1
|
Cieslak PE, Drabik S, Gugula A, Trenk A, Gorkowska M, Przybylska K, Szumiec L, Kreiner G, Rodriguez Parkitna J, Blasiak A. Dopamine Receptor-Expressing Neurons Are Differently Distributed throughout Layers of the Motor Cortex to Control Dexterity. eNeuro 2024; 11:ENEURO.0490-23.2023. [PMID: 38423792 DOI: 10.1523/eneuro.0490-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 03/02/2024] Open
Abstract
The motor cortex comprises the primary descending circuits for flexible control of voluntary movements and is critically involved in motor skill learning. Motor skill learning is impaired in patients with Parkinson's disease, but the precise mechanisms of motor control and skill learning are still not well understood. Here we have used transgenic mice, electrophysiology, in situ hybridization, and neural tract-tracing methods to target genetically defined cell types expressing D1 and D2 dopamine receptors in the motor cortex. We observed that putative D1 and D2 dopamine receptor-expressing neurons (D1+ and D2+, respectively) are organized in highly segregated, nonoverlapping populations. Moreover, based on ex vivo patch-clamp recordings, we showed that D1+ and D2+ cells have distinct morphological and electrophysiological properties. Finally, we observed that chemogenetic inhibition of D2+, but not D1+, neurons disrupts skilled forelimb reaching in adult mice. Overall, these results demonstrate that dopamine receptor-expressing cells in the motor cortex are highly segregated and play a specialized role in manual dexterity.
Collapse
Affiliation(s)
- Przemyslaw E Cieslak
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow 30-387, Poland
| | - Sylwia Drabik
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow 30-387, Poland
| | - Anna Gugula
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow 30-387, Poland
| | - Aleksandra Trenk
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow 30-387, Poland
| | - Martyna Gorkowska
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow 30-387, Poland
| | - Kinga Przybylska
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow 30-387, Poland
| | - Lukasz Szumiec
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow 31-343, Poland
| | - Grzegorz Kreiner
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow 31-343, Poland
| | - Jan Rodriguez Parkitna
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow 31-343, Poland
| | - Anna Blasiak
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow 30-387, Poland
| |
Collapse
|
2
|
Brown JA, Petersen N, Centanni SW, Jin AY, Yoon HJ, Cajigas SA, Bedenbaugh MN, Luchsinger JR, Patel S, Calipari ES, Simerly RB, Winder DG. An ensemble recruited by α 2a-adrenergic receptors is engaged in a stressor-specific manner in mice. Neuropsychopharmacology 2023; 48:1133-1143. [PMID: 36085168 PMCID: PMC10267140 DOI: 10.1038/s41386-022-01442-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/08/2022]
Abstract
α2a-adrenergic receptor (α2a-AR) agonists are candidate substance use disorder therapeutics due to their ability to recruit noradrenergic autoreceptors to dampen stress system engagement. However, we recently found that postsynaptic α2a-ARs are required for stress-induced reinstatement of cocaine-conditioned behavior. Understanding the ensembles recruited by these postsynaptic receptors (heteroceptors) is necessary to understand noradrenergic circuit control. We utilized a variety of approaches in FosTRAP (Targeted Recombination in Active Populations) mice to define an ensemble of cells activated by the α2a-AR partial agonist guanfacine ("Guansembles") in the bed nucleus of the stria terminalis (BST/BNST), a region key to stress-induced reinstatement of drug seeking. We define BNST "Guansembles" and show they differ from restraint stress-activated cells. Guanfacine produced inhibition of cAMP-dependent signaling in Guansembles, while chronic restraint stress increased cAMP-dependent signaling. Guanfacine both excited and inhibited aspects of Guansemble neuronal activity. Further, while some stressors produced overall reductions in Guansemble activity, active coping events during restraint stress and exposure to unexpected shocks were both associated with Guansemble recruitment. Using viral tracing, we define a BNST Guansemble afferent network that includes regions involved in the interplay of stress and homeostatic functions. Finally, we show that activation of Guansembles produces alterations in behavior on the elevated plus maze consistent with task-specific anxiety-like behavior. Overall, we define a population of BNST neurons recruited by α2a-AR signaling that opposes the behavioral action of canonical autoreceptor α2a-AR populations and which are differentially recruited by distinct stressors. Moreover, we demonstrate stressor-specific physiological responses in a specific neuronal population.
Collapse
Affiliation(s)
- Jordan A Brown
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Nicholas Petersen
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Samuel W Centanni
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Allie Y Jin
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Hye Jean Yoon
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Stephanie A Cajigas
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Michelle N Bedenbaugh
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Joseph R Luchsinger
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Sachin Patel
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Richard B Simerly
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Danny G Winder
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
3
|
SWI/SNF chromatin remodeler complex within the reward pathway is required for behavioral adaptations to stress. Nat Commun 2022; 13:1807. [PMID: 35379786 PMCID: PMC8980038 DOI: 10.1038/s41467-022-29380-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 02/22/2022] [Indexed: 01/01/2023] Open
Abstract
Enduring behavioral changes upon stress exposure involve changes in gene expression sustained by epigenetic modifications in brain circuits, including the mesocorticolimbic pathway. Brahma (BRM) and Brahma Related Gene 1 (BRG1) are ATPase subunits of the SWI/SNF complexes involved in chromatin remodeling, a process essential to enduring plastic changes in gene expression. Here, we show that in mice, social defeat induces changes in BRG1 nuclear distribution. The inactivation of the Brg1/Smarca4 gene within dopamine-innervated regions or the constitutive inactivation of the Brm/Smarca2 gene leads to resilience to repeated social defeat and decreases the behavioral responses to cocaine without impacting midbrain dopamine neurons activity. Within striatal medium spiny neurons, Brg1 gene inactivation reduces the expression of stress- and cocaine-induced immediate early genes, increases levels of heterochromatin and at a global scale decreases chromatin accessibility. Altogether these data demonstrate the pivotal function of SWI/SNF complexes in behavioral and transcriptional adaptations to salient environmental challenges. Repeated exposure to social stressors in rodents results in behavioural changes. Here the authors show that behavioural adaptations to stress are associated with nuclear organization changes through SWI/SNF chromatin remodeler in specific neuronal populations of the mesolimbic system.
