMC6613139 DOI: 10.1073/pnas.1901795116] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Leptin informs the brain about sufficiency of fuel stores. When insufficient, leptin levels fall, triggering compensatory increases in appetite. Falling leptin is first sensed by hypothalamic neurons, which then initiate adaptive responses. With regard to hunger, it is thought that leptin-sensing neurons work entirely via circuits within the central nervous system (CNS). Very unexpectedly, however, we now show this is not the case. Instead, stimulation of hunger requires an intervening endocrine step, namely activation of the hypothalamic-pituitary-adrenocortical (HPA) axis. Increased corticosterone then activates AgRP neurons to fully increase hunger. Importantly, this is true for 2 forms of low leptin-induced hunger, fasting and poorly controlled type 1 diabetes. Hypoglycemia, which also stimulates hunger by activating CNS neurons, albeit independently of leptin, similarly recruits and requires this pathway by which HPA axis activity stimulates AgRP neurons. Thus, HPA axis regulation of AgRP neurons is a previously underappreciated step in homeostatic regulation of hunger.
Collapse
Affiliation(s)
- Rachel J Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520
| | - Jon M Resch
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Amelia M Douglass
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Joseph C Madara
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Aviva Rabin-Court
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Hakan Kucukdereli
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Chen Wu
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Joongyu D Song
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215;
- Program in Neuroscience, Harvard Medical School, Boston, MA 02215
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520;
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
4
|
Lee SJ, Diener K, Kaufman S, Krieger JP, Pettersen KG, Jejelava N, Arnold M, Watts AG, Langhans W. Limiting glucocorticoid secretion increases the anorexigenic property of Exendin-4. Mol Metab 2016; 5:552-565. [PMID: 27408779 PMCID: PMC4921942 DOI: 10.1016/j.molmet.2016.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 04/14/2016] [Accepted: 04/27/2016] [Indexed: 12/25/2022] Open
Abstract
Objective Glucagon-like peptide-1 (GLP-1) analogs are attractive options for the treatment of type II diabetes and obesity because of their incretin and anorexigenic effects. Peripheral administration of the GLP-1R agonist Exendin-4 (Ex-4) also increases glucocorticoid secretion in rodents and humans, but whether the released glucocorticoids interact with Ex-4's anorexigenic effect remains unclear. Methods To test this, we used two experimental approaches that suppress corticosterone secretion and then assessed Ex-4 effects on eating in adult male rats. First, we combined acute and chronic low dose dexamethasone treatment with Ex-4. Second, we ablated hindbrain catecholamine neurons projecting to the hypothalamus with anti-dopamine-β-hydroxylase-saporin (DSAP) to block Ex-4-induced corticosterone secretion. Results Combining dexamethasone and Ex-4 produced a larger acute anorexigenic effect than Ex-4 alone. Likewise, chronic dexamethasone and Ex-4 co-treatment produced a synergistic effect on eating and greater body weight loss in diet-induced obese rats than Ex-4 alone. DSAP lesions not only blunted Ex-4's ability to increase corticosterone secretion, but potentiated the anorexigenic effect of Ex-4, indicating that Ex-4-dependent corticosterone secretion opposes Ex-4's actions. Consistent with the enhancement of Ex-4's anorexigenic effect, DSAP lesion altered Ex-4-dependent changes in neuropeptide Y, preproglucagon, and corticotropin releasing hormone gene expression involved in glucocorticoid feedback. Conclusions Our findings demonstrate that limiting glucocorticoid secretion and actions with low dose dexamethasone or DSAP lesion increases Ex-4's ability to reduce food intake and body weight. Novel glucocorticoid receptor based mechanisms, therefore, may help enhance GLP-1-based obesity therapies. Blocking HPA axis by low dose dexamethasone increased the anorexigenic property of Ex-4. Dexamethasone/Ex-4 co-treatment reduced food intake and body weight in diet-induced obese rats more than Ex-4 alone. A brain lesion model identified a potential central interaction between glucocorticoids and GLP-1 in food intake control.
Collapse
Affiliation(s)
- Shin J Lee
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland.
| | - Katharina Diener
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Sharon Kaufman
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | | | - Klaus G Pettersen
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Nino Jejelava
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Alan G Watts
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| |
Collapse
|
5
|
Barson JR, Leibowitz SF. Hypothalamic neuropeptide signaling in alcohol addiction. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:321-9. [PMID: 25689818 PMCID: PMC4537397 DOI: 10.1016/j.pnpbp.2015.02.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/30/2015] [Accepted: 02/09/2015] [Indexed: 11/27/2022]
Abstract
The hypothalamus is now known to regulate alcohol intake in addition to its established role in food intake, in part through neuromodulatory neurochemicals termed neuropeptides. Certain orexigenic neuropeptides act in the hypothalamus to promote alcohol drinking, although they affect different aspects of the drinking response. These neuropeptides, which include galanin, the endogenous opioid enkephalin, and orexin/hypocretin, appear to stimulate alcohol intake not only through mechanisms that promote food intake but also by enhancing reward and reinforcement from alcohol. Moreover, these neuropeptides participate in a positive feedback relationship with alcohol, whereby they are upregulated by alcohol intake to promote even further consumption. They contrast with other orexigenic neuropeptides, such as melanin-concentrating hormone and neuropeptide Y, which promote alcohol intake under limited circumstances, are not consistently stimulated by alcohol, and do not enhance reward. They also contrast with neuropeptides that can be anorexigenic, including the endogenous opioid dynorphin, corticotropin-releasing factor, and melanocortins, which act in the hypothalamus to inhibit alcohol drinking as well as reward and therefore counter the ingestive drive promoted by orexigenic neuropeptides. Thus, while multiple hypothalamic neuropeptides may work together to regulate different aspects of the alcohol drinking response, excessive signaling from orexigenic neuropeptides or inadequate signaling from anorexigenic neuropeptides can therefore allow alcohol drinking to become dysregulated.
Collapse
Affiliation(s)
- Jessica R. Barson
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, Box 278, New York, NY, 10065 USA
| | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, Box 278, New York, NY, 10065 USA
,Corresponding author at: Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, Box 278, New York, NY, 10065 USA. Tel.: +1 212 327 8378; fax: +1 212 327 8447
| |
Collapse
|
6
|
Tasan RO, Verma D, Wood J, Lach G, Hörmer B, de Lima TCM, Herzog H, Sperk G. The role of Neuropeptide Y in fear conditioning and extinction. Neuropeptides 2016; 55:111-26. [PMID: 26444585 DOI: 10.1016/j.npep.2015.09.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/10/2015] [Accepted: 09/10/2015] [Indexed: 12/23/2022]
Abstract
While anxiety disorders are the brain disorders with the highest prevalence and constitute a major burden for society, a considerable number of affected people are still treated insufficiently. Thus, in an attempt to identify potential new anxiolytic drug targets, neuropeptides have gained considerable attention in recent years. Compared to classical neurotransmitters they often have a regionally restricted distribution and may bind to several distinct receptor subtypes. Neuropeptide Y (NPY) is a highly conserved neuropeptide that is specifically concentrated in limbic brain areas and signals via at least 5 different G-protein-coupled receptors. It is involved in a variety of physiological processes including the modulation of emotional-affective behaviors. An anxiolytic and stress-reducing property of NPY is supported by many preclinical studies. Whether NPY may also interact with processing of learned fear and fear extinction is comparatively unknown. However, this has considerable relevance since pathological, inappropriate and generalized fear expression and impaired fear extinction are hallmarks of human post-traumatic stress disorder and a major reason for its treatment-resistance. Recent evidence from different laboratories emphasizes a fear-reducing role of NPY, predominantly mediated by exogenous NPY acting on Y1 receptors. Since a reduction of fear expression was also observed in Y1 receptor knockout mice, other Y receptors may be equally important. By acting on Y2 receptors, NPY promotes fear extinction and generates a long-term suppression of fear, two important preconditions that could support cognitive behavioral therapies in human patients. A similar effect has been demonstrated for the closely related pancreatic polypeptide (PP) when acting on Y4 receptors. Preliminary evidence suggests that NPY modulates fear in particular by activation of Y1 and Y2 receptors in the basolateral and central amygdala, respectively. In the basolateral amygdala, NPY signaling activates inhibitory G protein-coupled inwardly-rectifying potassium channels or suppresses hyperpolarization-induced I(h) currents in a Y1 receptor-dependent fashion, favoring a general suppression of neuronal activity. A more complex situation has been described for the central extended amygdala, where NPY reduces the frequency of inhibitory and excitatory postsynaptic currents. In particular the inhibition of long-range central amygdala output neurons may result in a Y2 receptor-dependent suppression of fear. The role of NPY in processes of learned fear and fear extinction is, however, only beginning to emerge, and multiple questions regarding the relevance of endogenous NPY and different receptor subtypes remain elusive. Y2 receptors may be of particular interest for future studies, since they are the most prominent Y receptor subtype in the human brain and thus among the most promising therapeutic drug targets when translating preclinical evidence to potential new therapies for human anxiety disorders.
Collapse
Affiliation(s)
- R O Tasan
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria.
| | - D Verma
- Institute of Physiology I, University of Münster, D-48149 Münster, Germany
| | - J Wood
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - G Lach
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria; Capes Foundation, Ministry of Education of Brazil, 70040-020 Brasília/DF, Brazil
| | - B Hörmer
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - T C M de Lima
- Department of Pharmacology, Federal University of Santa Catarina, 88049-970 Florianópolis, Brazil
| | - H Herzog
- Neuroscience Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - G Sperk
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
7
|
Cohen S, Vainer E, Matar MA, Kozlovsky N, Kaplan Z, Zohar J, Mathé AA, Cohen H. Diurnal fluctuations in HPA and neuropeptide Y-ergic systems underlie differences in vulnerability to traumatic stress responses at different zeitgeber times. Neuropsychopharmacology 2015; 40:774-90. [PMID: 25241802 PMCID: PMC4289967 DOI: 10.1038/npp.2014.257] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 09/15/2014] [Indexed: 11/09/2022]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis displays a characteristic circadian pattern of corticosterone release, with higher levels at the onset of the active phase and lower levels at the onset of the inactive phase. As corticosterone levels modify the response to stress and influence the susceptibility to and/or severity of stress-related sequelae, we examined the effects of an acute psychological trauma applied at different zeitgeber times (ZTs) on behavioral stress responses. Rats were exposed to stress either at the onset of the inactive-(light) phase (ZT=0) or at the onset of the active-(dark) phase (ZT=12). Their behavior in the elevated plus-maze and acoustic startle response paradigms were assessed 7 days post exposure for retrospective classification into behavioral response groups. Serum corticosterone levels and the dexamethasone suppression test were used to assess the stress response and feedback inhibition of the HPA axis. Immunoreactivity for neuropeptide Y (NPY) and NPY-Y1 receptor (Y1R) in the paraventricular (PVN) and arcuate (ARC) hypothalamic nuclei, hippocampus, and basolateral amygdala were measured. The behavioral effects of NPY/Y1R antagonist microinfused into the PVN 30 min before stress exposure during the inactive or active phase, respectively, were evaluated. PVN immunoreactivity for NPY and Y1R was measured 1 day after the behavioral tests. The time of day of the traumatic exposure markedly affected the pattern of the behavioral stress response and the prevalence of rats showing an extreme behavioral response. Rats exposed to the stressor at the onset of their inactive phase displayed a more traumatic behavioral response, faster post-exposure corticosterone decay, and a more pronounced stress-induced decline in NPY and Y1R expression in the PVN and arcuate hypothalamic nuclei. Blocking PVN Y1R before stress applied in the active phase, or administering NPY to the PVN before stress applied in the inactive phase, had a resounding behavioral effect. The time at which stress occurred significantly affected the behavioral stress response. Diurnal variations in HPA and NPY/Y1R significantly affect the behavioral response, conferring more resilience at the onset of the active phase and more vulnerability at the onset of the inactive phase, implying that NPY has a significant role in conferring resilience to stress-related psychopathology.
