1
|
Solár P, Brázda V, Bareš M, Zamani A, EmamiAref P, Joukal A, Kubíčková L, Kročka E, Hašanová K, Joukal M. Inflammatory changes in the choroid plexus following subarachnoid hemorrhage: the role of innate immune receptors and inflammatory molecules. Front Cell Neurosci 2025; 18:1525415. [PMID: 39839349 PMCID: PMC11747387 DOI: 10.3389/fncel.2024.1525415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction The choroid plexus is located in the cerebral ventricles. It consists of a stromal core and a single layer of cuboidal epithelial cells that forms the blood-cerebrospinal barrier. The main function of the choroid plexus is to produce cerebrospinal fluid. Subarachnoid hemorrhage due to aneurysm rupture is a devastating type of hemorrhagic stroke. Following subarachnoid hemorrhage, blood and the blood degradation products that disperse into the cerebrospinal fluid come in direct contact with choroid plexus epithelial cells. The aim of the current study was to elucidate the pathophysiological cascades responsible for the inflammatory reaction that is seen in the choroid plexus following subarachnoid hemorrhage. Methods Subarachnoid hemorrhage was induced in rats by injecting non-heparinized autologous blood to the cisterna magna. Increased intracranial pressure following subarachnoid hemorrhage was modeled by using artificial cerebrospinal fluid instead of blood. Subarachnoid hemorrhage and artificial cerebrospinal fluid animals were left to survive for 1, 3, 7 and 14 days. Immunohistochemical staining of TLR4, TLR9, FPR2, CCL2, TNFα, IL-1β, CCR2 and CX3CR1 was performed on the cryostat sections of choroid plexus tissue. The level of TLR4, TLR9, FPR2, CCL2, TNFα, IL-1β was detected by measuring immunofluorescence intensity in randomly selected epithelial cells. The number of CCR2 and CX3CR1 positive cells per choroid plexus area was manually counted. Immunohistochemical changes were confirmed by Western blot analyses. Results Immunohistochemical methods and Western blot showed increased levels of TLR9 and a slight increase in TLR4 and FRP2 following both subarachnoid hemorrhage as well as the application of artificial cerebrospinal fluid over time, although the individual periods were different. The levels of TNFα and IL-1β increased, while CCL2 level decreased slightly. Accumulation of macrophages positive for CCR2 and CX3CR1 was found in all periods after subarachnoid hemorrhage as well as after the application of artificial cerebrospinal fluid. Discussion Our results suggest that the inflammation develops in the choroid plexus and blood-cerebrospinal fluid barrier in response to blood components as well as acutely increased intracranial pressure following subarachnoid hemorrhage. These pro-inflammatory changes include accumulation in the choroid plexus of pro-inflammatory cytokines, innate immune receptors, and monocyte-derived macrophages.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Neurosurgery, St. Anne’s University Hospital, and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Václav Brázda
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia
| | - Martin Bareš
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Alemeh Zamani
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Parisa EmamiAref
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Andrea Joukal
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Lucie Kubíčková
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Erik Kročka
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Klaudia Hašanová
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marek Joukal
- Department of Anatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| |
Collapse
|
2
|
Mendes N, Zanesco A, Aguiar C, Rodrigues-Luiz GF, Silva D, Campos J, Camara NOS, Moraes-Vieira P, Araujo E, Velloso LA. CXCR3-expressing myeloid cells recruited to the hypothalamus protect against diet-induced body mass gain and metabolic dysfunction. eLife 2024; 13:RP95044. [PMID: 39535032 PMCID: PMC11560133 DOI: 10.7554/elife.95044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Microgliosis plays a critical role in diet-induced hypothalamic inflammation. A few hours after a high-fat diet (HFD), hypothalamic microglia shift to an inflammatory phenotype, and prolonged fat consumption leads to the recruitment of bone marrow-derived cells to the hypothalamus. However, the transcriptional signatures and functions of these cells remain unclear. Using dual-reporter mice, this study reveals that CX3CR1-positive microglia exhibit minimal changes in response to a HFD, while significant transcriptional differences emerge between microglia and CCR2-positive recruited myeloid cells, particularly affecting chemotaxis. These recruited cells also show sex-specific transcriptional differences impacting neurodegeneration and thermogenesis. The chemokine receptor CXCR3 is emphasized for its role in chemotaxis, displaying notable differences between recruited cells and resident microglia, requiring further investigation. Central immunoneutralization of CXCL10, a ligand for CXCR3, resulted in increased body mass and decreased energy expenditure, especially in females. Systemic chemical inhibition of CXCR3 led to significant metabolic changes, including increased body mass, reduced energy expenditure, elevated blood leptin, glucose intolerance, and decreased insulin levels. This study elucidates the transcriptional differences between hypothalamic microglia and CCR2-positive recruited myeloid cells in diet-induced inflammation and identifies CXCR3-expressing recruited immune cells as protective in metabolic outcomes linked to HFD consumption, establishing a new concept in obesity-related hypothalamic inflammation.
Collapse
Affiliation(s)
- Natalia Mendes
- School of Medical Sciences, Department of Translational Medicine (Section of Pharmacology), University of CampinasCampinasBrazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Ariane Zanesco
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Cristhiane Aguiar
- Laboratory of Immunometabolism, Institute of Biology - University of Campinas, BrazilCampinasBrazil
| | - Gabriela F Rodrigues-Luiz
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa CatarinaFlorianópolisBrazil
| | - Dayana Silva
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Jonathan Campos
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Niels Olsen Saraiva Camara
- Laboratory for Transplantation Immunobiology, Institute of Biomedical Sciences, University of Sao PauloSao PauloBrazil
| | - Pedro Moraes-Vieira
- Laboratory of Immunometabolism, Institute of Biology - University of Campinas, BrazilCampinasBrazil
| | - Eliana Araujo
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
- Faculty of Nursing, University of CampinasCampinasBrazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
- National Institute of Science and Technology on NeuroimmunomodulationRio de JaneiroBrazil
| |
Collapse
|
3
|
Davis AB, Lloyd KR, Bollinger JL, Wohleb ES, Reyes TM. Adolescent high fat diet alters the transcriptional response of microglia in the prefrontal cortex in response to stressors in both male and female mice. Stress 2024; 27:2365864. [PMID: 38912878 PMCID: PMC11228993 DOI: 10.1080/10253890.2024.2365864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/28/2024] [Indexed: 06/25/2024] Open
Abstract
Both obesity and high fat diets (HFD) have been associated with an increase in inflammatory gene expression within the brain. Microglia play an important role in early cortical development and may be responsive to HFD, particularly during sensitive windows, such as adolescence. We hypothesized that HFD during adolescence would increase proinflammatory gene expression in microglia at baseline and potentiate the microglial stress response. Two stressors were examined, a physiological stressor [lipopolysaccharide (LPS), IP] and a psychological stressor [15 min restraint (RST)]. From 3 to 7 weeks of age, male and female mice were fed standard control diet (SC, 20% energy from fat) or HFD (60% energy from fat). On P49, 1 h before sacrifice, mice were randomly assigned to either stressor exposure or control conditions. Microglia from the frontal cortex were enriched using a Percoll density gradient and isolated via fluorescence-activated cell sorting (FACS), followed by RNA expression analysis of 30 genes (27 target genes, three housekeeping genes) using Fluidigm, a medium throughput qPCR platform. We found that adolescent HFD induced sex-specific transcriptional response in cortical microglia, both at baseline and in response to a stressor. Contrary to our hypothesis, adolescent HFD did not potentiate the transcriptional response to stressors in males, but rather in some cases, resulted in a blunted or absent response to the stressor. This was most apparent in males treated with LPS. However, in females, potentiation of the LPS response was observed for select proinflammatory genes, including Tnfa and Socs3. Further, HFD increased the expression of Itgam, Ikbkb, and Apoe in cortical microglia of both sexes, while adrenergic receptor expression (Adrb1 and Adra2a) was changed in response to stressor exposure with no effect of diet. These data identify classes of genes that are uniquely affected by adolescent exposure to HFD and different stressor modalities in males and females.
Collapse
Affiliation(s)
- Alyshia B Davis
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Kelsey R Lloyd
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Teresa M Reyes
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
4
|
Campeau S, McNulty C, Stanley JT, Gerber AN, Sasse SK, Dowell RD. Determination of steady-state transcriptome modifications associated with repeated homotypic stress in the rat rostral posterior hypothalamic region. Front Neurosci 2023; 17:1173699. [PMID: 37360161 PMCID: PMC10288150 DOI: 10.3389/fnins.2023.1173699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Chronic stress is epidemiologically correlated with physical and psychiatric disorders. Whereas many animal models of chronic stress induce symptoms of psychopathology, repeated homotypic stressors to moderate intensity stimuli typically reduce stress-related responses with fewer, if any, pathological symptoms. Recent results indicate that the rostral posterior hypothalamic (rPH) region is a significant component of the brain circuitry underlying response reductions (habituation) associated with repeated homotypic stress. To test whether posterior hypothalamic transcriptional regulation associates with the neuroendocrine modifications induced by repeated homotypic stress, RNA-seq was performed in the rPH dissected from adult male rats that experienced either no stress, 1, 3, or 7 stressful loud noise exposures. Plasma samples displayed reliable increases of corticosterone in all stressed groups, with the smallest increase in the group exposed to 7 loud noises, indicating significant habituation compared to the other stressed groups. While few or no differentially expressed genes were detected 24-h after one or three loud noise exposures, relatively large numbers of transcripts were differentially expressed between the group exposed to 7 loud noises when compared to the control or 3-stress groups, respectively, which correlated with the corticosterone response habituation observed. Gene ontology analyses indicated multiple significant functional terms related to neuron differentiation, neural membrane potential, pre- and post-synaptic elements, chemical synaptic transmission, vesicles, axon guidance and projection, glutamatergic and GABAergic neurotransmission. Some of the differentially expressed genes (Myt1l, Zmat4, Dlx6, Csrnp3) encode transcription factors that were independently predicted by transcription factor enrichment analysis to target other differentially regulated genes in this study. A similar experiment employing in situ hybridization histochemical analysis in additional animals validated the direction of change of the 5 transcripts investigated (Camk4, Gabrb2, Gad1, Grin2a and Slc32a) with a high level of temporal and regional specificity for the rPH. In aggregate, the results suggest that distinct patterns of gene regulation are obtained in response to a repeated homotypic stress regimen; they also point to a significant reorganization of the rPH region that may critically contribute to the phenotypic modifications associated with repeated homotypic stress habituation.
