1
|
Lu X, Zhou M, Liu N, Zhang C, Zhao Z, Cai D. Synaptic Protein Phosphorylation Networks Are Associated With Electroacupuncture-Induced Circadian Control in the Suprachiasmatic Nucleus. Front Genet 2021; 12:762557. [PMID: 34976011 PMCID: PMC8717940 DOI: 10.3389/fgene.2021.762557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/29/2021] [Indexed: 11/23/2022] Open
Abstract
Phosphorylation is one of the most important posttranslational modifications and regulates the physiological process. While recent studies highlight a major role of phosphorylation in the regulation of sleep–wake cycles to a lesser extent, the phosphoproteome in the suprachiasmatic nucleus (SCN) is not well-understood. Herein, we reported that the EA treatment elicits partial reparation of circadian rhythmicity when mice were exposure to constant darkness for long time. We investigated the effects of EA on circadian rhythms in constant darkness between EA stimulation and free-running control. Next, mass spectrometry–based phosphoproteome was utilized to explore the molecular characteristics of EA-induced phosphorylation modification in the SCN. A total of 6,192 distinct phosphosites on 2,488 proteins were quantified. Functional annotation analysis and protein–protein interaction networks demonstrated the most significant enriched phosphor-proteins and phosphosites involved in postsynapse and glutamatergic synapse. The current data indicated that most of the altered molecules are structural proteins. The target proteins, NMDAR and CAMK2, were selected for verification, consistent with the results of LC–MS/MS. These findings revealed a complete profile of phosphorylation modification in response to EA.
Collapse
Affiliation(s)
| | | | | | | | | | - Dingjun Cai
- *Correspondence: Zhengyu Zhao, ; Dingjun Cai,
| |
Collapse
|
2
|
Potts EM, Coppotelli G, Ross JM. Histological-Based Stainings using Free-Floating Tissue Sections. J Vis Exp 2020. [PMID: 32925894 PMCID: PMC7743918 DOI: 10.3791/61622] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Immunohistochemistry is a widely used technique to visualize specific tissue structures as well as protein expression and localization. Two alternative approaches are widely used to handle the tissue sections during the staining procedure, one approach consists of mounting the sections directly on glass slides, while a second approach, the free-floating, allows for fixed sections to be maintained and stained while suspended in solution. Although slide-mounted and free-floating approaches may yield similar results, the free-floating technique allows for better antibody penetration and thus should be the method of choice when thicker sections are to be used for 3D reconstruction of the tissues, for example when the focus of the experiment is to gain information on dendritic and axonal projections in brain regions. In addition, since the sections are kept in solution, a single aliquot can easily accommodate 30 to 40 sections, handling of which is less laborious, particularly in large-scale biomedical studies. Here, we illustrate how to apply the free-floating method to fluorescent immunohistochemistry staining, with a major focus on brain sections. We will also discuss how the free-floating technique can easily be modified to fit the individual needs of researchers and adapted to other tissues as well as other histochemical-based stainings, such as hematoxylin and eosin and cresyl violet, as long as tissue samples are properly fixed, typically with paraformaldehyde or formalin.
Collapse
Affiliation(s)
- Emily M Potts
- George & Anne Ryan Institute for Neuroscience, College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island
| | - Giuseppe Coppotelli
- George & Anne Ryan Institute for Neuroscience, College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island
| | - Jaime M Ross
- George & Anne Ryan Institute for Neuroscience, College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island; Department of Neuroscience, Biomedicum, Karolinska Institutet;
| |
Collapse
|
3
|
Mercerón-Martínez D, Almaguer-Melian W, Alberti-Amador E, Calderón-Peña R, Bergado JA. Amygdala stimulation ameliorates memory impairments and promotes c-Fos activity in fimbria-fornix-lesioned rats. Synapse 2020; 74:e22179. [PMID: 32621298 DOI: 10.1002/syn.22179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/10/2020] [Accepted: 06/26/2020] [Indexed: 11/08/2022]
Abstract
Recently we provided data showing that amygdala stimulation can ameliorate spatial memory impairments in rats with lesion in the fimbria-fornix (FF). The mechanisms for this improvement involve early gene expression and synthesis of BDNF, MAP-2, and GAP43 in the hippocampus and prefrontal cortex. Now we have studied which brain structures are activated by the amygdala using c-Fos as a marker of neural activation. First, we studied neuronal activation after tetanic stimulation to the amygdala in intact rats. We then carried out a second study in FF-lesioned rats in which the amygdala was stimulated 15 min after daily spatial memory training in the water maze. Our results showed that amygdala stimulation produces widespread brain activation, that includes cortical, thalamic, and brain stem structures. Activation was particularly intense in the dentate gyrus and the prefrontal cortex. Training in the water maze increased c-Fos positive nuclei in the dentate gyrus of the hippocampus and in medial prefrontal cortex. Amygdala stimulation to trained FF-lesioned rats induced an increase of neural activity in the dentate gyrus and medial prefrontal cortex relative to the FF-lesioned, but not stimulated group, like the c-Fos activity seen in trained control rats. Based on these and previous results we explain the mechanisms of amygdala reinforcement of neural plasticity and the partial recovery of spatial memory deficits.
Collapse
Affiliation(s)
- Daymara Mercerón-Martínez
- Department of Experimental Neurophysiology, International Center for Neurological Restoration (CIREN), Havana, Cuba
| | - William Almaguer-Melian
- Department of Experimental Neurophysiology, International Center for Neurological Restoration (CIREN), Havana, Cuba
| | - Esteban Alberti-Amador
- Department of Experimental Neurophysiology, International Center for Neurological Restoration (CIREN), Havana, Cuba
| | | | - Jorge A Bergado
- Universidad del Sinú "Elías Bechara Zainum", Montería, Colombia
| |
Collapse
|
4
|
Huang YS, Fan CH, Hsu N, Chiu NH, Wu CY, Chang CY, Wu BH, Hong SR, Chang YC, Yan-Tang Wu A, Guo V, Chiang YC, Hsu WC, Chen L, Pin-Kuang Lai C, Yeh CK, Lin YC. Sonogenetic Modulation of Cellular Activities Using an Engineered Auditory-Sensing Protein. NANO LETTERS 2020; 20:1089-1100. [PMID: 31884787 DOI: 10.1021/acs.nanolett.9b04373] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Biomolecules that respond to different external stimuli enable the remote control of genetically modified cells. We report herein a sonogenetic approach that can manipulate target cell activities by focused ultrasound stimulation. This system requires an ultrasound-responsive protein derived from an engineered auditory-sensing protein prestin. Heterologous expression of mouse prestin containing two parallel amino acid substitutions, N7T and N308S, that frequently exist in prestins from echolocating species endowed transfected mammalian cells with the ability to sense ultrasound. An ultrasound pulse of low frequency and low pressure efficiently evoked cellular calcium responses after transfecting with prestin(N7T, N308S). Moreover, pulsed ultrasound can also noninvasively stimulate target neurons expressing prestin(N7T, N308S) in deep regions of mouse brains. Our study delineates how an engineered auditory-sensing protein can cause mammalian cells to sense ultrasound stimulation. Moreover, our sonogenetic tools will serve as new strategies for noninvasive therapy in deep tissues.