Collapse
Affiliation(s)
- Shlomi Cohen
- Faculty of Health Sciences, Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Ministry of Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Psychology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ella Vainer
- Faculty of Health Sciences, Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Ministry of Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Michael A Matar
- Faculty of Health Sciences, Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Ministry of Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nitsan Kozlovsky
- Faculty of Health Sciences, Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Ministry of Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Zeev Kaplan
- Faculty of Health Sciences, Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Ministry of Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Joseph Zohar
- Division of Psychiatry, State of Israel Ministry of Health, Chaim Sheba Medical Center, Sackler Medical School, Tel-Aviv University, Ramat-Gan, Israel
| | - Aleksander A Mathé
- Department of Neuroscience, Karolinska Institutet—Clinical Neuroscience, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Hagit Cohen
- Faculty of Health Sciences, Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Ministry of Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Psychology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
8
|
Steiner JL, Bardgett ME, Wolfgang L, Lang CH, Stocker SD. Glucocorticoids attenuate the central sympathoexcitatory actions of insulin. J Neurophysiol 2014; 112:2597-604. [PMID: 25185805 PMCID: PMC4233268 DOI: 10.1152/jn.00514.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/26/2014] [Indexed: 11/22/2022] Open
Abstract
Insulin acts within the central nervous system to regulate food intake and sympathetic nerve activity (SNA). Strong evidence indicates that glucocorticoids impair insulin-mediated glucose uptake and food intake. However, few data are available regarding whether glucocorticoids also modulate the sympathoexcitatory response to insulin. Therefore, the present study first confirmed that chronic administration of glucocorticoids attenuated insulin-induced increases in SNA and then investigated whether these effects were attributed to deficits in central insulin-mediated responses. Male Sprague-Dawley rats were given access to water or a drinking solution of the glucocorticoid agonist dexamethasone (0.3 μg/ml) for 7 days. A hyperinsulinemic-euglycemic clamp significantly increased lumbar SNA in control rats. This response was significantly attenuated in rats given access to dexamethasone for 7, but not 1, days. Similarly, injection of insulin into the lateral ventricle or locally within the arcuate nucleus (ARC) significantly increased lumbar SNA in control rats but this response was absent in rats given access to dexamethasone. The lack of a sympathetic response to insulin cannot be attributed to a generalized depression of sympathetic function or inactivation of ARC neurons as electrical activation of sciatic afferents or ARC injection of gabazine, respectively, produced similar increases in SNA between control and dexamethasone-treated rats. Western blot analysis indicates insulin produced similar activation of Akt Ser(473) and rpS6 Ser(240/244) in the ventral hypothalamus of control and dexamethasone-treated rats. Collectively, these findings suggest that dexamethasone attenuates the sympathoexcitatory actions of insulin through a disruption of ARC neuronal function downstream of Akt or mammalian target of rapamycin (mTOR) signaling.
Collapse
Affiliation(s)
- Jennifer L Steiner
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| | - Megan E Bardgett
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| | - Lawrence Wolfgang
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| | - Sean D Stocker
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania; Department of Neural and Behavioral Sciences, Pennsylvania State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
9
|
Bisschop PH, Dekker MJHJ, Osterthun W, Kwakkel J, Anink JJ, Boelen A, Unmehopa UA, Koper JW, Lamberts SWJ, Stewart PM, Swaab DF, Fliers E. Expression of 11β-hydroxysteroid dehydrogenase type 1 in the human hypothalamus. J Neuroendocrinol 2013; 25:425-32. [PMID: 23286317 DOI: 10.1111/jne.12017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 12/14/2012] [Accepted: 12/20/2012] [Indexed: 11/28/2022]
Abstract
The hypothalamus is a major target for glucocorticoids and a key structure for hypothalamic-pituitary-adrenal (HPA) axis setpoint regulation. The enzyme 11β hydroxysteroid dehydrogenase type 1 (11βHSD1) modulates glucocorticoid signalling in various tissues at the prereceptor level by converting biologically inactive cortisone to its active form cortisol. The present study aimed to assess 11βHSD1 expression in the human hypothalamus. We studied 11βHSD1 expression in five frozen and four formalin-fixed, paraffin-embedded human hypothalami (obtained from the Netherlands Brain Bank) by the polymerase chain reaction and immunocytochemistry, respectively. 11βHSD1 mRNA was expressed in the area of the suprachiasmatic nucleus, which is the biological clock of the brain, in the supraoptic nucleus and paraventricular nucleus (PVN), and in the infundibular nucleus, which is the human homologue of the rodent arcuate nucleus. 11βHSD1 was detected by immunocytochemistry in the same nuclei. In the PVN, neuronal 11βHSD1 immunoreactivity colocalised with corticotrophin-releasing hormone (CRH), arginine vasopressin and oxytocin, as shown by dual fluorescence staining. Our data demonstrate that 11βHSD1 is widely expressed in the human hypothalamus. Its colocalisation with CRH in the PVN suggests a role in modulation of glucocorticoid feedback of the HPA axis, whereas the expression of 11βHSD1 in additional and functionally diverse hypothalamic nuclei points to a role for the enzyme in the regulation of metabolism, appetite and circadian rhythms.
Collapse
Affiliation(s)
- P H Bisschop
- Department of Endocrinology and Metabolism, Academic Medical Centre, University of Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Evans SJ, Watson SJ, Akil H. Evaluation of sensitivity, performance and reproducibility of microarray technology in neuronal tissue. Integr Comp Biol 2012; 43:780-5. [PMID: 21680476 DOI: 10.1093/icb/43.6.780] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Microarray technology is a powerful technique that allows the simultaneous study of thousands of gene transcripts. During the past two years there has been an explosion of publications describing experiments utilizing microarray technology that range from original research findings from biological paradigms to mathematically modeled systems. However, neuroscientists using microarray technology face significant challenges due to high tissue complexity, low abundance transcripts, and small magnitude changes in transcript levels that have significant biological impact. This manuscript describes a series of studies designed to address issues regarding microarray sensitivity, ability of microarrays to detect subtle changes, and reproducibility of microarray experiments, all in the context of neuronal tissue. From the presentation of these studies, the authors argue that although microarray technology is limited with regards to sensitivity, the outcome of these experiments, if approached with appropriate skepticism, can be fruitful in the generation of hypotheses and seeding of future experiments.
Collapse
Affiliation(s)
- S J Evans
- Mental Health Research Institute, University of Michigan, Ann Arbor, Michigan 48109
| | | | | |
Collapse
|
11
|
Alkemade A, Yi CX, Pei L, Harakalova M, Swaab DF, la Fleur SE, Fliers E, Kalsbeek A. AgRP and NPY expression in the human hypothalamic infundibular nucleus correlate with body mass index, whereas changes in αMSH are related to type 2 diabetes. J Clin Endocrinol Metab 2012; 97:E925-33. [PMID: 22492775 DOI: 10.1210/jc.2011-3259] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Rodent data show that altered hypothalamic signaling contributes to the development of obesity and insulin resistance. OBJECTIVE To determine differences in hypothalamic expression levels of neuropeptide Y (NPY), agouti-related peptide (AgRP), and αMSH in the infundibular nucleus, the human equivalent of the arcuate nucleus, in relation to body mass index (BMI). In addition, the expression in the infundibular nucleus of eight subjects diagnosed with type 2 diabetes was measured to determine possible interference of type 2 diabetes with the association observed between neuropeptides and BMI. DESIGN We studied AgRP, NPY, and αMSH expression by means of quantitative immunocytochemistry in postmortem hypothalami of 30 subjects with known BMI. In separate experiments, we compared neuropeptide expression in eight subjects with type 2 diabetes with eight matched controls. RESULTS We found that AgRP immunoreactivity showed a U-shaped correlation with BMI. No evidence was found for possible influences of corticosteroid treatment. NPY immunoreactivity was significantly lower in overweight and obese subjects. αMSH did not correlate with BMI but was significantly lower in subjects with type 2 diabetes compared with controls. By contrast, NPY and AgRP expression was not affected in type 2 diabetes. CONCLUSION Our results indicate that the expression of AgRP and NPY are correlated with body weight changes, rather than the presence of type 2 diabetes, whereas changes in αMSH immunoreactivity are related to the presence of type 2 diabetes, indicating separate hypothalamic mechanisms.
Collapse
Affiliation(s)
- Anneke Alkemade
- Alan Turing Institute Almere, Louis Armstrongweg 84, 1311 RL Almere, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Ma Y, Wu X, Li X, Fu J, Shen J, Li X, Wang H. Corticosterone regulates the expression of neuropeptide Y and reelin in MLO-Y4 cells. Mol Cells 2012; 33:611-6. [PMID: 22610366 PMCID: PMC3887760 DOI: 10.1007/s10059-012-0053-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 04/18/2012] [Accepted: 04/19/2012] [Indexed: 12/13/2022] Open
Abstract
Osteocytes that have a dendritic appearance are widely believed to form a complex cellular network system and play crucial roles in mechanotransduction as a principal bone mechanosensor, which is the basis of their neuronallike biology, as previously reported. Neuropeptide Y (NPY) and reelin mRNA, which are brain-specific neurogenic markers, have been identified in osteocytes. However, changes in the production of NPY and reelin in response to specific biochemical stimulation are unknown. In this study, we investigated the in vitro effect of corticosterone, one of the endogenous glucocorticoids, on the expression of NPY and reelin in the MLO-Y4 osteocyte cell line. Cells were treated with corticosterone at different concentrations (10(-9) M-10(-5) M) for 1, 3, 6, 12 and 24 h. As revealed, corticosterone reduced the MLO-Y4 cell viability and proliferation in a dose- and time-dependent manner based on an MTT assay and a Vi-CELL analyzer. The cells were then incubated with corticosterone (10(-6) μM), and the NPY and reelin expression levels were detected at 1, 3, 6, 12 and 24 h using real-time PCR and Western blot analysis. These results demonstrated that at the gene and the protein levels, corticosterone significantly upregulated the NPY and reelin expression in a time-dependent manner. The application of a glucocorticoid receptor antagonist, RU486, reversed the reduced cell viability and the increased expression of NPY and reelin that were caused by corticosterone. To the best of our knowledge, this is the first report to verify that corticosterone regulates the NPY and reelin expression in osteocytes.