Collapse
Affiliation(s)
- Serge Campeau
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States
| | - Connor McNulty
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States
| | - Jacob T. Stanley
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States
- BioFrontiers Institute, University of Colorado, Boulder, CO, United States
| | - Anthony N. Gerber
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Sarah K. Sasse
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Robin D. Dowell
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States
- BioFrontiers Institute, University of Colorado, Boulder, CO, United States
- Department of Computer Science, University of Colorado, Boulder, CO, United States
| |
Collapse
|
5
|
Markov DD, Novosadova EV. Chronic Unpredictable Mild Stress Model of Depression: Possible Sources of Poor Reproducibility and Latent Variables. BIOLOGY 2022; 11:1621. [PMID: 36358321 PMCID: PMC9687170 DOI: 10.3390/biology11111621] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/29/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023]
Abstract
Major depressive disorder (MDD) is one of the most common mood disorders worldwide. A lack of understanding of the exact neurobiological mechanisms of depression complicates the search for new effective drugs. Animal models are an important tool in the search for new approaches to the treatment of this disorder. All animal models of depression have certain advantages and disadvantages. We often hear that the main drawback of the chronic unpredictable mild stress (CUMS) model of depression is its poor reproducibility, but rarely does anyone try to find the real causes and sources of such poor reproducibility. Analyzing the articles available in the PubMed database, we tried to identify the factors that may be the sources of the poor reproducibility of CUMS. Among such factors, there may be chronic sleep deprivation, painful stressors, social stress, the difference in sex and age of animals, different stress susceptibility of different animal strains, handling quality, habituation to stressful factors, various combinations of physical and psychological stressors in the CUMS protocol, the influence of olfactory and auditory stimuli on animals, as well as the possible influence of various other factors that are rarely taken into account by researchers. We assume that careful inspection of these factors will increase the reproducibility of the CUMS model between laboratories and allow to make the interpretation of the obtained results and their comparison between laboratories to be more adequate.
Collapse
|
6
|
Jiang Z, Chen C, Weiss GL, Fu X, Stelly CE, Sweeten BLW, Tirrell PS, Pursell I, Stevens CR, Fisher MO, Begley JC, Harrison LM, Tasker JG. Stress-induced glucocorticoid desensitizes adrenoreceptors to gate the neuroendocrine response to somatic stress in male mice. Cell Rep 2022; 41:111509. [PMID: 36261014 PMCID: PMC9635929 DOI: 10.1016/j.celrep.2022.111509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/05/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022] Open
Abstract
Noradrenergic afferents to hypothalamic corticotropin releasing hormone (CRH) neurons provide a major excitatory drive to the hypothalamic-pituitary-adrenal (HPA) axis via α1 adrenoreceptor activation. Noradrenergic afferents are recruited preferentially by somatic, rather than psychological, stress stimuli. Stress-induced glucocorticoids feed back onto the hypothalamus to negatively regulate the HPA axis, providing a critical autoregulatory constraint that prevents glucocorticoid overexposure and neuropathology. Whether negative feedback mechanisms target stress modality-specific HPA activation is not known. Here, we describe a desensitization of the α1 adrenoreceptor activation of the HPA axis following acute stress in male mice that is mediated by rapid glucocorticoid regulation of adrenoreceptor trafficking in CRH neurons. Glucocorticoid-induced α1 receptor trafficking desensitizes the HPA axis to a somatic but not a psychological stressor. Our findings demonstrate a rapid glucocorticoid suppression of adrenergic signaling in CRH neurons that is specific to somatic stress activation, and they reveal a rapid, stress modality-selective glucocorticoid negative feedback mechanism.
Collapse
Affiliation(s)
- Zhiying Jiang
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA
| | - Chun Chen
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA
| | - Grant L Weiss
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA
| | - Xin Fu
- Neuroscience Program, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Claire E Stelly
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Brook L W Sweeten
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Parker S Tirrell
- Neuroscience Program, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - India Pursell
- Neuroscience Program, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Carly R Stevens
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA
| | - Marc O Fisher
- Neuroscience Program, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - John C Begley
- Neuroscience Program, Tulane University, New Orleans, LA 70118, USA
| | - Laura M Harrison
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Jeffrey G Tasker
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
7
|
de Oliveira RP, de Andrade JS, Spina M, Chamon JV, Silva PHD, Werder AK, Ortolani D, Thomaz LDSC, Romariz S, Ribeiro DA, Longo BM, Spadari RC, Viana MDB, Melo-Thomas L, Céspedes IC, da Silva RCB. Clozapine prevented social interaction deficits and reduced c-Fos immunoreactivity expression in several brain areas of rats exposed to acute restraint stress. PLoS One 2022; 17:e0262728. [PMID: 35239670 PMCID: PMC8893644 DOI: 10.1371/journal.pone.0262728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/04/2022] [Indexed: 11/21/2022] Open
Abstract
In the present study, we evaluate the effect of acute restraint stress (15 min) of male Wistar rats on social interaction measurements and c-Fos immunoreactivity (c-Fos-ir) expression, a marker of neuronal activity, in areas involved with the modulation of acute physical restraint in rats, i.e., the paraventricular nucleus of the hypothalamus (PVN), median raphe nucleus (MnR), medial prefrontal cortex (mPFC), cingulate prefrontal cortex (cPFC), nucleus accumbens (NaC), hippocampus (CA3), lateral septum (LS) and medial amygdala (MeA). We considered the hypothesis that restraint stress exposure could promote social withdrawal induced by the activation of the hypothalamic-pituitary-adrenocortical (HPA) axis, and increase c-Fos expression in these limbic forebrain areas investigated. In addition, we investigated whether pretreatment with the atypical antipsychotic clozapine (5 mg/kg; I.P.) could attenuate or block the effects of restraint on these responses. We found that restraint stress induced social withdrawal, and increased c-Fos-ir in these areas, demonstrating that a single 15 min session of physical restraint of rats effectively activated the HPA axis, representing an effective tool for the investigation of neuronal activity in brain regions sensitive to stress. Conversely, pretreatment with clozapine, prevented social withdrawal and reduced c-Fos expression. We suggest that treatment with clozapine exerted a preventive effect in the social interaction deficit, at least in part, by blocking the effect of restraint stress in brain regions that are known to regulate the HPA-axis, including the cerebral cortex, hippocampus, hypothalamus, septum and amygdala. Further experiments will be done to confirm this hypothesis.
Collapse
Affiliation(s)
| | - José Simões de Andrade
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - Marianna Spina
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - João Vítor Chamon
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | | | - Ana Keyla Werder
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - Daniela Ortolani
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | | | - Simone Romariz
- Departamento de Fisiologia, Universidade Federal de São Paulo (UNIFESP/SP), São Paulo, Brazil
| | - Daniel Araki Ribeiro
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - Beatriz Monteiro Longo
- Departamento de Fisiologia, Universidade Federal de São Paulo (UNIFESP/SP), São Paulo, Brazil
| | - Regina Célia Spadari
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - Milena de Barros Viana
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
| | - Liana Melo-Thomas
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-Universityof Marburg, Marburg, Germany
- Marburg Center for Mind, Brain, and Behavior (MCMBB), Marburg, Marburg, Germany
| | - Isabel Cristina Céspedes
- Departamento de Morfologia e Genética, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Regina Cláudia Barbosa da Silva
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos (SP), Brazil
- Instituto de Neurociências e Comportamento (INeC), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
8
|
Bergamini G, Coloma P, Massinet H, Steiner MA. What evidence is there for implicating the brain orexin system in neuropsychiatric symptoms in dementia? Front Psychiatry 2022; 13:1052233. [PMID: 36506416 PMCID: PMC9732550 DOI: 10.3389/fpsyt.2022.1052233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/07/2022] [Indexed: 11/26/2022] Open
Abstract
Neuropsychiatric symptoms (NPS) affect people with dementia (PwD) almost universally across all stages of the disease, and regardless of its exact etiology. NPS lead to disability and reduced quality of life of PwD and their caregivers. NPS include hyperactivity (agitation and irritability), affective problems (anxiety and depression), psychosis (delusions and hallucinations), apathy, and sleep disturbances. Preclinical studies have shown that the orexin neuropeptide system modulates arousal and a wide range of behaviors via a network of axons projecting from the hypothalamus throughout almost the entire brain to multiple, even distant, regions. Orexin neurons integrate different types of incoming information (e.g., metabolic, circadian, sensory, emotional) and convert them into the required behavioral output coupled to the necessary arousal status. Here we present an overview of the behavioral domains influenced by the orexin system that may be relevant for the expression of some critical NPS in PwD. We also hypothesize on the potential effects of pharmacological interference with the orexin system in the context of NPS in PwD.
Collapse
Affiliation(s)
- Giorgio Bergamini
- CNS Pharmacology and Drug Discovery, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Preciosa Coloma
- Clinical Science, Global Clinical Development, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Helene Massinet
- CNS Pharmacology and Drug Discovery, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | |
Collapse
|
9
|
Färber N, Manuel J, May M, Foadi N, Beissner F. The Central Inflammatory Network: A Hypothalamic fMRI Study of Experimental Endotoxemia in Humans. Neuroimmunomodulation 2022; 29:231-247. [PMID: 34610606 PMCID: PMC9254315 DOI: 10.1159/000519061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/25/2021] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Inflammation is a mechanism of the immune system that is part of the reaction to pathogens or injury. The central nervous system closely regulates inflammation via neuroendocrine or direct neuroimmune mechanisms, but our current knowledge of the underlying circuitry is limited. Therefore, we aimed to identify hypothalamic centres involved in sensing or modulating inflammation and to study their association with known large-scale brain networks. METHODS Using high-resolution functional magnetic resonance imaging (fMRI), we recorded brain activity in healthy male subjects undergoing experimental inflammation from intravenous endotoxin. Four fMRI runs covered key phases of the developing inflammation: pre-inflammatory baseline, onset of endotoxemia, onset of pro-inflammatory cytokinemia, and peak of pro-inflammatory cytokinemia. Using masked independent component analysis, we identified functionally homogeneous subregions of the hypothalamus, which were further tested for changes in functional connectivity during inflammation and for temporal correlation with tumour necrosis factor and adrenocorticotropic hormone serum levels. We then studied the connection of these inflammation-associated hypothalamic subregions with known large-scale brain networks. RESULTS Our results show that there are at least 6 hypothalamic subregions associated with inflammation in humans including the paraventricular nucleus, supraoptic nucleus, dorsomedial hypothalamus, bed nucleus of the stria terminalis, lateral hypothalamic area, and supramammillary nucleus. They are functionally embedded in at least 3 different large-scale brain networks, namely a medial frontoparietal network, an occipital-pericentral network, and a midcingulo-insular network. CONCLUSION Measuring how the hypothalamus detects or modulates systemic inflammation is a first step to understand central nervous immunomodulation.
Collapse
Affiliation(s)
- Natalia Färber
- Somatosensory and Autonomic Therapy Research, Institute for Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hanover, Germany
- *Natalia Färber,
| | - Jorge Manuel
- Somatosensory and Autonomic Therapy Research, Institute for Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hanover, Germany
| | - Marcus May
- CRC Core Facility, Hannover Medical School, Hanover, Germany
| | - Nilufar Foadi
- Clinic for Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hanover, Germany
| | - Florian Beissner
- Somatosensory and Autonomic Therapy Research, Institute for Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hanover, Germany
- **Florian Beissner,
| |
Collapse
|
10
|
Nikbakhsh R, Nikbakhsh R, Radmard M, Tafazolimoghadam A, Haj-Mirzaian A, Pirri F, Noormohammady P, Sabouri M, Shababi N, Ziai SA, Dehpour AR. The possible role of nitric oxide in anti-convulsant effects of Naltrindole in seizure-induced by social isolation stress in male mice. Biomed Pharmacother 2020; 129:110453. [DOI: 10.1016/j.biopha.2020.110453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/11/2020] [Accepted: 06/23/2020] [Indexed: 01/06/2023] Open
|
11
|
Puleo C, Cotero V. Noninvasive Neuromodulation of Peripheral Nerve Pathways Using Ultrasound and Its Current Therapeutic Implications. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a034215. [PMID: 31138539 DOI: 10.1101/cshperspect.a034215] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review describes work from several research groups in which ultrasound is being used to target the peripheral nervous system and perform neuromodulation noninvasively. Although these techniques are in their infancy compared to implant-based and electrical nerve stimulation, if successful this new noninvasive method for neuromodulation could solve many of the challenges facing the field of bioelectronic medicine. The work outlined herein shows results in which two different (potentially therapeutic) targets are stimulated, a neuroimmune pathway within the spleen and a nutrient/sensory pathway within the liver. Both data and discussion are provided that compare this new noninvasive technique to implant-based nerve stimulation.