Collapse
Affiliation(s)
- Yao-Shen Huang
- Institute of Molecular Medicine , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering and Environmental Sciences , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Ning Hsu
- Institute of Molecular Medicine , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Nai-Hua Chiu
- Department of Biomedical Engineering and Environmental Sciences , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Chun-Yao Wu
- Department of Biomedical Engineering and Environmental Sciences , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Chu-Yuan Chang
- Institute of Molecular Medicine , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Bing-Huan Wu
- Institute of Molecular Medicine , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Shi-Rong Hong
- Institute of Molecular Medicine , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Ya-Chu Chang
- Institute of Molecular Medicine , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Anthony Yan-Tang Wu
- Institute of Atomic and Molecular Sciences , Academia Sinica , Taipei 106 , Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program , Academia Sinica , Taipei 106 , Taiwan
- Department and Graduate Institute of Pharmacology , National Taiwan University , Taipei 106 , Taiwan
| | - Vanessa Guo
- Institute of Atomic and Molecular Sciences , Academia Sinica , Taipei 106 , Taiwan
| | - Yueh-Chen Chiang
- Institute of Molecular Medicine , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Wei-Chia Hsu
- Institute of Molecular Medicine , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Linyi Chen
- Institute of Molecular Medicine , National Tsing Hua University , Hsinchu 300 , Taiwan
- Department of Medical Science , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Charles Pin-Kuang Lai
- Institute of Atomic and Molecular Sciences , Academia Sinica , Taipei 106 , Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program , Academia Sinica , Taipei 106 , Taiwan
- Genome and Systems Biology Degree Program , National Taiwan University and Academia Sinica , Taipei 106 , Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences , National Tsing Hua University , Hsinchu 300 , Taiwan
| | - Yu-Chun Lin
- Institute of Molecular Medicine , National Tsing Hua University , Hsinchu 300 , Taiwan
- Department of Medical Science , National Tsing Hua University , Hsinchu 300 , Taiwan
| |
Collapse
|
5
|
Sroor HM, Hassan AM, Zenz G, Valadez-Cosmes P, Farzi A, Holzer P, El-Sharif A, Gomaa FAZM, Kargl J, Reichmann F. Experimental colitis reduces microglial cell activation in the mouse brain without affecting microglial cell numbers. Sci Rep 2019; 9:20217. [PMID: 31882991 PMCID: PMC6934553 DOI: 10.1038/s41598-019-56859-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 12/16/2019] [Indexed: 12/25/2022] Open
Abstract
Inflammatory bowel disease (IBD) patients frequently suffer from anxiety disorders and depression, indicating that altered gut-brain axis signalling during gastrointestinal inflammation is a risk factor for psychiatric disease. Microglia, immune cells of the brain, is thought to be involved in a number of mental disorders, but their role in IBD is largely unknown. In the current work, we investigated whether colitis induced by dextran sulphate sodium (DSS), a murine model of IBD, alters microglial phenotypes in the brain. We found that colitis caused a reduction of Iba-1 and CD68 immunoreactivity, microglial activation markers, in specific brain regions of the limbic system such as the medial prefrontal cortex (mPFC), while other areas remained unaffected. Flow cytometry showed an increase of monocyte-derived macrophages during colitis and gene expression analysis in the mPFC showed pronounced changes of microglial markers including cluster of differentiation 86 (CD86), tumour necrosis factor-α, nitric oxide synthase 2, CD206 and chitinase-like protein 3 consistent with both M1 and M2 activation. Taken together, these findings suggest that experimental colitis-induced inflammation is propagated to the brain altering microglial function.
Collapse
Affiliation(s)
- Hoda M Sroor
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Centre for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria.,Microbiology and Immunology Department, Faculty of Pharmacy-Girls, Al-Azar University, Cairo, Egypt
| | - Ahmed M Hassan
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Centre for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Geraldine Zenz
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Centre for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Paulina Valadez-Cosmes
- Division of Pharmacology, Otto Loewi Research Centre for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Centre for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Peter Holzer
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Centre for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Amany El-Sharif
- Microbiology and Immunology Department, Faculty of Pharmacy-Girls, Al-Azar University, Cairo, Egypt
| | - Fatma Al-Zahraa M Gomaa
- Microbiology and Immunology Department, Faculty of Pharmacy-Girls, Al-Azar University, Cairo, Egypt.,Pharmacognosy and Medicinal Herbs Department, Faculty of Clinical Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Centre for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Division of Pharmacology, Otto Loewi Research Centre for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria.
| |
Collapse
|
6
|
Schizophrenia-like reduced sensorimotor gating in intact inbred and outbred rats is associated with decreased medial prefrontal cortex activity and volume. Neuropsychopharmacology 2019; 44:1975-1984. [PMID: 30986819 PMCID: PMC6784988 DOI: 10.1038/s41386-019-0392-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/02/2019] [Accepted: 04/07/2019] [Indexed: 02/08/2023]
Abstract
Prepulse inhibition (PPI) of startle response is a measure of sensorimotor gating that is impaired in schizophrenia and in many other clinical conditions. Rat models using pharmacological or surgical strategies reveal that PPI is modulated by the cortico-striatal-pallido-thalamic (CSPT) circuit. Here, we explore whether spontaneous variation in PPI in intact inbred and outbred rats is associated with functional and structural differences in the CSPT circuit. Inbred Roman High-(RHA) and Low-avoidance (RLA) and outbred heterogeneous stock (HS) rats were assessed for PPI, brain activity, and brain volume. Brain activity was assessed by c-Fos expression and brain volume by magnetic resonance imaging. Relevant structures of the CSPT circuit were evaluated, such as the medial prefrontal cortex (mPFC), cingulate cortex, hippocampus (HPC), amygdala, nucleus accumbens (NAc), and dorsal striatum. RHA showed lower PPI than RLA rats, while HS rats were stratified by their PPI levels in three groups. Reduced PPI was accompanied by decreased mPFC activity in Roman and HS rats and increased NAc shell activity in HS rats. Low PPI was also associated with decreased mPFC and HPC volumes in Roman and HS rats. This study reports a consistent relationship between decreased function and volume of the mPFC and spontaneous low-PPI levels in inbred and outbred intact rats. Moreover, our findings suggest that, apart from a hypoactive and smaller mPFC, a hyperactive NAc and smaller HPC may underlie reduced PPI levels. Our results support the notion that sensorimotor gating is modulated by forebrain structures and highlight the importance of the mPFC in its regulation.
Collapse
|
7
|
Harriott AM, Strother LC, Vila-Pueyo M, Holland PR. Animal models of migraine and experimental techniques used to examine trigeminal sensory processing. J Headache Pain 2019; 20:91. [PMID: 31464579 PMCID: PMC6734323 DOI: 10.1186/s10194-019-1043-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/19/2019] [Indexed: 12/12/2022] Open
Abstract
Background Migraine is a common debilitating condition whose main attributes are severe recurrent headaches with accompanying sensitivity to light and sound, nausea and vomiting. Migraine-related pain is a major cause of its accompanying disability and can encumber almost every aspect of daily life. Main body Advancements in our understanding of the neurobiology of migraine headache have come in large from basic science research utilizing small animal models of migraine-related pain. In this current review, we aim to describe several commonly utilized preclinical models of migraine. We will discuss the diverse array of methodologies for triggering and measuring migraine-related pain phenotypes and highlight briefly specific advantages and limitations therein. Finally, we will address potential future challenges/opportunities to refine existing and develop novel preclinical models of migraine that move beyond migraine-related pain and expand into alternate migraine-related phenotypes. Conclusion Several well validated animal models of pain relevant for headache exist, the researcher should consider the advantages and limitations of each model before selecting the most appropriate to answer the specific research question. Further, we should continually strive to refine existing and generate new animal and non-animal models that have the ability to advance our understanding of head pain as well as non-pain symptoms of primary headache disorders.
Collapse
Affiliation(s)
- Andrea M Harriott
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Lauren C Strother
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychology, Psychiatry and Neuroscience, King's College London, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Marta Vila-Pueyo
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychology, Psychiatry and Neuroscience, King's College London, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Philip R Holland
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychology, Psychiatry and Neuroscience, King's College London, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
8
|
García-Cabrerizo R, García-Fuster MJ. Adolescent cocaine exposure enhanced negative affect following drug re-exposure in adult rats: Attenuation of c-Fos activation. J Psychopharmacol 2019; 33:154-162. [PMID: 30484727 DOI: 10.1177/0269881118812353] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND The goal of the present study was to utilize the adolescent drug experience as an emerging vulnerability factor for developing psychiatric comorbidities in adulthood that could, in turn, help to elucidate and/or hypothesize possible mechanisms contributing to higher relapse rates. OUTCOMES The current results showed that adolescent cocaine exposure (15 mg/kg, intraperitoneally, seven days) during early-mid adolescence (postnatal days 33-39) enhanced negative affect in adulthood, by increasing behavioral despair following drug re-exposure and by increasing anhedonia. Thus, these behavioral data provided a good model to further ascertain the long-term cellular and molecular adaptations that might take place in the brain in response to adolescent cocaine exposure as well as the impact of drug re-exposure in adulthood. In this regard, the results showed that adolescent cocaine exposure did not modulate cell proliferation (Ki-67+ cells) or c-Fos protein activation in the dentate gyrus region of the hippocampus, but attenuated c-Fos activation in the dorsal striatum. CONCLUSIONS These results proved that a history of cocaine exposure during adolescence increased the vulnerability to induce negative affect (i.e. emergence of psychiatric comorbidity) in adulthood while it decreased neuronal activation in the dorsal striatum. Interestingly, these effects were only observed following cocaine re-exposure in adulthood, suggesting that avoiding drug contact in adulthood could prevent the long-term negative effects induced by adolescent cocaine.