Collapse
|
4
|
Local accumbens in vivo imaging during deep brain stimulation reveals a strategy-dependent amelioration of hedonic feeding. Proc Natl Acad Sci U S A 2022; 119:2109269118. [PMID: 34921100 PMCID: PMC8740575 DOI: 10.1073/pnas.2109269118] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 11/18/2022] Open
Abstract
Impulsive overeating is a common, disabling feature of eating disorders. Calcium imaging using fiber photometry has emerged as an in vivo methodology to measure neuronal population activity immune to electrical stimulation artifact from deep brain stimulation (DBS). Thus, when used simultaneously, calcium imaging can elucidate poorly understood DBS mechanisms. We show that nucleus accumbens D1 medial spiny calcium signaling increases in preparation of hedonic feeding of high-fat food. Further, responsive, over continuous, DBS strategies effectively disrupt this activity leading to decreased consumption. Implementation of this methodology to better understand mechanisms of these and other forms of neuromodulation for various indications may help advance the field to identify novel therapeutic targets with applications extending beyond obesity. Impulsive overeating is a common, disabling feature of eating disorders. Both continuous deep brain stimulation (DBS) and responsive DBS, which limits current delivery to pathological brain states, have emerged as potential therapies. We used in vivo fiber photometry in wild-type, Drd1-cre, and A2a-cre mice to 1) assay subtype-specific medium spiny neuron (MSN) activity of the nucleus accumbens (NAc) during hedonic feeding of high-fat food, and 2) examine DBS strategy-specific effects on NAc activity. D1, but not D2, NAc GCaMP activity increased immediately prior to high-fat food approach. Responsive DBS triggered a GCaMP surge throughout the stimulation period and durably reduced high-fat intake. However, with continuous DBS, this surge decayed, and high-fat intake reemerged. Our results argue for a stimulation strategy-dependent modulation of D1 MSNs with a more sustained decrease in consumption with responsive DBS. This study illustrates the important role in vivo imaging can play in understanding effects of such novel therapies.
Collapse
|
5
|
Allichon MC, Ortiz V, Pousinha P, Andrianarivelo A, Petitbon A, Heck N, Trifilieff P, Barik J, Vanhoutte P. Cell-Type-Specific Adaptions in Striatal Medium-Sized Spiny Neurons and Their Roles in Behavioral Responses to Drugs of Abuse. Front Synaptic Neurosci 2022; 13:799274. [PMID: 34970134 PMCID: PMC8712310 DOI: 10.3389/fnsyn.2021.799274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022] Open
Abstract
Drug addiction is defined as a compulsive pattern of drug-seeking- and taking- behavior, with recurrent episodes of abstinence and relapse, and a loss of control despite negative consequences. Addictive drugs promote reinforcement by increasing dopamine in the mesocorticolimbic system, which alters excitatory glutamate transmission within the reward circuitry, thereby hijacking reward processing. Within the reward circuitry, the striatum is a key target structure of drugs of abuse since it is at the crossroad of converging glutamate inputs from limbic, thalamic and cortical regions, encoding components of drug-associated stimuli and environment, and dopamine that mediates reward prediction error and incentive values. These signals are integrated by medium-sized spiny neurons (MSN), which receive glutamate and dopamine axons converging onto their dendritic spines. MSN primarily form two mostly distinct populations based on the expression of either DA-D1 (D1R) or DA-D2 (D2R) receptors. While a classical view is that the two MSN populations act in parallel, playing antagonistic functional roles, the picture seems much more complex. Herein, we review recent studies, based on the use of cell-type-specific manipulations, demonstrating that dopamine differentially modulates dendritic spine density and synapse formation, as well as glutamate transmission, at specific inputs projecting onto D1R-MSN and D2R-MSN to shape persistent pathological behavioral in response to drugs of abuse. We also discuss the identification of distinct molecular events underlying the detrimental interplay between dopamine and glutamate signaling in D1R-MSN and D2R-MSN and highlight the relevance of such cell-type-specific molecular studies for the development of innovative strategies with potential therapeutic value for addiction. Because drug addiction is highly prevalent in patients with other psychiatric disorders when compared to the general population, we last discuss the hypothesis that shared cellular and molecular adaptations within common circuits could explain the co-occurrence of addiction and depression. We will therefore conclude this review by examining how the nucleus accumbens (NAc) could constitute a key interface between addiction and depression.
Collapse
Affiliation(s)
- Marie-Charlotte Allichon
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Vanesa Ortiz
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Paula Pousinha
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Andry Andrianarivelo
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Anna Petitbon
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Nicolas Heck
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | - Pierre Trifilieff
- Université Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Jacques Barik
- Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
| | - Peter Vanhoutte
- CNRS, UMR 8246, Neuroscience Paris Seine, Paris, France.,INSERM, UMR-S 1130, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Université, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| |
Collapse
|
6
|
Reduced Dopamine Signaling Impacts Pyramidal Neuron Excitability in Mouse Motor Cortex. eNeuro 2021; 8:ENEURO.0548-19.2021. [PMID: 34556558 PMCID: PMC8525657 DOI: 10.1523/eneuro.0548-19.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 08/12/2021] [Accepted: 09/07/2021] [Indexed: 11/21/2022] Open
Abstract
Dopaminergic modulation is essential for the control of voluntary movement; however, the role of dopamine in regulating the neural excitability of the primary motor cortex (M1) is not well understood. Here, we investigated two modes by which dopamine influences the input/output function of M1 neurons. To test the direct regulation of M1 neurons by dopamine, we performed whole-cell recordings of excitatory neurons and measured excitability before and after local, acute dopamine receptor blockade. We then determined whether chronic depletion of dopaminergic input to the entire motor circuit, via a mouse model of Parkinson's disease, was sufficient to shift M1 neuron excitability. We show that D1 receptor (D1R) and D2R antagonism altered subthreshold and suprathreshold properties of M1 pyramidal neurons in a layer-specific fashion. The effects of D1R antagonism were primarily driven by changes to intrinsic properties, while the excitability shifts following D2R antagonism relied on synaptic transmission. In contrast, chronic depletion of dopamine to the motor circuit with 6-hydroxydopamine induced layer-specific synaptic transmission-dependent shifts in M1 neuron excitability that only partially overlapped with the effects of acute D1R antagonism. These results suggest that while acute and chronic changes in dopamine modulate the input/output function of M1 neurons, the mechanisms engaged are distinct depending on the duration and origin of the manipulation. Our study highlights the broad influence of dopamine on M1 excitability by demonstrating the consequences of local and global dopamine depletion on neuronal input/output function.