Collapse
|
12
|
Exposure to in utero inflammation increases locomotor activity, alters cognitive performance and drives vulnerability to cognitive performance deficits after acute immune activation. Brain Behav Immun 2019; 80:56-65. [PMID: 30797960 DOI: 10.1016/j.bbi.2019.02.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/31/2018] [Accepted: 02/20/2019] [Indexed: 12/24/2022] Open
Abstract
Fetal exposure to intrauterine inflammation (IUI) affects brain development. Using intrauterine lipopolysaccharide (LPS) administration to induce a localized, rather than a systemic, inflammation, we have previously shown that IUI increases cytokine expression and microglia number, and reduces white matter in the brains of exposed offspring. Clinical data suggest that IUI may increase the risk for cognitive and neurodevelopmental disorders, however, IUI is often found in the context of preterm birth, making it difficult to disentangle the adverse effects of inflammation from those related to prematurity. Therefore, using a mouse model of IUI that does not involve preterm birth, operant tasks were used to evaluate motivation, attention, impulsivity, and locomotion. IUI-exposed offspring were found to have increased locomotion and increased motivation (females only), and testing in the 5-choice serial reaction time task (5-CSRTT) showed that IUI-exposed offspring performed more trials and could respond accurately at a shorter stimulus length. We have previously shown that IUI animals have a potentiated cytokine response to a "second hit" (acute LPS injection) in adulthood, so animals' performance in the 5CSRTT was evaluated following an acute injection of LPS. As opposed to the improved performance observed under baseline conditions, IUI exposed animals demonstrated a greater decrease in performance after an acute LPS administration. To identify putative molecular mechanisms underlying this potentiated decline in cognitive performance, PFC samples were collected immediately after post-LPS cognitive testing and targeted gene expression analysis was correlated with specific measures of cognitive performance. Three receptors important for neuron-microglia crosstalk were found to correlate with task performance in the males following acute LPS administration. These data demonstrate that early life exposure to localized inflammation of the uterus, in the absence of prematurity, increases locomotor activity and improves some aspects of cognitive performance, but drives a vulnerability for adult cognitive performance deficits in response to acute infection.
Collapse
|
13
|
Noninvasive sub-organ ultrasound stimulation for targeted neuromodulation. Nat Commun 2019; 10:952. [PMID: 30862827 PMCID: PMC6414607 DOI: 10.1038/s41467-019-08750-9] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 01/22/2019] [Indexed: 12/20/2022] Open
Abstract
Tools for noninvasively modulating neural signaling in peripheral organs will advance the study of nerves and their effect on homeostasis and disease. Herein, we demonstrate a noninvasive method to modulate specific signaling pathways within organs using ultrasound (U/S). U/S is first applied to spleen to modulate the cholinergic anti-inflammatory pathway (CAP), and US stimulation is shown to reduce cytokine response to endotoxin to the same levels as implant-based vagus nerve stimulation (VNS). Next, hepatic U/S stimulation is shown to modulate pathways that regulate blood glucose and is as effective as VNS in suppressing the hyperglycemic effect of endotoxin exposure. This response to hepatic U/S is only found when targeting specific sub-organ locations known to contain glucose sensory neurons, and both molecular (i.e. neurotransmitter concentration and cFOS expression) and neuroimaging results indicate US induced signaling to metabolism-related hypothalamic sub-nuclei. These data demonstrate that U/S stimulation within organs provides a new method for site-selective neuromodulation to regulate specific physiological functions. Stimulation of peripheral nerve activity may be used to treat metabolic and inflammatory disorders, but current approaches need implanted devices. Here, the authors present a non-invasive approach, and show that ultrasound-mediated stimulation can be targeted to specific sub-organ locations in preclinical models and alter the response of metabolic and inflammatory neural pathways.
Collapse
|
14
|
Hwang KA, Hwang YJ, Hwang IG, Song J, Jun Kim Y. Low temperature-aged garlic extract suppresses psychological stress by modulation of stress hormones and oxidative stress response in brain. J Chin Med Assoc 2019; 82:191-195. [PMID: 30908412 DOI: 10.1097/jcma.0000000000000028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Garlic is a folk medicine known for its multiple physiological activities, but the neuro-modulatory effect of garlic against psychological stress has rarely been explored. The current study was conducted to determine the potential antipsychological stress effect of low temperature-aged garlic (LTAG). METHODS After acute restraint stress exposure, mice were administered with raw garlic (RG, 500 mg/kg, p.o.) or LTAG (500 mg/kg, p.o.). We investigated corticosterone, cortisol, and monoamines levels, and the mRNA expression of genes relevant to oxidative stress. RESULTS RG and LTAG treatment significantly decreased stress-related hormones such as corticotropin-releasing factor, adrenocorticotropic hormone, corticosterone, and cortisol. Moreover, RG and LTAG administration significantly restored acute restraint stress-induced changes in concentrations of brain neurotransmitters (serotonin, norepinephrine, dopamine, and epinephrine). In addition, RG and LTAG improved the antioxidant defense system by causing an increase in mRNA expression of superoxide dismutase, catalase, and glutathione peroxidase in the brain. CONCLUSION This study suggests an antipsychological stress and neuroprotective effect of RG and LTAG under stress conditions.
Collapse
Affiliation(s)
- Kyung-A Hwang
- Department of Agrofood Resources, National Institute of Agricultural Sciences, RDA, Wanju-Gun, Jeollabuk-do, Korea
| | - Yu-Jin Hwang
- Department of Agrofood Resources, National Institute of Agricultural Sciences, RDA, Wanju-Gun, Jeollabuk-do, Korea
| | - In-Guk Hwang
- Department of Agrofood Resources, National Institute of Agricultural Sciences, RDA, Wanju-Gun, Jeollabuk-do, Korea
| | - Jin Song
- Department of Agrofood Resources, National Institute of Agricultural Sciences, RDA, Wanju-Gun, Jeollabuk-do, Korea
| | - Young Jun Kim
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| |
Collapse
|
15
|
Zajdel J, Zager A, Blomqvist A, Engblom D, Shionoya K. Acute maternal separation potentiates the gene expression and corticosterone response induced by inflammation. Brain Behav Immun 2019; 77:141-149. [PMID: 30590109 DOI: 10.1016/j.bbi.2018.12.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 12/18/2018] [Accepted: 12/22/2018] [Indexed: 02/06/2023] Open
Abstract
Maternal care is crucial for infants and profoundly affects their responses to different kinds of stressors. Here, we examined how maternal separation affects inflammatory gene expression and the corticosterone response to an acute immune challenge induced by lipopolysaccharide (LPS; 40 µg/kg ip) in mouse pups, 8-9 days old. Maternal separation initially attenuated LPS-induced hypothalamic pro-inflammatory gene expression, but later, at 3 h after immune challenge, robustly augmented such gene expression and increased serum corticosterone levels. Providing the pups with a warm and soft object prevented the separation-induced augmented hypothalamic-pituitary-adrenal (HPA)-axis response. It also prevented the potentiated induction of some, but not all, inflammatory genes to a similar extent as did the dam. Our results show that maternal separation potentiates the inflammatory response and the resulting HPA-axis activation, which may have detrimental effects if separation is prolonged or repeated.
Collapse
Affiliation(s)
- Joanna Zajdel
- Center for Social and Affective Neuroscience and Faculty of Medicine and Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| | - Adriano Zager
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| | - Anders Blomqvist
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| | - David Engblom
- Center for Social and Affective Neuroscience and Faculty of Medicine and Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| | - Kiseko Shionoya
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, S-581 85 Linköping, Sweden.
| |
Collapse
|
16
|
Phan TX, Malkani RG. Sleep and circadian rhythm disruption and stress intersect in Alzheimer's disease. Neurobiol Stress 2019; 10:100133. [PMID: 30937343 PMCID: PMC6279965 DOI: 10.1016/j.ynstr.2018.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/12/2018] [Accepted: 10/13/2018] [Indexed: 01/12/2023] Open
Abstract
Alzheimer's disease (AD) was discovered and the pathological hallmarks were revealed more than a century ago. Subsequently, many remarkable discoveries and breakthroughs provided us with mechanistic insights into the pathogenesis of AD. The identification of the molecular underpinning of the disease not only provided the framework of AD pathogenesis but also targets for therapeutic inventions. Despite all the initial successes, no effective treatment for AD has emerged yet as all the late stages of clinical trials have failed. Many factors ranging from genetic to environmental factors have been critically appraised as the potential causes of AD. In particular, the role of stress on AD has been intensively studied while the relationship between sleep and circadian rhythm disruption (SCRD) and AD have recently emerged. SCRD has always been thought to be a corollary of AD pathologies until recently, multiple lines of evidence converge on the notion that SCRD might be a contributing factor in AD pathogenesis. More importantly, how stress and SCRD intersect and make their concerted contributions to AD phenotypes has not been reviewed. The goal of this literature review is to examine at multiple levels - molecular, cellular (e.g. microglia, gut microbiota) and holistic - how the interaction between stress and SCRD bi-directionally and synergistically exacerbate AD pathologies and cognitive impairment. AD, in turn, worsens stress and SCRD and forms the vicious cycle that perpetuates and amplifies AD.
Collapse
Affiliation(s)
- Trongha X. Phan
- Department of Neurology, Division of Sleep Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Circadian and Sleep Medicine, Northwestern University, Chicago, IL, USA
| | - Roneil G. Malkani
- Department of Neurology, Division of Sleep Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Circadian and Sleep Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
17
|
Sargin D. The role of the orexin system in stress response. Neuropharmacology 2018; 154:68-78. [PMID: 30266600 DOI: 10.1016/j.neuropharm.2018.09.034] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/18/2018] [Accepted: 09/21/2018] [Indexed: 11/30/2022]
Abstract
Orexins are neuropeptides that are exclusively produced by hypothalamic neurons, which project throughout the entire brain. Orexin, also known as hypocretins, were initially identified to play a fundamental role in food intake, arousal and the regulation of sleep and wakefulness. Recent studies identified orexins to be critical for diverse physiological processes including motivation, reward, attention, emotional regulation, stress and anxiety. Here, I review recent findings that indicate orexin has an important role in acute and chronic stress. I also summarize the recent optogenetic and chemogenetic studies that have advanced our understanding of the orexin system. I will conclude by discussing clinical studies that implicate orexins in mental health disorders. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.