Collapse
|
9
|
Jaglin M, Rhimi M, Philippe C, Pons N, Bruneau A, Goustard B, Daugé V, Maguin E, Naudon L, Rabot S. Indole, a Signaling Molecule Produced by the Gut Microbiota, Negatively Impacts Emotional Behaviors in Rats. Front Neurosci 2018; 12:216. [PMID: 29686603 PMCID: PMC5900047 DOI: 10.3389/fnins.2018.00216] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/19/2018] [Indexed: 12/22/2022] Open
Abstract
Gut microbiota produces a wide and diverse array of metabolites that are an integral part of the host metabolome. The emergence of the gut microbiome-brain axis concept has prompted investigations on the role of gut microbiota dysbioses in the pathophysiology of brain diseases. Specifically, the search for microbe-related metabolomic signatures in human patients and animal models of psychiatric disorders has pointed out the importance of the microbial metabolism of aromatic amino acids. Here, we investigated the effect of indole on brain and behavior in rats. Indole is produced by gut microbiota from tryptophan, through the tryptophanase enzyme encoded by the tnaA gene. First, we mimicked an acute and high overproduction of indole by injecting this compound in the cecum of conventional rats. This treatment led to a dramatic decrease of motor activity. The neurodepressant oxidized derivatives of indole, oxindole and isatin, accumulated in the brain. In addition, increase in eye blinking frequency and in c-Fos protein expression in the dorsal vagal complex denoted a vagus nerve activation. Second, we mimicked a chronic and moderate overproduction of indole by colonizing germ-free rats with the indole-producing bacterial species Escherichia coli. We compared emotional behaviors of these rats with those of germ-free rats colonized with a genetically-engineered counterpart strain unable to produce indole. Rats overproducing indole displayed higher helplessness in the tail suspension test, and enhanced anxiety-like behavior in the novelty, elevated plus maze and open-field tests. Vagus nerve activation was suggested by an increase in eye blinking frequency. However, unlike the conventional rats dosed with a high amount of indole, the motor activity was not altered and neither oxindole nor isatin could be detected in the brain. Further studies are required for a comprehensive understanding of the mechanisms supporting indole effects on emotional behaviors. As our findings suggest that people whose gut microbiota is highly prone to produce indole could be more likely to develop anxiety and mood disorders, we addressed the issue of the inter-individual variability of indole producing potential in humans. An in silico investigation of metagenomic data focused on the tnaA gene products definitively proved this inter-individual variability.
Collapse
Affiliation(s)
- Mathilde Jaglin
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Moez Rhimi
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Catherine Philippe
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Nicolas Pons
- MetaGenoPolis, Institut National de la Recherche Agronomique, Université Paris-Saclay, Jouy-en-Josas, France
| | - Aurélia Bruneau
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Bénédicte Goustard
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Valérie Daugé
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Centre National de la Recherche Scientifique, Université Paris-Saclay, Jouy-en-Josas, France
| | - Emmanuelle Maguin
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Laurent Naudon
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Centre National de la Recherche Scientifique, Université Paris-Saclay, Jouy-en-Josas, France
| | - Sylvie Rabot
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
10
|
Abstract
The majority of 20th century investigations into anesthetic effects on the nervous system have used electrophysiology. Yet some fundamental limitations to electrophysiologic recordings, including the invasiveness of the technique, the need to place (potentially several) electrodes in every site of interest, and the difficulty of selectively recording from individual cell types, have driven the development of alternative methods for detecting neuronal activation. Two such alternative methods with cellular scale resolution have matured in the last few decades and will be reviewed here: the transcription of immediate early genes, foremost c-fos, and the influx of calcium into neurons as reported by genetically encoded calcium indicators, foremost GCaMP6. Reporters of c-fos allow detection of transcriptional activation even in deep or distant nuclei, without requiring the accurate targeting of multiple electrodes at long distances. The temporal resolution of c-fos is limited due to its dependence upon the detection of transcriptional activation through immunohistochemical assays, though the development of RT-PCR probes has shifted the temporal resolution of the assay when tissues of interest can be isolated. GCaMP6 has several isoforms that trade-off temporal resolution for signal to noise, but the fastest are capable of resolving individual action potential events, provided the microscope used scans quickly enough. GCaMP6 expression can be selectively targeted to neuronal populations of interest, and potentially thousands of neurons can be captured within a single frame, allowing the neuron-by-neuron reporting of circuit dynamics on a scale that is difficult to capture with electrophysiology, as long as the populations are optically accessible.
Collapse
|
11
|
Li YW, Seager MA, Wojcik T, Heman K, Molski TF, Fernandes A, Langdon S, Pendri A, Gerritz S, Tian Y, Hong Y, Gallagher L, Merritt JR, Zhang C, Westphal R, Zaczek R, Macor JE, Bronson JJ, Lodge NJ. Biochemical and behavioral effects of PDE10A inhibitors: Relationship to target site occupancy. Neuropharmacology 2016; 102:121-35. [DOI: 10.1016/j.neuropharm.2015.10.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 10/14/2015] [Accepted: 10/26/2015] [Indexed: 12/21/2022]
|
12
|
Wilson JM, Ogden AML, Loomis S, Gilmour G, Baucum AJ, Belecky-Adams TL, Merchant KM. Phosphodiesterase 10A inhibitor, MP-10 (PF-2545920), produces greater induction of c-Fos in dopamine D2 neurons than in D1 neurons in the neostriatum. Neuropharmacology 2015; 99:379-86. [DOI: 10.1016/j.neuropharm.2015.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/31/2015] [Accepted: 08/04/2015] [Indexed: 11/16/2022]
|
13
|
Bastakis GG, Savvaki M, Stamatakis A, Vidaki M, Karagogeos D. Tag1 deficiency results in olfactory dysfunction through impaired migration of mitral cells. Development 2015; 142:4318-28. [PMID: 26525675 DOI: 10.1242/dev.123943] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 10/22/2015] [Indexed: 01/01/2023]
Abstract
The olfactory system provides mammals with the abilities to investigate, communicate and interact with their environment. These functions are achieved through a finely organized circuit starting from the nasal cavity, passing through the olfactory bulb and ending in various cortical areas. We show that the absence of transient axonal glycoprotein-1 (Tag1)/contactin-2 (Cntn2) in mice results in a significant and selective defect in the number of the main projection neurons in the olfactory bulb, namely the mitral cells. A subpopulation of these projection neurons is reduced in Tag1-deficient mice as a result of impaired migration. We demonstrate that the detected alterations in the number of mitral cells are well correlated with diminished odor discrimination ability and social long-term memory formation. Reduced neuronal activation in the olfactory bulb and the corresponding olfactory cortex suggest that Tag1 is crucial for the olfactory circuit formation in mice. Our results underpin the significance of a numerical defect in the mitral cell layer in the processing and integration of odorant information and subsequently in animal behavior.
