1
|
Yesmin R, Watanabe M, Sinha AS, Ishibashi M, Wang T, Fukuda A. A subpopulation of agouti-related peptide neurons exciting corticotropin-releasing hormone axon terminals in median eminence led to hypothalamic-pituitary-adrenal axis activation in response to food restriction. Front Mol Neurosci 2022; 15:990803. [PMID: 36245920 PMCID: PMC9557964 DOI: 10.3389/fnmol.2022.990803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
The excitatory action of gamma-aminobutyric-acid (GABA) in the median-eminence (ME) led to the steady-state release of corticotropin-releasing hormone (CRH) from CRH axon terminals, which modulates the hypothalamic-pituitary-adrenal (HPA) axis. However, in ME, the source of excitatory GABAergic input is unknown. We examined agouti-related peptide (AgRP) expressing neurons in the arcuate nucleus as a possible source for excitatory GABAergic input. Here, we show that a subpopulation of activated AgRP neurons directly project to the CRH axon terminals in ME elevates serum corticosterone levels in 60% food-restricted mice. This increase in serum corticosterone is not dependent on activation of CRH neuronal soma in the paraventricular nucleus. Furthermore, conditional deletion of Na+-K+-2Cl– cotransporter-1 (NKCC1), which promotes depolarizing GABA action, from the CRH axon terminals results in significantly lower corticosterone levels in response to food restriction. These findings highlight the important role of a subset of AgRP neurons in HPA axis modulation via NKCC1-dependent GABAergic excitation in ME.
Collapse
Affiliation(s)
- Ruksana Yesmin
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Adya Saran Sinha
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Masaru Ishibashi
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Tianying Wang
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
- Advanced Research Facilities and Services, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
- *Correspondence: Atsuo Fukuda,
| |
Collapse
|
2
|
|
3
|
Lee TJ, Kinzig KP. Reprint of "Repeated adolescent activity-based anorexia influences central estrogen signaling and adulthood anxiety-like behaviors in rats". Physiol Behav 2017; 178:179-186. [PMID: 28341321 DOI: 10.1016/j.physbeh.2017.02.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/28/2016] [Accepted: 12/17/2016] [Indexed: 11/18/2022]
Abstract
Anorexia nervosa (AN) typically presents in adolescence and is highly comorbid with anxiety and depression, which often persist after elimination of AN symptomology. The activity-based anorexia (ABA) paradigm allows for evaluation of behavioral and neuroendocrine consequences of AN-like behaviors, including voluntary anorexia, hyperactivity, and disruption of the hypothalamic-pituitary-gonadal (HPG) and the hypothalamic pituitary adrenal (HPA) axis. Because ABA in adolescent females results in increased anxiety-like behavior in adulthood and the estrogen signaling system has been shown to play a role in anxiety and food intake, we investigated the role of ovarian hormones in adolescent ABA-treated rats, and long-term effects of mid- and late adolescent ABA exposure on behavior and estrogen signaling. While previous research demonstrated that two bouts of ABA during adolescence resulted in decreased time in the open arm of the elevated plus maze (EPM) and increased activity of the HPA axis in response to a novel stressor, here we show that one bout of ABA in mid-or late-adolescence did not result in the same behavioral outcome. Two exposures to ABA during adolescence were necessary to produce long-term anxiety-like behavior on the EPM. Finally, removal of ovarian hormones by ovariectomy (OVX) prior to puberty did not attenuate long-term behavioral consequences of ABA in adolescence, and estrogen receptor β (ERβ) expression level in the amygdala of ABA rats was significantly lower than control subjects. Taken together, these studies identify enduring effects of ABA in adolescent females that may be mediated by ABA-induced changes to CNS ERβ signaling that increase anxiety-like behaviors.
Collapse
Affiliation(s)
- Tien-Jui Lee
- Department of Psychological Sciences, Purdue University, 703 Third Street, West Lafayette, IN 47907, United States.
| | - Kimberly P Kinzig
- Department of Psychological Sciences, Purdue University, 703 Third Street, West Lafayette, IN 47907, United States.
| |
Collapse
|
4
|
Lee TJ, Kinzig KP. Repeated adolescent activity-based anorexia influences central estrogen signaling and adulthood anxiety-like behaviors in rats. Physiol Behav 2017; 171:199-206. [PMID: 28069464 DOI: 10.1016/j.physbeh.2016.12.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/28/2016] [Accepted: 12/17/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Tien-Jui Lee
- Department of Psychological Sciences, Purdue University, 703 Third Street, West Lafayette, IN 47907, United States.
| | - Kimberly P Kinzig
- Department of Psychological Sciences, Purdue University, 703 Third Street, West Lafayette, IN 47907, United States.
| |
Collapse
|
5
|
Rabaglino MB, Chang EI, Richards EM, James MO, Keller-Wood M, Wood CE. Genomic Effect of Triclosan on the Fetal Hypothalamus: Evidence for Altered Neuropeptide Regulation. Endocrinology 2016; 157:2686-97. [PMID: 27145008 PMCID: PMC4929550 DOI: 10.1210/en.2016-1080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Triclosan (TCS), an antibacterial compound commonly added to personal care products, could be an endocrine disruptor at low doses. Although TCS has been shown to alter fetal physiology, its effects in the developing fetal brain are unknown. We hypothesize that exposure to TCS during fetal life could affect fetal hypothalamic gene expression. The objective of this study was to use transcriptomics and systems analysis to identify significantly altered biological processes in the late gestation ovine fetal hypothalamus after direct or indirect exposure to low doses of TCS. For direct TCS exposure, chronically catheterized late gestation fetal sheep were infused with vehicle (n = 4) or TCS (250 μg/d; n = 4) iv. For indirect TCS exposure, TCS (100 μg/kg · d; n = 3) or vehicle (n = 3) was infused into the maternal circulation. Fetal hypothalami were collected after 2 days of infusion, and gene expression was measured through microarray. Hierarchical clustering of all samples according to gene expression profiles showed that samples from the TCS-treated animals clustered apart from the controls. Gene set enrichment analysis revealed that fetal hypothalamic genes stimulated by maternal and fetal TCS infusion were significantly enriching for cell cycle, reproductive process, and feeding behavior, whereas the inhibited genes were significantly enriching for chromatin modification and metabolism of steroids, lipoproteins, fatty acids, and glucose (P < .05). In conclusion, short-term infusion of TCS induces vigorous changes in the fetal hypothalamic transcriptomics, which are mainly related to food intake pathways and metabolism. If these changes persist to postnatal life, they could result in adverse consequences in adulthood.