Collapse
Affiliation(s)
- Derya Sargin
- Hotchkiss Brain Institute and the Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
18
|
Mumtaz F, Khan MI, Zubair M, Dehpour AR. Neurobiology and consequences of social isolation stress in animal model-A comprehensive review. Biomed Pharmacother 2018; 105:1205-1222. [PMID: 30021357 DOI: 10.1016/j.biopha.2018.05.086] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 05/10/2018] [Accepted: 05/18/2018] [Indexed: 12/09/2022] Open
Abstract
The brain is a vital organ, susceptible to alterations under genetic influences and environmental experiences. Social isolation (SI) acts as a stressor which results in alterations in reactivity to stress, social behavior, function of neurochemical and neuroendocrine system, physiological, anatomical and behavioral changes in both animal and humans. During early stages of life, acute or chronic SIS has been proposed to show signs and symptoms of psychiatric and neurological disorders such as anxiety, depression, schizophrenia, epilepsy and memory loss. Exposure to social isolation stress induces a variety of endocrinological changes including the activation of hypothalamic-pituitary-adrenal (HPA) axis, culminating in the release of glucocorticoids (GCs), release of catecholamines, activation of the sympatho-adrenomedullary system, release of Oxytocin and vasopressin. In several regions of the central nervous system (CNS), SIS alters the level of neurotransmitter such as dopamine, serotonin, gamma aminobutyric acid (GABA), glutamate, nitrergic system and adrenaline as well as leads to alteration in receptor sensitivity of N-methyl-D-aspartate (NMDA) and opioid system. A change in the function of oxidative and nitrosative stress (O&NS) mediated mitochondrial dysfunction, inflammatory factors, neurotrophins and neurotrophicfactors (NTFs), early growth response transcription factor genes (Egr) and C-Fos expression are also involved as a pathophysiological consequences of SIS which induce neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Faiza Mumtaz
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Muhammad Imran Khan
- Department of Pharmacy, Kohat University of Science and Technology, 26000 Kohat, KPK, Pakistan; Drug Detoxification Health Welfare Research Center, Bannu, KPK, Pakistan
| | - Muhammad Zubair
- Key Laboratory of Integrated Management of Crop Diseases and Pests, College of Plant Protection, Nanjing Agriculture University, Nanjing, 210095, PR China
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Khan AM, Grant AH, Martinez A, Burns GAPC, Thatcher BS, Anekonda VT, Thompson BW, Roberts ZS, Moralejo DH, Blevins JE. Mapping Molecular Datasets Back to the Brain Regions They are Extracted from: Remembering the Native Countries of Hypothalamic Expatriates and Refugees. ADVANCES IN NEUROBIOLOGY 2018; 21:101-193. [PMID: 30334222 PMCID: PMC6310046 DOI: 10.1007/978-3-319-94593-4_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This article focuses on approaches to link transcriptomic, proteomic, and peptidomic datasets mined from brain tissue to the original locations within the brain that they are derived from using digital atlas mapping techniques. We use, as an example, the transcriptomic, proteomic and peptidomic analyses conducted in the mammalian hypothalamus. Following a brief historical overview, we highlight studies that have mined biochemical and molecular information from the hypothalamus and then lay out a strategy for how these data can be linked spatially to the mapped locations in a canonical brain atlas where the data come from, thereby allowing researchers to integrate these data with other datasets across multiple scales. A key methodology that enables atlas-based mapping of extracted datasets-laser-capture microdissection-is discussed in detail, with a view of how this technology is a bridge between systems biology and systems neuroscience.
Collapse
Affiliation(s)
- Arshad M Khan
- UTEP Systems Neuroscience Laboratory, University of Texas at El Paso, El Paso, TX, USA.
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA.
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA.
| | - Alice H Grant
- UTEP Systems Neuroscience Laboratory, University of Texas at El Paso, El Paso, TX, USA
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
- Graduate Program in Pathobiology, University of Texas at El Paso, El Paso, TX, USA
| | - Anais Martinez
- UTEP Systems Neuroscience Laboratory, University of Texas at El Paso, El Paso, TX, USA
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
- Graduate Program in Pathobiology, University of Texas at El Paso, El Paso, TX, USA
| | - Gully A P C Burns
- Information Sciences Institute, Viterbi School of Engineering, University of Southern California, Marina del Rey, CA, USA
| | - Brendan S Thatcher
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, USA
| | - Vishwanath T Anekonda
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, USA
| | - Benjamin W Thompson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, USA
| | - Zachary S Roberts
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, USA
| | - Daniel H Moralejo
- Division of Neonatology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - James E Blevins
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, USA
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
20
|
Matsuwaki T, Shionoya K, Ihnatko R, Eskilsson A, Kakuta S, Dufour S, Schwaninger M, Waisman A, Müller W, Pinteaux E, Engblom D, Blomqvist A. Involvement of interleukin-1 type 1 receptors in lipopolysaccharide-induced sickness responses. Brain Behav Immun 2017; 66:165-176. [PMID: 28655587 DOI: 10.1016/j.bbi.2017.06.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/15/2017] [Accepted: 06/23/2017] [Indexed: 12/17/2022] Open
Abstract
Sickness responses to lipopolysaccharide (LPS) were examined in mice with deletion of the interleukin (IL)-1 type 1 receptor (IL-1R1). IL-1R1 knockout (KO) mice displayed intact anorexia and HPA-axis activation to intraperitoneally injected LPS (anorexia: 10 or 120µg/kg; HPA-axis: 120µg/kg), but showed attenuated but not extinguished fever (120µg/kg). Brain PGE2 synthesis was attenuated, but Cox-2 induction remained intact. Neither the tumor necrosis factor-α (TNFα) inhibitor etanercept nor the IL-6 receptor antibody tocilizumab abolished the LPS induced fever in IL-1R1 KO mice. Deletion of IL-1R1 specifically in brain endothelial cells attenuated the LPS induced fever, but only during the late, 3rd phase of fever, whereas deletion of IL-1R1 on neural cells or on peripheral nerves had little or no effect on the febrile response. We conclude that while IL-1 signaling is not critical for LPS induced anorexia or stress hormone release, IL-1R1, expressed on brain endothelial cells, contributes to the febrile response to LPS. However, also in the absence of IL-1R1, LPS evokes a febrile response, although this is attenuated. This remaining fever seems not to be mediated by IL-6 receptors or TNFα, but by some yet unidentified pyrogenic factor.
Collapse
Affiliation(s)
- Takashi Matsuwaki
- Department of Clinical and Experimental Medicine, Linköping University, 581 85 Linköping, Sweden; Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kiseko Shionoya
- Department of Clinical and Experimental Medicine, Linköping University, 581 85 Linköping, Sweden
| | - Robert Ihnatko
- Department of Clinical and Experimental Medicine, Linköping University, 581 85 Linköping, Sweden
| | - Anna Eskilsson
- Department of Clinical and Experimental Medicine, Linköping University, 581 85 Linköping, Sweden
| | - Shigeru Kakuta
- Department of Biomedical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, 23538 Lübeck, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Werner Müller
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - David Engblom
- Department of Clinical and Experimental Medicine, Linköping University, 581 85 Linköping, Sweden
| | - Anders Blomqvist
- Department of Clinical and Experimental Medicine, Linköping University, 581 85 Linköping, Sweden.
| |
Collapse
|
21
|
Deak T, Kudinova A, Lovelock DF, Gibb BE, Hennessy MB. A multispecies approach for understanding neuroimmune mechanisms of stress. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 28566946 PMCID: PMC5442363 DOI: 10.31887/dcns.2017.19.1/tdeak] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The relationship between stress challenges and adverse health outcomes, particularly for the development of affective disorders, is now well established. The highly conserved neuroimmune mechanisms through which responses to stressors are transcribed into effects on males and females have recently garnered much attention from researchers and clinicians alike. The use of animal models, from mice to guinea pigs to primates, has greatly increased our understanding of these mechanisms on the molecular, cellular, and behavioral levels, and research in humans has identified particular brain regions and connections of interest, as well as associations between stress-induced inflammation and psychiatric disorders. This review brings together findings from multiple species in order to better understand how the mechanisms of the neuroimmune response to stress contribute to stress-related psychopathologies, such as major depressive disorder, schizophrenia, and bipolar disorder.
Collapse
Affiliation(s)
- Terrence Deak
- Center for Affective Science and Department of Psychology, Binghamton University-State University of New York (SUNY), Binghamton, New York, USA
| | - Anastacia Kudinova
- Center for Affective Science and Department of Psychology, Binghamton University-State University of New York (SUNY), Binghamton, New York, USA
| | - Dennis F Lovelock
- Center for Affective Science and Department of Psychology, Binghamton University-State University of New York (SUNY), Binghamton, New York, USA
| | - Brandon E Gibb
- Center for Affective Science and Department of Psychology, Binghamton University-State University of New York (SUNY), Binghamton, New York, USA
| | | |
Collapse
|
22
|
Fioravante M, Bombassaro B, Ramalho AF, Dragano NR, Morari J, Solon C, Tobar N, Ramos CD, Velloso LA. Inhibition of hypothalamic leukemia inhibitory factor exacerbates diet-induced obesity phenotype. J Neuroinflammation 2017; 14:178. [PMID: 28865476 PMCID: PMC5581454 DOI: 10.1186/s12974-017-0956-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/28/2017] [Indexed: 12/17/2022] Open
Abstract
Background The consumption of large amounts of dietary fats can trigger an inflammatory response in the hypothalamus and contribute to the dysfunctional control of caloric intake and energy expenditure commonly present in obesity. The objective of this study was to identify chemokine-related transcripts that could be involved in the early stages of diet-induced hypothalamic inflammation. Methods We used immunoblot, PCR array, real-time PCR, immunofluorescence staining, glucose and insulin tolerance tests, and determination of general metabolic parameters to evaluate markers of inflammation, body mass variation, and glucose tolerance in mice fed a high-fat diet. Results Using a real-time PCR array, we identified leukemia inhibitory factor as a chemokine/cytokine undergoing a rapid increase in the hypothalamus of obesity-resistant and a rapid decrease in the hypothalamus of obesity-prone mice fed a high-fat diet for 1 day. We hypothesized that the increased hypothalamic expression of leukemia inhibitory factor could contribute to the protective phenotype of obesity-resistant mice. To test this hypothesis, we immunoneutralized hypothalamic leukemia inhibitory factor and evaluated inflammatory and metabolic parameters. The immunoneutralization of leukemia inhibitory factor in the hypothalamus of obesity-resistant mice resulted in increased body mass gain and increased adiposity. Body mass gain was mostly due to increased caloric intake and reduced spontaneous physical activity. This modification in the phenotype was accompanied by increased expression of inflammatory cytokines in the hypothalamus. In addition, the inhibition of hypothalamic leukemia inhibitory factor was accompanied by glucose intolerance and insulin resistance. Conclusion Hypothalamic expression of leukemia inhibitory factor may protect mice from the development of diet-induced obesity; the inhibition of this protein in the hypothalamus transforms obesity-resistant into obesity-prone mice. Electronic supplementary material The online version of this article (10.1186/s12974-017-0956-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Milena Fioravante
- Laboratory of Cell Signaling, University of Campinas, Campinas, São Paulo, 13084-970, Brazil
| | - Bruna Bombassaro
- Laboratory of Cell Signaling, University of Campinas, Campinas, São Paulo, 13084-970, Brazil
| | - Albina F Ramalho
- Laboratory of Cell Signaling, University of Campinas, Campinas, São Paulo, 13084-970, Brazil
| | - Nathalia R Dragano
- Laboratory of Cell Signaling, University of Campinas, Campinas, São Paulo, 13084-970, Brazil
| | - Joseane Morari
- Laboratory of Cell Signaling, University of Campinas, Campinas, São Paulo, 13084-970, Brazil
| | - Carina Solon
- Laboratory of Cell Signaling, University of Campinas, Campinas, São Paulo, 13084-970, Brazil
| | - Natalia Tobar
- Department of Radiology, University of Campinas, Campinas, São Paulo, 13084-970, Brazil
| | - Celso D Ramos
- Department of Radiology, University of Campinas, Campinas, São Paulo, 13084-970, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, University of Campinas, Campinas, São Paulo, 13084-970, Brazil.