Collapse
|
7
|
Crittenden JR, Zhai S, Sauvage M, Kitsukawa T, Burguière E, Thomsen M, Zhang H, Costa C, Martella G, Ghiglieri V, Picconi B, Pescatore KA, Unterwald EM, Jackson WS, Housman DE, Caine SB, Sulzer D, Calabresi P, Smith AC, Surmeier DJ, Graybiel AM. CalDAG-GEFI mediates striatal cholinergic modulation of dendritic excitability, synaptic plasticity and psychomotor behaviors. Neurobiol Dis 2021; 158:105473. [PMID: 34371144 PMCID: PMC8486000 DOI: 10.1016/j.nbd.2021.105473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 01/19/2023] Open
Abstract
CalDAG-GEFI (CDGI) is a protein highly enriched in the striatum, particularly in the principal spiny projection neurons (SPNs). CDGI is strongly down-regulated in two hyperkinetic conditions related to striatal dysfunction: Huntington’s disease and levodopa-induced dyskinesia in Parkinson’s disease. We demonstrate that genetic deletion of CDGI in mice disrupts dendritic, but not somatic, M1 muscarinic receptors (M1Rs) signaling in indirect pathway SPNs. Loss of CDGI reduced temporal integration of excitatory postsynaptic potentials at dendritic glutamatergic synapses and impaired the induction of activity-dependent long-term potentiation. CDGI deletion selectively increased psychostimulant-induced repetitive behaviors, disrupted sequence learning, and eliminated M1R blockade of cocaine self-administration. These findings place CDGI as a major, but previously unrecognized, mediator of cholinergic signaling in the striatum. The effects of CDGI deletion on the self-administration of drugs of abuse and its marked alterations in hyperkinetic extrapyramidal disorders highlight CDGI’s therapeutic potential.
Collapse
Affiliation(s)
- Jill R Crittenden
- McGovern Institute for Brain Research and Dept. of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Shenyu Zhai
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Magdalena Sauvage
- McGovern Institute for Brain Research and Dept. of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Leibniz Institute for Neurobiology, Functional Architecture of Memory Dept., Magdeburg, Germany
| | - Takashi Kitsukawa
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Eric Burguière
- McGovern Institute for Brain Research and Dept. of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Brain and Spine Institute (ICM), CNRS UMR 7225, INSERM U 1127, UPMC-P6 UMR S, 1127, Hôpital de la Pitié-Salpêtrière, 47 boulevard de l'hôpital, Paris, France
| | - Morgane Thomsen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen and University, DK-2100, Copenhagen, Denmark; Basic Neuroscience Division, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Hui Zhang
- Departments of Psychiatry, Pharmacology, Neurology, Columbia University, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Cinzia Costa
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della misericordia, University of Perugia, 06100 Perugia, Italy
| | - Giuseppina Martella
- Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | | | | | - Karen A Pescatore
- Department of Pharmacology and Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Ellen M Unterwald
- Department of Pharmacology and Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Walker S Jackson
- Wallenberg Center for Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - David E Housman
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - S Barak Caine
- Basic Neuroscience Division, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - David Sulzer
- Departments of Psychiatry, Pharmacology, Neurology, Columbia University, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Paolo Calabresi
- Neurological Clinic, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; Department of Neuroscience, Faculty of Medicine, Università Cattolica del "Sacro Cuore", 00168 Rome, Italy
| | - Anne C Smith
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85724, USA
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ann M Graybiel
- McGovern Institute for Brain Research and Dept. of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
8
|
Ganley RP, Werder K, Wildner H, Zeilhofer HU. Spinally projecting noradrenergic neurons of the locus coeruleus display resistance to AAV2retro-mediated transduction. Mol Pain 2021; 17:17448069211037887. [PMID: 34344259 PMCID: PMC8351027 DOI: 10.1177/17448069211037887] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The locus coeruleus (LC) is the principal source of noradrenaline (NA) in the central nervous system. Projection neurons in the ventral portion of the LC project to the spinal cord and are considered the main source of spinal NA. To understand the precise physiology of this pathway, it is important to have tools that allow specific genetic access to these descending projections. AAV2retro serotype vectors are a potential tool to transduce these neurons via their axon terminals in the spinal cord, and thereby limit the expression of genetic material to the spinal projections from the LC. Here, we assess the suitability of AAV2retro to target these neurons and investigate strategies to increase their labelling efficiency. RESULTS We show that the neurons in the LC that project to the spinal dorsal horn are largely resistant to transduction with AAV2retro serotype vectors. Compared to Cholera toxin B (CTb) tracing, AAV2retro.eGFP labelled far fewer neurons within the LC and surrounding regions, particularly within neurons that express tyrosine hydroxylase (TH), the rate-limiting enzyme for NA synthesis. We also show that the sensitivity for transduction of this projection can be increased using AAV2retro.eGFP.cre in ROSA26tdTom reporter mice (23% increase), with a higher proportion of the newly revealed neurons expressing TH compared to those directly labelled with AAV2retro containing an eGFP expression sequence. CONCLUSION These tracing studies identify limitations in AAV2retro-mediated retrograde transduction of a subset of projection neurons, specifically those that express NA and project to the spinal cord. This is likely to have implications for the study of NA-containing projections as well as other types of projection neuron in the central nervous system.