Collapse
Affiliation(s)
- George G Bastakis
- Department of Basic Science, Faculty of Medicine, University of Crete and Institute of Molecular Biology and Biotechnology-FoRTH, Vassilika Vouton, Heraklion, Crete 71110, Greece
| | - Maria Savvaki
- Department of Basic Science, Faculty of Medicine, University of Crete and Institute of Molecular Biology and Biotechnology-FoRTH, Vassilika Vouton, Heraklion, Crete 71110, Greece
| | - Antonis Stamatakis
- Laboratory of Biology, Faculty of Nursing, School of Health Sciences, University of Athens, Papadiamantopoulou 123, Athens GR11527, Greece
| | - Marina Vidaki
- Department of Basic Science, Faculty of Medicine, University of Crete and Institute of Molecular Biology and Biotechnology-FoRTH, Vassilika Vouton, Heraklion, Crete 71110, Greece
| | - Domna Karagogeos
- Department of Basic Science, Faculty of Medicine, University of Crete and Institute of Molecular Biology and Biotechnology-FoRTH, Vassilika Vouton, Heraklion, Crete 71110, Greece
| |
Collapse
|
14
|
A novel UCS memory retrieval-extinction procedure to inhibit relapse to drug seeking. Nat Commun 2015; 6:7675. [PMID: 26169171 PMCID: PMC4510700 DOI: 10.1038/ncomms8675] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/01/2015] [Indexed: 02/08/2023] Open
Abstract
We recently reported that a conditioned stimulus (CS) memory retrieval-extinction procedure decreases reinstatement of cocaine and heroin seeking in rats and heroin craving in humans. Here we show that non-contingent cocaine or methylphenidate injections (UCS retrieval) 1 h before the extinction sessions decreases cocaine-priming-induced reinstatement, spontaneous recovery, and renewal of cocaine seeking in rats. Unlike the CS-based memory retrieval-extinction procedure, the UCS memory retrieval manipulation decreases renewal and reinstatement of cocaine seeking in the presence of cocaine cues that were not present during extinction training and also decreases cocaine seeking when the procedure commences after 28 days of abstinence. The inhibitory effect of the UCS retrieval manipulation on cocaine-priming-induced reinstatement is mediated by regulation of AMPA-receptor endocytosis in the basolateral amygdala. The UCS memory retrieval-extinction procedure has superior relapse prevention characteristics than the CS memory retrieval-extinction procedure and could be a promising method for decreasing relapse in human addicts. Cue-based therapies for treating drug addiction have proven to be only partially effective. Here the authors demonstrate a new memory retrieval based treatment protocol for drug addiction that results in long-lasting inhibition of drug seeking behavior in rodents.
Collapse
|
15
|
Farzi A, Reichmann F, Meinitzer A, Mayerhofer R, Jain P, Hassan AM, Fröhlich EE, Wagner K, Painsipp E, Rinner B, Holzer P. Synergistic effects of NOD1 or NOD2 and TLR4 activation on mouse sickness behavior in relation to immune and brain activity markers. Brain Behav Immun 2015; 44:106-20. [PMID: 25218901 PMCID: PMC4295938 DOI: 10.1016/j.bbi.2014.08.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 08/29/2014] [Accepted: 08/30/2014] [Indexed: 01/09/2023] Open
Abstract
Toll-like receptors (TLRs) and nuclear-binding domain (NOD)-like receptors (NLRs) are sensors of bacterial cell wall components to trigger an immune response. The TLR4 agonist lipopolysaccharide (LPS) is a strong immune activator leading to sickness and depressed mood. NOD agonists are less active but can prime immune cells to augment LPS-induced cytokine production. Since the impact of NOD and TLR co-activation in vivo has been little studied, the effects of the NOD1 agonist FK565 and the NOD2 agonist muramyl dipeptide (MDP), alone and in combination with LPS, on immune activation, brain function and sickness behavior were investigated in male C57BL/6N mice. Intraperitoneal injection of FK565 (0.001 or 0.003mg/kg) or MDP (1 or 3mg/kg) 4h before LPS (0.1 or 0.83mg/kg) significantly aggravated and prolonged the LPS-evoked sickness behavior as deduced from a decrease in locomotion, exploration, food intake and temperature. When given alone, FK565 and MDP had only minor effects. The exacerbation of sickness behavior induced by FK565 or MDP in combination with LPS was paralleled by enhanced plasma protein and cerebral mRNA levels of proinflammatory cytokines (IFN-γ, IL-1β, IL-6, TNF-α) as well as enhanced plasma levels of kynurenine. Immunohistochemical visualization of c-Fos in the brain revealed that NOD2 synergism with TLR4 resulted in increased activation of cerebral nuclei relevant to sickness. These data show that NOD1 or NOD2 synergizes with TLR4 in exacerbating the immune, sickness and brain responses to peripheral immune stimulation. Our findings demonstrate that the known interactions of NLRs and TLRs at the immune cell level extend to interactions affecting brain function and behavior.
Collapse
Affiliation(s)
- Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria.
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Raphaela Mayerhofer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Piyush Jain
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Ahmed M. Hassan
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Esther E. Fröhlich
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Karin Wagner
- Core Facility Molecular Biology, Center for Medical Research, Medical University of Graz, Stiftingtalstrasse 24/1, 8010 Graz, Austria
| | - Evelin Painsipp
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| | - Beate Rinner
- Core Facility Flow Cytometry, Center for Medical Research, Medical University of Graz, Stiftingtalstrasse 24/1, 8010 Graz, Austria
| | - Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| |
Collapse
|
16
|
Neuropeptide trefoil factor 3 attenuates naloxone-precipitated withdrawal in morphine-dependent mice. Psychopharmacology (Berl) 2014; 231:4659-68. [PMID: 24825609 DOI: 10.1007/s00213-014-3615-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/28/2014] [Indexed: 02/06/2023]
Abstract
RATIONALE The persistence of physical dependence and craving in addicts is considered to contribute to relapse. Increasing evidence indicates that neuropeptide systems are associated with several phases of drug addiction, but little is known about whether the neuropeptide trefoil factor affects withdrawal symptoms. OBJECTIVES This study aims to investigate the potential effects of the neuropeptide trefoil factor 3 (TFF3) on naloxone-precipitated withdrawal symptoms in morphine-dependent mice. RESULTS Mice received increasing doses of morphine over 3 days. On day 4, the mice were injected with TFF3 (1.0 mg/kg, i.p.) 30 min after the last dose of morphine. Thirty minutes after TFF3 treatment, naloxone (1 mg/kg, i.p.) was injected, and body weight, jumping behavior, wet-dog shakes, and locomotor activity were assessed 30 min later. Naloxone caused significant weight loss and increased jumping behavior and wet-dog shakes in morphine-dependent mice. TFF3 (1.0 mg/kg) reversed these behavioral symptoms caused by morphine withdrawal, suggesting that TFF3 might ameliorate physical dependence associated with opiate addiction. Furthermore, TFF3 pretreatment significantly reduced morphine withdrawal-induced increases in plasma corticosterone and adrenocorticotropic hormone levels. The glucocorticoid receptor agonist RU486 blocked the behavioral effects of TFF3 on morphine withdrawal symptoms. Finally, Fos expression in the medial prefrontal cortex which was decreased during morphine withdrawal was increased by TFF3 pretreatment. CONCLUSION These findings indicate that TFF3 might be a potential therapeutic candidate for opiate addiction by regulating glucocorticoid secretion and neuronal activation in the prefrontal cortex.
Collapse
|
17
|
Perreault ML, Shen MYF, Fan T, George SR. Regulation of c-fos expression by the dopamine D1-D2 receptor heteromer. Neuroscience 2014; 285:194-203. [PMID: 25446350 DOI: 10.1016/j.neuroscience.2014.11.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 11/03/2014] [Accepted: 11/10/2014] [Indexed: 11/24/2022]
Abstract
The dopamine D1 and D2 receptors form the D1-D2 receptor heteromer in a subset of neurons and couple to the Gq protein to regulate intracellular calcium signaling. In the present study the effect of D1-D2 heteromer activation and disruption on neuronal activation in the rat brain was mapped. This was accomplished using the dopamine agonist SKF 83959 to activate the D1-D2 heteromer in combination with a TAT-D1 disrupting peptide we developed, and which has been shown to disrupt the D1/D2 receptor interaction and antagonize D1-D2 heteromer-induced cell signaling and behavior. Acute SKF 83959 administration to rats induced significant c-fos expression in the nucleus accumbens that was significantly inhibited by TAT-D1 pretreatment. No effects of SKF 83959 were seen in caudate putamen. D1-D2 heteromer disruption by TAT-D1 did not have any effects in any striatal subregions, but induced significant c-fos immunoreactivity in a number of cortical regions including the orbitofrontal cortex, prelimbic and infralimbic cortices and piriform cortex. The induction of c-fos by TAT-D1 was also evident in the anterior olfactory nucleus, as well as the lateral habenula and thalamic nuclei. These findings show for the first time that the D1-D2 heteromer can differentially regulate c-fos expression in a region-dependent manner either through its activation or through tonic inhibition of neuronal activity.