Collapse
Affiliation(s)
- Yuanyuan Ma
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041,
China
- Department of Prosthodontics, West China College of Stomatology, Sichuan University, Chengdu 610041,
China
| | - Xiangnan Wu
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041,
China
- Department of Prosthodontics, West China College of Stomatology, Sichuan University, Chengdu 610041,
China
| | - Xianxian Li
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041,
China
- Department of Prosthodontics, West China College of Stomatology, Sichuan University, Chengdu 610041,
China
| | - Jing Fu
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041,
China
- Department of Prosthodontics, West China College of Stomatology, Sichuan University, Chengdu 610041,
China
| | - Jiefei Shen
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041,
China
- Department of Prosthodontics, West China College of Stomatology, Sichuan University, Chengdu 610041,
China
| | - Xiaoyu Li
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041,
China
| | - Hang Wang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041,
China
- Department of Prosthodontics, West China College of Stomatology, Sichuan University, Chengdu 610041,
China
| |
Collapse
|
13
|
Yi CX, Foppen E, Abplanalp W, Gao Y, Alkemade A, la Fleur SE, Serlie MJ, Fliers E, Buijs RM, Tschöp MH, Kalsbeek A. Glucocorticoid signaling in the arcuate nucleus modulates hepatic insulin sensitivity. Diabetes 2012; 61:339-45. [PMID: 22210324 PMCID: PMC3266416 DOI: 10.2337/db11-1239] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Glucocorticoid receptors are highly expressed in the hypothalamic paraventricular nucleus (PVN) and arcuate nucleus (ARC). As glucocorticoids have pronounced effects on neuropeptide Y (NPY) expression and as NPY neurons projecting from the ARC to the PVN are pivotal for balancing feeding behavior and glucose metabolism, we investigated the effect of glucocorticoid signaling in these areas on endogenous glucose production (EGP) and insulin sensitivity by local retrodialysis of the glucocorticoid receptor agonist dexamethasone into the ARC or the PVN, in combination with isotope dilution and hyperinsulinemic-euglycemic clamp techniques. Retrodialysis of dexamethasone for 90 min into the ARC or the PVN did not have significant effects on basal plasma glucose concentration. During the hyperinsulinemic-euglycemic clamp, retrodialysis of dexamethasone into the ARC largely prevented the suppressive effect of hyperinsulinemia on EGP. Antagonizing the NPY1 receptors by intracerebroventricular infusion of its antagonist largely blocked the hepatic insulin resistance induced by dexamethasone in the ARC. The dexamethasone-ARC-induced inhibition of hepatic insulin sensitivity was also prevented by hepatic sympathetic denervation. These data suggest that glucocorticoid signaling specifically in the ARC neurons modulates hepatic insulin responsiveness via NPY and the sympathetic system, which may add to our understanding of the metabolic impact of clinical conditions associated with hypercortisolism.
Collapse
Affiliation(s)
- Chun-Xia Yi
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Gyengesi E, Liu ZW, D'Agostino G, Gan G, Horvath TL, Gao XB, Diano S. Corticosterone regulates synaptic input organization of POMC and NPY/AgRP neurons in adult mice. Endocrinology 2010; 151:5395-402. [PMID: 20843996 PMCID: PMC2954711 DOI: 10.1210/en.2010-0681] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Changes in circulating hormones, such as leptin and ghrelin, induce alterations in synaptic input organization and electrophysiological properties of neurons of the arcuate nucleus of the hypothalamus. To assess whether changes in circulating glucocorticoids also alter synaptic arrangement and membrane potential properties, we studied the effect of adrenalectomy (ADX) and corticosterone replacement in mice on the proopiomelanocortin (POMC) and neuropeptide Y (NPY)/agouti-related protein (AgRP) neurons of the hypothalamic arcuate nucleus. ADX reduced the number of symmetric, putative inhibitory synapses onto POMC neurons and the number of asymmetric, putative excitatory synapses onto NPY/AgRP neurons. Corticosterone replacement in ADX mice to levels similar to sham-operated animals restored the number of synapses onto POMC and NPY/AgRP neurons to that seen in sham-operated controls. The alterations in the synaptic arrangement in ADX mice were not due to their decrease in food intake as evidenced by the synaptic analysis of the pair-fed control animals. In line with the altered synaptic input organization, a depolarization of POMC membrane potential and a hyperpolarization of NPY/AgRP membrane potential were observed in ADX mice compared with their sham-operated controls. All of these changes reverted upon corticosterone replacement. These results reveal that the known orexigenic action of corticosteroids is mediated, at least in part, by synaptic changes and altered excitability of the melanocortin system.
Collapse
Affiliation(s)
- Erika Gyengesi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, P.O. Box 208063, New Haven, Connecticut 06520-208063, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Gaysinskaya VA, Karatayev O, Shuluk J, Leibowitz SF. Hyperphagia induced by sucrose: relation to circulating and CSF glucose and corticosterone and orexigenic peptides in the arcuate nucleus. Pharmacol Biochem Behav 2010; 97:521-30. [PMID: 21036188 DOI: 10.1016/j.pbb.2010.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 10/21/2010] [Accepted: 10/23/2010] [Indexed: 01/12/2023]
Abstract
Sucrose-rich diets compared to starch-rich diets are known to stimulate overeating under chronic conditions. The present study in normal-weight rats established an acute "preload-to-test meal" paradigm for demonstrating sucrose-induced hyperphagia and investigating possible mechanisms that mediate this behavioral phenomenon. In this acute paradigm, the rats were first given a small (15 kcal) sucrose preload (30% sucrose) for 30 min compared to an equicaloric, starch preload (25% starch with 5% sucrose) and then allowed to freely consume a subsequent test meal of lab chow. The sucrose preload, when compared to a starch preload equal in energy density and palatability, consistently increased food intake in the subsequent test meal occurring between 60 and 120 min after the end of the preload. Measurements of hormones, metabolites and hypothalamic peptides immediately preceding this hyperphagia revealed marked differences between the sucrose vs starch groups that could contribute to the increase in food intake. Whereas the sucrose group compared to the starch group immediately after the preload (at 10 min) had elevated levels of glucose in serum and cerebrospinal fluid (CSF) along with reduced expressions of neuropeptide Y (NPY) and agouti-related protein (AgRP) in the arcuate nucleus (ARC), the subsequent effects (at 30-60 min) just preceding the test meal hyperphagia were the reverse. Along with lower levels of glucose, they included markedly elevated serum and CSF levels of corticosterone and mRNA levels of NPY and AgRP in the ARC. In addition to establishing an animal model for sucrose-induced hyperphagia, these results demonstrate peripheral and central mechanisms that may mediate this behavioral phenomenon.
Collapse
Affiliation(s)
- V A Gaysinskaya
- The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | | | | | | |
Collapse
|
16
|
Brown LM, Clegg DJ. Central effects of estradiol in the regulation of food intake, body weight, and adiposity. J Steroid Biochem Mol Biol 2010; 122:65-73. [PMID: 20035866 PMCID: PMC2889220 DOI: 10.1016/j.jsbmb.2009.12.005] [Citation(s) in RCA: 224] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2009] [Revised: 11/19/2009] [Accepted: 12/15/2009] [Indexed: 12/14/2022]
Abstract
In recent years, obesity and its associated health disorders and costs have increased. Accumulation of adipose tissue, or fat, in the intra-abdominal adipose depot is associated with an increased risk of developing cardiovascular problems, type-2 diabetes mellitus, certain cancers, and other disorders like the metabolic syndrome. Males and females differ in terms of how and where their body fat is stored, in their hormonal secretions, and in their neural responses to signals regulating weight and body fat distribution. Men and post-menopausal women accumulate more fat in their intra-abdominal depots than pre-menopausal women, resulting in a greater risk of developing complications associated with obesity. The goal of this review is to discuss the current literature on sexual dimorphisms in body weight regulation, adipose tissue accrual and deposition.
Collapse
Affiliation(s)
- LM Brown
- Department of Nutrition, University of North Carolina Greensboro, Greensboro, NC 27412
| | - DJ Clegg
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390-8854
- Corresponding author at: Deborah J. Clegg, RD, PhD, Assistant Professor, Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, 5323 Harry Hines Blvd., K5.252, Dallas, TX 75390-8854, Tel: 214-648-3401, Fax: 214-648-8720, (D. Clegg)
| |
Collapse
|
17
|
Effects of corticosterone treatment on responses to fasting in Japanese quail. Comp Biochem Physiol A Mol Integr Physiol 2009; 154:211-5. [DOI: 10.1016/j.cbpa.2009.06.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 06/08/2009] [Accepted: 06/09/2009] [Indexed: 11/20/2022]
|
18
|
Maternal high-fat diet and fetal programming: increased proliferation of hypothalamic peptide-producing neurons that increase risk for overeating and obesity. J Neurosci 2009; 28:12107-19. [PMID: 19005075 DOI: 10.1523/jneurosci.2642-08.2008] [Citation(s) in RCA: 281] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Recent studies in adult and weanling rats show that dietary fat, in close association with circulating lipids, can stimulate expression of hypothalamic peptides involved in controlling food intake and body weight. In the present study, we examined the possibility that a fat-rich diet during pregnancy alters the development of these peptide systems in utero, producing neuronal changes in the offspring that persist postnatally in the absence of the diet and have long-term consequences. The offspring of dams on a high-fat diet (HFD) versus balanced diet (BD), from embryonic day 6 to postnatal day 15 (P15), showed increased expression of orexigenic peptides, galanin, enkephalin, and dynorphin, in the paraventricular nucleus and orexin and melanin-concentrating hormone in the perifornical lateral hypothalamus. The increased density of these peptide-expressing neurons, evident in newborn offspring as well as P15 offspring cross-fostered at birth to dams on the BD, led us to examine events that might be occurring in utero. During gestation, the HFD stimulated the proliferation of neuroepithelial and neuronal precursor cells of the embryonic hypothalamic third ventricle. It also stimulated the proliferation and differentiation of neurons and their migration toward hypothalamic areas where ultimately a greater proportion of the new neurons expressed the orexigenic peptides. This increase in neurogenesis, closely associated with a marked increase in lipids in the blood, may have a role in producing the long-term behavioral and physiological changes observed in offspring after weaning, including an increase in food intake, preference for fat, hyperlipidemia, and higher body weight.
Collapse
|
19
|
Velkoska E, Cole TJ, Dean RG, Burrell LM, Morris MJ. Early undernutrition leads to long-lasting reductions in body weight and adiposity whereas increased intake increases cardiac fibrosis in male rats. J Nutr 2008; 138:1622-7. [PMID: 18716160 DOI: 10.1093/jn/138.9.1622] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Previous studies suggest that both overfeeding and undernutrition during development increase the risk of obesity and hypertension in adulthood. In this study, we examined both short- (24 d) and long- (16 wk) term effects of early postnatal over- and underfeeding in rats on body weight, body composition, plasma hormones, adiposity markers, and hypothalamic neuropeptide Y content. Cardiovascular changes were also examined by measuring blood pressure and cardiac fibrosis. Rats raised in litters of 3, 12, or 18 pups per mother were used to model early onset overfeeding, control, and underfeeding, respectively. At 24 d of age, pups raised in small litters (SL) were 10% heavier than pups from normal litters, accompanied by increased organ mass and fat mass, elevated plasma leptin, corticosterone, and uncoupling protein-1 mRNA in brown adipose tissue. On the other hand, pups raised in large litters were 17% lighter with no significant changes in plasma leptin. Overfeeding during the first 3 wk of life led to increased plasma leptin concentration in adulthood, whereas underfed rats remained significantly lighter throughout the study, with no evidence of catch-up growth. Rats raised in SL were more susceptible to developing cardiac fibrosis with a 22% increase in collagen deposition compared with control rats at 16 wk of age (P < 0.05). This was independent of any changes in blood pressure. This study demonstrates that nutritional changes early in postnatal development can have long-lasting effects on body weight, adiposity, and some mediators involved in energy homeostasis and can also lead to structural changes in the heart in adulthood. This highlights the importance of identifying potential early life risk factors involved in the modulation of childhood nutrition.