Collapse
Affiliation(s)
- Robert P Ganley
- Institute for Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Kira Werder
- Institute for Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Hendrik Wildner
- Institute for Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Hanns Ulrich Zeilhofer
- Institute for Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland.,Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland.,Neuroscience Center Zurich, Zürich, Switzerland
| |
Collapse
|
9
|
Oliveira da Cruz JF, Busquets-Garcia A, Zhao Z, Varilh M, Lavanco G, Bellocchio L, Robin L, Cannich A, Julio-Kalajzić F, Lesté-Lasserre T, Maître M, Drago F, Marsicano G, Soria-Gómez E. Specific Hippocampal Interneurons Shape Consolidation of Recognition Memory. Cell Rep 2021; 32:108046. [PMID: 32814049 PMCID: PMC7443618 DOI: 10.1016/j.celrep.2020.108046] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 06/15/2020] [Accepted: 07/27/2020] [Indexed: 12/27/2022] Open
Abstract
A complex array of inhibitory interneurons tightly controls hippocampal activity, but how such diversity specifically affects memory processes is not well understood. We find that a small subclass of type 1 cannabinoid receptor (CB1R)-expressing hippocampal interneurons determines episodic-like memory consolidation by linking dopamine D1 receptor (D1R) signaling to GABAergic transmission. Mice lacking CB1Rs in D1-positive cells (D1-CB1-KO) display impairment in long-term, but not short-term, novel object recognition memory (NOR). Re-expression of CB1Rs in hippocampal D1R-positive cells rescues this NOR deficit. Learning induces an enhancement of in vivo hippocampal long-term potentiation (LTP), which is absent in mutant mice. CB1R-mediated NOR and the associated LTP facilitation involve local control of GABAergic inhibition in a D1-dependent manner. This study reveals that hippocampal CB1R-/D1R-expressing interneurons control NOR memory, identifying a mechanism linking the diversity of hippocampal interneurons to specific behavioral outcomes. CB1Rs are present in hippocampal D1R-positive interneurons CB1R/D1R-positive interneurons control the late phase of recognition memory CB1R/D1R-positive interneurons control learning-induced facilitation of LTP
Collapse
Affiliation(s)
- Jose F Oliveira da Cruz
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France; New York University, Center for Neural Science, New York, NY 10003, USA
| | - Arnau Busquets-Garcia
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France; Integrative Pharmacology and System Neuroscience, IMIM-Hospital del Mar Medical Research Institute, Barcelona 08003, Spain
| | - Zhe Zhao
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France
| | - Marjorie Varilh
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France
| | - Gianluca Lavanco
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France; Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania 95124, Italy
| | - Luigi Bellocchio
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France
| | - Laurie Robin
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France
| | - Astrid Cannich
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France
| | - Francisca Julio-Kalajzić
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France
| | - Thierry Lesté-Lasserre
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France
| | - Marlène Maître
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France
| | - Filippo Drago
- Integrative Pharmacology and System Neuroscience, IMIM-Hospital del Mar Medical Research Institute, Barcelona 08003, Spain
| | - Giovanni Marsicano
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France.
| | - Edgar Soria-Gómez
- INSERM U1215, NeuroCentre Magendie, Bordeaux 33300, France; University of Bordeaux, Bordeaux 33300, France; Ikerbasque-Basque Foundation for Science, Bilbao 48013, Spain; Department of Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU) Leioa 48940, Spain; Achucarro Basque Center for Neuroscience, Leioa 48940, Spain.
| |
Collapse
|
10
|
Ibrayeva A, Bay M, Pu E, Jörg DJ, Peng L, Jun H, Zhang N, Aaron D, Lin C, Resler G, Hidalgo A, Jang MH, Simons BD, Bonaguidi MA. Early stem cell aging in the mature brain. Cell Stem Cell 2021; 28:955-966.e7. [PMID: 33848469 PMCID: PMC10069280 DOI: 10.1016/j.stem.2021.03.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/19/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
Stem cell dysfunction drives many age-related disorders. Identifying mechanisms that initially compromise stem cell behavior represent early targets to promote tissue function later in life. Here, we pinpoint multiple factors that disrupt neural stem cell (NSC) behavior in the adult hippocampus. Clonal tracing showed that NSCs exhibit asynchronous depletion by identifying short-term NSCs (ST-NSCs) and long-term NSCs (LT-NSCs). ST-NSCs divide rapidly to generate neurons and deplete in the young brain. Meanwhile, multipotent LT-NSCs are maintained for months but are pushed out of homeostasis by lengthening quiescence. Single-cell transcriptome analysis of deep NSC quiescence revealed several hallmarks of molecular aging in the mature brain and identified tyrosine-protein kinase Abl1 as an NSC aging factor. Treatment with the Abl inhibitor imatinib increased NSC activation without impairing NSC maintenance in the middle-aged brain. Our study indicates that hippocampal NSCs are particularly vulnerable and adaptable to cellular aging.
Collapse
Affiliation(s)
- Albina Ibrayeva
- Eli and Edythe Broad Center for Regenerative Medicine & Stem Cell Research at USC, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA; USC Davis School - Buck Institute Graduate Program in the Biology of Aging, University of Southern California, Los Angeles, CA 90033, USA
| | - Maxwell Bay
- Eli and Edythe Broad Center for Regenerative Medicine & Stem Cell Research at USC, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA; Neuroscience Graduate Program, W. M. Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Elbert Pu
- Eli and Edythe Broad Center for Regenerative Medicine & Stem Cell Research at USC, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - David J Jörg
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK; Gurdon Institute, University of Cambridge, Cambridge CB3 0HE, UK
| | - Lei Peng
- Eli and Edythe Broad Center for Regenerative Medicine & Stem Cell Research at USC, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA; Neuroscience Graduate Program, W. M. Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Heechul Jun
- Department of Neurological Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Naibo Zhang
- Eli and Edythe Broad Center for Regenerative Medicine & Stem Cell Research at USC, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Daniel Aaron
- Eli and Edythe Broad Center for Regenerative Medicine & Stem Cell Research at USC, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Congrui Lin
- Eli and Edythe Broad Center for Regenerative Medicine & Stem Cell Research at USC, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Galen Resler
- Eli and Edythe Broad Center for Regenerative Medicine & Stem Cell Research at USC, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Axel Hidalgo
- Eli and Edythe Broad Center for Regenerative Medicine & Stem Cell Research at USC, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Mi-Hyeon Jang
- Department of Neurological Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Benjamin D Simons
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK; Gurdon Institute, University of Cambridge, Cambridge CB3 0HE, UK
| | - Michael A Bonaguidi
- Eli and Edythe Broad Center for Regenerative Medicine & Stem Cell Research at USC, University of Southern California, Los Angeles, CA 90033, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA; USC Davis School - Buck Institute Graduate Program in the Biology of Aging, University of Southern California, Los Angeles, CA 90033, USA; Neuroscience Graduate Program, W. M. Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA; Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90033, USA; Davis School of Gerontology, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
11
|
Soria-Gomez E, Pagano Zottola AC, Mariani Y, Desprez T, Barresi M, Bonilla-del Río I, Muguruza C, Le Bon-Jego M, Julio-Kalajzić F, Flynn R, Terral G, Fernández-Moncada I, Robin LM, Oliveira da Cruz JF, Corinti S, Amer YO, Goncalves J, Varilh M, Cannich A, Redon B, Zhao Z, Lesté-Lasserre T, Vincent P, Tolentino-Cortes T, Busquets-García A, Puente N, Bains JS, Hebert-Chatelain E, Barreda-Gómez G, Chaouloff F, Lohman AW, Callado LF, Grandes P, Baufreton J, Marsicano G, Bellocchio L. Subcellular specificity of cannabinoid effects in striatonigral circuits. Neuron 2021; 109:1513-1526.e11. [DOI: 10.1016/j.neuron.2021.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/09/2021] [Accepted: 03/04/2021] [Indexed: 12/14/2022]
|
12
|
Blázquez C, Ruiz-Calvo A, Bajo-Grañeras R, Baufreton JM, Resel E, Varilh M, Pagano Zottola AC, Mariani Y, Cannich A, Rodríguez-Navarro JA, Marsicano G, Galve-Roperh I, Bellocchio L, Guzmán M. Inhibition of striatonigral autophagy as a link between cannabinoid intoxication and impairment of motor coordination. eLife 2020; 9:56811. [PMID: 32773031 PMCID: PMC7417168 DOI: 10.7554/elife.56811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022] Open
Abstract
The use of cannabis is rapidly expanding worldwide. Thus, innovative studies aimed to identify, understand and potentially reduce cannabis-evoked harms are warranted. Here, we found that Δ9-tetrahydrocannabinol, the psychoactive ingredient of cannabis, disrupts autophagy selectively in the striatum, a brain area that controls motor behavior, both in vitro and in vivo. Boosting autophagy, either pharmacologically (with temsirolimus) or by dietary intervention (with trehalose), rescued the Δ9-tetrahydrocannabinol-induced impairment of motor coordination in mice. The combination of conditional knockout mouse models and viral vector-mediated autophagy-modulating strategies in vivo showed that cannabinoid CB1 receptors located on neurons belonging to the direct (striatonigral) pathway are required for the motor-impairing activity of Δ9-tetrahydrocannabinol by inhibiting local autophagy. Taken together, these findings identify inhibition of autophagy as an unprecedented mechanistic link between cannabinoids and motor performance, and suggest that activators of autophagy might be considered as potential therapeutic tools to treat specific cannabinoid-evoked behavioral alterations.