Collapse
Affiliation(s)
- M L Perreault
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - M Y F Shen
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - T Fan
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - S R George
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
18
|
Qin J, Zhang RX, Li JL, Wang JX, Hou J, Yang X, Zhu WL, Shi J, Lu L. cRGD mediated liposomes enhanced antidepressant-like effects of edaravone in rats. Eur J Pharm Sci 2014; 58:63-71. [PMID: 24704101 DOI: 10.1016/j.ejps.2014.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 03/13/2014] [Accepted: 03/25/2014] [Indexed: 11/28/2022]
Abstract
The delayed onset of therapeutic outcomes is a major drawback of the current antidepressants. The blood-brain barrier is the most important bottleneck impeding drug transport into the brain. Therefore, development of novel antidepressant medications with rapid onset and sustained activity is urgent. RGD liposomes showed an excellent effect of brain-targeting drug delivery and increased the entering rate to the brain. In the present study, we prepared cyclic RGD liposomes loaded with edaravone (cRGD-ERLs) and evaluated the potential antidepressant-like effects of this drug delivery system in rats. The results showed single injection of cRGD-ERLs produced significant antidepressant-like effects in both forced swim and novelty suppressed feeding test. Moreover, acute cRGD-ERLs increased the expression of c-fos in the medial prefrontal cortex, suggesting that cRGD-ERLs could activate the neuronal function. Furthermore, cRGD-ERLs reversed the increase of lipopolysaccharides (LPS)-induced plasma cytokine IL-1β and IL-6, suggesting that normalization of cytokine level might be involved in the behavioral response of cRGD-ERLs. Finally, cRGD-ERLs prevented the increase of immobility induced by LPS in the forced swim test. Overall, the current data revealed a novel brain-target drug delivery system, which can be used to improve the therapeutic outcomes of antidepressants by increase of crossing rate to the brain.
Collapse
Affiliation(s)
- Jing Qin
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Ruo-Xi Zhang
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Jia-Li Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Jian-Xin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jia Hou
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Xu Yang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Wei-Li Zhu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China.
| | - Jie Shi
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Lin Lu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China
| |
Collapse
|
19
|
Reichmann F, Painsipp E, Holzer P. Environmental enrichment and gut inflammation modify stress-induced c-Fos expression in the mouse corticolimbic system. PLoS One 2013; 8:e54811. [PMID: 23349972 PMCID: PMC3547954 DOI: 10.1371/journal.pone.0054811] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 12/17/2012] [Indexed: 12/20/2022] Open
Abstract
Environmental enrichment (EE) has a beneficial effect on rodent behaviour, neuronal plasticity and brain function. Although it may also improve stress coping, it is not known whether EE influences the brain response to an external (psychological) stressor such as water avoidance stress (WAS) or an internal (systemic) stressor such as gastrointestinal inflammation. This study hence explored whether EE modifies WAS-induced activation of the mouse corticolimbic system and whether this stress response is altered by gastritis or colitis. Male C67BL/6N mice were housed under standard or enriched environment for 9 weeks, after which they were subjected to a 1-week treatment with oral iodoacetamide to induce gastritis or oral dextran sulfate sodium to induce colitis. Following exposure to WAS the expression of c-Fos, a marker of neuronal activation, was measured by immunocytochemistry. EE aggravated experimentally induced colitis, but not gastritis, as shown by an increase in the disease activity score and the colonic myeloperoxidase content. In the brain, EE enhanced the WAS-induced activation of the dentate gyrus and unmasked an inhibitory effect of gastritis and colitis on WAS-evoked c-Fos expression within this part of the hippocampus. Conversely, EE inhibited the WAS-evoked activation of the central amygdala and prevented the inhibitory effect of gastritis and colitis on WAS-evoked c-Fos expression in this region. EE, in addition, blunted the WAS-induced activation of the infralimbic cortex and attenuated the inhibitory effect of gastritis and colitis on WAS-evoked c-Fos expression in this area. These data reveal that EE has a region-specific effect on stress-induced c-Fos expression in the corticolimbic system, which is likely to improve stress resilience. The response of the prefrontal cortex – amygdala – hippocampus circuitry to psychological stress is also modified by the systemic stress of gut inflammation, and this interaction between external and internal stressors is modulated by the housing environment.
Collapse
Affiliation(s)
- Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria.
| | | | | |
Collapse
|
20
|
Shi HS, Zhu WL, Liu JF, Luo YX, Si JJ, Wang SJ, Xue YX, Ding ZB, Shi J, Lu L. PI3K/Akt signaling pathway in the basolateral amygdala mediates the rapid antidepressant-like effects of trefoil factor 3. Neuropsychopharmacology 2012; 37:2671-83. [PMID: 22828749 PMCID: PMC3473333 DOI: 10.1038/npp.2012.131] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Depression is one of the most common and debilitating psychiatric illnesses around the world, but the current antidepressants used to treat depression have many limitations. Progressively more studies have shown that neuropeptide systems are potential novel therapeutic targets for depression. However, whether the neuropeptide trefoil factor 3 (TFF3) participates in the development of depression has not been examined. In the current experiments, we assessed the antidepressant effects of TFF3 using the forced swim test (FST), tail suspension test (TST), and chronic mild stress (CMS) paradigm. Furthermore, we determined the mechanism that underlies the antidepressant-like effects of TFF3 in the rat FST. TFF3 dose-dependently reduced immobility time in both FST and TST. CMS elevated plasma TFF3 and decreased basolateral amygdala (BLA) TFF3 levels in rats, and acute TFF3 (0.1 mg/kg, i.p.) treatment reversed the depressive-like behaviors induced by CMS. Furthermore, TFF3 (0.1 mg/kg, i.p.) significantly increased Fos expression in the BLA, medial prefrontal cortex, and hypothalamus in rats subjected to the FST. Intra-BLA infusions of TFF3 (1 ng/side) exerted rapid antidepressant-like effects in the rat FST. Additionally, acute systemic TFF3 administration increased the level of phosphorylated-Akt (p-Akt) in the BLA. Finally, intra-BLA infusions of LY294002 (5 mM/side), a specific phosphatidylinositol 3-kinase (PI3K) inhibitor, significantly blocked the antidepressant-like effect of TFF3. Our results demonstrated that TFF3 exerts antidepressant-like effects that might be mediated by the PI3K/Akt signaling pathway in the BLA. These findings suggest a novel neuropeptide system target in the development of new antidepressants.
Collapse
Affiliation(s)
- Hai-Shui Shi
- National Institute on Drug Dependence, Peking University, Beijing, China,Department of Biochemistry and Molecular Biology, Basic Medical College, Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang, China
| | - Wei-Li Zhu
- National Institute on Drug Dependence, Peking University, Beijing, China,National Institute on Drug Dependence, Peking University, 38, Xue Yuan Road, Hai Dian District, Beijing 100191, China, Tel: +86 10 82802456, Fax: +86 10 62032624, E-mail: (W-LZ) or (LL)
| | - Jian-Feng Liu
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Yi-Xiao Luo
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Ji-Jian Si
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Shen-Jun Wang
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Yan-Xue Xue
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Zeng-Bo Ding
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Jie Shi
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Lin Lu
- National Institute on Drug Dependence, Peking University, Beijing, China,Key Lab for Neuroscience, Ministry of Education/Ministry of Health, Beijing, China,National Institute on Drug Dependence, Peking University, 38, Xue Yuan Road, Hai Dian District, Beijing 100191, China, Tel: +86 10 82802456, Fax: +86 10 62032624, E-mail: (W-LZ) or (LL)
| |
Collapse
|
21
|
Leger M, Quiedeville A, Paizanis E, Natkunarajah S, Freret T, Boulouard M, Schumann-Bard P. Environmental enrichment enhances episodic-like memory in association with a modified neuronal activation profile in adult mice. PLoS One 2012; 7:e48043. [PMID: 23110171 PMCID: PMC3478271 DOI: 10.1371/journal.pone.0048043] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 09/20/2012] [Indexed: 12/27/2022] Open
Abstract
Although environmental enrichment is well known to improve learning and memory in rodents, the underlying neuronal networks' plasticity remains poorly described. Modifications of the brain activation pattern by enriched condition (EC), especially in the frontal cortex and the baso-lateral amygdala, have been reported during an aversive memory task in rodents. The aims of our study were to examine 1) whether EC modulates episodic-like memory in an object recognition task and 2) whether EC modulates the task-induced neuronal networks. To this end, adult male mice were housed either in standard condition (SC) or in EC for three weeks before behavioral experiments (n = 12/group). Memory performances were examined in an object recognition task performed in a Y-maze with a 2-hour or 24-hour delay between presentation and test (inter-session intervals, ISI). To characterize the mechanisms underlying the promnesiant effect of EC, the brain activation profile was assessed after either the presentation or the test sessions using immunohistochemical techniques with c-Fos as a neuronal activation marker. EC did not modulate memory performances after a 2 h-ISI, but extended object recognition memory to a 24 h-ISI. In contrast, SC mice did not discriminate the novel object at this ISI. Compared to SC mice, no activation related to the presentation session was found in selected brain regions of EC mice (in particular, no effect was found in the hippocampus and the perirhinal cortex and a reduced activation was found in the baso-lateral amygdala). On the other hand, an activation of the hippocampus and the infralimbic cortex was observed after the test session for EC, but not SC mice. These results suggest that the persistence of object recognition memory in EC could be related to a reorganization of neuronal networks occurring as early as the memory encoding.