| |
Collapse
|
23
|
Grissom NM, George R, Reyes TM. Suboptimal nutrition in early life affects the inflammatory gene expression profile and behavioral responses to stressors. Brain Behav Immun 2017; 63:115-126. [PMID: 27756624 DOI: 10.1016/j.bbi.2016.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/06/2016] [Accepted: 10/14/2016] [Indexed: 12/15/2022] Open
Abstract
Nutritional conditions in early life can have a lasting impact on health and disease risk, though the underlying mechanisms are incompletely understood. In the healthy individual, physiological and behavioral responses to stress are coordinated in such a way as to mobilize resources necessary to respond to the stressor and to terminate the stress response at the appropriate time. Induction of proinflammatory gene expression within the brain is one such example that is initiated in response to both physiological and psychological stressors, and is the focus of the current study. We tested the hypothesis that early life nutrition would impact the proinflammatory transcriptional response to a stressor. Pregnant and lactating dams were fed one of three diets; a low-protein diet, a high fat diet, or the control diet through pregnancy and lactation. Adult male offspring were then challenged with either a physiological stressor (acute lipopolysaccharide injection, IP) or a psychological stressor (15 min restraint). Expression of 20 proinflammatory and stress-related genes was evaluated in hypothalamus, prefrontal cortex, amygdala and ventral tegmental area. In a second cohort, behavioral responses (food intake, locomotor activity, metabolic rate) were evaluated. Offspring from low protein fed dams showed a generally reduced transcriptional response, particularly to LPS, and resistance to behavioral changes associated with restraint, while HF offspring showed an exacerbated transcriptional response within the PFC, a reduced transcriptional response in hypothalamus and amygdala, and an exacerbation of the LPS-induced reduction of locomotor activity. The present data identify differential proinflammatory transcriptional responses throughout the brain driven by perinatal diet as an important variable that may affect risk or resilience to stressors.
Collapse
Affiliation(s)
- Nicola M Grissom
- University of Minnesota, Department of Psychology, Minneapolis, MN, USA
| | - Robert George
- University of Pennsylvania, Department of Pharmacology, Philadelphia, PA, USA
| | - Teresa M Reyes
- University of Cincinnati, Department of Psychiatry and Behavioral Neuroscience, Cincinnati, OH, USA.
| |
Collapse
|
24
|
Inhalation of a racemic mixture (R,S)-linalool by rats experiencing restraint stress alters neuropeptide and MHC class I gene expression in the hypothalamus. Neurosci Lett 2017; 653:314-319. [PMID: 28595953 DOI: 10.1016/j.neulet.2017.05.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/16/2017] [Accepted: 05/22/2017] [Indexed: 12/30/2022]
Abstract
Some odorants have physiological and psychological effects on organisms. However, little is known about the effects of inhaling them, particularly on the central nervous system. Using DNA microarray analysis, we obtained gene expression profiles of the hypothalamus from restraint stressed rats exposed to racemic (R,S)-linalool. Hierarchical clustering across all probe sets showed that this inhalation of (R,S)-linalool influenced the expression levels of a wide range of genes in the hypothalamus. A comparison of transcription levels revealed that the inhalation of (R,S)-linalool restored the expression of 560 stress-induced probe sets to a normal status. Gene Ontology (GO) analysis showed that these genes were associated with synaptic transmission via neurotransmitters including anxiolytic neuropeptides such as oxytocin and neuropeptide Y. These genes also included several major histocompatibility complex (MHC) class I molecules necessary for neural development and plasticity. Moreover, Upstream Regulator Analysis predicted that the hormone prolactin would be activated by the inhalation of (R,S)-linalool under stress. Our results reveal some of the molecular mechanisms associated with odor inhalation in the hypothalamus in organisms under stress.
Collapse
|
25
|
Pro-inflammatory immune-to-brain signaling is involved in neuroendocrine responses to acute emotional stress. Brain Behav Immun 2017; 62:53-63. [PMID: 28179107 DOI: 10.1016/j.bbi.2017.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 01/31/2017] [Accepted: 02/02/2017] [Indexed: 11/23/2022] Open
Abstract
Activation of the hypothalamo-pituitary-adrenal (HPA) axis by inflammatory stressors (e.g., bacterial lipopolysaccharide) is thought to involve vascular transduction of circulating cytokines, with perivascular macrophages (PVMs) along with endothelia, effecting activation of HPA control circuitry via inducible (cyclooxygenase-2- or COX-2-dependent) prostaglandin synthesis. To test the stressor-specificity of this mechanism, we examined whether ablation of PVMs or pharmacologic blockade of COX activity affected HPA responses to a representative emotional stressor, restraint. Exposing rats to a single 30min acute restraint episode provoked increased plasma levels of at least one proinflammatory cytokine, IL-6, microglial activation and multiple indices of cerebrovascular activation, including COX-2 expression and increased brain prostaglandin E2 levels at 0-2h after stress. Pretreatment with the nonselective COX inhibitor, indomethacin, either icv (10μg in 5μl) or iv (1mg/kg) significantly reduced restraint-induced Fos expression in the paraventricular hypothalamic nucleus (PVH) by 45%, relative to vehicle-injected controls. A 75% reduction of the PVH activational response was seen in rats exposed to acute restraint 5-7days after ablation of brain PVMs by icv injection of liposomes encapsulating the bisphosphonate drug, clodronate. Basal plasma levels of ACTH and corticosterone were not altered in clodronate liposome-injected rats, but the peak magnitude of restraint-induced HPA secretory responses was substantially reduced, relative to animals pretreated with saline-filled liposomes. These findings support an unexpectedly prominent role for inducible prostaglandin synthesis by PVMs in HPA responses to acute restraint, a prototypic emotional stressor.
Collapse
|
26
|
Villéga F, Delpech JC, Griton M, André C, Franconi JM, Miraux S, Konsman JP. Circulating bacterial lipopolysaccharide-induced inflammation reduces flow in brain-irrigating arteries independently from cerebrovascular prostaglandin production. Neuroscience 2017; 346:160-172. [DOI: 10.1016/j.neuroscience.2017.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 01/07/2017] [Accepted: 01/11/2017] [Indexed: 12/24/2022]
|
27
|
Deak T. A multispecies approach for understanding neuroimmune mechanisms of stress. DIALOGUES IN CLINICAL NEUROSCIENCE 2017; 19:37-53. [PMID: 28566946 PMCID: PMC5442363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2023]
Abstract
The relationship between stress challenges and adverse health outcomes, particularly for the development of affective disorders, is now well established. The highly conserved neuroimmune mechanisms through which responses to stressors are transcribed into effects on males and females have recently garnered much attention from researchers and clinicians alike. The use of animal models, from mice to guinea pigs to primates, has greatly increased our understanding of these mechanisms on the molecular, cellular, and behavioral levels, and research in humans has identified particular brain regions and connections of interest, as well as associations between stress-induced inflammation and psychiatric disorders. This review brings together findings from multiple species in order to better understand how the mechanisms of the neuroimmune response to stress contribute to stress-related psychopathologies, such as major depressive disorder, schizophrenia, and bipolar disorder.
Collapse
Affiliation(s)
- Terrence Deak
- Center for Affective Science and Department of Psychology, Binghamton University-State University of New York (SUNY), Binghamton, New York, USA
| |
Collapse
|
28
|
Grissom N, George R, Reyes T. The hypothalamic transcriptional response to stress is severely impaired in offspring exposed to adverse nutrition during gestation. Neuroscience 2017. [DOI: 10.1016/j.neuroscience.2015.07.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
29
|
Cyclooxygenase Isoform Exchange Blocks Brain-Mediated Inflammatory Symptoms. PLoS One 2016; 11:e0166153. [PMID: 27861574 PMCID: PMC5115700 DOI: 10.1371/journal.pone.0166153] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 10/24/2016] [Indexed: 02/02/2023] Open
Abstract
Cyclooxygenase-2 (COX-2) is the main source of inducible prostaglandin E2 production and mediates inflammatory symptoms including fever, loss of appetite and hyperalgesia. COX-1 is dispensable for fever, anorexia and hyperalgesia but is important for several other functions both under basal conditions and during inflammation. The differential functionality of the COX isoforms could be due to differences in the regulatory regions of the genes, leading to different expression patterns, or to differences in the coding sequence, resulting in distinct functional properties of the proteins. To study the molecular underpinnings of the functional differences between the two isoforms in the context of inflammatory symptoms, we used mice in which the coding sequence of COX-2 was replaced by the corresponding sequence of COX-1. In these mice, COX-1 mRNA was induced by inflammation but COX-1 protein expression did not fully mimic inflammation-induced COX-2 expression. Just like mice globally lacking COX-2, these mice showed a complete lack of fever and inflammation-induced anorexia as well as an impaired response to inflammatory pain. However, as previously reported, they displayed close to normal survival rates, which contrasts to the high fetal mortality in COX-2 knockout mice. This shows that the COX activity generated from the hybrid gene was strong enough to allow survival but not strong enough to mediate the inflammatory symptoms studied, making the line an interesting alternative to COX-2 knockouts for the study of inflammation. Our results also show that the functional differences between COX-1 and COX-2 in the context of inflammatory symptoms are not only dependent on the features of the promoter regions. Instead they indicate that there are fundamental differences between the isoforms at translational or posttranslational levels.
Collapse
|
30
|
Carlin JL, Grissom N, Ying Z, Gomez-Pinilla F, Reyes TM. Voluntary exercise blocks Western diet-induced gene expression of the chemokines CXCL10 and CCL2 in the prefrontal cortex. Brain Behav Immun 2016; 58:82-90. [PMID: 27492632 PMCID: PMC5352157 DOI: 10.1016/j.bbi.2016.07.161] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/14/2016] [Accepted: 07/29/2016] [Indexed: 12/12/2022] Open
Abstract
Obesity increases inflammation, both peripherally and centrally, and exercise can ameliorate some of the negative health outcomes associated with obesity. Within the brain, the effect of obesity on inflammation has been well characterized in the hypothalamus and hippocampus, but has been relatively understudied in other brain regions. The current study was designed to address two primary questions; (1) whether western diet (high fat/high sucrose) consumption would increase markers of inflammation in the prefrontal cortex and (2) whether concurrent voluntary wheel running would ameliorate any inflammation. Adult male mice were exposed to a western diet or a control diet for 8weeks. Concurrently, half the animals were given running wheels in their home cages, while half did not have access to wheels. At the conclusion of the study, prefrontal cortex was removed and expression of 18 proinflammatory genes was assayed. Expression of a number of proinflammatory molecules was upregulated by consumption of the western diet. For two chemokines, chemokine (C-C motif) ligand 2 (CCL2) and C-X-C motif chemokine 10 (CXCL10), voluntary exercise blocked the increase in the expression of these genes. Cluster analysis confirmed that the majority of the tested genes were upregulated by western diet, and identified another small cluster of genes that were downregulated by either diet or exercise. These data identify a proinflammatory phenotype within the prefrontal cortex of mice fed a western diet, and indicate that chemokine induction can be blocked by voluntary exercise.