Collapse
Affiliation(s)
- Cristina Blázquez
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Universitario de Investigación Neuroquímica (IUIN) and Department of Biochemistry and Molecular Biology, Complutense University, Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Andrea Ruiz-Calvo
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Universitario de Investigación Neuroquímica (IUIN) and Department of Biochemistry and Molecular Biology, Complutense University, Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Raquel Bajo-Grañeras
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Universitario de Investigación Neuroquímica (IUIN) and Department of Biochemistry and Molecular Biology, Complutense University, Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Jérôme M Baufreton
- Centre National de la Recherche Scientifique (CNRS) and University of Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Eva Resel
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Universitario de Investigación Neuroquímica (IUIN) and Department of Biochemistry and Molecular Biology, Complutense University, Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Marjorie Varilh
- Institut National de la Santé et de la Recherche Médicale (INSERM) and University of Bordeaux, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, Bordeaux, France
| | - Antonio C Pagano Zottola
- Institut National de la Santé et de la Recherche Médicale (INSERM) and University of Bordeaux, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, Bordeaux, France
| | - Yamuna Mariani
- Institut National de la Santé et de la Recherche Médicale (INSERM) and University of Bordeaux, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, Bordeaux, France
| | - Astrid Cannich
- Institut National de la Santé et de la Recherche Médicale (INSERM) and University of Bordeaux, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, Bordeaux, France
| | | | - Giovanni Marsicano
- Institut National de la Santé et de la Recherche Médicale (INSERM) and University of Bordeaux, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, Bordeaux, France
| | - Ismael Galve-Roperh
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Universitario de Investigación Neuroquímica (IUIN) and Department of Biochemistry and Molecular Biology, Complutense University, Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Luigi Bellocchio
- Institut National de la Santé et de la Recherche Médicale (INSERM) and University of Bordeaux, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, Bordeaux, France
| | - Manuel Guzmán
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Universitario de Investigación Neuroquímica (IUIN) and Department of Biochemistry and Molecular Biology, Complutense University, Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| |
Collapse
|
13
|
Grauer SM, Sanoja R, Poulin D, Rashid H, Jochnowitz N, Calhoun M, Zwilling D, Varty GB, Rosahl TW, Meziane H, Mittlelhaeuser C, Mazzola R, Morrow J, Smith SM, Henze D, Marcus J. Antinociceptive effects of potent, selective and brain penetrant muscarinic M4 positive allosteric modulators in rodent pain models. Brain Res 2020; 1737:146814. [DOI: 10.1016/j.brainres.2020.146814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 03/06/2020] [Accepted: 03/27/2020] [Indexed: 11/27/2022]
|
14
|
Salery M, Trifilieff P, Caboche J, Vanhoutte P. From Signaling Molecules to Circuits and Behaviors: Cell-Type-Specific Adaptations to Psychostimulant Exposure in the Striatum. Biol Psychiatry 2020; 87:944-953. [PMID: 31928716 DOI: 10.1016/j.biopsych.2019.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
Addiction is characterized by a compulsive pattern of drug seeking and consumption and a high risk of relapse after withdrawal that are thought to result from persistent adaptations within brain reward circuits. Drugs of abuse increase dopamine (DA) concentration in these brain areas, including the striatum, which shapes an abnormal memory trace of drug consumption that virtually highjacks reward processing. Long-term neuronal adaptations of gamma-aminobutyric acidergic striatal projection neurons (SPNs) evoked by drugs of abuse are critical for the development of addiction. These neurons form two mostly segregated populations, depending on the DA receptor they express and their output projections, constituting the so-called direct (D1 receptor) and indirect (D2 receptor) SPN pathways. Both SPN subtypes receive converging glutamate inputs from limbic and cortical regions, encoding contextual and emotional information, together with DA, which mediates reward prediction and incentive values. DA differentially modulates the efficacy of glutamate synapses onto direct and indirect SPN pathways by recruiting distinct striatal signaling pathways, epigenetic and genetic responses likely involved in the transition from casual drug use to addiction. Herein we focus on recent studies that have assessed psychostimulant-induced alterations in a cell-type-specific manner, from remodeling of input projections to the characterization of specific molecular events in each SPN subtype and their impact on long-lasting behavioral adaptations. We discuss recent evidence revealing the complex and concerted action of both SPN populations on drug-induced behavioral responses, as these studies can contribute to the design of future strategies to alleviate specific behavioral components of addiction.