Collapse
Affiliation(s)
- Marianne Leger
- Groupe Mémoire et Plasticité Comportementale-GMPc, Université de Caen Basse-Normandie, EA 4259, Caen, France.
| | | | | | | | | | | | | |
Collapse
|
22
|
Panhelainen AE, Korpi ER. Evidence for a role of inhibition of orexinergic neurons in the anxiolytic and sedative effects of diazepam: A c-Fos study. Pharmacol Biochem Behav 2011; 101:115-24. [PMID: 22210490 DOI: 10.1016/j.pbb.2011.12.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 12/09/2011] [Accepted: 12/14/2011] [Indexed: 12/22/2022]
Abstract
The classical benzodiazepine diazepam (DZ) induces anxiolysis at low doses and sedation and hypnosis at higher doses. Different brain areas and neuronal populations most likely mediate these different behavioral effects. We used c-Fos immunohistochemistry as an indirect way to study neuronal activation or inhibition induced by DZ at anxiolytic and sedative doses (0.5 and 5mg/kg, respectively) in various brain areas involved in anxiety, arousal, sedation and addiction in C57BL/6J mice. We also focused on the two neuronal populations, orexinergic and dopaminergic neuronal populations, with the help of double-immunohistochemistry using c-Fos and orexin-A antibodies and c-Fos and tyrosine hydroxylase antibodies. We found that different brain areas of unhabituated mice reacted differently to the mild stress induced by vehicle injection. Also the response to anxiolytic or sedative doses of DZ differed between the areas, suggesting that distinct brain areas mediate the behavioral effects of low and high DZ doses. Our findings propose a role for inhibition of orexin neurons in the anxiolytic and sleep-promoting effects of DZ. In addition, the activation of central amygdala neurons by DZ treatment was associated with anxiolytic and sedative effects. On the other hand, the ventral hippocampus, basolateral amygdala, ventral tegmental area and prefrontal cortex were sensitive even to the mild injection stress, but not to the anxiolytic dose of DZ.
Collapse
Affiliation(s)
- Anne E Panhelainen
- Institute of Biomedicine, Pharmacology, Biomedicum Helsinki, POB 63 (Haartmaninkatu 8), FI-00014 University of Helsinki, Finland.
| | | |
Collapse
|
23
|
Schwartz CP, Smotherman MS. Mapping vocalization-related immediate early gene expression in echolocating bats. Behav Brain Res 2011; 224:358-68. [PMID: 21726584 DOI: 10.1016/j.bbr.2011.06.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 05/20/2011] [Accepted: 06/14/2011] [Indexed: 12/26/2022]
Abstract
Recent studies of spontaneously vocalizing primates, cetaceans, bats and rodents suggest these animals possess a limited but meaningful capacity to manipulate the timing and acoustic structure of their vocalizations, yet the neural substrate for even the simplest forms of vocal modulation in mammals remains unknown. Echolocating bats rapidly and routinely manipulate the acoustic structure of their outgoing vocalizations to improve echolocation efficiency, reflecting cognitive rather than limbic control of the vocal motor pathways. In this study, we used immunohistochemical localization of immediate early gene (c-fos) expression to map neural activity in the brains of spontaneously echolocating stationary Mexican free-tailed bats. Our results support the current model of vocal control obtained largely through microstimulation studies, but also provide evidence for the contributions of two novel regions, the dorsolateral caudate nucleus and mediodorsal thalamic nucleus, which together suggest a striatothalamic feedback loop may be involved in the control of echolocation pulse production. Additionally, we found evidence of a motivation pathway, including the lateral habenula, substantia nigra pars compacta, and raphe nuclei. These data provide novel insights into where and how mammalian vocalizations may be regulated by sensory, contextual and motivational cues.
Collapse
Affiliation(s)
- Christine P Schwartz
- Department of Biology, Texas A&M University, College Station, TX 77843-3258, USA.
| | | |
Collapse
|
24
|
Procaccini C, Aitta-aho T, Jaako-Movits K, Zharkovsky A, Panhelainen A, Sprengel R, Linden AM, Korpi ER. Excessive novelty-induced c-Fos expression and altered neurogenesis in the hippocampus of GluA1 knockout mice. Eur J Neurosci 2010; 33:161-74. [PMID: 21073553 DOI: 10.1111/j.1460-9568.2010.07485.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor GluA1 subunit-deficient (GluA1-/-) mice display novelty-induced hyperactivity, cognitive and social defects and may model psychiatric disorders, such as schizophrenia and depression/mania. We used c-Fos expression in GluA1-/- mice to identify brain regions responsible for novelty-induced hyperlocomotion. Exposure to a novel cage for 2 h significantly increased c-Fos expression in many brain regions in both wild-type and knockout mice. Interestingly, the clearest genotype effect was observed in the hippocampus and its main input region, the entorhinal cortex, where the novelty-induced c-Fos expression was more strongly enhanced in GluA1-/- mice. Their novelty-induced hyperlocomotion partly depended on the activity of AMPA receptors, as it was diminished by the AMPA receptor antagonist 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]quinoxaline-7-sulphonamide (NBQX) and unaffected by the AMPA receptor potentiator 2,3-dihydro-1,4-benzodioxin-6-yl-1-piperidinylmethanone (CX546). The hyperlocomotion of GluA1-/- mice was normalised to the level of wild-type mice within 5-6 h, after which their locomotion followed normal circadian rhythm and was not affected by acute or chronic treatments with the selective serotonin reuptake inhibitor escitalopram. We propose that hippocampal dysfunction, as evidenced by the excessive c-Fos response to novelty, is the major contributor to novelty-induced hyperlocomotion in GluA1-/- mice. Hippocampal dysfunction was also indicated by changes in proliferation and survival of adult-born dentate gyrus cells in the knockout mice. These results suggest focusing on the functions of hippocampal formation, such as novelty detection, when using the GluA1-/- mouse line as a model for neuropsychiatric and cognitive disorders.
Collapse
Affiliation(s)
- Chiara Procaccini
- Institute of Biomedicine/Pharmacology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Phase misalignment between suprachiasmatic neuronal oscillators impairs photic behavioral phase shifts but not photic induction of gene expression. J Neurosci 2010; 30:13150-6. [PMID: 20881133 DOI: 10.1523/jneurosci.1853-10.2010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The ability of the circadian pacemaker within the suprachiasmatic nucleus (SCN) to respond to light stimulation in a phase-specific manner constitutes the basis for photic entrainment of circadian rhythms. The neural basis for this phase specificity is unclear. We asked whether a lack of synchrony between SCN neurons, as reflected in phase misalignment between dorsomedial (dmSCN) and ventrolateral (vlSCN) neuronal oscillators in the rat, would impact the ability of the pacemaker to respond to phase-resetting light pulses. Light pulses delivered at maximal phase misalignment between the vlSCN and dmSCN oscillators increased expression of Per1 mRNA, regardless of the circadian phase of the dmSCN. However, phase shifts of locomotor activity were only observed when the vlSCN and dmSCN were phase aligned at the time of stimulation. Our results fit a model in which a vlSCN oscillator phase gates its own response to light and in turn relays light information to a dmSCN oscillator. This model predicts that the phase misalignment that results from circadian internal desynchronization could preserve the ability of light to induce gene expression within the master circadian clock but impair its ability to induce behavioral phase shifts.