Collapse
Affiliation(s)
- Jesse L. Carlin
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Nicola Grissom
- Department of Psychiatry and Behavioral Neurosciences, College of Medicine, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Zhe Ying
- Departments of Neurosurgery, and Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, United States
| | - Fernando Gomez-Pinilla
- Departments of Neurosurgery, and Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, United States
| | - Teresa M. Reyes
- Department of Psychiatry and Behavioral Neurosciences, College of Medicine, University of Cincinnati, Cincinnati, OH 45237, United States,Corresponding author at: University of Cincinnati, College of Medicine, Dept of Psychiatry and Behavioral Neuroscience, 2120 East Galbraith Road, A-129 Cincinnati, OH 45237-1625, United States. (T.M. Reyes)
| |
Collapse
|
31
|
Lateral hypothalamus orexinergic system modulates the stress effect on pentylenetetrazol induced seizures through corticotropin releasing hormone receptor type 1. Neuropharmacology 2016; 110:15-24. [PMID: 27395784 DOI: 10.1016/j.neuropharm.2016.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/22/2016] [Accepted: 07/05/2016] [Indexed: 01/06/2023]
Abstract
Stress is a trigger factor for seizure initiation which activates hypothalamic pituitary adrenal (HPA) axis as well other brain areas. In this respect, corticotropin releasing hormone (CRH) and lateral hypothalamus (LH) orexinergic system are involved in seizure occurrence. In this study, we investigated the role of LH area and orexin expression in (mediation of) stress effect on pentylenetetrazol (PTZ) -induced seizures with hippocampal involvement. Two mild foot shock stresses were applied to intact and adrenalectomized animals; with or without CRHr1 blocking (NBI 27914) in the LH area. Then, changes in orexin production were evaluated by RT-PCR. Intravenous PTZ infusion (25 mg/ml) -induced convulsions were scored upon modified Racine scale. Finally, hippocampal glutamate and GABA were evaluated to study excitability changes. We demonstrated that the duration and severity of convulsions in stress-induced as well as adrenalectomized group were increased. Plasma corticosterone (CRT) level and orexin mRNA expression were built up in the stress and/or seizure groups. Furthermore, glutamate and GABA content was increased and decreased respectively due to stress and seizures. In contrast, rats receiving CRHr1 inhibitor showed reduced severity and duration of seizures, increased GABA, decreased glutamate and corticosterone and also orexin mRNA compared to the inhibitor free rats. Stress and adrenalectomy induced augmenting effect on seizure severity and duration and the subsequent reduction due to CRHr1 blocking with parallel orexin mRNA changes, indicated the likely involvement of CRH1r induced orexin expression of the LH in gating stress effect on convulsions.
Collapse
|
32
|
Abstract
The innate immune response is a coordinated set of reactions involving cells of myeloid lineage and a network of signaling molecules. Such a response takes place in the CNS during trauma, stroke, spinal cord injury, and neurodegenerative diseases, suggesting that macrophages/microglia are the cells that perpetuate the progressive neuronal damage. However, there is accumulating evidence that these cells and their secreted proinflammatory molecules have more beneficial effects than detrimental consequences for the neuronal elements. Indeed, a timely controlled innate immune response may limit toxicity in swiftly eliminating foreign materials and debris that are known to interfere with recovery and regeneration. Each step of the immune cascade is under the tight control of stimulatory and inhibitory signals. Glucocorticoids (GCs) act as the critical negative feedback on all myeloid cells, including those present within the brain parenchyma. Because too little is like too much, both an inappropriate feedback of GCs on microglia and high circulating GC levels in stressed individuals have been associated with deleterious consequences for the brain. In this review, the authors discuss both sides of the story with a particular emphasis on the neuro-protective role of endogenous GCs during immune challenges and the problems in determining whether GCs can be a good therapy for the treatment of neuropathological conditions.
Collapse
Affiliation(s)
- Isaias Glezer
- Laboratory of Molecular Endocrinology, CHUL Research Center, Department of Anatomy and Physiology, Laval University, Québec, Canada
| | | |
Collapse
|
33
|
Liu Y, Huang Y, Liu T, Wu H, Cui H, Gautron L. Lipopolysacharide Rapidly and Completely Suppresses AgRP Neuron-Mediated Food Intake in Male Mice. Endocrinology 2016; 157:2380-92. [PMID: 27111742 PMCID: PMC4891783 DOI: 10.1210/en.2015-2081] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although Agouti-related peptide (AgRP) neurons play a key role in the regulation of food intake, their contribution to the anorexia caused by proinflammatory insults has yet to be identified. Using a combination of neuroanatomical and pharmacogenetics experiments, this study sought to investigate the importance of AgRP neurons and downstream targets in the anorexia caused by the peripheral administration of a moderate dose of lipopolysaccharide (LPS) (100 μg/kg, ip). First, in the C57/Bl6 mouse, we demonstrated that LPS induced c-fos in select AgRP-innervated brain sites involved in feeding but not in any arcuate proopiomelanocortin neurons. Double immunohistochemistry further showed that LPS selectively induced c-Fos in a large subset of melanocortin 4 receptor-expressing neurons in the lateral parabrachial nucleus. Secondly, we used pharmacogenetics to stimulate the activity of AgRP neurons during the course of LPS-induced anorexia. In AgRP-Cre mice expressing the designer receptor hM3Dq-Gq only in AgRP neurons, the administration of the designer drug clozapine-N-oxide (CNO) induced robust food intake. Strikingly, CNO-mediated food intake was rapidly and completely blunted by the coadministration of LPS. Neuroanatomical experiments further indicated that LPS did not interfere with the ability of CNO to stimulate c-Fos in AgRP neurons. In summary, our findings combined together support the view that the stimulation of select AgRP-innervated brain sites and target neurons, rather than the inhibition of AgRP neurons themselves, is likely to contribute to the rapid suppression of food intake observed during acute bacterial endotoxemia.
Collapse
Affiliation(s)
- Yang Liu
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| | - Ying Huang
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| | - Tiemin Liu
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| | - Hua Wu
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| | - Huxing Cui
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| | - Laurent Gautron
- Division of Hypothalamic Research and Department of Internal Medicine (Y.L., Y.H., T.L., L.G.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Department of Orthopedics (Y.L., H.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China 430030; and Department of Pharmacology (H.C.), Center for Hypertension Research, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242
| |
Collapse
|
34
|
Haj-Mirzaian A, Kordjazy N, Ostadhadi S, Amiri S, Haj-Mirzaian A, Dehpour A. Fluoxetine reverses the behavioral despair induced by neurogenic stress in mice: role of N-methyl-d-aspartate and opioid receptors. Can J Physiol Pharmacol 2016; 94:599-612. [DOI: 10.1139/cjpp-2015-0429] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Opioid and N-methyl-d-aspartate (NMDA) receptors mediate different effects of fluoxetine. We investigated whether opioid and NMDA receptors are involved in the protective effect of fluoxetine against the behavioral despair induced by acute physical stress in male mice. We used the forced swimming test (FST), tail suspension test (TST), and open-field test (OFT) for behavioral evaluation. We used fluoxetine, naltrexone (opioid receptor antagonist), MK-801 (NMDA receptor antagonist), morphine (opioid receptor agonist), and NMDA (NMDA receptor agonist). Acute foot-shock stress (FSS) significantly induced behavioral despair (depressive-like) and anxiety-like behaviors in tests. Fluoxetine (5 mg/kg) reversed the depressant-like effect of FSS, but it did not alter the locomotion and anxiety-like behavior in animals. Acute administration of subeffective doses of naltrexone (0.3 mg/kg) or MK-801 (0.01 mg/kg) potentiated the antidepressant-like effect of fluoxetine, while subeffective doses of morphine (1 mg/kg) and NMDA (75 mg/kg) abolished this effect of fluoxetine. Also, co-administration of subeffective doses of naltrexone (0.05 mg/kg) and MK-801 (0.003 mg/kg) with fluoxetine (1 mg/kg) induced a significant decrease in the immobility time in FST and TST. Our results showed that opioid and NMDA receptors (alone or in combination) are involved in the antidepressant-like effect of fluoxetine against physical stress.
Collapse
Affiliation(s)
- Arya Haj-Mirzaian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Nastaran Kordjazy
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Sattar Ostadhadi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shayan Amiri
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Arvin Haj-Mirzaian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - AhmadReza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| |
Collapse
|
35
|
Motta SC, Canteras NS. Restraint stress and social defeat: What they have in common. Physiol Behav 2016; 146:105-110. [PMID: 26066716 DOI: 10.1016/j.physbeh.2015.03.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 11/28/2022]
Abstract
Bob Blanchard was a great inspiration for our studies on the neural basis of social defense. In the present study, we compared the hypothalamic pattern of activation between social defeat and restraint stress. As important stress situations, both defeated and immobilized animals displayed a substantial increase in Fos in the parvicellular part of the paraventricular nucleus,mostly in the region that contains the CRH neurons. In addition, socially defeated animals, but not restrained animals, recruited elements of the medial hypothalamic conspecific-responsive circuit, a region also engaged in other forms of social behavior. Of particular interest, both defeated and immobilized animals presented a robust increase in Fos expression in specific regions of the lateral hypothalamic area (i.e., juxtaparaventricular and juxtadorsomedial regions) likely to convey septo-hippocampal information encoding the environmental boundary restriction observed in both forms of stress, and in the dorsomedial part of the dorsal premammillary nucleus which seems to work as a key player for the expression of, at least, part of the behavioral responses during both restraint and social defeat. These results indicate interesting commonalities between social defeat and restraint stress, suggesting, for the first time, a septo-hippocampal–hypothalamic path likely to respond to the environmental boundary restriction that may act as common stressor component for both types of stress. Moreover, the comparison of the neural circuits mediating physical restraint and social defense revealed a possible path for encoding the entrapment component during social confrontation.
Collapse
|
36
|
Removal of high-fat diet after chronic exposure drives binge behavior and dopaminergic dysregulation in female mice. Neuroscience 2016; 326:170-179. [PMID: 27063418 DOI: 10.1016/j.neuroscience.2016.04.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 03/08/2016] [Accepted: 04/01/2016] [Indexed: 12/28/2022]
Abstract
A significant contributor to the obesity epidemic is the overconsumption of highly palatable, energy dense foods. Chronic intake of palatable foods is associated with neuroadaptations within the mesocorticolimbic dopamine system adaptations which may lead to behavioral changes, such as overconsumption or bingeing. We examined behavioral and molecular outcomes in mice that were given chronic exposure to a high-fat diet (HFD; 12weeks), with the onset of the diet either in adolescence or adulthood. To examine whether observed effects could be reversed upon removal of the HFD, animals were also studied 4weeks after a return to chow feeding. Most notably, female mice, particularly those exposed to HFD starting in adolescence, demonstrated the emergence of binge-like behavior when given restricted access to a palatable food. Further, changes in dopamine-related gene expression and dopamine content in the prefrontal cortex were observed. Some of these HFD-driven phenotypes reversed upon removal of the diet, whereas others were initiated by removal of the diet. These findings have implications for obesity management and interventions, as both pharmacological and behavioral therapies are often combined with dietary interventions (e.g., reduction in calorie dense foods).