Collapse
Affiliation(s)
- Elena Velkoska
- Department of Pharmacology, University of Melbourne, Parkville, 3010, Australia
| | | | | | | | | |
Collapse
|
20
|
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
|
21
|
Grundemar L. Editorial Central & Peripheral Nervous Systems: Will neuropeptide Y receptor antagonists offer new therapeutic approaches? Expert Opin Ther Pat 2008. [DOI: 10.1517/13543776.5.10.1007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
22
|
Beck B. Neuropeptide Y in normal eating and in genetic and dietary-induced obesity. Philos Trans R Soc Lond B Biol Sci 2007; 361:1159-85. [PMID: 16874931 PMCID: PMC1642692 DOI: 10.1098/rstb.2006.1855] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neuropeptide Y (NPY) is one the most potent orexigenic peptides found in the brain. It stimulates food intake with a preferential effect on carbohydrate intake. It decreases latency to eat, increases motivation to eat and delays satiety by augmenting meal size. The effects on feeding are mediated through at least two receptors, the Y1 and Y5 receptors. The NPY system for feeding regulation is mostly located in the hypothalamus. It is formed of the arcuate nucleus (ARC), where the peptide is synthesized, and the paraventricular (PVN), dorsomedial (DMN) and ventromedial (VMN) nuclei and perifornical area where it is active. This activity is modulated by the hindbrain and limbic structures. It is dependent on energy availability, e.g. upregulation with food deprivation or restriction, and return to baseline with refeeding. It is also sensitive to diet composition with variable effects of carbohydrates and fats. Leptin signalling and glucose sensing which are directly linked to diet type are the most important factors involved in its regulation. Absence of leptin signalling in obesity models due to gene mutation either at the receptor level, as in the Zucker rat, the Koletsky rat or the db/db mouse, or at the peptide level, as in ob/ob mouse, is associated with increased mRNA abundance, peptide content and/or release in the ARC or PVN. Other genetic obesity models, such as the Otsuka-Long-Evans-Tokushima Fatty rat, the agouti mouse or the tubby mouse, are characterized by a diminution in NPY expression in the ARC nucleus and by a significant increase in the DMN. Further studies are necessary to determine the exact role of NPY in these latter models. Long-term exposure to high-fat or high-energy palatable diets leads to the development of adiposity and is associated with a decrease in hypothalamic NPY content or expression, consistent with the existence of a counter-regulatory mechanism to diminish energy intake and limit obesity development. On the other hand, an overactive NPY system (increased mRNA expression in the ARC associated with an upregulation of the receptors) is characteristic of rats or rodent strains sensitive to dietary-induced obesity. Finally, NPY appears to play an important role in body weight and feeding regulation, and while it does not constitute the only target for drug treatment of obesity, it may nevertheless provide a useful target in conjunction with others.
Collapse
Affiliation(s)
- B Beck
- Université Henri Poincaré, Neurocal, Nancy, France.
| |
Collapse
|
23
|
Pecoraro N, Dallman MF, Warne JP, Ginsberg AB, Laugero KD, la Fleur SE, Houshyar H, Gomez F, Bhargava A, Akana SF. From Malthus to motive: how the HPA axis engineers the phenotype, yoking needs to wants. Prog Neurobiol 2006; 79:247-340. [PMID: 16982128 DOI: 10.1016/j.pneurobio.2006.07.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Revised: 07/17/2006] [Accepted: 07/24/2006] [Indexed: 01/28/2023]
Abstract
The hypothalamo-pituitary-adrenal (HPA) axis is the critical mediator of the vertebrate stress response system, responding to environmental stressors by maintaining internal homeostasis and coupling the needs of the body to the wants of the mind. The HPA axis has numerous complex drivers and highly flexible operating characterisitics. Major drivers include two circadian drivers, two extra-hypothalamic networks controlling top-down (psychogenic) and bottom-up (systemic) threats, and two intra-hypothalamic networks coordinating behavioral, autonomic, and neuroendocrine outflows. These various networks jointly and flexibly control HPA axis output of periodic (oscillatory) functions and a range of adventitious systemic or psychological threats, including predictable daily cycles of energy flow, actual metabolic deficits over many time scales, predicted metabolic deficits, and the state-dependent management of post-prandial responses to feeding. Evidence is provided that reparation of metabolic derangement by either food or glucocorticoids results in a metabolic signal that inhibits HPA activity. In short, the HPA axis is intimately involved in managing and remodeling peripheral energy fluxes, which appear to provide an unidentified metabolic inhibitory feedback signal to the HPA axis via glucocorticoids. In a complementary and perhaps a less appreciated role, adrenocortical hormones also act on brain to provide not only feedback, but feedforward control over the HPA axis itself and its various drivers, as well as coordinating behavioral and autonomic outflows, and mounting central incentive and memorial networks that are adaptive in both appetitive and aversive motivational modes. By centrally remodeling the phenotype, the HPA axis provides ballistic and predictive control over motor outflows relevant to the type of stressor. Evidence is examined concerning the global hypothesis that the HPA axis comprehensively induces integrative phenotypic plasticity, thus remodeling the body and its governor, the brain, to yoke the needs of the body to the wants of the mind. Adverse side effects of this yoking under conditions of glucocorticoid excess are discussed.
Collapse
Affiliation(s)
- Norman Pecoraro
- Department of Physiology, University of California, San Francisco, CA 94143-0444, United States.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Eva C, Serra M, Mele P, Panzica G, Oberto A. Physiology and gene regulation of the brain NPY Y1 receptor. Front Neuroendocrinol 2006; 27:308-39. [PMID: 16989896 DOI: 10.1016/j.yfrne.2006.07.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2005] [Revised: 07/18/2006] [Accepted: 07/25/2006] [Indexed: 10/24/2022]
Abstract
Neuropeptide Y (NPY) is one of the most prominent and abundant neuropeptides in the mammalian brain where it interacts with a family of G-protein coupled receptors, including the Y(1) receptor subtype (Y(1)R). NPY-Y(1)R signalling plays a prominent role in the regulation of several behavioural and physiological functions including feeding behaviour and energy balance, sexual hormone secretion, stress response, emotional behaviour, neuronal excitability and ethanol drinking. Y(1)R expression is regulated by neuronal activity and peripheral hormones. The Y(1)R gene has been isolated from rodents and humans and it contains multiple regulatory elements that may participate in the regulation of its expression. Y(1)R expression in the hypothalamus is modulated by changes in energetic balance induced by a wide variety of conditions (fasting, pregnancy, hyperglycaemic challenge, hypophagia, diet induced obesity). Estrogens up-regulate responsiveness to NPY to stimulate preovulatory GnRH and gonadotropin surges by increasing Y(1)R gene expression both in the hypothalamus and the pituitary. Y(1)R expression is modulated by different kinds of brain insults, such as stress and seizure activity, and alteration in its expression may contribute to antidepressant action. Chronic modulation of GABA(A) receptor function by benzodiazepines or neuroactive steroids also affects Y(1)R expression in the amygdala, suggesting that a functional interaction between the GABA(A) receptor and Y(1)R mediated signalling may contribute to the regulation of emotional behaviour. In this paper, we review the state of the art concerning Y(1)R function and gene expression, including our personal contribution to many of the subjects mentioned above.
Collapse
Affiliation(s)
- Carola Eva
- Sezione di Farmacologia, Dipartimento di Anatomia, Farmacologia e Medicina Legale, Università di Torino, Italy; Centro Rita Levi Montalcini, Università di Torino, Italy.
| | | | | | | | | |
Collapse
|
25
|
Watts AG. Glucocorticoid regulation of peptide genes in neuroendocrine CRH neurons: a complexity beyond negative feedback. Front Neuroendocrinol 2005; 26:109-30. [PMID: 16289311 DOI: 10.1016/j.yfrne.2005.09.001] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Accepted: 09/14/2005] [Indexed: 11/19/2022]
Abstract
This review will examine our current knowledge of a fundamental property of CRH neuroendocrine neurons: how the major endpoint of the HPA axis--adrenal glucocorticoids--interacts with the mechanisms controlling the expression of the genes that encode ACTH secretogogues. A great deal of work over the past 25 years has led to the notion that this question has an ostensibly simple answer: glucocorticoids inhibit peptide gene expression using "negative feedback" at the CRH neuron and elsewhere. However, closely examining how glucocorticoids act in different physiological circumstances reveals a much more complex set of answers, particularly if we consider how the processes that control peptide synthesis and release are coupled. Out of this examination emerges a more flexible and complex framework for examining the integrative mechanisms controlling the CRH neuron. Although we will mostly focus on the Crh gene, relevant aspects of the vasopressin (Avp) and pro-enkephalin (pEnk) gene regulatory mechanisms will also be discussed.
Collapse
Affiliation(s)
- Alan G Watts
- The Neuroscience Research Institute, and The Department of Biological Sciences, USC College, University of Southern California, Los Angeles, USA.
| |
Collapse
|
26
|
Ford GK, Al-Barazanji KA, Wilson S, Jones DNC, Harbuz MS, Jessop DS. Orexin expression and function: glucocorticoid manipulation, stress, and feeding studies. Endocrinology 2005; 146:3724-31. [PMID: 15961555 DOI: 10.1210/en.2005-0496] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We investigated the effects of glucocorticoid manipulation on orexin-A-induced feeding and prepro-orexin mRNA levels in the lateral hypothalamic area (LHA) of the rat brain. Adrenalectomy (ADX) reduced orexin-A-induced feeding over 4 h by about 60%, compared with shams, an effect that was reversed by corticosterone (CORT) replacement. ADX had no effect on prepro-orexin mRNA levels in the LHA in either the morning or the evening; however, message was up-regulated by CORT in the morning but not the evening. An increased number of emulsion grains per cell in the LHA suggests that this is a specific increase in prepro-orexin mRNA and is not due to an increased number of cells expressing message. Prepro-orexin mRNA levels in the LHA were elevated 4 h after injection of lipopolysaccharide, compared with saline-injected controls. Partial but not complete abolition of orexin-A-induced feeding by ADX suggests that orexin-A-induced feeding may be mediated through glucocorticoid-dependent and glucocorticoid-independent pathways. In the morning increased prepro-orexin mRNA after CORT replacement demonstrates that orexin expression is sensitive to increased concentrations of glucocorticoids. However, the lack of effect of ADX on prepro-orexin mRNA levels suggests that endogenous glucocorticoids are not involved in tonic regulation of basal prepro-orexin expression. Overall our data constitute a body of evidence for an integrated relationship between central orexin expression, stress, glucocorticoid manipulation, and feeding patterns in the rat.
Collapse
Affiliation(s)
- Gemma K Ford
- Henry Wellcome Laboratory for Integrative Neuroscience and Endocrinology, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom
| | | | | | | | | | | |
Collapse
|
27
|
Velísek L. Prenatal corticosteroid impact on hippocampus: implications for postnatal outcomes. Epilepsy Behav 2005; 7:57-67. [PMID: 15975854 PMCID: PMC1432088 DOI: 10.1016/j.yebeh.2005.04.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Accepted: 04/20/2005] [Indexed: 10/25/2022]
Abstract
Prenatal administration of corticosteroids is common in obstetrics to improve the outcome of premature deliveries. Many pregnant women receive multiple corticosteroid courses. Long-term follow-up studies in humans are limited, but those available suggest detrimental effects on the behavior of those children. Animal data also show adverse effects of prenatal corticosteroids mainly in the hippocampus, a structure sensitive to corticosteroid action. Several molecules involved in neuronal survival, seizure susceptibility, and behavior have been identified as possible targets of prenatal corticosteroid effects. These molecules include hippocampal glucocorticoid receptors, brain-derived neurotrophic factor, corticotropin-releasing hormone, and neuropeptide Y. Prenatal corticosteroid treatment permanently reprograms expression of these molecules. The future goals of research in this area include development of specific antagonists of corticosteroid activation pathways that would help differentiate between positive main effects and undesired adverse effects of prenatally administered corticosteroids.