Collapse
Affiliation(s)
- Marine Salery
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pierre Trifilieff
- NutriNeuro, Unité Mixte de Recherche (UMR) 1286, Institut National de la Recherche Agronomique, Bordeaux Institut Polytechnique, University of Bordeaux, Bordeaux, France
| | - Jocelyne Caboche
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France.
| | - Peter Vanhoutte
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Sorbonne Université, Faculty of Sciences, Paris, France; Centre National de la Recherche Scientifique, UMR8246, Paris, France; Institut National de la Santé et de la Recherche Médicale, U1130, Paris France
| |
Collapse
|
15
|
Papilloud A, Weger M, Bacq A, Zalachoras I, Hollis F, Larrieu T, Battivelli D, Grosse J, Zanoletti O, Parnaudeau S, Tronche F, Sandi C. The glucocorticoid receptor in the nucleus accumbens plays a crucial role in social rank attainment in rodents. Psychoneuroendocrinology 2020; 112:104538. [PMID: 31841985 DOI: 10.1016/j.psyneuen.2019.104538] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/01/2019] [Accepted: 11/29/2019] [Indexed: 01/05/2023]
Abstract
Social hierarchy in social species is usually established through competitive encounters with conspecifics. It determines the access to limited resources and, thus, leads to reduced fights among individuals within a group. Despite the known importance of social rank for health and well-being, the knowledge about the processes underlying rank attainment remains limited. Previous studies have highlighted the nucleus accumbens (NAc) as a key brain region in the attainment of social hierarchies in rodents. In addition, glucocorticoids and the glucocorticoid receptor (GR) have been implicated in the establishment of social hierarchies and social aversion. However, whether GR in the NAc is involved in social dominance is not yet known. To address this question, we first established that expression levels of GR in the NAc of high anxious, submissive-prone rats are lower than that of their low anxious, dominant-prone counterparts. Furthermore, virally-induced downregulation of GR expression in the NAc in rats led to an improvement of social dominance rank. We found a similar result in a cell-specific mouse model lacking GR in dopaminoceptive neurons (i.e., neurons containing dopamine receptors). Indeed, when cohabitating in dyads of mixed genotypes, mice deficient for GR in dopaminoceptive neurons had a higher probability to become dominant than wild-type mice. Overall, our results highlight GR in the NAc and in dopaminoceptive neurons as an important regulator of social rank attainment.
Collapse
Affiliation(s)
- Aurélie Papilloud
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Meltem Weger
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Alexandre Bacq
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Ioannis Zalachoras
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Fiona Hollis
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Thomas Larrieu
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Dorian Battivelli
- Neuroscience Paris-Seine, Gene Regulation and Adaptive Behaviors Team, CNRS UMR8246 - INSERM U1130 - Sorbonne Université, Institut De Biologie Paris-Seine, Paris, France
| | - Jocelyn Grosse
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Olivia Zanoletti
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Sébastien Parnaudeau
- Neuroscience Paris-Seine, Gene Regulation and Adaptive Behaviors Team, CNRS UMR8246 - INSERM U1130 - Sorbonne Université, Institut De Biologie Paris-Seine, Paris, France
| | - François Tronche
- Neuroscience Paris-Seine, Gene Regulation and Adaptive Behaviors Team, CNRS UMR8246 - INSERM U1130 - Sorbonne Université, Institut De Biologie Paris-Seine, Paris, France
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland.
| |
Collapse
|
16
|
Mustafa R, Kreiner G, Kamińska K, Wood AEJ, Kirsch J, Tucker KL, Parlato R. Targeted Depletion of Primary Cilia in Dopaminoceptive Neurons in a Preclinical Mouse Model of Huntington's Disease. Front Cell Neurosci 2019; 13:565. [PMID: 31920562 PMCID: PMC6936315 DOI: 10.3389/fncel.2019.00565] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/05/2019] [Indexed: 12/22/2022] Open
Abstract
Multiple pathomechanisms triggered by mutant Huntingtin (mHTT) underlie progressive degeneration of dopaminoceptive striatal neurons in Huntington’s disease (HD). The primary cilium is a membrane compartment that functions as a hub for various pathways that are dysregulated in HD, for example, dopamine (DA) receptor transmission and the mechanistic target of rapamycin (mTOR) pathway. The roles of primary cilia (PC) for the maintenance of striatal neurons and in HD progression remain unknown. Here, we investigated PC defects in vulnerable striatal neurons in a progressive model of HD, the mHTT-expressing knock-in zQ175 mice. We found that PC length is affected in striatal but not in cortical neurons, in association with the accumulation of mHTT. To explore the role of PC, we generated conditional mutant mice lacking IFT88, a component of the anterograde intraflagellar transport-B complex lacking PC in dopaminoceptive neurons. This mutation preserved the expression of the dopamine 1 receptor (D1R), and the survival of striatal neurons, but resulted in a mild increase of DA metabolites in the striatum, suggesting an imbalance of ciliary DA receptor transmission. Conditional loss of PC in zQ175 mice did not trigger astrogliosis, however, mTOR signaling was more active and resulted in a more pronounced accumulation of nuclear inclusions containing mHTT. Further studies will be required of aged mice to determine the role of aberrant ciliary function in more advanced stages of HD.
Collapse
Affiliation(s)
- Rasem Mustafa
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,Institute of Anatomy and Cell Biology, Medical Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Grzegorz Kreiner
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Katarzyna Kamińska
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland.,Jagiellonian Center for Experimental Therapeutics, Jagiellonian University, Kraków, Poland
| | - Amelia-Elise J Wood
- Department of Biomedical Sciences, Center for Excellence in the Neurosciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, United States
| | - Joachim Kirsch
- Institute of Anatomy and Cell Biology, Medical Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Kerry L Tucker
- Department of Biomedical Sciences, Center for Excellence in the Neurosciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, United States
| | - Rosanna Parlato
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,Institute of Anatomy and Cell Biology, Medical Cell Biology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
17
|
Kreiner G, Sönmez A, Liss B, Parlato R. Integration of the Deacetylase SIRT1 in the Response to Nucleolar Stress: Metabolic Implications for Neurodegenerative Diseases. Front Mol Neurosci 2019; 12:106. [PMID: 31110473 PMCID: PMC6499230 DOI: 10.3389/fnmol.2019.00106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/09/2019] [Indexed: 01/10/2023] Open
Abstract
Understanding underlying mechanisms of neurodegenerative diseases is fundamental to develop effective therapeutic intervention. Yet they remain largely elusive, but metabolic, and transcriptional dysregulation are common events. Sirtuin 1 (SIRT1) is a nicotinamide adenine dinucleotide (NAD+)-dependent lysine deacetylase, regulating transcription, and critical for the cellular adaptations to metabolic stress. SIRT1 regulates the transcription of ribosomal RNA (rRNA), connecting the energetic state with cell growth and function. The activity of the transcription initiation factor-IA (TIF-IA) is important for the transcriptional regulation of ribosomal DNA (rDNA) genes in the nucleolus, and is also sensitive to changes in the cellular energetic state. Moreover, TIF-IA is responsive to nutrient-deprivation, neurotrophic stimulation, and oxidative stress. Hence, both SIRT1 and TIF-IA connect changes in cellular stress with transcriptional regulation and metabolic adaptation. Moreover, they finely tune the activity of the transcription factor p53, maintain mitochondrial function, and oxidative stress responses. Here we reviewed and discussed evidence that SIRT1 and TIF-IA are regulated by shared pathways and their activities preserve neuronal homeostasis in response to metabolic stressors. We provide evidence that loss of rDNA transcription due to altered TIF-IA function alters SIRT1 expression and propose a model of interdependent feedback mechanisms. An imbalance of this signaling might be a critical common event in neurodegenerative diseases. In conclusion, we provide a novel perspective for the prediction of the therapeutic benefits of the modulation of SIRT1- and nucleolar-dependent pathways in metabolic and neurodegenerative diseases.