Collapse
|
37
|
Ozsoy S, Olguner Eker O, Abdulrezzak U, Esel E. Relationship between orexin A and childhood maltreatment in female patients with depression and anxiety. Soc Neurosci 2016; 12:330-336. [DOI: 10.1080/17470919.2016.1169216] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
38
|
Cui L, Ding Y, Feng Y, Chen S, Xu Y, Li M, Hu M, Qiu Z, Ding M. MiRNAs are involved in chronic electroacupuncture tolerance in the rat hypothalamus. Mol Neurobiol 2016; 54:1429-1439. [PMID: 26846282 DOI: 10.1007/s12035-016-9759-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 01/26/2016] [Indexed: 02/06/2023]
Abstract
Acupuncture tolerance is the gradual decrease in analgesic effect due to its prolonged application. However, its mechanism in terms of miRNA is still unknown. To explore the role of miRNAs in electroacupuncture (EA) tolerance of rats using deep sequencing, rats with more than a 50 % increase in tail flick latency (TFL) in response to EA were selected for this experiment. EA tolerance was induced by EA once daily for eight consecutive days. The hypothalami were harvested for deep sequencing. As a result, 49 differentially expressed miRNAs were identified and validated by real-time PCR. Of them, let-7b-5p, miR-148a-3p, miR-124-3p, miR-107-3p, and miR-370-3p were further confirmed to be related to EA tolerance by an intracerebroventricular injection of agomirs or antagomirs of these miRNAs. Potential targets of the 49 miRNAs were enriched in 9 pathways and 282 gene ontology (GO) terms. Five miRNAs were confirmed to participate in EA tolerance probably through the functional categories related to nerve impulse transmission, receptor signal pathways, and gene expression regulation, as well as pathways related to MAPK, neurotrophin, fatty acid metabolism, lysosome, and the degradation of valine, leucine, and isoleucine. Our findings reveal a characterized panel of the differentially expressed miRNAs in the hypothalamus in response to EA and thus provide a solid experimental framework for future analysis of the mechanisms underlying EA-induced tolerance.
Collapse
Affiliation(s)
- Luying Cui
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yi Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yan Feng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shuhuai Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yingqing Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Meng Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Manli Hu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhengying Qiu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mingxing Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
39
|
Qadri F, Rimmele F, Mallis L, Häuser W, Dendorfer A, Jöhren O, Dominiak P, Leeb-Lundberg LF, Bader M. Acute hypothalamo-pituitary-adrenal axis response to LPS-induced endotoxemia: expression pattern of kinin type B1 and B2 receptors. Biol Chem 2016; 397:97-109. [DOI: 10.1515/hsz-2015-0206] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 10/07/2015] [Indexed: 11/15/2022]
Abstract
Abstract
Bradykinin (BK) and des-Arg9-BK are pro-inflammatory mediators acting via B2 (B2R) and B1 (B1R) receptors, respectively. We investigated the role of B2R and B1R in lipopolysaccharide (LPS)-induced hypothalamo-pituitary-adrenal (HPA) axis activation in SD rats. LPS given intraperitoneally (ip) up-regulated B1R mRNA in the hypothalamus, both B1R and B2R were up-regulated in pituitary and adrenal glands. Receptor localization was performed using immunofluorescence staining. B1R was localized in the endothelial cells, nucleus supraopticus (SON), adenohypophysis and adrenal cortex. B2R was localized nucleus paraventricularis (PVN) and SON, pituitary and adrenal medulla. Blockade of B1R prior to LPS further increased ACTH release and blockade of B1R 1 h after LPS decreased its release. In addition, we evaluated if blockade of central kinin receptors influence the LPS-induced stimulation of hypothalamic neurons. Blockade of both B1R and B2R reduced the LPS-induced c-Fos immunoreactivity in the hypothalamus. Our data demonstrate that a single injection of LPS induced a differential expression pattern of kinin B1R and B2R in the HPA axis. The tissue specific cellular localization of these receptors indicates that they may play a crucial role in the maintenance of body homeostasis during endotoxemia.
Collapse
|
40
|
Vecchiarelli HA, Gandhi CP, Gray JM, Morena M, Hassan KI, Hill MN. Divergent responses of inflammatory mediators within the amygdala and medial prefrontal cortex to acute psychological stress. Brain Behav Immun 2016; 51:70-91. [PMID: 26260453 DOI: 10.1016/j.bbi.2015.07.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/23/2015] [Accepted: 07/29/2015] [Indexed: 01/03/2023] Open
Abstract
There is now a growing body of literature that indicates that stress can initiate inflammatory processes, both in the periphery and brain; however, the spatiotemporal nature of this response is not well characterized. The aim of this study was to examine the effects of an acute psychological stress on changes in mRNA and protein levels of a wide range of inflammatory mediators across a broad temporal range, in key corticolimbic brain regions involved in the regulation of the stress response (amygdala, hippocampus, hypothalamus, medial prefrontal cortex). mRNA levels of inflammatory mediators were analyzed immediately following 30min or 120min of acute restraint stress and protein levels were examined 0h through 24h post-termination of 120min of acute restraint stress using both multiplex and ELISA methods. Our data demonstrate, for the first time, that exposure to acute psychological stress results in an increase in the protein level of several inflammatory mediators in the amygdala while concomitantly producing a decrease in the protein level of multiple inflammatory mediators within the medial prefrontal cortex. This pattern of changes seemed largely restricted to the amygdala and medial prefrontal cortex, with stress producing few changes in the mRNA or protein levels of inflammatory mediators within the hippocampus or hypothalamus. Consistent with previous research, stress resulted in a general elevation in multiple inflammatory mediators within the circulation. These data indicate that neuroinflammatory responses to stress do not appear to be generalized across brain structures and exhibit a high degree of spatiotemporal specificity. Given the impact of inflammatory signaling on neural excitability and emotional behavior, these data may provide a platform with which to explore the importance of inflammatory signaling within the prefrontocortical-amygdala circuit in the regulation of the neurobehavioral responses to stress.
Collapse
Affiliation(s)
- Haley A Vecchiarelli
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Department of Neuroscience, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada
| | - Chaitanya P Gandhi
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Department of Neuroscience, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada
| | - J Megan Gray
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada
| | - Maria Morena
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada
| | - Kowther I Hassan
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; Department of Psychiatry, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
41
|
Wang H, Ji J, Yu Y, Wei X, Chai S, Liu D, Huang D, Li Q, Dong Z, Xiao X. Neonatal Overfeeding in Female Mice Predisposes the Development of Obesity in their Male Offspring via Altered Central Leptin Signalling. J Neuroendocrinol 2015; 27:600-8. [PMID: 25855235 DOI: 10.1111/jne.12281] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 03/15/2015] [Accepted: 03/31/2015] [Indexed: 02/01/2023]
Abstract
The prevalence of obesity among child-bearing women has increased significantly. The adverse consequences of maternal obesity on the descendants have been well accepted, although few studies have examined the underlying mechanisms. We investigated whether neonatal overfeeding in female mice alters metabolic phenotypes in the offspring and whether hypothalamic leptin signalling is involved. Neonatal overfeeding was induced by reducing the litter size to three pups per litter, in contrast to normal litter size of 10 pups per litter. Normal and neonatally overfed female mice were bred with normal male mice, and offspring of overfeeding mothers (OOM) and control mothers (OCM) were generated. We examined body weight, daily food intake, leptin responsiveness and the number of positive neurones for phosphorylated-signal transducer and activator of transcription 3 (pSTAT3) along with neuropeptide Y (NPY) in the arcuate nucleus of the hypothalamus (ARH) and NPY in the nucleus tractus solitarius (NTS) of the brain stem. The body weight and daily food intake of OOM were significantly higher than those of OCM. Leptin significantly reduced food intake and increased the number of pSTAT3 positive neurones in the ARH of OCM mice, whereas no significant changes in food intake and pSTAT3 neurones were found in leptin-treated OOM mice. The number of NPY neurones in the ARH and NTS of the OOM mice was significantly higher than that of OCM mice. The results of the present study indicate that the obese phenotype from mothers can be passed onto the subsequent generation, which is possibly associated with hypothalamic leptin resistance.
Collapse
Affiliation(s)
- H Wang
- Laboratory of Lipid & Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China
| | - J Ji
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Y Yu
- Laboratory of Lipid & Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - X Wei
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China
| | - S Chai
- Laboratory of Lipid & Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China
| | - D Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China
| | - D Huang
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China
| | - Q Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Z Dong
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - X Xiao
- Laboratory of Lipid & Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
42
|
Neuropharmacological effect of novel 5-HT3 receptor antagonist, N-n-propyl-3-ethoxyquinoxaline-2-carboxamide (6n) on chronic unpredictable mild stress-induced molecular and cellular response: Behavioural and biochemical evidences. Pharmacol Rep 2014; 66:804-10. [DOI: 10.1016/j.pharep.2014.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/06/2014] [Accepted: 05/09/2014] [Indexed: 11/21/2022]
|
43
|
Paul ED, Johnson PL, Shekhar A, Lowry CA. The Deakin/Graeff hypothesis: focus on serotonergic inhibition of panic. Neurosci Biobehav Rev 2014; 46 Pt 3:379-96. [PMID: 24661986 PMCID: PMC4170046 DOI: 10.1016/j.neubiorev.2014.03.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 02/15/2014] [Accepted: 03/01/2014] [Indexed: 12/20/2022]
Abstract
The Deakin/Graeff hypothesis proposes that different subpopulations of serotonergic neurons through topographically organized projections to forebrain and brainstem structures modulate the response to acute and chronic stressors, and that dysfunction of these neurons increases vulnerability to affective and anxiety disorders, including panic disorder. We outline evidence supporting the existence of a serotonergic system originally discussed by Deakin/Graeff that is implicated in the inhibition of panic-like behavioral and physiological responses. Evidence supporting this panic inhibition system comes from the following observations: (1) serotonergic neurons located in the 'ventrolateral dorsal raphe nucleus' (DRVL) as well as the ventrolateral periaqueductal gray (VLPAG) inhibit dorsal periaqueductal gray-elicited panic-like responses; (2) chronic, but not acute, antidepressant treatment potentiates serotonin's panicolytic effect; (3) contextual fear activates a central nucleus of the amygdala-DRVL/VLPAG circuit implicated in mediating freezing and inhibiting panic-like escape behaviors; (4) DRVL/VLPAG serotonergic neurons are central chemoreceptors and modulate the behavioral and cardiorespiratory response to panicogenic agents such as sodium lactate and CO2. Implications of the panic inhibition system are discussed.