Collapse
Affiliation(s)
- Libor Velísek
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, Einstein/Montefiore Epilepsy Management Center, Bronx, NY 10461, USA.
| |
Collapse
|
28
|
Coll AP, Challis BG, López M, Piper S, Yeo GSH, O'Rahilly S. Proopiomelanocortin-deficient mice are hypersensitive to the adverse metabolic effects of glucocorticoids. Diabetes 2005; 54:2269-76. [PMID: 16046291 DOI: 10.2337/diabetes.54.8.2269] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Congenital lack of proopiomelanocortin (POMC) causes obesity and glucocorticoid deficiency. The responses of Pomc-/- and wild-type mice to the administration of corticosterone were compared. In study 1, mice were given corticosterone-supplemented water (CORT) for 10 days, resulting in plasma CORT levels within the physiological range, with partial suppression of hypothalamic corticotropin-releasing hormone expression to a similar degree between genotypes. Body weight, fat mass, and food intake increased in CORT-treated Pomc-/- but not wild-type mice. CORT increased plasma insulin levels 50-fold in Pomc-/- versus 14-fold in wild-type mice (P < 0.01) and increased hypothalamic agouti-related protein (AgRP) expression by more than 200% in Pomc-/- versus 40% in wild type (P < 0.05). In study 2, mice were given CORT from weaning, and Pomc-/- but not wild-type mice developed hyperglycemia, ketonuria, and hepatic steatosis by 8-12 weeks. Thus, Pomc-/- mice are hypersensitive to the adverse metabolic effects of glucocorticoids. Additionally, as the levels of plasma CORT achieved, especially in study 1, were not grossly supraphysiological, we conclude that glucocorticoid deficiency may afford Pomc-/- mice some protection from the full adverse consequences of melanocortin deficiency. This may occur through a mechanism involving the suppression of AgRP by the hypoadrenal state.
Collapse
Affiliation(s)
- Anthony P Coll
- University Department of Medicine,Addenbrooke's Hospital, Cambridge CB2 2QR, UK
| | | | | | | | | | | |
Collapse
|
29
|
Chang GQ, Karatayev O, Davydova Z, Wortley K, Leibowitz SF. Glucose injection reduces neuropeptide Y and agouti-related protein expression in the arcuate nucleus: A possible physiological role in eating behavior. ACTA ACUST UNITED AC 2005; 135:69-80. [PMID: 15857670 DOI: 10.1016/j.molbrainres.2004.12.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2004] [Revised: 11/29/2004] [Accepted: 12/05/2004] [Indexed: 12/25/2022]
Abstract
Evidence suggests that neuropeptide Y (NPY) and agouti-related protein (AgRP) in the arcuate nucleus (ARC) are modulated by glucoregulatory hormones and involved in maintaining normal eating patterns and glucose homeostasis in states of energy deficiency. This study investigated whether these peptides respond to glucose itself under conditions, e.g., before the nocturnal feeding cycle, when carbohydrate stores are low. After removal of food 3 h before dark onset, Sprague-Dawley rats were given a single, intraperitoneal (i.p.) injection of saline or 10% glucose (0.13 g/kg) and were sacrificed at different intervals, from 3.5 to 90 min later, for measurements of circulating hormones and metabolites or of NPY and AgRP mRNA in the ARC. With no change in insulin, leptin, or triglycerides, glucose injection produced a 1.8-mM rise in circulating glucose during the first 15 min, followed by a 30-60% reduction in NPY and AgRP mRNA at 30 and 60 min post-injection. A similar effect was observed with intraventricular administration of 5% glucose. At 90 min, however, this suppressive effect of i.p. glucose relative to saline was lost and actually reversed into a 50% increase in NPY and AgRP, possibly attributed to a decline in circulating glucose followed by a 50% rise in corticosterone at 60 min. These biphasic shifts over a 90-min period may reflect mechanisms underlying natural eating patterns at the onset of the nocturnal cycle, when spontaneous meals are approximately 90 min apart and rich in carbohydrate, glucose levels are low, and corticosterone and ARC peptides naturally peak.
Collapse
Affiliation(s)
- Guo-Qing Chang
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
30
|
Leibowitz SF, Sepiashvili K, Akabayashi A, Karatayev O, Davydova Z, Alexander JT, Wang J, Chang GQ. Function of neuropeptide Y and agouti-related protein at weaning: relation to corticosterone, dietary carbohydrate and body weight. Brain Res 2005; 1036:180-91. [PMID: 15725416 DOI: 10.1016/j.brainres.2004.12.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2004] [Revised: 12/14/2004] [Accepted: 12/17/2004] [Indexed: 11/18/2022]
Abstract
Neuropeptide Y (NPY) and agouti-related protein (AgRP), potent stimulants of feeding, have been linked in adult rats to both corticosterone (CORT) and dietary carbohydrate. To understand the significance of this relationship early in life, measurements were taken of these parameters at different ages around weaning, in rats given a choice of macronutrient diets or maintained on a carbohydrate-rich diet. The results demonstrate that, in both male and female rat pups, the expression and production of NPY and AgRP in the arcuate nucleus (ARC) peak on postnatal day 21 (P21), compared to P15 before weaning and P27 after weaning. These elevated levels of peptide were associated with peak levels of CORT and glucose and also a strong, natural preference for carbohydrate at weaning, which accounted for 55-65% of the pups' total diet. In subgroups defined by their body weight at these stages, rats with as little as 4% lower body weight (compared to higher weight pups) had 30-60% greater expression of NPY and AgRP in the ARC and elevated levels of CORT, with no difference in leptin or insulin. This response was significantly more pronounced at P21 than at P15 or P27. The importance of carbohydrate during this stage was suggested by additional results showing elevated NPY expression, CORT levels, body weight and inguinal fat pad weights in P27 pups raised on a 65% carbohydrate diet vs. 45% carbohydrate. These results suggest that hypothalamic NPY and AgRP, together with CORT, have glucoregulatory as well as feeding stimulatory functions that help mediate the transition from suckling of a fat-rich diet to independent feeding of a carbohydrate-rich diet. During this critical period, the carbohydrate together with the peptides and CORT provide the important signals, including elevated glucose, that promote de novo lipogenesis and enable weanling animals to survive periods of food deprivation.
Collapse
Affiliation(s)
- Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Bugarith K, Dinh TT, Li AJ, Speth RC, Ritter S. Basomedial hypothalamic injections of neuropeptide Y conjugated to saporin selectively disrupt hypothalamic controls of food intake. Endocrinology 2005; 146:1179-91. [PMID: 15604214 DOI: 10.1210/en.2004-1166] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neuropeptide Y (NPY) conjugated to saporin (NPY-SAP), a ribosomal inactivating toxin, is a newly developed compound designed to selectively target and lesion NPY receptor-expressing cells. We injected NPY-SAP into the basomedial hypothalamus (BMH), just dorsal to the arcuate nucleus (ARC), to investigate its neurotoxicity and to determine whether ARC NPY neurons are required for glucoprivic feeding. We found that NPY-SAP profoundly reduced NPY Y1 receptor and alpha MSH immunoreactivity, as well as NPY, Agouti gene-related protein (AGRP), and cocaine and amphetamine-related transcript mRNA expression in the BMH. NPY-SAP lesions were localized to the injection site with no evidence of retrograde transport by hindbrain NPY neurons with BMH terminals. These lesions impaired responses to intracerebroventricular (icv) leptin (5 microg/5 microl x d) and ghrelin (2 microg/5 microl), which are thought to alter feeding primarily by actions on ARC NPY/AGRP and proopiomelanocortin/cocaine and amphetamine-related transcript neurons. However, the hypothesis that NPY/AGRP neurons are required downstream mediators of glucoprivic feeding was not supported. Although NPY/AGRP neurons were destroyed by NPY-SAP, the lesion did not impair either the feeding or the hyperglycemic response to 2-deoxy-D-glucose-induced blockade of glycolysis use. Similarly, responses to glucagon-like peptide-1 (GLP-1, 5 microg/3 microl icv), NPY (5 microg/3 microl icv), cholecystokinin octapeptide (4 microg/kg ip), and beta-mercaptoacetate (68 mg/kg ip) were not altered by the NPY-SAP lesion. Thus, NPY-SAP destroyed NPY receptor-expressing neurons in the ARC and selectively disrupted controls of feeding dependent on those neurons but did not disrupt peptidergic or metabolic controls dependent upon circuitry outside the BMH.
Collapse
Affiliation(s)
- Kishor Bugarith
- Programs in Neuroscience, Washington State University, Pullman, Washington 99164-6520, USA
| | | | | | | | | |
Collapse
|
32
|
Thorsell A, Slawecki CJ, Ehlers CL. Effects of neuropeptide Y and corticotropin-releasing factor on ethanol intake in Wistar rats: interaction with chronic ethanol exposure. Behav Brain Res 2005; 161:133-40. [PMID: 15904720 DOI: 10.1016/j.bbr.2005.01.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2005] [Accepted: 01/31/2005] [Indexed: 11/25/2022]
Abstract
Neuropeptide Y (NPY) and corticotropin-releasing factor (CRF) have opposing effects on stress-associated and consummatory behaviors in rodents. Recent studies also suggest that both peptides influence ethanol intake. In the present study, the effects of administration of CRF and NPY into the lateral ventricle on ethanol intake in naive and ethanol-vapor-exposed Wistar rats were examined. A limited access paradigm was used to measure intake of a 10% (v/v) ethanol solution in Wistar rats trained to drink using a sucrose fading procedure. Ethanol vapor exposure for 8 weeks significantly elevated ethanol intake in this limited access paradigm relative to pre-exposure levels. The effects of icv administration of CRF (1 microg), NPY (10 microg) or NPY/CRF combined (10 and 1 microg, respectively) on ethanol intake were then assessed. In non-vapor-exposed subjects, icv infusion of NPY had no effect on ethanol intake, while a significant suppression of drinking was seen following icv administration of CRF. Administration of NPY in combination with CRF had no effect on ethanol intake in non-ethanol-vapor-exposed rats. In vapor-exposed subjects, both NPY and CRF reduced ethanol intake, but when given in combination, no difference from vehicle was detected. Locomotor activity was measured during drinking sessions and was unaffected by peptide administration. These studies underscore the importance of a history of exposure to chronic ethanol vapor in the regulation of ethanol intake by NPY. Furthermore, the results presented here suggest that a balance between the stress-related peptides NPY and CRF may be involved in the regulation of ethanol intake.