Collapse
Affiliation(s)
- Grzegorz Kreiner
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Aynur Sönmez
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,New College, Oxford University, Oxford, United Kingdom
| | - Rosanna Parlato
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
18
|
Extinction of avoidance behavior by safety learning depends on endocannabinoid signaling in the hippocampus. J Psychiatr Res 2017; 90:46-59. [PMID: 28222356 DOI: 10.1016/j.jpsychires.2017.02.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/24/2017] [Accepted: 02/02/2017] [Indexed: 12/29/2022]
Abstract
The development of exaggerated avoidance behavior is largely responsible for the decreased quality of life in patients suffering from anxiety disorders. Studies using animal models have contributed to the understanding of the neural mechanisms underlying the acquisition of avoidance responses. However, much less is known about its extinction. Here we provide evidence in mice that learning about the safety of an environment (i.e., safety learning) rather than repeated execution of the avoided response in absence of negative consequences (i.e., response extinction) allowed the animals to overcome their avoidance behavior in a step-down avoidance task. This process was context-dependent and could be blocked by pharmacological (3 mg/kg, s.c.; SR141716) or genetic (lack of cannabinoid CB1 receptors in neurons expressing dopamine D1 receptors) inactivation of CB1 receptors. In turn, the endocannabinoid reuptake inhibitor AM404 (3 mg/kg, i.p.) facilitated safety learning in a CB1-dependent manner and attenuated the relapse of avoidance behavior 28 days after conditioning. Safety learning crucially depended on endocannabinoid signaling at level of the hippocampus, since intrahippocampal SR141716 treatment impaired, whereas AM404 facilitated safety learning. Other than AM404, treatment with diazepam (1 mg/kg, i.p.) impaired safety learning. Drug effects on behavior were directly mirrored by drug effects on evoked activity propagation through the hippocampal trisynaptic circuit in brain slices: As revealed by voltage-sensitive dye imaging, diazepam impaired whereas AM404 facilitated activity propagation to CA1 in a CB1-dependent manner. In line with this, systemic AM404 enhanced safety learning-induced expression of Egr1 at level of CA1. Together, our data render it likely that AM404 promotes safety learning by enhancing information flow through the trisynaptic circuit to CA1.
Collapse
|
19
|
Local and Long-Range Circuit Connections to Hilar Mossy Cells in the Dentate Gyrus. eNeuro 2017; 4:eN-NWR-0097-17. [PMID: 28451637 PMCID: PMC5396130 DOI: 10.1523/eneuro.0097-17.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 01/04/2023] Open
Abstract
Hilar mossy cells are the prominent glutamatergic cell type in the dentate hilus of the dentate gyrus (DG); they have been proposed to have critical roles in the DG network. To better understand how mossy cells contribute to DG function, we have applied new viral genetic and functional circuit mapping approaches to quantitatively map and compare local and long-range circuit connections of mossy cells and dentate granule cells in the mouse. The great majority of inputs to mossy cells consist of two parallel inputs from within the DG: an excitatory input pathway from dentate granule cells and an inhibitory input pathway from local DG inhibitory neurons. Mossy cells also receive a moderate degree of excitatory and inhibitory CA3 input from proximal CA3 subfields. Long range inputs to mossy cells are numerically sparse, and they are only identified readily from the medial septum and the septofimbrial nucleus. In comparison, dentate granule cells receive most of their inputs from the entorhinal cortex. The granule cells receive significant synaptic inputs from the hilus and the medial septum, and they also receive direct inputs from both distal and proximal CA3 subfields, which has been underdescribed in the existing literature. Our slice-based physiological mapping studies further supported the identified circuit connections of mossy cells and granule cells. Together, our data suggest that hilar mossy cells are major local circuit integrators and they exert modulation of the activity of dentate granule cells as well as the CA3 region through "back-projection" pathways.
Collapse
|
20
|
Bradley SJ, Tobin AB. Design of Next-Generation G Protein-Coupled Receptor Drugs: Linking Novel Pharmacology and In Vivo Animal Models. Annu Rev Pharmacol Toxicol 2016; 56:535-59. [PMID: 26738479 DOI: 10.1146/annurev-pharmtox-011613-140012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite the fact that G protein-coupled receptors (GPCRs) are the most successful drug targets in history, this supergene family of cell surface receptors has yet to be fully exploited as targets in the treatment of human disease. Here, we present optimism that this may change in the future by reviewing the substantial progress made in the understanding of GPCR molecular pharmacology that has generated an extensive toolbox of ligand types that include orthosteric, allosteric, and bitopic ligands, many of which show signaling bias. We discuss how combining these advances with recently described transgenic, chemical genetic, and optogenetic animal models will provide the framework to allow for the rational design of next-generation GPCR drugs that possess increased therapeutic efficacy and decreased adverse/toxic responses.