Collapse
Affiliation(s)
- Maria Belen Rabaglino
- Centro de Excelencia en Procesos y Productos de Córdoba (M.B.R.), National Scientific and Technical Research Council, Córdoba, Argentina X5164; Department of Physiology and Functional Genomics (E.I.C., C.E.W.), College of Medicine, University of Florida, Gainesville, Florida 32610; and Departments of Medicinal Chemistry (M.O.J.) and Pharmacodynamics (E.M.R., M.K.-W.), College of Pharmacy, University of Florida, Gainesville, Florida 32610
| | - Eileen I Chang
- Centro de Excelencia en Procesos y Productos de Córdoba (M.B.R.), National Scientific and Technical Research Council, Córdoba, Argentina X5164; Department of Physiology and Functional Genomics (E.I.C., C.E.W.), College of Medicine, University of Florida, Gainesville, Florida 32610; and Departments of Medicinal Chemistry (M.O.J.) and Pharmacodynamics (E.M.R., M.K.-W.), College of Pharmacy, University of Florida, Gainesville, Florida 32610
| | - Elaine M Richards
- Centro de Excelencia en Procesos y Productos de Córdoba (M.B.R.), National Scientific and Technical Research Council, Córdoba, Argentina X5164; Department of Physiology and Functional Genomics (E.I.C., C.E.W.), College of Medicine, University of Florida, Gainesville, Florida 32610; and Departments of Medicinal Chemistry (M.O.J.) and Pharmacodynamics (E.M.R., M.K.-W.), College of Pharmacy, University of Florida, Gainesville, Florida 32610
| | - Margaret O James
- Centro de Excelencia en Procesos y Productos de Córdoba (M.B.R.), National Scientific and Technical Research Council, Córdoba, Argentina X5164; Department of Physiology and Functional Genomics (E.I.C., C.E.W.), College of Medicine, University of Florida, Gainesville, Florida 32610; and Departments of Medicinal Chemistry (M.O.J.) and Pharmacodynamics (E.M.R., M.K.-W.), College of Pharmacy, University of Florida, Gainesville, Florida 32610
| | - Maureen Keller-Wood
- Centro de Excelencia en Procesos y Productos de Córdoba (M.B.R.), National Scientific and Technical Research Council, Córdoba, Argentina X5164; Department of Physiology and Functional Genomics (E.I.C., C.E.W.), College of Medicine, University of Florida, Gainesville, Florida 32610; and Departments of Medicinal Chemistry (M.O.J.) and Pharmacodynamics (E.M.R., M.K.-W.), College of Pharmacy, University of Florida, Gainesville, Florida 32610
| | - Charles E Wood
- Centro de Excelencia en Procesos y Productos de Córdoba (M.B.R.), National Scientific and Technical Research Council, Córdoba, Argentina X5164; Department of Physiology and Functional Genomics (E.I.C., C.E.W.), College of Medicine, University of Florida, Gainesville, Florida 32610; and Departments of Medicinal Chemistry (M.O.J.) and Pharmacodynamics (E.M.R., M.K.-W.), College of Pharmacy, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
6
|
Paternal calorie restriction prior to conception alters anxiety-like behavior of the adult rat progeny. Psychoneuroendocrinology 2016; 64:1-11. [PMID: 26571216 DOI: 10.1016/j.psyneuen.2015.10.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 10/26/2015] [Indexed: 11/22/2022]
Abstract
The maternal environment influences a broad range of phenotypic outcomes for offspring, with anxiety-like behavior being particularly susceptible to maternal environmental perturbations. Much less is known regarding paternal environmental influences. To investigate this, adult male rats were exposed to 25% calorie restriction (CR) or glucocorticoid elevation (CORT; 200 μg/ml of corticosterone in drinking water) for ∼ 6 weeks prior to breeding. Elevated plus maze (EPM), open field (OF), predator odor (cat urine), and acoustic startle/pre-pulse inhibition (AS/PPI) were characterised in the adult male offspring. Plasma concentrations of corticotrophin-releasing hormone (CRF), adrenocorticotropin hormone (ACTH), and serum leptin were characterised in both sires and offspring. Maternal care received by litters was additionally observed. Expectedly, CR and CORT treatment attenuated weight gain, whilst only CR induced anxiolytic behavior in the EPM. The adult offspring sired by CR males also demonstrated a reduction in weight gain, food intake and serum leptin levels when compared to controls. Moreover, CR offspring demonstrated an anxiolytic-like profile in the EPM and OF, enhanced habituation to the AS pulse, reduced PPI, but no alteration to predator odor induced defensiveness compared to control. CORT offspring failed to demonstrate any behavioral differences from controls, however, exhibited a trend towards reduced ACTH and leptin concentration. Collectively, the results indicate that a reduction in calories in males prior to conception can affect the behavior of adult offspring. The phenotypic transmission of CR experiences from fathers to the progeny could potentially be mediated epigenetically. The role of glucocorticoid elevation and maternal care are also discussed.
Collapse
|
7
|
Ibrahim BA, Alenazi FSH, Briski KP. Energy status determines hindbrain signal transduction pathway transcriptional reactivity to AMPK in the estradiol-treated ovariectomized female rat. Neuroscience 2014; 284:888-899. [PMID: 25446360 DOI: 10.1016/j.neuroscience.2014.10.068] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/13/2014] [Accepted: 10/31/2014] [Indexed: 10/24/2022]
Abstract
Dorsal vagal complex (DVC) AMPK regulation of food intake in the estradiol-treated ovariectomized (OVX) female rat is energy state-dependent. Here, RT-PCR array technology was used to identify estradiol-sensitive AMPK-regulated DVC signal transduction pathways that exhibit differential reactivity to sensor activation during energy balance versus imbalance. The AMP mimetic AICAR correspondingly reduced or stimulated cDVC phosphoAMPK (pAMPK) and estrogen receptor-beta (ERβ) proteins in full-fed (F) versus 12-h food-deprived (D) estradiol-treated ovariectomized (OVX) rats, but elevated ER-alpha (ERα) in F only. Estradiol suppressed DVC ERβ protein and hypoxia, NFκB, STAT3, STAT6, and Hedgehog signaling pathway marker genes against oil-implanted OVX controls. F+(A)ICAR and D+(S)aline groups each exhibited further inhibition of NFκB, STAT3, and Hedgehog pathway genes, and diminished PPAR, Notch, and STAT5 transcripts versus F+S. Conversely, genes in these six pathways were up-regulated by AICAR treatment of D. Results show that in this animal model, acute AMP augmentation or feeding cessation each inhibit both pAMPK and ERβ expression, but in combination increase these protein profiles. pAMPK protein and DVC TNF (NFκB), SOCS3 (JAK/STAT), WNT6 (Hedgehog), and FABP1 (PPAR) mRNAs were down- or upregulated in parallel by AICAR in F versus D states, respectively. Further research is needed to determine the impact of ERβ on opposing directionality of these responses, and to characterize the role of the aforementioned signaling pathways in hyperphagic responses in the female to AICAR-induced DVC AMPK activation during acute interruption of feeding.
Collapse
Affiliation(s)
- B A Ibrahim
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, United States
| | - F S H Alenazi
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, United States
| | - K P Briski
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, United States.
| |
Collapse
|
8
|
Kenny R, Dinan T, Cai G, Spencer SJ. Effects of mild calorie restriction on anxiety and hypothalamic-pituitary-adrenal axis responses to stress in the male rat. Physiol Rep 2014; 2:e00265. [PMID: 24760519 PMCID: PMC4002245 DOI: 10.1002/phy2.265] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chronic calorie restriction (CR) is one of the few interventions to improve longevity and quality of life in a variety of species. It also reduces behavioral indices of anxiety and influences some stress hormones under basal conditions. However, it is not known how CR influences hypothalamic–pituitary–adrenal (HPA) axis function or if those on a CR diet have heightened HPA axis responses to stress. We hypothesized elevated basal glucocorticoid levels induced by CR would lead to exacerbated HPA axis responses to the psychological stress, restraint, in the male rat. We first confirmed rats fed 75% of their normal calorie intake for 3 weeks were less anxious than ad libitum‐fed (AD) rats in the elevated plus maze test for anxiety. The anxiolytic effect was mild, with only grooming significantly attenuated in the open field and no measured behavior affected in the light/dark box. Despite elevated basal glucocorticoids, CR rats had very similar hormonal and central responses to 15‐min restraint to the AD rats. Both CR and AD rats responded to restraint stress with a robust increase in glucocorticoids that was resolved by 60 min. Both groups also showed robust neuronal activation in the paraventricular nucleus of the hypothalamus and in other stress‐ and feeding‐sensitive brain regions that was not substantially affected by calorie intake. Our findings thus demonstrate chronic mild CR is subtly anxiolytic and is not likely to affect HPA axis responses to psychological stress. These findings support research suggesting a beneficial effect of mild CR.
Collapse
Affiliation(s)
- Rachel Kenny
- School of Health Sciences and Health Innovations Research Institute (HIRi), RMIT University, Melbourne, Vic., Australia
| | | | | | | |
Collapse
|
9
|
Ganella DE, Callander GE, Ma S, Bye CR, Gundlach AL, Bathgate RAD. Modulation of feeding by chronic rAAV expression of a relaxin-3 peptide agonist in rat hypothalamus. Gene Ther 2012; 20:703-16. [PMID: 23135160 DOI: 10.1038/gt.2012.83] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 08/30/2012] [Accepted: 09/20/2012] [Indexed: 11/09/2022]
Abstract
Relaxin-3 is a neuropeptide that is abundantly expressed by discrete brainstem neuron populations that broadly innervate forebrain areas rich in the relaxin-3 G-protein-coupled-receptor, RXFP3. Acute and subchronic central administration of synthetic relaxin-3 or an RXFP3-selective agonist peptide, R3/I5, increase feeding and body weight in rats. Intrahypothalamic injection of relaxin-3 also increases feeding. In this study, we developed a recombinant adeno-associated virus 1/2 (rAAV1/2) vector that drives expression and constitutive secretion of bioactive R3/I5 and assessed the effect of intrahypothalamic injections on daily food intake and body weight gain in adult male rats over 8 weeks. In vitro testing revealed that the vector rAAV1/2-fibronectin (FIB)-R3/I5 directs the constitutive secretion of bioactive R3/I5 peptide. Bilateral injection of rAAV1/2-FIB-R3/I5 vector into the paraventricular nucleus produced an increase in daily food intake and body weight gain (P<0.01, ~23%, respectively), relative to control treatment. In a separate cohort of rats, quantitative polymerase chain reaction analysis of hypothalamic mRNA revealed strong expression of R3/I5 transgene at 3 months post-rAAV1/2-FIB-R3/I5 infusion. Levels of mRNA transcripts for the relaxin-3 receptor RXFP3, the hypothalamic 'feeding' peptides neuropeptide Y, AgRP and POMC, and the reproductive hormone, GnRH, were all similar to control, whereas vasopressin and oxytocin (OT) mRNA levels were reduced by ~25% (P=0.051) and ~50% (P<0.005), respectively, in rAAV1/2-FIB-R3/I5-treated rats (at 12 weeks, n=9/8 rats per group). These data demonstrate for the first time that R3/I5 is effective in modulating feeding in the rat by chronic hypothalamic RXFP3 activation and suggest a potential underlying mechanism involving altered OT signalling. Importantly, there was no desensitization of the feeding response over the treatment period and no apparent deleterious health effects, indicating that targeting the relaxin-3-RXFP3 system may be an effective long-term therapy for eating disorders.