Collapse
|
26
|
Goulton CS, Patten AR, Kerr JR, Kerr DS. Pharmacological Preconditioning with GYKI 52466: A Prophylactic Approach to Neuroprotection. Front Neurosci 2010; 4. [PMID: 20953290 PMCID: PMC2955399 DOI: 10.3389/fnins.2010.00054] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 07/02/2010] [Indexed: 11/13/2022] Open
Abstract
Some toxins and drugs can trigger lasting neuroprotective mechanisms that enable neurons to resist a subsequent severe insult. This “pharmacological preconditioning” has far-reaching implications for conditions in which blood flow to the brain is interrupted. We have previously shown that in vitro preconditioning with the AMPA receptor antagonist GYKI 52466 induces tolerance to kainic acid (KA) toxicity in hippocampus. This effect persists well after washout of the drug and may be mediated via inverse agonism of G-protein coupled receptors (GPCRs). Given the amplifying nature of metabotropic modulation, we hypothesized that GYKI 52466 may be effective in reducing seizure severity at doses well below those normally associated with adverse side effects. Here we report that pharmacological preconditioning with low-dose GYKI imparts a significant protection against KA-induced seizures in vivo. GYKI (3 mg/kg, s.c.), 90–180 min prior to high-dose KA, markedly reduced seizure scores, virtually abolished all level 3 and level 4 seizures, and completely suppressed KA-induced hippocampal c-FOS expression. In addition, preconditioned animals exhibited significant reductions in high frequency/high amplitude spiking and ECoG power in the delta, theta, alpha, and beta bands during KA. Adverse behaviors often associated with higher doses of GYKI were not evident during preconditioning. The fact that GYKI is effective at doses well-below, and at pre-administration intervals well-beyond previous studies, suggests that a classical blockade of ionotropic AMPA receptors does not underlie anticonvulsant effects. Low-dose GYKI preconditioning may represent a novel, prophylactic strategy for neuroprotection in a field almost completely devoid of effective pharmaceuticals.
Collapse
Affiliation(s)
- Chelsea S Goulton
- Department of Pharmacology and Toxicology, School of Medical Sciences, University of Otago Dunedin, New Zealand
| | | | | | | |
Collapse
|
27
|
Gardiner JV, Bataveljic A, Patel NA, Bewick GA, Roy D, Campbell D, Greenwood HC, Murphy KG, Hameed S, Jethwa PH, Ebling FJ, Vickers SP, Cheetham S, Ghatei MA, Bloom SR, Dhillo WS. Prokineticin 2 is a hypothalamic neuropeptide that potently inhibits food intake. Diabetes 2010; 59:397-406. [PMID: 19933997 PMCID: PMC2809973 DOI: 10.2337/db09-1198] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Prokineticin 2 (PK2) is a hypothalamic neuropeptide expressed in central nervous system areas known to be involved in food intake. We therefore hypothesized that PK2 plays a role in energy homeostasis. RESEARCH DESIGN AND METHODS We investigated the effect of nutritional status on hypothalamic PK2 expression and effects of PK2 on the regulation of food intake by intracerebroventricular (ICV) injection of PK2 and anti-PK2 antibody. Subsequently, we investigated the potential mechanism of action by determining sites of neuronal activation after ICV injection of PK2, the hypothalamic site of action of PK2, and interaction between PK2 and other hypothalamic neuropeptides regulating energy homeostasis. To investigate PK2's potential as a therapeutic target, we investigated the effect of chronic administration in lean and obese mice. RESULTS Hypothalamic PK2 expression was reduced by fasting. ICV administration of PK2 to rats potently inhibited food intake, whereas anti-PK2 antibody increased food intake, suggesting that PK2 is an anorectic neuropeptide. ICV administration of PK2 increased c-fos expression in proopiomelanocortin neurons of the arcuate nucleus (ARC) of the hypothalamus. In keeping with this, PK2 administration into the ARC reduced food intake and PK2 increased the release of alpha-melanocyte-stimulating hormone (alpha-MSH) from ex vivo hypothalamic explants. In addition, ICV coadministration of the alpha-MSH antagonist agouti-related peptide blocked the anorexigenic effects of PK2. Chronic peripheral administration of PK2 reduced food and body weight in lean and obese mice. CONCLUSIONS This is the first report showing that PK2 has a role in appetite regulation and its anorectic effect is mediated partly via the melanocortin system.
Collapse
Affiliation(s)
- James V. Gardiner
- Department of Investigative Medicine, Imperial College London, London, U.K
| | - Attia Bataveljic
- Department of Investigative Medicine, Imperial College London, London, U.K
| | - Neekhil A. Patel
- Department of Investigative Medicine, Imperial College London, London, U.K
| | - Gavin A. Bewick
- Department of Investigative Medicine, Imperial College London, London, U.K
| | - Debabrata Roy
- Department of Investigative Medicine, Imperial College London, London, U.K
| | - Daniel Campbell
- Department of Investigative Medicine, Imperial College London, London, U.K
| | | | - Kevin G. Murphy
- Department of Investigative Medicine, Imperial College London, London, U.K
| | - Saira Hameed
- Department of Investigative Medicine, Imperial College London, London, U.K
| | - Preeti H. Jethwa
- School of Biomedical Sciences, University of Nottingham, Nottingham, U.K
| | | | | | | | - Mohammad A. Ghatei
- Department of Investigative Medicine, Imperial College London, London, U.K
| | - Stephen R. Bloom
- Department of Investigative Medicine, Imperial College London, London, U.K
- Corresponding author: Stephen R. Bloom,
| | - Waljit S. Dhillo
- Department of Investigative Medicine, Imperial College London, London, U.K
| |
Collapse
|
28
|
Xu A, Sakurai E, Kuramasu A, Zhang J, Li J, Okamura N, Zhang D, Yoshikawa T, Watanabe T, Yanai K. Roles of Hypothalamic Subgroup Histamine and Orexin Neurons on Behavioral Responses to Sleep Deprivation Induced by the Treadmill Method in Adolescent Rats. J Pharmacol Sci 2010; 114:444-53. [DOI: 10.1254/jphs.10177fp] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
29
|
Tan LA, Xu K, Vaccarino FJ, Lovejoy DA, Rotzinger S. Teneurin C-terminal associated peptide (TCAP)-1 attenuates corticotropin-releasing factor (CRF)-induced c-Fos expression in the limbic system and modulates anxiety behavior in male Wistar rats. Behav Brain Res 2009; 201:198-206. [DOI: 10.1016/j.bbr.2009.02.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Revised: 02/04/2009] [Accepted: 02/10/2009] [Indexed: 11/28/2022]
|
30
|
Hwang BH, Chang HM, Gu ZH, Suzuki R. c-fos gene expression is increased in the paraventricular hypothalamic nucleus of Sprague-Dawley rats with visceral pain induced by acetic acid without detectable changes of corticotrophin-releasing factor mRNA: a quantitative approach with an image analysis system. Anat Rec (Hoboken) 2007; 290:406-13. [PMID: 17514764 DOI: 10.1002/ar.20495] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This study is the first of its kind to demonstrate that c-Fos immunoreactivity (ir) together with c-fos mRNA in their immediately adjacent tissue sections of a discrete brain region can be reliably measured. The c-fos gene expression in the paraventricular hypothalamic nucleus (PVN) of Sprague-Dawley rats for an animal model for visceral or somatovisceral pain induced by 2% acetic acid (AA) was used in this study. Specifically, c-fos mRNA signals were measured by quantitative autoradiography after in situ hybridization using c-fos oligodeoxynucleotide probe, and c-Fos-ir signals were represented by c-Fos immunostaining, as detected using c-Fos antibody in a regular immunohistochemistry. Signals from both c-Fos-ir and c-fos mRNA in the PVN were measured from their immediately adjacent cryostat sections. For the measurement of c-Fos-ir, it was carried out by reading 10 rectangles (1,000 microm(2)/rectangle) on each PVN section with c-Fos immunostaining. Specific signals were obtained from subtracting the nonspecific background signal from the total signals using a computer-assisted image analysis system. Results indicated that the AA treatment induced a significant increase of both c-Fos-ir and c-fos mRNA in the PVN. Interestingly, there was no increase of corticotrophin-releasing factor (CRF) mRNA expression in the PVN and central nucleus of the amygdala of Sprague-Dawley rats subjected to the AA treatment. In summary, this study has demonstrated that c-Fos-ir in the PVN with an anatomical resolution can be semiquantitatively measured after immunohistochemistry using an image analysis system, and that increased c-fos mRNA in the PVN 1 hr after the AA treatment is associated with no changes of the CRF mRNA expression.