Collapse
Affiliation(s)
- Evan D Paul
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Philip L Johnson
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Anantha Shekhar
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | - Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| |
Collapse
|
44
|
Haj-Mirzaian A, Ostadhadi S, Kordjazy N, Dehpour AR, Ejtemaei Mehr S. Opioid/NMDA receptors blockade reverses the depressant-like behavior of foot shock stress in the mouse forced swimming test. Eur J Pharmacol 2014; 735:26-31. [DOI: 10.1016/j.ejphar.2014.03.053] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 03/15/2014] [Accepted: 03/26/2014] [Indexed: 10/25/2022]
|
45
|
Basharat S, Parker JA, Murphy KG, Bloom SR, Buckingham JC, John CD. Leptin fails to blunt the lipopolysaccharide-induced activation of the hypothalamic-pituitary-adrenal axis in rats. J Endocrinol 2014; 221:229-34. [PMID: 24578293 PMCID: PMC4045222 DOI: 10.1530/joe-13-0249] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Obesity is a risk factor for sepsis morbidity and mortality, whereas the hypothalamic-pituitary-adrenal (HPA) axis plays a protective role in the body's defence against sepsis. Sepsis induces a profound systemic immune response and cytokines serve as excellent markers for sepsis as they act as mediators of the immune response. Evidence suggests that the adipokine leptin may play a pathogenic role in sepsis. Mouse endotoxaemic models present with elevated leptin levels and exogenously added leptin increased mortality whereas human septic patients have elevated circulating levels of the soluble leptin receptor (Ob-Re). Evidence suggests that leptin can inhibit the regulation of the HPA axis. Thus, leptin may suppress the HPA axis, impairing its protective role in sepsis. We hypothesised that leptin would attenuate the HPA axis response to sepsis. We investigated the direct effects of an i.p. injection of 2 mg/kg leptin on the HPA axis response to intraperitoneally injected 25 μg/kg lipopolysaccharide (LPS) in the male Wistar rat. We found that LPS potently activated the HPA axis, as shown by significantly increased plasma stress hormones, ACTH and corticosterone, and increased plasma interleukin 1β (IL1β) levels, 2 h after administration. Pre-treatment with leptin, 2 h before LPS administration, did not influence the HPA axis response to LPS. In turn, LPS did not affect plasma leptin levels. Our findings suggest that leptin does not influence HPA function or IL1β secretion in a rat model of LPS-induced sepsis, and thus that leptin is unlikely to be involved in the acute-phase endocrine response to bacterial infection in rats.
Collapse
Affiliation(s)
| | - Jennifer A Parker
- University College London, Institute of NeurologyQueen Square House, Queen Square, London, WC1N 3BGUK
| | | | | | - Julia C Buckingham
- Brunel UniversityWilfred Brown Building, Kingston Lane, Uxbridge, UB8 3PHUK
| | | |
Collapse
|
46
|
Patel AX, Miller SR, Nathan PJ, Kanakaraj P, Napolitano A, Lawrence P, Koch A, Bullmore ET. Neuroendocrine and sympathetic responses to an orexin receptor antagonist, SB-649868, and alprazolam following insulin-induced hypoglycemia in humans. Psychopharmacology (Berl) 2014; 231:3817-28. [PMID: 24770625 PMCID: PMC4159598 DOI: 10.1007/s00213-014-3520-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/24/2014] [Indexed: 02/02/2023]
Abstract
RATIONALE The orexin-hypocretin system is important for translating peripheral metabolic signals and central neuronal inputs to a diverse range of behaviors, from feeding, motivation and arousal, to sleep and wakefulness. Orexin signaling is thus an exciting potential therapeutic target for disorders of sleep, feeding, addiction, and stress. OBJECTIVES/METHODS Here, we investigated the low dose pharmacology of orexin receptor antagonist, SB-649868, on neuroendocrine, sympathetic nervous system, and behavioral responses to insulin-induced hypoglycemic stress, in 24 healthy male subjects (aged 18-45 years; BMI 19.0-25.9 kg/m(2)), using a randomized, double-blind, placebo-controlled, within-subject crossover design. Alprazolam, a licensed benzodiazepine anxiolytic, was used as a positive comparator, as it has previously been validated using the insulin tolerance test (ITT) model in humans. RESULTS Of the primary endpoints, ITT induced defined increases in pulse rate, plasma cortisol, and adrenocorticotropic hormone in the placebo condition, but these responses were not significantly impacted by alprazolam or SB-649868 pre-treatment. Of the secondary endpoints, ITT induced a defined increase in plasma concentrations of adrenaline, noradrenaline, growth hormone (GH), and prolactin in the placebo condition. Alprazolam pre-treatment significantly reduced the GH response to ITT (p < 0.003), the peak electromyography (p < 0.0001) and galvanic skin response (GSR, p = 0.04) to acoustic startle, the resting GSR (p = 0.01), and increased appetite following ITT (p < 0.0005). SB-649868 pre-treatment produced no significant results. CONCLUSION We concluded that the ITT model may be informative for assessing the effects of drugs directly acting on the neuroendocrine or sympathetic nervous systems, but could not be validated for studying low dose orexin antagonist activity.
Collapse
Affiliation(s)
- Ameera X. Patel
- Brain Mapping Unit, Behavioral and Clinical Neuroscience Institute, University of Cambridge, Cambridge, CB2 3EB UK
| | - Sam R. Miller
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK
| | - Pradeep J. Nathan
- Brain Mapping Unit, Behavioral and Clinical Neuroscience Institute, University of Cambridge, Cambridge, CB2 3EB UK ,Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK ,School of Psychology and Psychiatry, Monash University, Melbourne, Australia
| | - Ponmani Kanakaraj
- Quantitative Sciences India, GlaxoSmithKline Pharmaceuticals Ltd, Bangalore, India
| | - Antonella Napolitano
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK
| | - Philip Lawrence
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK
| | - Annelize Koch
- Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK
| | - Edward T. Bullmore
- Brain Mapping Unit, Behavioral and Clinical Neuroscience Institute, University of Cambridge, Cambridge, CB2 3EB UK ,Clinical Unit Cambridge, GlaxoSmithKline, Addenbrooke’s Centre for Clinical Investigation, Cambridge, UK
| |
Collapse
|
47
|
Carlin J, Hill-Smith TE, Lucki I, Reyes TM. Reversal of dopamine system dysfunction in response to high-fat diet. Obesity (Silver Spring) 2013; 21:2513-21. [PMID: 23512420 PMCID: PMC3700634 DOI: 10.1002/oby.20374] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 12/31/2012] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To test whether high-fat diet (HFD) decreases dopaminergic tone in reward regions of the brain and evaluate whether these changes reverse after removal of the HFD. DESIGN AND METHODS Male and female mice were fed a 60% HFD for 12 weeks. An additional group was evaluated 4 weeks after removal of the HFD. These groups were compared with control fed, age-matched controls. Sucrose and saccharin preference was measured along with mRNA expression of dopamine (DA)-related genes by Real Time-quantitative PCR (RT-qPCR). DA and 3,4-dihydroxyphenylacetic acid (DOPAC) were measured using high-performance liquid chromatography. DNA methylation of the dopamine transporter (DAT) promoter was measured by methylated DNA immunoprecipitation and RT-qPCR. RESULTS After chronic HFD, sucrose preference was reduced, and then normalized after removal of the HFD. Decreased expression of DA genes, decreased DA content and alterations in DAT promoter methylation, was observed. Importantly, response to HFD and the persistence of changes depended on sex and brain region. CONCLUSIONS These data identify diminished DA tone after early-life chronic HFD with a complex pattern of reversal and persistence that varies by both sex and brain region. Central nervous system changes that did not reverse after HFD withdrawal may contribute to the difficulty in maintaining weight-loss after diet intervention.
Collapse
Affiliation(s)
- Jesselea Carlin
- Department of Pharmacology, Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | |
Collapse
|
48
|
Hueston CM, Deak T. The inflamed axis: the interaction between stress, hormones, and the expression of inflammatory-related genes within key structures comprising the hypothalamic-pituitary-adrenal axis. Physiol Behav 2013; 124:77-91. [PMID: 24184413 DOI: 10.1016/j.physbeh.2013.10.035] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 10/23/2013] [Indexed: 12/13/2022]
Abstract
Acute stress increases the expression of cytokines and other inflammatory-related factors in the CNS, plasma, and endocrine glands, and activation of inflammatory signaling pathways within the hypothalamic-pituitary-adrenal (HPA) axis may play a key role in later stress sensitization. In addition to providing a summary of stress effects on neuroimmune changes within the CNS, we present a series of experiments that characterize stress effects on members of the interleukin-1β (IL-1) super-family and other inflammatory-related genes in key structures comprising the HPA axis (PVN, pituitary and adrenal glands), followed by a series of experiments examining the impact of exogenous hormone administration (CRH and ACTH) and dexamethasone on the expression of inflammatory-related genes in adult male Sprague-Dawley rats. The results demonstrated robust, time-dependent, and asynchronous expression patterns for IL-1 and IL-1R2 in the PVN, with substantial increases in IL-6 and COX-2 in the adrenal glands emerging as key findings. The effects of exogenous CRH and ACTH were predominantly isolated within the adrenals. Finally, pretreatment with dexamethasone severely blunted neuroimmune changes in the adrenal glands, but not in the PVN. These findings provide novel insight into the relationship between stress, the expression of inflammatory signaling factors within key structures comprising the HPA axis, and their interaction with HPA hormones, and provide a foundation for better understanding the role of cytokines as modulators of hypothalamic, pituitary and adrenal sensitivity.
Collapse
Affiliation(s)
- Cara M Hueston
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | | |
Collapse
|
49
|
Liu HW, Srinivasan M, Mahmood S, Smiraglia DJ, Patel MS. Adult-onset obesity induced by early life overnutrition could be reversed by moderate caloric restriction. Am J Physiol Endocrinol Metab 2013; 305:E785-94. [PMID: 23900419 PMCID: PMC3798704 DOI: 10.1152/ajpendo.00280.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Overnutrition during the suckling period (small litter, SL) results in the development of adult-onset obesity. Our aim was to investigate whether two levels of caloric restriction (CR) in the early postweaning period can reverse obese phenotype in SL rats. The normal litter (NL) had 12 pups/dam and SL had 3 male pups/dam from the postnatal day 3 until day 21. After weaning, rats consumed lab chow as indicated: 1) NL and SL groups were on ad libitum regimen up to day 140, 2) another SL group was pair-fed (SL/PF) to NL(∼14% reduction), 3) SL/PF/AL group was pair-fed up to day 94 and then switched to ad libitum feeding, 4) SL/CR group received 24% reduction (moderate CR) in food intake compared with SL, and 5) SL/CR/AL group was on 24% CR up to day 94 and then switched to ad libitum feeding. Pair-feeding reduced body weight gains and serum insulin and leptin levels compared with SL rats, but these parameters were restored to SL levels in the SL/PF/AL rats after switching to ad libitum feeding. Interestingly, the moderate CR normalized these parameters in SL/CR and SL/CR/AL rats compared with NL. The expression of neuropeptide Y, proopiomelanocortin, and leptin receptor returned to control levels in hypothalami from SL/CR and SL/CR/AL rats. These results indicate that appropriate manipulation of energy intake during the early postweaning period could lead to longer-lasting effects on the regulation of body weight homeostasis via reversal of the early preweaning programming effects on the hypothalamic appetite regulation mechanism.
Collapse
Affiliation(s)
- Hung-Wen Liu
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | | | | | | | | |
Collapse
|
50
|
Stankiewicz AM, Swiergiel AH, Lisowski P. Epigenetics of stress adaptations in the brain. Brain Res Bull 2013; 98:76-92. [DOI: 10.1016/j.brainresbull.2013.07.003] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 07/04/2013] [Accepted: 07/06/2013] [Indexed: 02/07/2023]
|