Collapse
Affiliation(s)
- D E Ganella
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
10
|
Olszewski PK, Rozman J, Jacobsson JA, Rathkolb B, Strömberg S, Hans W, Klockars A, Alsiö J, Risérus U, Becker L, Hölter SM, Elvert R, Ehrhardt N, Gailus-Durner V, Fuchs H, Fredriksson R, Wolf E, Klopstock T, Wurst W, Levine AS, Marcus C, Hrabě de Angelis M, Klingenspor M, Schiöth HB, Kilimann MW. Neurobeachin, a regulator of synaptic protein targeting, is associated with body fat mass and feeding behavior in mice and body-mass index in humans. PLoS Genet 2012; 8:e1002568. [PMID: 22438821 PMCID: PMC3305408 DOI: 10.1371/journal.pgen.1002568] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 01/16/2012] [Indexed: 01/08/2023] Open
Abstract
Neurobeachin (Nbea) regulates neuronal membrane protein trafficking and is required for the development and functioning of central and neuromuscular synapses. In homozygous knockout (KO) mice, Nbea deficiency causes perinatal death. Here, we report that heterozygous KO mice haploinsufficient for Nbea have higher body weight due to increased adipose tissue mass. In several feeding paradigms, heterozygous KO mice consumed more food than wild-type (WT) controls, and this consumption was primarily driven by calories rather than palatability. Expression analysis of feeding-related genes in the hypothalamus and brainstem with real-time PCR showed differential expression of a subset of neuropeptide or neuropeptide receptor mRNAs between WT and Nbea+/− mice in the sated state and in response to food deprivation, but not to feeding reward. In humans, we identified two intronic NBEA single-nucleotide polymorphisms (SNPs) that are significantly associated with body-mass index (BMI) in adult and juvenile cohorts. Overall, data obtained in mice and humans suggest that variation of Nbea abundance or activity critically affects body weight, presumably by influencing the activity of feeding-related neural circuits. Our study emphasizes the importance of neural mechanisms in body weight control and points out NBEA as a potential risk gene in human obesity. Body weight and energy balance are under very complex neural, endocrine, and metabolic control. Correspondingly, recent research suggests that hundreds of genes contribute to human obesity and that only a small proportion of them have as yet been identified. Neurobeachin (Nbea) is a protein specifically expressed in nerve and endocrine cells and is important for neurotransmission, apparently by influencing the synaptic targeting of membrane proteins. Here, we show that heterozygous knockout mice, expressing Nbea at 50% of normal levels, display increased adipose tissue mass, abnormal feeding behavior, and modified expression of specific genes in the brainstem and hypothalamus known to be important for body weight control. Moreover, we find that NBEA gene polymorphisms are associated with body-mass index in adult and juvenile human cohorts. Our results demonstrate that variation of Nbea activity critically affects body weight, presumably by influencing the activity of feeding-related neural circuits. They emphasize the importance of neural mechanisms in body weight control, and they identify NBEA as a potential genetic risk factor in human obesity.
Collapse
Affiliation(s)
- Pawel K. Olszewski
- Functional Pharmacology Unit, Department of Neuroscience, Uppsala University, Uppsala, Sweden
- Minnesota Obesity Center, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center and ZIEL Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Josefin A. Jacobsson
- Functional Pharmacology Unit, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, München, Germany
| | - Siv Strömberg
- Molecular Cell Biology Unit, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Wolfgang Hans
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Anica Klockars
- Functional Pharmacology Unit, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Johan Alsiö
- Functional Pharmacology Unit, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Lore Becker
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Neurology, Friedrich-Baur-Institut, Ludwig-Maximilians-Universität München, München, Germany
| | - Sabine M. Hölter
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Ralf Elvert
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Nicole Ehrhardt
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Valérie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Robert Fredriksson
- Functional Pharmacology Unit, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, München, Germany
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institut, Ludwig-Maximilians-Universität München, München, Germany
| | - Wolfgang Wurst
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
- Max-Planck-Institute for Psychiatry, München, Germany
- Technical University München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik c/o Helmholtz Zentrum München, Neuherberg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Munich, Germany
| | - Allen S. Levine
- Minnesota Obesity Center, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Claude Marcus
- Department for Clinical Science, Intervention, and Technology, Karolinska Institutet, Division of Pediatrics, National Childhood Obesity Centre, Stockholm, Sweden
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Freising-Weihenstephan, Germany
| | - Martin Klingenspor
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center and ZIEL Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Helgi B. Schiöth
- Functional Pharmacology Unit, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Manfred W. Kilimann
- Molecular Cell Biology Unit, Department of Neuroscience, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
11
|
Trinko R, Gan G, Gao XB, Sears RM, Guarnieri DJ, DiLeone RJ. Erk1/2 mediates leptin receptor signaling in the ventral tegmental area. PLoS One 2011; 6:e27180. [PMID: 22076135 PMCID: PMC3208604 DOI: 10.1371/journal.pone.0027180] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2011] [Accepted: 10/11/2011] [Indexed: 12/15/2022] Open
Abstract
Leptin acts on the ventral tegmental area (VTA) to modulate neuronal function and feeding behavior in rats and mice. To identify the intracellular effectors of the leptin receptor (Lepr), downstream signal transduction events were assessed for regulation by direct leptin infusion. Phosphorylated signal transducer and activator of transcription 3 (pSTAT3) and phosphorylated extracellular signal-regulated kinase-1 and -2 (pERK1/2) were increased in the VTA while phospho-AKT (pAKT) was unaffected. Pretreatment of brain slices with the mitogen-activated protein kinase kinase -1 and -2 (MEK1/2) inhibitor U0126 blocked the leptin-mediated decrease in firing frequency of VTA dopamine neurons. The anorexigenic effects of VTA-administered leptin were also blocked by pretreatment with U0126, which effectively blocked phosphorylation of ERK1/2 but not STAT3. These data demonstrate that pERK1/2 may have a critical role in mediating both the electrophysiogical and behavioral effects of leptin receptor signaling in the VTA.
Collapse
Affiliation(s)
- Richard Trinko
- Division of Molecular Psychiatry, Ribicoff Research Facilities, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Geliang Gan
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Xiao-Bing Gao
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Robert M. Sears
- Division of Molecular Psychiatry, Ribicoff Research Facilities, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Douglas J. Guarnieri
- Division of Molecular Psychiatry, Ribicoff Research Facilities, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Ralph J. DiLeone
- Division of Molecular Psychiatry, Ribicoff Research Facilities, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
12
|
Association of TMEM18 variants with BMI and waist circumference in children and correlation of mRNA expression in the PFC with body weight in rats. Eur J Hum Genet 2011; 20:192-7. [PMID: 21952719 DOI: 10.1038/ejhg.2011.176] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Genome-wide association studies have shown a strong association of single-nucleotide polymorphisms (SNPs) in the near vicinity of the TMEM18 gene. The effects of the TMEM18-associated variants are more readily observed in children. TMEM18 encodes a 3TM protein, which locates to the nuclear membrane. The functional context of TMEM18 and the effects of its associated variants are as of yet undetermined. To further explore the effects of near-TMEM18 variants, we have genotyped two TMEM18-associated SNPs, rs6548238 and rs4854344, in a cohort of 2352 Greek children (Healthy Growth Study). Included in this study are data on anthropomorphic traits body weight, BMI z-score and waist circumference. Also included are dietary energy and macronutrient intake as measured via 24-h recall interviews. Major alleles of rs6548238 and rs4854344 were significantly associated with an increased risk of obesity (odds ratio = 1.489 (1.161-1.910) and 1.494 (1.165-1.917), respectively), and positively correlated to body weight (P = 0.017, P = 0.010) and waist circumference (P = 0.003, P = 0.003). An association to energy and macronutrient intake was not observed in this cohort. We also correlated food intake and body weight in a food choice model in rats to Tmem18 expression in central regions involved in feeding behavior. We observed a strong positive correlation between TMEM18 expression and body weight in the prefrontal cortex (PFC) (r = 0.5694, P = 0.0003) indicating a potential role for TMEM18 in higher functions related to feeding involving the PFC.