Collapse
Affiliation(s)
- Annika Thorsell
- The Scripps Research Institute, Department of Neuropharmacology, Mail Drop CVN-14, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
33
|
Vicentic A, Dominguez G, Hunter RG, Philpot K, Wilson M, Kuhar MJ. Cocaine- and amphetamine-regulated transcript peptide levels in blood exhibit a diurnal rhythm: regulation by glucocorticoids. Endocrinology 2004; 145:4119-24. [PMID: 15155577 DOI: 10.1210/en.2003-1648] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cocaine- and amphetamine-regulated transcript (CART) peptides are novel neurotransmitters that are implicated in several physiological functions such as control of feeding behavior, drug reward, sensory processing, stress, and development. Although a majority of studies have examined the role of CART in the brain, less is known about its function in the periphery. Therefore, the goals of this study were to examine the levels and species of CART peptides in blood, to determine whether they undergo diurnal rhythms, and to elucidate their sources and regulatory factors. RIA showed that CART peptides are present in the blood of rats and monkeys and that they exhibit a diurnal variation. Western blotting confirmed the pattern of diurnal variation in rats and, additionally, showed that CART immunoreactivity was due to a single predominant fragment with an apparent molecular weight in the range of the active CART 55-102 peptide. Adrenalectomy caused a 70% reduction in CART peptide levels in rat blood, and this was reversed by corticosterone replacement. CART levels paralleled glucocorticoid levels in rat and monkey blood. Control of CART levels by corticosterone suggests the possibility that CART peptides in blood may be influenced by hypothalamic-pituitary-adrenal interactions and that they may play a role in glucocorticoid-related processes such as stress.
Collapse
Affiliation(s)
- A Vicentic
- Yerkes National Primate Research Center of Emory University, 954 Gatewood Road NE, Atlanta, Georgia 30329, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Energy balance is maintained via a homeostatic system involving both the brain and the periphery. A key component of this system is the hypothalamus. Over the past two decades, major advances have been made in identifying an increasing number of peptides within the hypothalamus that contribute to the process of energy homeostasis. Under stable conditions, equilibrium exists between anabolic peptides that stimulate feeding behavior, as well as decrease energy expenditure and lipid utilization in favor of fat storage, and catabolic peptides that attenuate food intake, while stimulating sympathetic nervous system (SNS) activity and restricting fat deposition by increasing lipid metabolism. The equilibrium between these neuropeptides is dynamic in nature. It shifts across the day-night cycle and from day to day and also in response to dietary challenges as well as peripheral energy stores. These shifts occur in close relation to circulating levels of the hormones, leptin, insulin, ghrelin and corticosterone, and also the nutrients, glucose and lipids. These circulating factors together with neural processes are primary signals relaying information regarding the availability of fuels needed for current cellular demand, in addition to the level of stored fuels needed for long-term use. Together, these signals have profound impact on the expression and production of neuropeptides that, in turn, initiate the appropriate anabolic or catabolic responses for restoring equilibrium. In this review, we summarize the evidence obtained on nine peptides in the hypothalamus that have emerged as key players in this process. Data from behavioral, physiological, pharmacological and genetic studies are described and consolidated in an attempt to formulate a clear statement on the underlying function of each of these peptides and also on how they work together to create and maintain energy homeostasis.
Collapse
Affiliation(s)
- Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA.
| | | |
Collapse
|
35
|
Fraley GS, Ritter S. Immunolesion of norepinephrine and epinephrine afferents to medial hypothalamus alters basal and 2-deoxy-D-glucose-induced neuropeptide Y and agouti gene-related protein messenger ribonucleic acid expression in the arcuate nucleus. Endocrinology 2003; 144:75-83. [PMID: 12488332 DOI: 10.1210/en.2002-220659] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neuropeptide Y (NPY) and agouti gene-related protein (AGRP) are orexigenic peptides of special importance for control of food intake. In situ hybridization studies have shown that NPY and AGRP mRNAs are increased in the arcuate nucleus of the hypothalamus (ARC) by glucoprivation. Other work has shown that glucoprivation stimulates food intake by activation of hindbrain glucoreceptor cells and requires the participation of rostrally projecting norepinephrine (NE) or epinephrine (E) neurons. Here we determine the role of hindbrain catecholamine afferents in glucoprivation-induced increase in ARC NPY and AGRP gene expression. The selective NE/E immunotoxin saporin-conjugated antidopamine-beta-hydroxylase (anti-dbetah) was microinjected into the medial hypothalamus and expression of AGRP and NPY mRNA was analyzed subsequently in the ARC under basal and glucoprivic conditions using (33)P-labeled in situ hybridization. Saporin-conjugated anti-dbetah virtually eliminated dbetah-immunoreactive terminals in the ARC without causing nonspecific damage. These lesions significantly increased basal but eliminated 2-deoxy-D-glucose-induced increases in AGRP and NPY mRNA expression. Results indicate that hindbrain catecholaminergic neurons contribute to basal NPY and AGRP gene expression and mediate the responsiveness of NPY and AGRP neurons to glucose deficit. Our results also suggest that catecholamine neurons couple potent orexigenic neural circuitry within the hypothalamus with hindbrain glucose sensors that monitor brain glucose supply.
Collapse
Affiliation(s)
- G S Fraley
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology, Washington State University, Pullman, Washington 99164-6520, USA
| | | |
Collapse
|
36
|
Sainsbury A, Schwarzer C, Couzens M, Herzog H. Y2 receptor deletion attenuates the type 2 diabetic syndrome of ob/ob mice. Diabetes 2002; 51:3420-7. [PMID: 12453895 DOI: 10.2337/diabetes.51.12.3420] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Hypothalamic neuropeptide Y (NPY) is implicated in the regulation of a variety of physiological functions, notably energy homeostasis and reproduction. Chronically elevated NPY levels in the hypothalamus, as in genetically obese ob/ob mice, are associated with obesity, a syndrome of type 2 diabetes, and infertility. However, it is not known which of the five cloned Y receptors mediate these effects. Here, we show that crossing the Y2 receptor knockout mouse (Y2(-/-)) onto the ob/ob background attenuates the increased adiposity, hyperinsulinemia, hyperglycemia, and increased hypothalamo-pituitary-adrenal (HPA) axis activity of ob/ob mice. Compared with lean controls, ob/ob mice had elevated expression of NPY and agouti-related protein (AgRP) mRNA in the arcuate nucleus and decreased expression of proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) mRNA. Y2 deletion in ob/ob mice significantly increased the hypothalamic POMC mRNA expression, with no effect on NPY, AgRP, or CART expression. [Y2(-/-)ob/ob] mice were no different from ob/ob littermates with respect to food intake and body weight, and Y2 receptor deficiency had no beneficial effect on the infertility or the reduced hypothalamo-pituitary-gonadotropic function of ob/ob mice. These data demonstrate that Y2 receptors mediate the obese type 2 diabetes phenotype of ob/ob mice, possibly via alterations in melanocortin tonus in the arcuate nucleus and/or effects on the HPA axis.
Collapse
Affiliation(s)
- Amanda Sainsbury
- Neurobiology Program, Garvan Institute of Medical Research, St. Vincent's Hospital, 384 Victoria Street, Darlinghurst NSW 2010, Sydney, Australia
| | | | | | | |
Collapse
|
37
|
Lu XY, Shieh KR, Kabbaj M, Barsh GS, Akil H, Watson SJ. Diurnal rhythm of agouti-related protein and its relation to corticosterone and food intake. Endocrinology 2002; 143:3905-15. [PMID: 12239102 DOI: 10.1210/en.2002-220150] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In the present study we examined the diurnal patterns of agouti-related protein (AGRP) and proopiomelanocortin (POMC) mRNA expression in the arcuate nucleus and their relation to circulating glucocorticoids and food intake. Animals were killed at 4-h intervals throughout the 24-h diurnal cycle, and the expression of AGRP and POMC mRNA was evaluated by semiquantitative in situ hybridization analysis. We observed a significant diurnal rhythm in AGRP mRNA expression, with a marked peak at 2200 h (4 h after lights off) and a trough at 1000 h (4 h after lights on), consistent with the overall day-night rhythm of food intake. In contrast, POMC mRNA levels did not show a significant fluctuation across the diurnal cycle, although there was a tendency for levels to decrease after the onset of the dark cycle. Corticosterone secretion temporally coincided with the rising phase of AGRP mRNA expression. Depletion of corticosterone by adrenalectomy abolished the AGRP diurnal rhythm by suppressing the nighttime expression, but did not alter the feeding rhythm. Exposure of adrenalectomized rats to constant corticosterone replacement (10 or 50 mg continuous release corticosterone pellet) resulted in fixed AGRP mRNA expression throughout the 12-h light, 12-h dark cycle. A relatively high level of corticosterone (50 mg) significantly increased AGRP mRNA expression, with a positive correlation between these two measures. These results indicate that 1) the diurnal expression of AGRP mRNA is regulated by corticosterone independently of the light/dark cue; and 2) a normal endogenous corticosterone rhythm is required for generating the diurnal AGRP rhythm.
Collapse
Affiliation(s)
- Xin-Yun Lu
- University of Michigan School of Medicine, Mental Health Research Institute, Ann Arbor 48109, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Bray GA, York DA. Obesity. Compr Physiol 2001. [DOI: 10.1002/cphy.cp070234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
39
|
Ainslie DA, Morris MJ, Wittert G, Turnbull H, Proietto J, Thorburn AW. Estrogen deficiency causes central leptin insensitivity and increased hypothalamic neuropeptide Y. Int J Obes (Lond) 2001; 25:1680-8. [PMID: 11753591 DOI: 10.1038/sj.ijo.0801806] [Citation(s) in RCA: 158] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2000] [Revised: 03/13/2001] [Accepted: 05/02/2001] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Altered fat distribution is a consequence of menopause, but the mechanisms responsible are unknown. Estrogen insufficiency in humans can be modeled using ovariectomized rats. We have shown that increased adiposity in these rats is due to reduced physical activity and transient hyperphagia, and can be reversed with 17beta-estradiol treatment. The aims of this study were to examine whether this altered energy balance is associated with circulating leptin insufficiency, central leptin insensitivity, decreased hypothalamic leptin receptor (Ob-Rb) expression, and/or increased hypothalamic neuropeptide Y (NPY). METHODS Plasma leptin levels, adipose tissue ob gene expression, energy balance responses to i.c.v. leptin, hypothalamic Ob-Rb expression and NPY concentration in five separate hypothalamic regions were measured in adult female rats after either ovariectomy or sham operations. RESULTS Obesity was not associated with hypoleptinemia or decreased ob gene expression in ovariectomized rats; however, it was associated with insensitivity to central leptin administration. Food intake was less suppressed and spontaneous physical activity was less stimulated by leptin. This was not due to decreased hypothalamic Ob-Rb expression. NPY concentration in the paraventricular nucleus of the hypothalamus was elevated in the ovariectomized rats, consistent with leptin insensitivity; however this effect was transient and disappeared as body fat and leptin levels increased further and hyperphagia normalized. CONCLUSION Impaired central leptin sensitivity and overproduction of NPY may contribute to excess fat accumulation caused by estrogen deficiency.
Collapse
Affiliation(s)
- D A Ainslie
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
40
|
Sainsbury A, Wilks D, Cooney GJ. Central but not peripheral glucocorticoid infusion in adrenalectomized male rats increases basal and substrate-induced insulinemia through a parasympathetic pathway. OBESITY RESEARCH 2001; 9:274-81. [PMID: 11331432 DOI: 10.1038/oby.2001.33] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Glucocorticoids acting through the central nervous system are postulated to play a role in the hyperinsulinemia and increased adiposity of obesity. We investigated the role of parasympathetic activation in glucocorticoid-induced hyperinsulinemia. RESEARCH METHODS AND PROCEDURES Plasma pancreatic polypeptide (PP) levels were used as an index of parasympathetic output. Insulinemia and plasma PP levels were measured basally and after intravenous glucose injection (300 mg/kg) in adrenalectomized male rats infused with dexamethasone (7.5 microg/kg per day) intracerebroventricularly (ICV) or subcutaneously (SC) for 3 to 6 days in the presence or absence of acute atropine blockade (1.0 mg/kg). Food intake was controlled between groups. RESULTS Compared with normal rats, adrenalectomy decreased white adipose tissue depot weights and leptinemia, and these were restored to normal values by ICV but not SC dexamethasone infusion. Adrenalectomy significantly reduced insulinemia below normal levels, which was restored by SC dexamethasone replacement. However, ICV dexamethasone replacement increased insulinemia of adrenalectomized rats to levels higher than normal control values (basal, 500 +/- 40 pM vs. 280 +/- 40 pM; 1-minute postglucose, 2500 +/- 180 pM vs. 1240 +/- 260 pM; p < 0.0001) and increased plasma PP levels, which were correlated with insulinemia. Atropine significantly reduced plasma insulin and PP to levels similar to normal controls but had no effect in any other group. DISCUSSION These data show that glucocorticoids act within the brain to increase insulinemia, most likely through activation of parasympathetic efferent fibers. Such an affect would contribute to the adipogenic effects of central glucocorticoids.