Collapse
Affiliation(s)
- Bang H Hwang
- Department of Anatomy and Cell Biology, School of Medicine, Indiana University, Indianapolis, Indiana 46202, USA.
| | | | | | | |
Collapse
|
31
|
Linden AM, Baez M, Bergeron M, Schoepp DD. Effects of mGlu2 or mGlu3 receptor deletions on mGlu2/3 receptor agonist (LY354740)-induced brain c-Fos expression: Specific roles for mGlu2 in the amygdala and subcortical nuclei, and mGlu3 in the hippocampus. Neuropharmacology 2006; 51:213-28. [PMID: 16733060 DOI: 10.1016/j.neuropharm.2006.03.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2006] [Revised: 03/08/2006] [Accepted: 03/10/2006] [Indexed: 11/29/2022]
Abstract
LY354740 is a potent and selective mGlu2/3 receptor agonist with activity in models of psychiatric disorders (anxiety, psychosis), and early clinical studies in anxiety patients. However, the specific receptor subtypes and brain regions which mediate mGlu2/3 receptor agonist pharmacology/efficacy are not well understood. Here we investigate the effects of deleting mGlu2 or mGlu3 receptors on basal and LY354740-regulated c-Fos expression in mouse brain using mGlu2 or mGlu3 knockout mice. Consistent with our earlier findings, LY354740 administration (20 mg/kg, i.p.) to wild-type mice increased c-Fos expression in specific limbic (central amygdala, bed nucleus of the stria terminalis, midline thalamic nuclei) and non-limbic (thalamic dorsolateral geniculate nucleus, superior colliculus, Edinger-Westphal) structures, while modestly suppressing hippocampal c-Fos expression. The LY354740-induced increases in c-Fos expression in all the above regions were abolished by mGlu2, but not mGlu3, receptor deletion. Interestingly, basal c-Fos expression was significantly increased in the hippocampus of mGlu3, but not mGlu2, receptor knockouts compared to wild-type mice. Moreover, this increase was not suppressed by LY354740, such that in the CA3 region LY354740 now increased c-Fos expression in the mGlu3 knockouts. These results demonstrate that the LY354740-induced increases of c-Fos expression in specific brain regions, including the central and extended amygdala are specifically linked to mGlu2 receptors, and LY354740 suppressions of neuronal activity in the hippocampus are linked to mGlu3 receptors.
Collapse
Affiliation(s)
- Anni-Maija Linden
- Neuroscience Discovery, Lilly Research Laboratories, Central Nervous System Research, Eli Lilly and Company, Lilly Corporate Center, drop code 0510, Indianapolis, IN 46285, USA
| | | | | | | |
Collapse
|
32
|
Rasmussen K, Benvenga MJ, Bymaster FP, Calligaro DO, Cohen IR, Falcone JF, Hemrick-Luecke SK, Martin FM, Moore NA, Nisenbaum LK, Schaus JM, Sundquist SJ, Tupper DE, Wiernicki TR, Nelson DL. Preclinical Pharmacology of FMPD [6-Fluoro-10-[3-(2-methoxyethyl)-4-methyl-piperazin-1-yl]-2-methyl-4H-3-thia-4,9-diaza-benzo[f]azulene]: A Potential Novel Antipsychotic with Lower Histamine H1 Receptor Affinity Than Olanzapine. J Pharmacol Exp Ther 2005; 315:1265-77. [PMID: 16141369 DOI: 10.1124/jpet.105.089326] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
FMPD [6-fluoro-10-[3-(2-methoxyethyl)-4-methyl-piperazin-1-yl]-2-methyl-4H-3-thia-4,9-diaza-benzo[f]azulene] is a potential novel antipsychotic with high affinity for dopamine D2 (Ki= 6.3 nM), 5-HT(2A) (Ki= 7.3 nM), and 5-HT6 (Ki= 8.0 nM) human recombinant receptors and lower affinity for histamine H1 (Ki= 30 nM) and 5-HT2C (Ki= 102 nM) human recombinant receptors than olanzapine. Oral administration of FMPD increased rat nucleus accumbens 3,4-dihyroxyphenylacetic acid concentrations (ED200 = 6 mg/kg), blocked 5-HT2A agonist-induced increases in rat serum corticosterone levels (ED50= 1.8 mg/kg), and inhibited the ex vivo binding of [125I]SB-258585 [4-iodo-N-[4-methoxy-3-(4-methyl-piperazin-1-yl)-phenyl]-benzenesulfonamide] to striatal 5-HT6 receptors (ED50= 10 mg/kg) but failed to inhibit ex vivo binding of [3H]pyrilamine to hypothalamic histamine H1 receptors at doses of up to 30 mg/kg. In electrophysiology studies, acute administration of FMPD selectively elevated the number of spontaneously active A10 (versus A9) dopamine neurons and chronic administration selectively decreased the number of spontaneously active A10 (versus A9) dopamine neurons. FMPD did not produce catalepsy at doses lower than 25 mg/kg p.o. In Fos-induction studies, FMPD had an atypical antipsychotic profile in the striatum and nucleus accumbens and increased Fos expression in orexin-containing neurons of the hypothalamus. FMPD produced only a transient elevation of prolactin levels. These data indicate that FMPD is an orally available potent antagonist of dopamine D2, 5-HT2A, and 5-HT6 receptors and a weak antagonist of H1 and 5-HT2C receptors. FMPD has the potential to have efficacy in treating schizophrenia and bipolar mania with a low risk of treatment-emergent extrapyramidal symptoms, prolactin elevation, and weight gain. Clinical trials are needed to test these hypotheses.
Collapse
Affiliation(s)
- Kurt Rasmussen
- Lilly Research Laboratories, Eli Lilly and Co., Lilly Corporate Center, Indianapolis, IN 46285, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Rensing N, Ouyang Y, Yang XF, Yamada KA, Rothman SM, Wong M. In vivo imaging of dendritic spines during electrographic seizures. Ann Neurol 2005; 58:888-98. [PMID: 16240365 DOI: 10.1002/ana.20658] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Epilepsy is associated with significant neurological morbidity, including learning disabilities, motor deficits, and behavioral problems. Although the causes of neurological dysfunction in epilepsy are multifactorial, accumulating evidence indicates that seizures in themselves may directly cause brain injury. Although it is clear that seizures can result in neuronal death, it is likely that under some circumstances seizures can induce more subtle functional or structural alterations in neurons. We induced focal neocortical seizures with 4-aminopyridine in transgenic mice expressing green fluorescent protein in cortical neurons and sequentially imaged individual dendrites in living animals with two-photon laser-scanning microscopy to determine whether these seizures caused acute alterations in dendritic spine morphology. No dendritic alterations were observed in anesthetized animals during electrographic seizures over a 3-hour period. Similarly, in unanesthetized mice, low-stage, clinical electrographic seizures had minimal effect on dendritic spines. More severe, high-stage seizures in unanesthetized mice were associated with a moderate loss of spines and dendritic swelling, but this effect may have been contingent on a synergistic action of phototoxicity from the imaging method itself. Overall, our results suggest that most neocortical seizures have minimal acute effects on dendrites over several hours, but may predispose to dendritic injury under extreme conditions.
Collapse
Affiliation(s)
- Nicholas Rensing
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|