Collapse
|
13
|
MacDonald L, Radler M, Paolini AG, Kent S. Calorie restriction attenuates LPS-induced sickness behavior and shifts hypothalamic signaling pathways to an anti-inflammatory bias. Am J Physiol Regul Integr Comp Physiol 2011; 301:R172-84. [PMID: 21525175 DOI: 10.1152/ajpregu.00057.2011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Calorie restriction (CR) has been demonstrated to alter cytokine levels; however, its potential to modify sickness behavior (fever, anorexia, cachexia) has not. The effect of CR on sickness behavior was examined in male C57BL/6J mice fed ad libitum or restricted 25% (CR25%) or restricted 50% (CR50%) in food intake for 28 days and injected with 50 μg/kg of LPS on day 29. Changes in body temperature, locomotor activity, body weight, and food intake were determined. A separate cohort of mice were fed ad libitum or CR50% for 28 days, and hypothalamic mRNA expression of inhibitory factor κB-α (IκB-α), cyclooxygenase-2 (COX-2), prostaglandin E(2) (PGE(2)), suppressor of cytokine signaling 3 (SOCS3), IL-10, neuropeptide Y (NPY), leptin, proopiomelanocortin (POMC), and corticotrophin-releasing hormone (CRH) were determined at 0, 2, and 4 h post-LPS. CR50% mice did not develop fevers, whereas the CR25% mice displayed a fever shorter in duration but with the same peak as the controls. Both CR25% and CR50% mice showed no sign of anorexia and reduced cachexia after LPS administration. Hypothalamic mRNA expression of NPY and CRH were both increased by severalfold in CR50% animals preinjection compared with controls. The CR50% mice did not demonstrate the expected rise in hypothalamic mRNA expression of COX-2, microsomal prostaglandin E synthase-1, POMC, or CRH 2 h post-LPS, and leptin expression was decreased at this time point. Increases in SOCS3, IL-10, and IκB-α expression in CR50% animals were enhanced compared with ad libitum-fed controls at 4 h post-LPS. CR results in a suppression of sickness behavior in a dose-dependent manner, which may be due to CR attenuating proinflammatory pathways and enhancing anti-inflammatory pathways.
Collapse
Affiliation(s)
- Leah MacDonald
- School of Psychological Science, La Trobe University, Bundoora, VIC 3086, Australia
| | | | | | | |
Collapse
|
14
|
Olszewski PK, Fredriksson R, Eriksson JD, Mitra A, Radomska KJ, Gosnell BA, Solvang MN, Levine AS, Schiöth HB. Fto colocalizes with a satiety mediator oxytocin in the brain and upregulates oxytocin gene expression. Biochem Biophys Res Commun 2011; 408:422-6. [PMID: 21514276 DOI: 10.1016/j.bbrc.2011.04.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/07/2011] [Indexed: 12/31/2022]
Abstract
Single nucleotide polymorphisms in the fat mass and obesity-associated (FTO) gene have been associated with obesity in humans. Alterations in Fto expression in transgenic animals affect body weight, energy expenditure and food intake. Fto, a nuclear protein and proposed transcription co-factor, has been speculated to affect energy balance through a functional relationship with specific genes encoding feeding-related peptides. Herein, we employed double immunohistochemistry and showed that the majority of neurons synthesizing a satiety mediator, oxytocin, coexpress Fto in the brain of male and female mice. We then overexpressed Fto in a murine hypothalamic cell line and, using qPCR, detected a 50% increase in the level of oxytocin mRNA. Expression levels of several other feeding-related genes, including neuropeptide Y (NPY) and Agouti-related protein (AgRP), were unaffected by the FTO transfection. Addition of 10 and 100 nmol oxytocin to the cell culture medium did not affect Fto expression in hypothalamic cells. We conclude that Fto, a proposed transcription co-factor, influences expression of the gene encoding a satiety mediator, oxytocin.
Collapse
Affiliation(s)
- Pawel K Olszewski
- Department of Neuroscience, Functional Pharmacology, Uppsala University, 75124 Uppsala, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kuhla B, Albrecht D, Bruckmaier R, Viergutz T, Nürnberg G, Metges CC. Proteome and radioimmunoassay analyses of pituitary hormones and proteins in response to feed restriction of dairy cows. Proteomics 2010; 10:4491-500. [DOI: 10.1002/pmic.201000383] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
16
|
Calorie restriction at increasing levels leads to augmented concentrations of corticosterone and decreasing concentrations of testosterone in rats. Nutr Res 2010; 30:366-73. [PMID: 20579529 DOI: 10.1016/j.nutres.2010.05.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 04/25/2010] [Accepted: 05/03/2010] [Indexed: 11/20/2022]
Abstract
The influence of calorie restriction (CR) on increasing life span, enhancing immunocompetence, and reducing the incidence of age-related diseases is well established. Evidence points to the involvement of neuroendocrine alterations in these beneficial effects. Accordingly, we hypothesized that CR will result in significant alterations to the hormones investigated. Little attention has been directed toward ascertaining the doses of CR required to obtain such alterations and, indeed, whether a dose-response exists. Adult rats were subjected to 1 of 5 dietary regimens: control, CR12.5%, CR25%, CR37.5%, or CR50%. Rats were decapitated 3 weeks following the onset of restriction; and trunk blood was collected and assayed for concentrations of serum adrenocorticotropic hormone, corticosterone, and testosterone, as well as plasma concentrations of noradrenalin and adrenalin. No effect was found as a result of dietary manipulation for serum concentrations of adrenocorticotropic hormone. However, all doses of CR resulted in increased serum corticosterone in a dose-response trend. A dose-response was also observed for serum testosterone, with higher doses of CR associated with lower testosterone. Concentrations of noradrenalin were not found to be altered by any CR dose, although a trend toward a down-regulation at CR50% was observed. Plasma adrenalin displayed a biphasic distribution with reductions observed at CR25% and CR50%, although the down-regulations only attained statistical significance relative to the CR37.5% and not the control group. As well as reporting the effect of CR on multiple hormones within individual animals, these results go some way in determining the optimal levels of CR needed to induce neuroendocrinologic alterations.
Collapse
|
17
|
Olszewski PK, Klockars A, Olszewska AM, Fredriksson R, Schiöth HB, Levine AS. Molecular, immunohistochemical, and pharmacological evidence of oxytocin's role as inhibitor of carbohydrate but not fat intake. Endocrinology 2010; 151:4736-44. [PMID: 20685878 PMCID: PMC2946140 DOI: 10.1210/en.2010-0151] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Oxytocin (OT) facilitates feeding termination stemming from high osmolality, stomach distention, and malaise. Recent knockout (KO) studies suggested a crucial function for OT in carbohydrate intake: OT-/- mice had increased preference for carbohydrates, including sucrose, but not fat (Intralipid). In striking contrast, sugar appetite was unaffected in the OT receptor KO mouse; data from wild-type animals have been insufficient. Therefore, we examined the involvement of OT in the regulation of sucrose vs. fat intake in C57BL/6 mice that served as a background KO strain. We exposed mice to a meal of sucrose or Intralipid and determined that the percentage of c-Fos-immunoreactive paraventricular hypothalamic OT neurons was elevated at termination of intake of either of the tastants, but this increase was 2-fold higher in sucrose-fed mice. A 48-h exposure to sucrose compared with Intralipid caused up-regulation of OT mRNA, whereas inherent individual preferences for sucrose vs. fat were not associated with differences in baseline OT expression as established with quantitative PCR. We found that L-368,899, an OT receptor antagonist, increased sugar intake when sucrose was presented alone or concurrently with Intralipid; it had no effect on Intralipid or total calorie consumption. L-368,899 affected Fos immunoreactivity in the paraventricular hypothalamus, arcuate nucleus, amygdala, and nucleus of the solitary tract, areas involved in aversion, satiety, and reward. This pattern serves as neuroanatomical basis of OT's complex role in food intake, including sucrose intake. The current findings expand our knowledge on OT and suggest that it acts as a carbohydrate-specific inhibitor of feeding.
Collapse
Affiliation(s)
- Pawel K Olszewski
- Minnesota Obesity Center, University of Minnesota, Department of Food Science and Nutrition, 1334 Eckles Avenue, Saint Paul, Minnesota 55108, USA
| | | | | | | | | | | |
Collapse
|
18
|
Stevens A, Begum G, Cook A, Connor K, Rumball C, Oliver M, Challis J, Bloomfield F, White A. Epigenetic changes in the hypothalamic proopiomelanocortin and glucocorticoid receptor genes in the ovine fetus after periconceptional undernutrition. Endocrinology 2010; 151:3652-64. [PMID: 20573728 DOI: 10.1210/en.2010-0094] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Maternal food restriction is associated with the development of obesity in offspring. This study examined how maternal undernutrition in sheep affects the fetal hypothalamic glucocorticoid receptor (GR) and the appetite-regulating neuropeptides, proopiomelanocortin (POMC) and neuropeptide Y, which it regulates. In fetuses from ewes undernourished from -60 to +30 d around conception, there was increased histone H3K9 acetylation (1.63-fold) and marked hypomethylation (62% decrease) of the POMC gene promoter but no change in POMC expression. In the same group, acetylation of histone H3K9 associated with the hypothalamic GR gene was increased 1.60-fold and the GR promoter region was hypomethylated (53% decrease). In addition, there was a 4.7-fold increase in hypothalamic GR expression but no change in methylation of GR gene expression in the anterior pituitary or hippocampus. Interestingly, hypomethylation of both POMC and GR promoter markers in fetal hypothalami was also identified after maternal undernutrition from -60 to 0 d and -2 to +30 d. In comparison, the Oct4 gene, was hypermethylated in both control and underfed groups. Periconceptional undernutrition is therefore associated with marked epigenetic changes in hypothalamic genes. Increase in GR expression in the undernourished group may contribute to fetal programming of a predisposition to obesity, via altered GR regulation of POMC and neuropeptide Y. These epigenetic changes in GR and POMC in the hypothalamus may also predispose the offspring to altered regulation of food intake, energy expenditure, and glucose homeostasis later in life.