Collapse
Affiliation(s)
- A Sainsbury
- Diabetes Research Group, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia.
| | | | | |
Collapse
|
41
|
Sainsbury A, Herzog H. Inhibitory effects of central neuropeptide Y on the somatotropic and gonadotropic axes in male rats are independent of adrenal hormones. Peptides 2001; 22:467-71. [PMID: 11287103 DOI: 10.1016/s0196-9781(01)00342-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neuropeptide Y (NPY) in the hypothalamus exerts multiple physiological functions including stimulation of adipogenic pathways such as feeding and insulin secretion as well as inhibition of the somatotropic and gonadotropic axes. Since hypothalamic NPY-ergic activity is increased by negative energy balance, NPY enables coordinated regulation of growth and reproduction in parallel with energy availability. Chronic pathological increases in central NPY-ergic activity contribute to obesity. Many of the adipogenic effects of NPY are specifically dependent on adrenal glucocorticoids. However, in the current study we show that central NPY does not require adrenal hormones to inhibit the somatotropic and gonadotropic axes in rats. Male adrenalectomized and sham-operated normal rats were intracerebroventricularly (ICV) infused with NPY (15 microg/day) or saline for 5-7 days, and plasma leptin, insulin-like growth factor (IGF-1) and testosterone were assayed, and epididymal white adipose tissue (WATe) was weighed. In normal intact rats, WATe weight and leptinemia were significantly increased by NPY, and these effects were prevented by adrenalectomy. In normal rats, NPY markedly reduced plasma IGF-1 levels (470 +/- 40 versus 1260 +/- 90 ng/ml) and testosterone (0.53 +/- 0.28 versus 5.4 +/- 0.80 nmol/l in saline-infused controls, p < 0.0001). Adrenalectomy decreased plasma IGF-1 concentrations to 290 +/- 30 (p < 0.0001 versus normal rats), which were significantly reduced further by NPY. However, adrenalectomy had no significant effect on basal nor on NPY-induced plasma testosterone concentrations. In conclusion unlike the stimulatory effects of NPY on fat mass and leptinemia, NPY-induced inhibition of the somatotropic and gonadotropic axes in male rats do not require adrenal hormones.
Collapse
Affiliation(s)
- A Sainsbury
- Diabetes Research Group, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst NSW 2010, Sydney, Australia
| | | |
Collapse
|
42
|
Rioux KP, Le T, Swain MG. Decreased orexigenic response to neuropeptide Y in rats with obstructive cholestasis. Am J Physiol Gastrointest Liver Physiol 2001; 280:G449-56. [PMID: 11171627 DOI: 10.1152/ajpgi.2001.280.3.g449] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neuropeptide Y (NPY) is a key factor in the neurochemical control of food intake, and obstructive cholestasis can be associated with disturbances in food intake. Our aim in this study was to determine whether obstructive cholestasis in the rat is associated with defective central responsiveness to NPY. Cholestasis was induced in rats by surgical bile duct resection. Rats with obstructive cholestasis exhibited a 20% reduction in food intake 2 days after laparotomy (compared with sham-resected controls) that had resolved by 4 days after surgery. Responsiveness to the orexigenic action of NPY was tested by measuring food intake after intracerebroventricular injection of NPY. In sham-resected rats, NPY infusion strikingly increased food intake, whereas bile duct-resected (BDR) rats showed a consistent significantly impaired feeding response to NPY at postlaparotomy days 2, 4, and 7. Separate experiments measured specific binding of [(3)H]NPY to hypothalamic receptors. Fos protein expression was measured in the hypothalamic paraventricular nucleus (PVN) as a marker of NPY-induced neuronal activation. The decreased orexigenic responsiveness to NPY was not caused by altered NPY binding at hypothalamic receptors or its ability to activate neurons in the PVN. Therefore, cholestatic rats demonstrate an attenuated NPY-induced orexigenic drive that occurs early after biliary obstruction, when cholestatic rats exhibit reduced food intake, and persists despite the return of food intake to normal levels and the presence of intact central NPY-related neuronal pathways.
Collapse
Affiliation(s)
- K P Rioux
- Liver Unit, Gastrointestinal Research Group, University of Calgary, Health Sciences Centre, Calgary, Alberta, Canada T2N 4N1
| | | | | |
Collapse
|
43
|
Makino S, Baker RA, Smith MA, Gold PW. Differential regulation of neuropeptide Y mRNA expression in the arcuate nucleus and locus coeruleus by stress and antidepressants. J Neuroendocrinol 2000; 12:387-95. [PMID: 10792576 DOI: 10.1046/j.1365-2826.2000.00451.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In rats, circulating corticosterone and insulin are involved in regulation of the hypothalamic neuropeptide Y (NPY) system, which in turn, is involved in regulation of the hypothalamic-pituitary-adrenal (HPA) axis. Since the HPA axis and stress responsivity is altered in diseases such as depression, we investigated interactions between the effects of stress and antidepressant drug treatment on arcuate nucleus and locus coeruleus NPY mRNA expressions using in-situ hybridization histochemistry. After acute (2 h) and repeated immobilization (2 h daily, for 14 days), plasma concentrations of corticosterone increased, and those of insulin decreased. The expression of NPY mRNA was significantly increased in the arcuate nucleus, but was unchanged in the locus coeruleus following acute and repeated immobilization. Adrenalectomized rats with systemic corticosterone replacement (ADX+CORT), whose corticosterone concentration was maintained at approximately 50-100 ng/ml during repeated stress, showed a decrease in plasma insulin and an increase in arcuate nucleus NPY mRNA similar to that observed in sham rats, suggesting that changes in NPY mRNA levels are more closely tied to circulating insulin than to circulating corticosterone. In contrast, locus coeruleus NPY mRNA expressions in ADX+CORT rats were significantly higher than those in sham rats after repeated stress. Desmethylimipramine (DMI) treatment for 24 days did not affect basal plasma concentrations of corticosterone or insulin, or arcuate nucleus NPY mRNA expressions, but significantly decreased basal levels of locus coeruleus NPY mRNA compared to saline-treated rats. After repeated immobilization (2 h daily, for 4 days), DMI significantly reduced the stress-induced rise in locus coeruleus NPY mRNA levels, but potentiated the stress-induced rise in arcuate nucleus NPY mRNA expression. These results demonstrate that: (1) the increase in arcuate nucleus NPY mRNA expressions in stressed rats closely follows the decrease in plasma concentrations of insulin; (2) increases in NPY mRNA expressions occur in the absence of changes in plasma corticosterone; and (3) desipramine treatment potentiated the effect of stress on arcuate nucleus NPY mRNA expressions, but blocked the repeated stress-induced increase in locus coeruleus NPY mRNA expressions. Thus, NPY mRNA expression in the arcuate nucleus and the locus coeruleus is sensitive to the effects of stress and to the antidepressant drug desipramine, but the arcuate nucleus NPY system is regulated by different mechanisms than the locus coeruleus NPY system. The results provide further evidence for the importance of circulating insulin in the regulation of the arcuate nucleus NPY system.
Collapse
Affiliation(s)
- S Makino
- Clinical Neuroendocrinology Branch; Biological Psychiatry Branch, National Institute of Mental Health, Bethesda, MD 20892-1284, USA
| | | | | | | |
Collapse
|
44
|
Wisialowski T, Parker R, Preston E, Sainsbury A, Kraegen E, Herzog H, Cooney G. Adrenalectomy reduces neuropeptide Y-induced insulin release and NPY receptor expression in the rat ventromedial hypothalamus. J Clin Invest 2000; 105:1253-9. [PMID: 10792000 PMCID: PMC315443 DOI: 10.1172/jci8695] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Chronic central administration of neuropeptide Y (NPY) causes hyperphagia, hyperinsulinemia, and obesity, a response that is prevented by prior adrenalectomy (ADX) in rats. The basis of NPY's effect and how the acute responses to this peptide are affected by ADX remain unknown. This study investigates the role of glucocorticoids in acute NPY-stimulated food intake, acute NPY-induced insulin release, and hypothalamic NPY-receptor mRNA expression levels. NPY-induced food intake was similar in ADX and control rats after acute intracerebroventricular injection of NPY. Injection of NPY caused a significant increase in plasma insulin in control rats, but this effect was completely absent in ADX rats in which basal plasma insulin levels were also lower than controls. In addition, ADX significantly reduced the number of neurons expressing NPY receptor Y(1) and Y(5) mRNAs in the ventromedial hypothalamus (VMH), without affecting Y(1)- or Y(5)-mRNA expression in the paraventricular hypothalamus or the arcuate nucleus. These data indicate that glucocorticoids are necessary for acute NPY-mediated insulin release and suggest that the mechanisms involve glucocorticoid regulation of Y(1) and Y(5) receptors specifically within the VMH nucleus.
Collapse
Affiliation(s)
- T Wisialowski
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, Australia
| | | | | | | | | | | | | |
Collapse
|
45
|
Wang J, Dourmashkin JT, Yun R, Leibowitz SF. Rapid changes in hypothalamic neuropeptide Y produced by carbohydrate-rich meals that enhance corticosterone and glucose levels. Brain Res 1999; 848:124-36. [PMID: 10612704 DOI: 10.1016/s0006-8993(99)02040-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Prior studies have demonstrated that chronic consumption over several weeks of a high-carbohydrate (65%) diet, compared to a moderate-carbohydrate (45%) or low-carbohydrate (15%) diet, potentiates the expression, synthesis and release of hypothalamic NPY. This effect occurs specifically in neurons of the arcuate nucleus (ARC) which project to the paraventricular nucleus (PVN). In the present experiments, tests involving acute manipulations were conducted to determine whether such diet-induced changes in NPY can occur rapidly, perhaps within 1-2 h, and whether these effects can be linked to specific changes in circulating glucoregulatory hormones or glucose itself., In adult, albino rats maintained on lab chow, the acute manipulations included the presentation of either a high-carbohydrate, moderate-carbohydrate or high-fat diet for 90 min at the onset of the natural feeding cycle. They also involved manipulations of glucose itself, either through the ingestion of a glucose (20%) solution in a drinking tube or intraperitoneal injection of a glucose solution (10%). After a high-carbohydrate meal compared to a moderate-carbohydrate or high-fat meal, NPY gene expression examined via in situ hybridization is found to be significantly enhanced in the ARC. The high-carbohydrate meal also potentiates NPY immunoreactivity in the ARC and PVN but has little effect on NPY in other hypothalamic areas examined and actually causes a reduction in the feeding-stimulatory peptide, galanin, specifically in the PVN. The meal-induced increase in NPY is associated with specific endocrine patterns, as revealed by measurements in serum collected from trunk blood or from rats implanted with a chronic jugular catheter. After a high-carbohydrate meal, levels of glucose, together with corticosterone and insulin, are significantly elevated, while non-esterified fatty acids are reduced. A possible effect of circulating glucose on hypothalamic NPY is further suggested by the finding that the consumption or a single injection of a glucose solution at the onset of the feeding cycle similarly elevates NPY mRNA and peptide immunoreactivity in the ARC and PVN. These results demonstrate that hypothalamic NPY can change rapidly in response to dietary carbohydrate. They also suggest that this effect may be related to changes in circulating CORT as well as to the availability or utilization of glucose.