Collapse
Affiliation(s)
- Adam Stevens
- Faculties of Life Sciences and Medical and Human Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M13 9PT, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Olszewski PK, Grace MK, Fard SS, Le Grevès M, Klockars A, Massi M, Schiöth HB, Levine AS. Central nociceptin/orphanin FQ system elevates food consumption by both increasing energy intake and reducing aversive responsiveness. Am J Physiol Regul Integr Comp Physiol 2010; 299:R655-63. [PMID: 20427724 PMCID: PMC3774471 DOI: 10.1152/ajpregu.00556.2009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 04/23/2010] [Indexed: 11/22/2022]
Abstract
Nociceptin/orphanin FQ (N/OFQ), the nociceptin opioid peptide (NOP) receptor ligand, increases feeding when injected centrally. Initial data suggest that N/OFQ blocks the development of a conditioned taste aversion (CTA). The current project further characterized the involvement of N/OFQ in the regulation of hunger vs. aversive responses in rats by employing behavioral, immunohistochemical, and real-time PCR methodology. We determined that the same low dose of the NOP antagonist [Nphe(1)]N/OFQ(1-13)NH(2) delivered via the lateral ventricle diminishes both N/OFQ- and deprivation-induced feeding. This anorexigenic effect did not stem from aversive consequences, as the antagonist did not cause the development of a CTA. When [Nphe(1)]N/OFQ(1-13)NH(2) was administered with LiCl, it moderately delayed extinction of the LiCl-induced CTA. Injection of LiCl + antagonist compared with LiCl alone generated an increase in c-Fos immunoreactivity in the central nucleus of the amygdala. The antagonist alone elevated Fos immunoreactivity in the paraventricular nucleus of the hypothalamus, nucleus of the solitary tract, and central nucleus of the amygdala. Hypothalamic NOP mRNA levels were decreased during energy intake restriction induced by aversion, as well as in non-CTA rats food-restricted to match CTA-reduced consumption. Brain stem NOP was upregulated only in aversion. Prepro-N/OFQ mRNA showed a trend toward upregulation in restricted rats (P = 0.068). We conclude that the N/OFQ system promotes feeding by affecting the need to replenish lacking calories and by reducing aversive responsiveness. It may belong to mechanisms that shift a balance between the drive to ingest energy and avoidance of potentially tainted food.
Collapse
|
20
|
Sugizaki MM, Leopoldo AS, Okoshi MP, Bruno A, Conde SJ, Lima-Leopoldo AP, Padovani CR, Carvalho RF, Nascimento AFD, Campos DHSD, Nogueira CR, Cicogna AC. Severe food restriction induces myocardial dysfunction related to SERCA2 activity. Can J Physiol Pharmacol 2009; 87:666-73. [DOI: 10.1139/y09-060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies have shown that food restriction promotes myocardial dysfunction in rats. However, the molecular mechanisms that are responsible are unclear. We investigated the role of sarcoplasmic reticulum Ca2+-ATPase (SERCA2) on myocardial performance in food-restricted rats. Male Wistar–Kyoto rats, 60 days old, were fed a control or restricted diet (daily energy intake reduced to 50% of the control) for 90 days. Expression of Serca2a, phospholamban (PLB), Na+/Ca2+ exchanger (NCX), and thyroid hormone receptor (TRα1, TRβ1) mRNA was determined by quantitative PCR. SERCA2 activity was measured by using 20 µmol/L cyclopiazonic acid (CPA) in a left ventricular papillary muscle preparation during isometric contraction in basal conditions and during post-rest contraction. Serum concentrations of thyroxine (T4) and thyrotropin (TSH) were also determined. The 50%-restricted diet reduced body and ventricular weight and serum T4 and TSH levels. The interaction of CPA and food restriction reduced peak developed tension and maximum rate of tension decline (–dT/dt), but increased the resting tension intensity response during post-rest contraction. PLB and NCX mRNA were upregulated and TRα1 mRNA was downregulated by food restriction. These results suggest that food restriction promotes myocardial dysfunction related to impairment of sarcoplasmic reticulum Ca2+ uptake as a result of a hypothyroid state.
Collapse
Affiliation(s)
- Mário Mateus Sugizaki
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - André Soares Leopoldo
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - Marina Politi Okoshi
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - Alessandro Bruno
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - Sandro José Conde
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - Ana Paula Lima-Leopoldo
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - Carlos Roberto Padovani
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - Robson Francisco Carvalho
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - André Ferreira do Nascimento
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - Dijon Henrique Salomé de Campos
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - Célia Regina Nogueira
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| | - Antonio Carlos Cicogna
- Department of Clinical and Cardiology, School of Medicine, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Morphology, Institute of Biological Sciences, UNESP State University Júlio Mesquita Filho, Botucatu, São Paulo, Brazil
- Department of Physical Education, FIB, Faculdades Integradas de Bauru, São Paulo, Brazil
| |
Collapse
|
21
|
C6ORF192 Forms a Unique Evolutionary Branch Among Solute Carriers (SLC16, SLC17, and SLC18) and Is Abundantly Expressed in Several Brain Regions. J Mol Neurosci 2009; 41:230-42. [DOI: 10.1007/s12031-009-9222-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 07/13/2009] [Indexed: 10/20/2022]
|
22
|
Alsiö J, Birgner C, Björkblom L, Isaksson P, Bergström L, Schiöth HB, Lindblom J. Impact of nandrolone decanoate on gene expression in endocrine systems related to the adverse effects of anabolic androgenic steroids. Basic Clin Pharmacol Toxicol 2009; 105:307-14. [PMID: 19549128 DOI: 10.1111/j.1742-7843.2009.00439.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Elite athletes, body builders and adolescents misuse anabolic-androgenic steroids (AAS) in order to increase muscle mass or to enhance physical endurance and braveness. The high doses misused are associated with numerous adverse effects. The purpose of this study was to evaluate the impact of chronic supratherapeutic AAS treatment on circulating hormones and gene expression in peripheral tissues related to such adverse effects. Quantitative real-time PCR was used to measure expression levels of in total 37 genes (including peptide hormones, cell membrane receptors, nuclear receptors, steroid synthesising enzymes and other enzymes) in the pituitary, testes, adrenals, adipose tissue, kidneys and liver of male Sprague-Dawley rats after 14-day administration of the AAS nandrolone decanoate, 3 or 15 mg/kg. Plasma glucose and levels of adrenocorticotropic hormone (ACTH), adiponectin, corticosterone, ghrelin, insulin and leptin were also measured. We found several expected effects on the hypothalamic-pituitary-gonadal axis, while the treatment also caused a number of other not previously identified changes in circulating factors and gene transcription levels such as the dose-dependent reduction of the beta(3)-adrenergic receptor in adipose tissue, reduction of both circulating and mRNA levels of adiponectin, up-regulation of both hydroxymethylglutaryl-CoA-reductase, the rate-limiting enzyme in de novo synthesis of cholesterol, and the receptor for ACTH in the adrenals. The results provide evidence for wide ranging effects of AAS on the hypothalamic-pituitary-adrenal axis, adipose tissue and substrates of the renal control of blood pressure.
Collapse
Affiliation(s)
- Johan Alsiö
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Uppsala, Sweden.
| | | | | | | | | | | | | |
Collapse
|
23
|
De Tomasi LC, Bruno A, Sugizaki MM, Lima-Leopoldo AP, Nascimento AF, Júnior SADO, Pinotti MF, Padovani CR, Leopoldo AS, Cicogna AC. Food restriction promotes downregulation of myocardial L-type Ca2+ channels. Can J Physiol Pharmacol 2009; 87:426-31. [DOI: 10.1139/y09-025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Food restriction (FR) has been shown to impair myocardial performance. However, the mechanisms behind these changes in myocardial function due to FR remain unknown. Since myocardial L-type Ca2+ channels may contribute to the cardiac dysfunction, we examined the influence of FR on L-type Ca2+ channels. Male 60-day-old Wistar rats were fed a control or a restricted diet (daily intake reduced to 50% of the amount of food consumed by the control group) for 90 days. Myocardial performance was evaluated in isolated left ventricular papillary muscles. The function of myocardial L-type Ca2+ channels was determined by using a pharmacological Ca2+ channel blocker, and changes in the number of channels were evaluated by mRNA and protein expression. FR decreased final body weights, as well as weights of the left and right ventricles. The Ca2+ channel blocker diltiazem promoted a higher blockade on developed tension in FR groups than in controls. The protein content of L-type Ca2+ channels was significantly diminished in FR rats, whereas the mRNA expression was similar between groups. These results suggest that the myocardial dysfunction observed in previous studies with FR animals could be caused by downregulation of L-type Ca2+ channels.
Collapse
Affiliation(s)
- Loreta Casquel De Tomasi
- Department of Clinical Cardiology, School of Medicine, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
| | - Alessandro Bruno
- Department of Clinical Cardiology, School of Medicine, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
| | - Mário Mateus Sugizaki
- Department of Clinical Cardiology, School of Medicine, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
| | - Ana Paula Lima-Leopoldo
- Department of Clinical Cardiology, School of Medicine, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
| | - André Ferreira Nascimento
- Department of Clinical Cardiology, School of Medicine, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
| | - Silvio Assis de Oliveira Júnior
- Department of Clinical Cardiology, School of Medicine, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
| | - Matheus Fécchio Pinotti
- Department of Clinical Cardiology, School of Medicine, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
| | - Carlos Roberto Padovani
- Department of Clinical Cardiology, School of Medicine, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
| | - André Soares Leopoldo
- Department of Clinical Cardiology, School of Medicine, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
| | - Antonio Carlos Cicogna
- Department of Clinical Cardiology, School of Medicine, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
- Department of Biostatistics, Institute of Biological Sciences, UNESP – State University Júlio Mesquita Filho, Rubião Júnior, S/N, Cep. 18.618-000 Botucatu, São Paulo, Brazil
| |
Collapse
|
24
|
Alsiö J, Roman E, Olszewski PK, Jonsson P, Fredriksson R, Levine AS, Meyerson BJ, Hulting AL, Lindblom J, Schiöth HB. Inverse association of high-fat diet preference and anxiety-like behavior: a putative role for urocortin 2. GENES BRAIN AND BEHAVIOR 2008; 8:193-202. [PMID: 19077174 DOI: 10.1111/j.1601-183x.2008.00464.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The aim of this study was to investigate whether the preference for a palatable high-fat diet (HFD) is associated with response to novelty and with anxiety-like behavior in rats and whether such fat preference correlates with gene expression of hypothalamic neuropeptides related to feeding. We subjected male rats to two tests of exploration of novel environments: the multivariate concentric square field (MCSF) and the elevated plus maze (EPM). The rats were then exposed to a 5-day test of preference for a palatable HFD versus reference diets. Messenger RNA (mRNA) levels of 21 neuropeptides were investigated by quantitative polymerase chain reaction. We found a strong positive correlation of HFD preference and open-arm activity in the EPM (% open-arm time, r(s) = 0.629, df = 26, P < 0.001). Thus, HFD preference was inversely associated with anxiety-like behavior. The same association was found for HFD preference and behavior in the MCSF (bridge entries, r(s) = 0.399, df = 23, P = 0.048). In addition, the HFD preference was positively correlated (r(s) = 0.433, df = 25, P = 0.021) with hypothalamic mRNA levels of urocortin 2 (Ucn 2). Moreover, behavior in the EPM was significantly correlated with expression levels of the receptor for Ucn 2, the corticotropin-releasing factor receptor 2, in the hypothalamus (r(s) = 0.382, df = 33, P = 0.022, pituitary (r(s) = 0.494, df = 31, P = 0.004) and amygdala (r(s) = 0.381, df = 30, P = 0.032). We conclude that preference for palatable HFD is inversely associated with anxiety and propose that Ucn 2 signaling may play a role in this association.
Collapse
Affiliation(s)
- J Alsiö
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Johansson A, Fredriksson R, Winnergren S, Hulting AL, Schiöth HB, Lindblom J. The relative impact of chronic food restriction and acute food deprivation on plasma hormone levels and hypothalamic neuropeptide expression. Peptides 2008; 29:1588-95. [PMID: 18550224 DOI: 10.1016/j.peptides.2008.04.018] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 04/30/2008] [Accepted: 04/30/2008] [Indexed: 12/24/2022]
Abstract
Our understanding of the central regulation of food intake and body weight has increased tremendously through implication of a high number of neuropeptides. However, lack of all-embracing studies have made comparison difficult in the past. The objective of this study was to demonstrate the relative importance of the different neuropeptides in terms of involvement in appetite regulatory mechanisms. We quantified expression levels of 21 hypothalamic neuropeptides and circulating levels of leptin, insulin, corticosterone, adrenocorticotropic hormone, ghrelin and adiponectin in rats after acute food deprivation and chronic food restriction using validated quantitative real-time PCR and hormone measurements. Body weight, insulin and leptin were reduced whereas corticosterone was increased by both acute food deprivation and chronic food restriction. Our results confirmed the relative importance in body weight homeostasis of neuropeptide Y and proopiomelanocortin, which were increased and decreased as predicted. The expression of other neuropeptides previously attributed central roles in body weight homeostasis, e.g. melanin-concentrating hormone and orexin, appeared to be less affected by the treatments. Moreover, the expression of dynorphin, galanin-like peptide and neuropeptide B was dramatically reduced after both treatments. This suggests that the latter neuropeptides--although previously known to be involved in body weight homeostasis--may be of unexpected importance in states of negative energy balance.
Collapse
|
26
|
Sirotkin AV, Chrenková M, Nitrayová S, Patras P, Darlak K, Valenzuela F, Pinilla L, Tena-Sempere M. Effects of chronic food restriction and treatments with leptin or ghrelin on different reproductive parameters of male rats. Peptides 2008; 29:1362-8. [PMID: 18455268 DOI: 10.1016/j.peptides.2008.03.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Revised: 03/12/2008] [Accepted: 03/12/2008] [Indexed: 12/15/2022]
Abstract
The existence of a close relationship between energy status and reproductive function is well-documented, especially in females, but its underlying mechanisms remain to be fully unfolded. This study aimed to examine the effects of restriction of daily calorie intake, as well as chronic treatments with the metabolic hormones leptin and ghrelin, on the secretion of different reproductive hormones, namely pituitary gonadotropins and prolactin, as well as testosterone, in male rats. Restriction (50%) in daily food intake for 20 days significantly reduced body weight as well as plasma PRL and T levels, without affecting basal LH and FSH concentrations and testicular weight. Chronic administration of leptin to rats fed ad libitum increased plasma PRL levels and decreased circulating T, while it did not alter other hormonal parameters under analysis. In contrast, in rats subjected to 50% calorie restriction, leptin administration increased plasma T levels and reduced testis weight. Conversely, ghrelin failed to induce major hormonal changes but tended to increase testicular weight in fed animals, while repeated ghrelin injections in food-restricted males dramatically decreased plasma LH and T concentrations and reduced testis weight. In sum, we document herein the isolated and combined effects of metabolic stress (50% food restriction) and leptin or ghrelin treatments on several reproductive hormones in adult male rats. Overall, our results further stress the impact and complex way of action of different metabolic cues, such as energy status and key hormones, in reproductive function also in the male.
Collapse
Affiliation(s)
- Alexander V Sirotkin
- Research Institute of Animal Production, Slovak Agricultural Research Centre, Hlohovska 2, 949 92 Nitra, Slovakia
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Kindlundh-Högberg AMS, Blomqvist A, Malki R, Schiöth HB. Extensive neuroadaptive changes in cortical gene-transcript expressions of the glutamate system in response to repeated intermittent MDMA administration in adolescent rats. BMC Neurosci 2008; 9:39. [PMID: 18419818 PMCID: PMC2358913 DOI: 10.1186/1471-2202-9-39] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Accepted: 04/17/2008] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Many studies have focused on the implication of the serotonin and dopamine systems in neuroadaptive responses to the recreational drug 3,4-methylenedioxy-metamphetamine (MDMA). Less attention has been given to the major excitatory neurotransmitter glutamate known to be implicated in schizophrenia and drug addiction. The aim of the present study was to investigate the effect of repeated intermittent MDMA administration upon gene-transcript expression of the glutamate transporters (EAAT1, EAAT2-1, EAAT2-2), the glutamate receptor subunits of AMPA (GluR1, GluR2, GluR3), the glutamate receptor subunits of NMDA (NR1, NR2A and NR2B), as well as metabotropic glutamate receptors (mGluR1, mGluR2, mGluR3, mGluR5) in six different brain regions. Adolescent male Sprague Dawley rats received MDMA at the doses of 3 x 1 and 3 x 5 mg/kg/day, or 3x vehicle 3 hours apart, every 7th day for 4 weeks. The gene-transcript levels were assessed using real-time PCR validated with a range of housekeeping genes. RESULTS The findings showed pronounced enhancements in gene-transcript expression of GluR2, mGluR1, mGluR5, NR1, NR2A, NR2B, EAAT1, and EAAT2-2 in the cortex at bregma +1.6. In the caudate putamen, mRNA levels of GluR3, NR2A, and NR2B receptor subunits were significantly increased. In contrast, the gene-transcript expression of GluR1 was reduced in the hippocampus. In the hypothalamus, there was a significant increase of GluR1, GluR3, mGluR1, and mGluR3 gene-transcript expressions. CONCLUSION Repeated intermittent MDMA administration induces neuroadaptive changes in gene-transcript expressions of glutamatergic NMDA and AMPA receptor subunits, metabotropic receptors and transporters in regions of the brain regulating reward-related associative learning, cognition, and memory and neuro-endocrine functions.