Collapse
Affiliation(s)
- J Wang
- The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | | | | | | |
Collapse
|
46
|
Krukoff TL, MacTavish D, Jhamandas JH. Effects of restraint stress and spontaneous hypertension on neuropeptide Y neurones in the brainstem and arcuate nucleus. J Neuroendocrinol 1999; 11:715-23. [PMID: 10447810 DOI: 10.1046/j.1365-2826.1999.00391.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Neuropeptide Y (NPY) is found in autonomic neurones and participates in regulation of autonomic functions. To investigate the role of NPY in the stress response in normo- and hypertensive rats, activation of brainstem and arcuate nucleus (ARC) NPY neurones and levels of NPY mRNA in the ARC were measured in response to restraint stress in adult spontaneously hypertensive rats (SHRs) and two strains of normotensive rats. Controls from each strain were not restrained. Sections of the brain were prepared for Fos immunohistochemistry and NPY in-situ hybridization to identify activated NPY neurones in the nucleus of the tractus solitarii (NTS), ventrolateral medulla (VLM), and ARC. NPY mRNA levels were quantified in the ARC. In the NTS and VLM of restrained rats, approximately 33% and 75%, respectively, of NPY neurones were activated. No differences among strains were found. In the ARC, about 36% of neurones activated by restraint contained NPY mRNA with no differences found among strains. In unrestrained rats, NPY mRNA levels were significantly elevated in SHRs compared to the normotensive rats. Restraint led to significant decreases in mRNA levels in all strains and mRNA levels among strains were no longer different from one another. These data show that NPY likely participates as a neurotransmitter in the autonomic pathways utilized during stress and originating in the NTS, VLM, and ARC. On the other hand, the decreased gene expression of NPY in the ARC in response to restraint stress argues against a role for activation of autonomic pathways or the hypothalamo-pituitary-adrenal (HPA) axis by NPY from the ARC of stressed rats. The elevated NPY gene expression in resting SHRs compared to normotensive rats is abrogated after restraint, suggesting that this gene is differentially regulated in SHRs compared to normotensive rats.
Collapse
Affiliation(s)
- T L Krukoff
- Department of Cell Biology and Anatomy, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| | | | | |
Collapse
|
47
|
Bailey AR, Giles M, Brown CH, Bull PM, Macdonald LP, Smith LC, Smith RG, Leng G, Dickson SL. Chronic central infusion of growth hormone secretagogues: effects on fos expression and peptide gene expression in the rat arcuate nucleus. Neuroendocrinology 1999; 70:83-92. [PMID: 10461022 DOI: 10.1159/000054462] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Growth hormone (GH) secretagogues induce GH release, in part, by direct actions upon anterior pituitary somatotropes and, in part, by actions upon the neuroendocrine circuitry that regulates GH secretion. In particular, acute systemic administration of GH secretagogues results in increased neuronal activity and Fos protein expression in the arcuate nucleus of the hypothalamus. Prolonged administration of GH secretagogues has been reported to have long-lasting effects upon GH release, promoting increased pulsatile secretion. Here, we investigated how chronic central infusion of GH secretagogues affects the response of arcuate nucleus neurons to acute systemic administration of GH secretagogues. In male rats, after central infusion of GH secretagogues for 5 days, there was no sustained expression of Fos in the arcuate nucleus, no significant induction of Fos expression in response to acute GH secretagogue challenge, and a greatly attenuated secretion of GH in response to acute GH secretagogue challenge, all reflecting loss of funtional responsiveness to GH secretagogues. In situ hybridisation revealed that, in the arcuate nucleus of GH secretagogue-infused rats, mRNA levels for GH-releasing hormone, neuropeptide Y and somatostatin were not different than in saline-infused animals. However, somatostatin mRNA levels in the periventricular nuclei of GH secretagogue-infused rats were significantly higher than those of saline-infused rats, indicating that this nucleus may play an important role in mediating the effects of chronic GH secretagogue administration.
Collapse
Affiliation(s)
- A R Bailey
- Department of Biomedical Sciences, University Medical School, Edinburgh, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kalra SP, Dube MG, Pu S, Xu B, Horvath TL, Kalra PS. Interacting appetite-regulating pathways in the hypothalamic regulation of body weight. Endocr Rev 1999; 20:68-100. [PMID: 10047974 DOI: 10.1210/edrv.20.1.0357] [Citation(s) in RCA: 245] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Various aspects of the complex spatio-temporal patterning of hypothalamic signaling that leads to the development of synchronized nocturnal feeding in the rat are critically examined. Undoubtedly, as depicted in Fig. 7, a distinct ARN in the hypothalamus is involved in the control of nocturnal appetite. At least four basic elements operate within this ARN. These are: 1) A discrete appetite-driving or orexigenic network of NPY, NE, GABA, GAL, EOP, and orexin transduces and releases appetite-stimulating signals. 2) Similarly, anorexigenic signal-producing pathways (e.g., CRH, GLP-1, alpha MSH, and CART) orchestrate neural events for dissipation of appetite and to terminate feeding, possibly by interrupting NPY efflux and action at a postsynaptic level within the hypothalamus. It is possible that some of these may represent the physiologically relevant "off" switches under the influence of GABA alone, or AgrP alone, or in combination with NPY released from the NPY-, GABA-, and AgrP-coproducing neurons. 3) Recent evidence shows that neural elements in the VMN-DMN complex tonically restrain the orexigenic signals during the intermeal interval; the restraint is greatly aided by leptin's action via diminution of orexigenic (NPY) and augmentation of anorexigenic (GLP-1, alpha MSH, and CART) signals. Since interruption of neurotransmission in the VMN resulted in hyperphagia and development of leptin resistance, it seems likely that the VMN is an effector site for the restraint exercised by leptin. The daily rhythms in leptin synthesis and release are temporally dissociable because the onset of daily rise in leptin gene expression in adipocytes precedes that in leptin secretion. Nevertheless, these rhythms are in phase with daily ingestive behavior because the peak in circulating leptin levels occurs during the middle of the feeding period. These observations, coupled with the fact that circulating levels of leptin are directly related to adiposity, pose a new challenge for elucidating the precise role of leptin in daily patterning of feeding in the rat. 4) A neural timing mechanism also operates upstream from the ARN in the daily management of energy homeostasis. Although the precise anatomical boundaries are not clearly defined, this device is likely to be composed of a group of neurons that integrate incoming internal and external information for the timely onset of the drive to eat. Evidently, this network operates independently in primates, but it is entrained to the circadian time keeper in the SCN of rodents. Apart from its role in the onset of drive to eat, the circadian patterns of gene expression of NPY, GAL, and POMC denote independent control of the timing device on the synthesis and availability for release of orexigenic signals. The VMN-DMN-PVN complex is apparently an integrated constituent of the timing mechanism in this context, because lesions in each of these sites result in loss of regulated feeding. The accumulated evidence points to the PVN and surrounding neural sites within this framework as the primary sites of release and action of various orexigenic and anorexigenic signals. A novel finding is the identification of the interconnected wiring of the DMN-mPVN axis that may mediate leptin restraint on NPY-induced feeding. The chemical phenotypes of leptin and NPY target neurons in this axis remain to be identified. These multiple orexigenic and anorexigenic pathways in the hypothalamic ARN appear to represent redundancy, a characteristic of regulated biological systems to provide a "fail-safe" neural mechanism to meet an organism's constant energy needs for growth and maintenance. Within this formulation, the coexisting orexigenic signals (NPY, NE, GAL, GABA, and AgrP) represent either another level of redundancy or it is possible that these signals operate within the ARN as reinforcing agents to varying degrees under different circumstances. (ABSTRACT TRUNCATED)
Collapse
Affiliation(s)
- S P Kalra
- Department of Neuroscience, University of Florida Brain Institute, University of Florida College of Medicine, Gainesville 32610, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Leibowitz SF. Differential functions of hypothalamic galanin cell grows in the regulation of eating and body weight. Ann N Y Acad Sci 1998; 863:206-20. [PMID: 9928172 DOI: 10.1111/j.1749-6632.1998.tb10696.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Evidence suggests that hypothalamic galanin (GAL) has a variety of functions related to energy and nutrient balance, reproduction, water balance, and neuroendocrine regulation. The focus of this chapter is the role of GAL in eating and body weight regulation. Findings described herein demonstrate that GAL, in a cell group of the anterior region of the paraventricular nucleus (aPVN) that projects to the median eminence, has a role in the control of fat intake, fat metabolism, and body fat. This function of aPVN GAL neurons is carried out in close relation to circulating insulin and glucose. Galanin-expressing perikarya in the medial preoptic area (MPOA) have a similar function, although GAL here operates in association with the female steroids estrogen and progesterone. These GAL cell groups of the aPVN and MPOA contrast with those in the arcuate nucleus as well as the magnocellular vasopressin-containing neurons of the PVN and supraoptic nucleus, which show no relation to fat balance. This evidence reveals differential functions for the distinct GAL neuronal cell groups of the hypothalamus.
Collapse
Affiliation(s)
- S F Leibowitz
- Rockefeller University, New York, New York 10021, USA
| |
Collapse
|
50
|
Thorsell A, Svensson P, Wiklund L, Sommer W, Ekman R, Heilig M. Suppressed neuropeptide Y (NPY) mRNA in rat amygdala following restraint stress. REGULATORY PEPTIDES 1998; 75-76:247-54. [PMID: 9802416 DOI: 10.1016/s0167-0115(98)00075-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We have previously demonstrated that NPY produces anxiolytic-like effects through actions in the amygdala, and that anxiogenic-like effects of restraint stress are mediated through this structure. Here, we examined the effects of restraint stress on NPY mRNA levels in amygdala and several other brain regions. A sensitive solution hybridization-RNase protection assay (RPA) was developed, employing a combination of internal and external standards, which allowed absolute quantitation of NPY mRNA in tissue-samples of less than 10 mg. NPY mRNA levels were determined, following a 1-h restraint stress, in homogenates of tissue from the amygdala, neocortex, striatum and hypothalamus, and the time course of these effects was examined. A highly significant decrease in NPY-mRNA levels was seen in the amygdala at 1 h and 2 h following restraint, with levels returning to normal within 10 h. A similar effect was seen in the neocortex, but was less pronounced and slower in onset. Striatal and hypothalamic NPY expression was not significantly affected. Tissue levels of NPY-peptide were modestly decreased in the amygdala at 1 h following restraint and had returned to normal within 4 h. The present findings support the hypothesis that anxiety related behavioral effects of stress may in part be mediated through modulation of NPY function in the amygdala.
Collapse
Affiliation(s)
- A Thorsell
- Department of Clinical Neuroscience and Family Medicine, Karolinska Institute, Addiction Center South, M67 Huddinge University Hospital, Sweden
| | | | | | | | | | | |
Collapse
|