Collapse
Affiliation(s)
- Anna MS Kindlundh-Högberg
- Department of Neuroscience, Division of Functional Pharmacology, Uppsala University, 751 24 Uppsala, Sweden
| | - Anna Blomqvist
- Department of Neuroscience, Division of Functional Pharmacology, Uppsala University, 751 24 Uppsala, Sweden
| | - Rana Malki
- Department of Neuroscience, Division of Functional Pharmacology, Uppsala University, 751 24 Uppsala, Sweden
| | - Helgi B Schiöth
- Department of Neuroscience, Division of Functional Pharmacology, Uppsala University, 751 24 Uppsala, Sweden
| |
Collapse
|
28
|
Pickering C, Avesson L, Liljequist S, Lindblom J, Schiöth HB. The role of hypothalamic peptide gene expression in alcohol self-administration behavior. Peptides 2007; 28:2361-71. [PMID: 17976860 DOI: 10.1016/j.peptides.2007.09.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Revised: 09/18/2007] [Accepted: 09/19/2007] [Indexed: 11/19/2022]
Abstract
Self-administration of ethanol and food share many common features and Richter hypothesized that an increase in ethanol consumption would decrease feeding to balance the excess calories contained in the ethanol. Previously, we have shown that individual alcohol consumption correlates with neurotransmitter gene expression, especially in the prefrontal cortex. To test the hypothesis of Richter, we measured hypothalamic gene expression of receptors or neuropeptides of known relevance for the regulation of food intake using qPCR and correlated this to individual ethanol consumption in Wistar rats. For validation, gene expression was first correlated with body weight. We found a correlation of dynorphin, somatostatin, melanocortin-4 receptor and serotonin 5-HT(2C) with body weight and trends to correlation for CART, thus confirming the established role of the hypothalamus in the regulation of weight. For ethanol consumption, correlations were found for CRH receptors 1 and 2 and vasopressin while strong trends were observed for galanin receptor 1, orexin receptor 1, MCH and adrenoceptor alpha(1B). Therefore, alcohol consumption does seem to involve several hypothalamic systems which also mediate feeding responses and suggests that the hypothalamus, together with the prefrontal cortex, may determine the 'stopping point' of an individual.
Collapse
Affiliation(s)
- Chris Pickering
- Uppsala University, Department of Neuroscience, Division of Pharmacology, BMC, Box 593, SE-75124 Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
29
|
Levay EA, Govic A, Penman J, Paolini AG, Kent S. Effects of adult-onset calorie restriction on anxiety-like behavior in rats. Physiol Behav 2007; 92:889-96. [PMID: 17673267 DOI: 10.1016/j.physbeh.2007.06.018] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 05/25/2007] [Accepted: 06/25/2007] [Indexed: 11/20/2022]
Abstract
Calorie restriction (CR) has consistently been shown to increase lifespan and ameliorate disease outcomes. Its effects on behavior are less clear, although anxiolytic-like effects have been observed. Rats were subjected to 1 of 4 dietary regimens: control, CR25%, CR50% and, an acute episode of CR and tested in 3 tests of anxiety: the open field test, the elevated plus maze, and the modified open field test. In the open field test, the CR25% and CR50% groups made more central zone entries than the control and Acute groups, which was primarily due to differences in the initial 5 min of the test. Moreover, both CR groups engaged in greater exploration of the central zone than the control group in the initial 5 min of the test. The Acute group also exhibited significantly longer latencies to leave the central zone at test onset than the control and CR50% group. In the elevated plus maze, the Acute group also displayed longer latencies to open arm entry as compared to the control and CR50% group and showed a lower ratio of open to total arm entries compared to all other groups. There were no effects of CR on any variable of the modified open field test. Possible neurochemical mechanisms underlying the anxiolytic-like effect of CR are discussed.
Collapse
Affiliation(s)
- Elizabeth A Levay
- School of Psychological Science, La Trobe University, Bundoora, VIC 3086, Australia
| | | | | | | | | |
Collapse
|
30
|
Leriche M, Cote-Vélez A, Méndez M. Presence of pro-opiomelanocortin mRNA in the rat medial prefrontal cortex, nucleus accumbens and ventral tegmental area: studies by RT-PCR and in situ hybridization techniques. Neuropeptides 2007; 41:421-31. [PMID: 17980426 DOI: 10.1016/j.npep.2007.08.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 08/01/2007] [Accepted: 08/30/2007] [Indexed: 11/24/2022]
Abstract
Pro-opiomelanocortin (POMC) is a large proteic precursor which originates several biologically actives neuropeptides, such as beta-lipotropin (beta-LPH), beta-endorphin (beta-END), adenocorticotropic hormone (ACTH) and alpha-melanocyte-stimulating hormone (alpha-MSH). The arcuate nucleus of the hypothalamus is the main POMC producing cell group in brain and innervates several areas of the limbic system and brainstem. POMC-derived neuropeptides have been related to several motivated and rewarding behaviours, including sexual facilitation, feeding, and drug addiction. However, POMC mRNA has not been detected in regions of the dopaminergic mesocorticolimbic system, which represents the most important reward pathway. The aim of this work was to investigate if POMC mRNA is expressed in the medial prefrontal cortex (mPFC), the nucleus accumbens (NAcc) and the ventral tegmental area (VTA) of the rat. We used the reverse transcriptase reaction coupled to the polymerase chain reaction (RT-PCR). We also used the in situ hybridization technique to study the regional distribution of POMC mRNA in the same regions. We report that RT-PCR amplification of extracted RNA with two different pairs of primers generates the predicted 94bp and 678bp POMC-PCR products. Both the amplification of RNA obtained from the rat glial C-6 cell line (which does not express POMC mRNA) and the omission of reverse transcriptase from the RT reaction of rat brain samples showed no amplification products. We have shown for the first time that the rat medial prefrontal cortex, the nucleus accumbens and the ventral tegmental area contain POMC mRNA. This mRNA is in low concentration, ranging from 21% to 31% with respect to the hypothalamus. In situ hybridization experiments showed that POMC mRNA is homogeneously distributed in these areas. The presence of POMC mRNA in regions of the mesocorticolimbic system could have functional implications in motivated behaviours.
Collapse
Affiliation(s)
- Mariana Leriche
- Instituto Nacional de Psiquiatría Ramón de la Fuente, Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, 14370 México DF, Mexico
| | | | | |
Collapse
|
31
|
Sirotkin AV, Chrenková M, Nitrayová S, Patraš P. Restricted food intake promotes accumulation of proliferation-, apoptosis-, and anti–apoptotic-related peptides in rat testicular cells. Nutr Res 2007. [DOI: 10.1016/j.nutres.2007.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
32
|
Tanic N, Perovic M, Mladenovic A, Ruzdijic S, Kanazir S. Effects of aging, dietary restriction and glucocorticoid treatment on housekeeping gene expression in rat cortex and hippocampus-evaluation by real time RT-PCR. J Mol Neurosci 2007; 32:38-46. [PMID: 17873286 DOI: 10.1007/s12031-007-0006-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 01/02/2006] [Indexed: 10/23/2022]
Abstract
Accurate normalization is the prerequisite for obtaining reliable results in the quantification of gene expression. Using TaqMan Real Time RT-PCR, we carried out an extensive evaluation of five most commonly used endogenous controls, gapdh, beta-actin, 18S rRNA, hprt and cypB, for their presumed stability of expression, in rat cortex and hippocampus, during aging, under dietary restriction and dexamethasone treatment. Valid reference genes (HKGs) were identified using GeNorm and NormFinder software packages and by direct comparison of Ct values. Analysis revealed gapdh and beta-actin as the most stable HKGs for all treatments analyzed, combined or separately, in the cortex, while in the hippocampus gapdh/hprt and beta-actin/hprt are the combination of choice for the single or combined effects of dietary restriction/dexamethasone, respectively. All treatments significantly influenced expression of 18S rRNA and cypB in both structures. In addition, we used gapdh and normalization factor, calculated by GeNorm, to compare the expression of alpha-syn in the cortex. Our results demonstrate the importance of the right choice of HKG and suggest the appropriate endogenous control to be used for TaqMan RT-PCR analysis of mRNA expression in rat cortex and hippocampus for selected experimental paradigms.
Collapse
Affiliation(s)
- Nikola Tanic
- Institute for Biological Research, Bul. Despota Stefana 142, 11 060 Belgrade, Serbia
| | | | | | | | | |
Collapse
|
33
|
Kuhla B, Kuhla S, Rudolph PE, Albrecht D, Metges CC. Proteomics analysis of hypothalamic response to energy restriction in dairy cows. Proteomics 2007; 7:3602-17. [PMID: 17907270 DOI: 10.1002/pmic.200700248] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The hypothalamus is the central regulatory unit that balances a number of body functions including metabolic rate, hunger, and satiety signals. Hypothalamic neurons monitor and respond to alterations of circulating nutrients and hormones that reflect the peripheral energy status. These extracellular signals are integrated within the cell at the ATP:AMP ratio and at the level of ROS, triggering gene expression associated with glucose and lipid metabolism. In order to identify new molecular factors potentially associated with the control of energy homeostasis, metabolic adaptation, and regulation of feed intake, hypothalami from ad libitum fed and energy restricted cows were characterized using 2-DE and MALDI-TOF-MS. Among 189 different protein spots identified, nine proteins were found to be differentially expressed between groups. Beside the 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase, stress-induced phosphoprotein-1, heat shock protein 70 kDa-protein-5, dihydropyrimidinase-related protein-2, [Cu-Zn]-superoxide dismutase, ubiquitin carboxy-terminal hydrolase-L1, and inorganic pyrophosphatase were found to be up-regulated, whereas glyceraldehyde 3-phosphate dehydrogenase and aconitase-2 were down-regulated in the restricted group. In conclusion, differentially expressed proteins are related to energy and nucleotide metabolism and cellular stress under conditions of dietary energy deficiency. These proteins may be new candidate molecules that are potentially involved in signaling for maintaining energy homeostasis.
Collapse
Affiliation(s)
- Björn Kuhla
- Research Unit Nutritional Physiology Oskar Kellner, Research Institute for the Biology of Farm Animals (FBN), Dummerstorf, Germany
| | | | | | | | | |
Collapse
|
34
|
Jacobsson JA, Haitina T, Lindblom J, Fredriksson R. Identification of six putative human transporters with structural similarity to the drug transporter SLC22 family. Genomics 2007; 90:595-609. [PMID: 17714910 DOI: 10.1016/j.ygeno.2007.03.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 03/21/2007] [Accepted: 03/24/2007] [Indexed: 01/11/2023]
Abstract
The solute carrier family 22 (SLC22) is a large family of organic cation and anion transporters. These are transmembrane proteins expressed predominantly in kidneys and liver and mediate the uptake and excretion of environmental toxins, endogenous substances, and drugs from the body. Through a comprehensive database search we identified six human proteins not yet cloned or annotated in the reference sequence databases. Five of these belong to the SLC22 family, SLC22A20, SLC22A23, SLC22A24, SLC22A25, and SPNS3, and the sixth gene, SVOPL, is a paralog to the synaptic vesicle protein SVOP. We identified the orthologs for these genes in mouse and rat and additional homologous proteins and performed the first phylogenetic analysis on the entire SLC22 family in human, mouse, and rat. In addition, we performed a phylogenetic analysis which showed that SVOP and SV2A-C are, in a comparison with all vertebrate proteins, most similar to the SLC22 family. Finally, we performed a tissue localization study on 15 genes on a panel of 30 rat tissues using quantitative real-time polymerase chain reaction.
Collapse
Affiliation(s)
- Josefin A Jacobsson
- Department of Neuroscience, Unit of Pharmacology, Uppsala University, BMC, Uppsala SE 75124, Sweden
| | | | | | | |
Collapse
|
35
|
Fetoui H, Bouaziz H, Mahjoubi-Samet A, Soussia L, Guermazi F, Zeghal N. Food restriction induced thyroid changes and their reversal after refeeding in female rats and their pups. ACTA BIOLOGICA HUNGARICA 2006; 57:391-402. [PMID: 17278701 DOI: 10.1556/abiol.57.2006.4.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In the present study, two groups of pregnant female rats were submitted to food restriction (24 h fast versus 24 h diet intake) from the 14th day of pregnancy until either the 14th day (group B) or the 4th day after parturition (group C). All pups and their mothers were sacrificed on day 14 after delivery. The body weight of the 14-day-old pups (group B) was 46% less than the controls (group A). Free thyroxine and free triiodothyronine levels in the plasma were reduced by 44 and 16% in pups and by 20 and 36% in their mothers, respectively. These reductions were correlated with a decrease in thyroid iodine content of the pups (-50%) and their mothers (-24%). Radioiodine uptake (131I) by the thyroid gland of pups was significantly increased by 27%. Plasma TSH levels were decreased by 38% in pups and by 44% in dams. Morphological changes in thyroid glands were observed in energy restricted dams and in their pups. Some of follicles in pups were empty. Moroever in dams, we noted the presence of peripheral resorbed vacuoles, sign of thyroid hyperactivity. After a refeeding (group C) period of ten days, total recovery occurred in plasma thyroid hormone levels (FT4 and FT3) and in thyroid iodine contents of pups in spite of a partial recovery of body weights and plasma TSH levels. In dams, a partial recovery occurred in plasma thyroid hormone levels in spite of total recovery in thyroid iodine contents, while plasma TSH levels exceeded control values. A significant amelioration in thyroid histological aspects was observed in pups and their dams.
Collapse
Affiliation(s)
- Hamadi Fetoui
- Animal Physiology Laboratory, Sciences Faculty of Sfax, Sfax, Tunisia
| | | | | | | | | | | |
Collapse
|
36
|
Lindblom J, Johansson A, Holmgren A, Grandin E, Nedergård C, Fredriksson R, Schiöth HB. Increased mRNA levels of tyrosine hydroxylase and dopamine transporter in the VTA of male rats after chronic food restriction. Eur J Neurosci 2006; 23:180-6. [PMID: 16420427 DOI: 10.1111/j.1460-9568.2005.04531.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dieting as a strategy to reduce body weight often fails as it causes food cravings leading to bingeing and weight regain. Evidence from several lines of research suggests the presence of shared elements for neural regulation of food and drug craving. We quantified the expression of eight genes involved in dopamine signalling in brain regions related to the mesolimbic and nigrostriatal dopamine system in male rats subjected to chronic food restriction using quantitative real-time polymerase chain reaction. Food restriction strongly increased mRNA levels of tyrosine hydroxylase and the dopamine transporter in the ventral tegmental area. Quantitative autoradiography indicated that the dopamine transporter was also upregulated at the protein level in the shell of the nucleus accumbens. However, these effects were not observed after acute food deprivation. We suggest that the results reflect a sensitization of the mesolimbic dopamine pathway characterized by increased clearance of extracellular dopamine in the nucleus accumbens shell. Such sensitization of the mesolimbic dopamine system may be one of the underlying causes for the food cravings that interfere with dietary compliance.
Collapse
Affiliation(s)
- Jonas Lindblom
- Department of Neuroscience, Uppsala University, Box 593 BMC, S-751 24 Uppsala, Sweden.
| | | | | | | | | | | | | |
Collapse
|
37
|
Lindblom J, Petrovska R, Hallberg M, Magnusson K, Nyberg F, Uhlén S. Nandrolone treatment decreases the α1B-adrenoceptor mRNA level in rat kidney, but not the density of α1B-adrenoceptors in cultured MDCK-D1 kidney cells. Eur J Pharmacol 2005; 527:157-62. [PMID: 16309668 DOI: 10.1016/j.ejphar.2005.09.062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Revised: 05/12/2005] [Accepted: 09/01/2005] [Indexed: 10/25/2022]
Abstract
We have previously shown that treatment of rats with the anabolic androgen steroid nandrolone decreased the density of alpha1B-adrenoceptors in the rat kidney [Uhlén, S., Lindblom, J., Kindlundh, A., Muhisha, P., Nyberg, F., (2003). Nandrolone treatment decreases the level of rat kidney alpha1B-adrenoceptors. Naunyn-Schmiedeberg's Arch. Pharmacol. 368, 91-98]. This effect may have been caused either by decreased de novo synthesis of alpha1B-adrenoceptors or by increased degradation of alpha1B-adrenoceptors [corrected] In the present study, we show that treatment of rats with nandrolone decreases the level of mRNA for the alpha1B-adrenoceptor in the kidneys, implying decreased synthesis of alpha1B-adrenoceptors. On the other hand, nandrolone did not decrease the density of alpha1B-adrenoceptors in Madin-Darby Canine Kidney (MDCK) cells, even though the sub-cell line tested, MDCK-D1, expresses both the androgen receptor and the alpha1B-adrenoceptor. It is concluded that the regulation of alpha1B-adrenoceptor expression by anabolic androgenic steroids is intricate and cell-type specific.
Collapse
Affiliation(s)
- Jonas Lindblom
- Department of Neuroscience, Division of Pharmacology, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|