Collapse
Affiliation(s)
- Sophie J Bradley
- MRC Toxicology Unit, University of Leicester, Leicester LE1 9HN United Kingdom; ,
| | - Andrew B Tobin
- MRC Toxicology Unit, University of Leicester, Leicester LE1 9HN United Kingdom; ,
| |
Collapse
|
21
|
Dopamine D2 receptors in striatal output neurons enable the psychomotor effects of cocaine. Proc Natl Acad Sci U S A 2016; 113:11609-11614. [PMID: 27671625 DOI: 10.1073/pnas.1608362113] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The psychomotor effects of cocaine are mediated by dopamine (DA) through stimulation of striatal circuits. Gabaergic striatal medium spiny neurons (MSNs) are the only output of this pivotal structure in the control of movements. The majority of MSNs express either the DA D1 or D2 receptors (D1R, D2R). Studies have shown that the motor effect of cocaine depends on the DA-mediated stimulation of D1R-expressing MSNs (dMSNs), which is mirrored at the cellular level by stimulation of signaling pathways leading to phosphorylation of ERKs and induction of c-fos Nevertheless, activation of dMSNs by cocaine is necessary but not sufficient, and D2R signaling is required for the behavioral and cellular effects of cocaine. Indeed, cocaine motor effects and activation of signaling in dMSNs are blunted in mice with the constitutive knockout of D2R (D2RKO). Using mouse lines with a cell-specific knockout of D2R either in MSNs (MSN-D2RKO) or in dopaminergic neurons (DA-D2RKO), we show that D2R signaling in MSNs is required and permissive for the motor stimulant effects of cocaine and the activation of signaling in dMSNs. MSN-D2RKO mice show the same phenotype as constitutive D2RKO mice both at the behavioral and cellular levels. Importantly, activation of signaling in dMSNs by cocaine is rescued by intrastriatal injection of the GABA antagonist, bicuculline. These results are in support of intrastriatal connections of D2R+-MSNs (iMSNs) with dMSNs and indicate that D2R signaling in MSNs is critical for the function of intrastriatal circuits.
Collapse
|
22
|
Blank T, Prinz M. CatacLysMic specificity when targeting myeloid cells? Eur J Immunol 2016; 46:1340-2. [PMID: 27198084 DOI: 10.1002/eji.201646437] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 04/14/2016] [Accepted: 04/19/2016] [Indexed: 11/09/2022]
Abstract
The antibacterial enzyme lysozyme M (LysM) encoded by the Lyz2 gene is broadly expressed in myeloblasts, macrophages, and neutrophils, and thus has been used for a long time as a cell-specific marker for myeloid cells in mice. In order to delete loxP-site flanked genes in myeloid cells, a Cre-recombinase (Cre) expressing mouse line was created by inserting Cre-coding sequence into the translational start site of the LysM gene. In this issue of the European Journal of Immunology [2016. 46: 1529-1532], Orthgiess et al. verify, with the help of tdTomato and YFP reporter mouse lines, LysM-driven recombination. Unexpectedly, the authors also describe major expression of the tdTomato reporter protein in brain neurons of the central nervous system (CNS), with only a very small percentage of gene recombination in myeloid cells of the brain, called microglia. These findings cause justified concerns regarding the efficient and specific targeting of microglia and peripheral myeloid cells using LysM-Cre mice and should stimulate thoughts on conclusions drawn from past experiments on the diseased CNS employing this Cre/loxP-deleter line.
Collapse
Affiliation(s)
- Thomas Blank
- Institute of Neuropathology, Freiburg University Medical Centre, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Freiburg University Medical Centre, Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
23
|
Aelvoet SA, Pascual-Brazo J, Libbrecht S, Reumers V, Gijsbers R, Van den Haute C, Baekelandt V. Long-Term Fate Mapping Using Conditional Lentiviral Vectors Reveals a Continuous Contribution of Radial Glia-Like Cells to Adult Hippocampal Neurogenesis in Mice. PLoS One 2015; 10:e0143772. [PMID: 26600383 PMCID: PMC4658138 DOI: 10.1371/journal.pone.0143772] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 11/09/2015] [Indexed: 11/21/2022] Open
Abstract
Newborn neurons are generated throughout life in two neurogenic regions, the subventricular zone and the hippocampal dentate gyrus. Stimulation of adult neurogenesis is considered as an attractive endogenous repair mechanism to treat different neurological disorders. Although tremendous progress has been made in our understanding of adult hippocampal neurogenesis, important questions remain unanswered, regarding the identity and the behavior of neural stem cells in the dentate gyrus. We previously showed that conditional Cre-Flex lentiviral vectors can be used to label neural stem cells in the subventricular zone and to track the migration of their progeny with non-invasive bioluminescence imaging. Here, we applied these Cre-Flex lentiviral vectors to study neurogenesis in the dentate gyrus with bioluminescence imaging and histological techniques. Stereotactic injection of the Cre-Flex vectors into the dentate gyrus of transgenic Nestin-Cre mice resulted in specific labeling of the nestin-positive neural stem cells. The labeled cell population could be detected with bioluminescence imaging until 9 months post injection, but no significant increase in the number of labeled cells over time was observed with this imaging technique. Nevertheless, the specific labeling of the nestin-positive neural stem cells, combined with histological analysis at different time points, allowed detailed analysis of their neurogenic potential. This long-term fate mapping revealed that a stable pool of labeled nestin-positive neural stem cells continuously contributes to the generation of newborn neurons in the mouse brain until 9 months post injection. In conclusion, the Cre-Flex technology is a valuable tool to address remaining questions regarding neural stem cell identity and behavior in the dentate gyrus.
Collapse
Affiliation(s)
- Sarah-Ann Aelvoet
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Flanders, Belgium
| | - Jesus Pascual-Brazo
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Flanders, Belgium
| | - Sarah Libbrecht
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Flanders, Belgium
| | - Veerle Reumers
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Flanders, Belgium
| | - Rik Gijsbers
- Leuven Viral Vector Core, KU Leuven, Leuven, Flanders, Belgium
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Flanders, Belgium.,Leuven Viral Vector Core, KU Leuven, Leuven, Flanders, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Flanders, Belgium
| |
Collapse
|
24
|
Chiken S, Sato A, Ohta C, Kurokawa M, Arai S, Maeshima J, Sunayama-Morita T, Sasaoka T, Nambu A. Dopamine D1 Receptor-Mediated Transmission Maintains Information Flow Through the Cortico-Striato-Entopeduncular Direct Pathway to Release Movements. Cereb Cortex 2015; 25:4885-97. [PMID: 26443442 PMCID: PMC4635926 DOI: 10.1093/cercor/bhv209] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In the basal ganglia (BG), dopamine plays a pivotal role in motor control, and dopamine deficiency results in severe motor dysfunctions as seen in Parkinson's disease. According to the well-accepted model of the BG, dopamine activates striatal direct pathway neurons that directly project to the output nuclei of the BG through D1 receptors (D1Rs), whereas dopamine inhibits striatal indirect pathway neurons that project to the external pallidum (GPe) through D2 receptors. To clarify the exact role of dopaminergic transmission via D1Rs in vivo, we developed novel D1R knockdown mice in which D1Rs can be conditionally and reversibly regulated. Suppression of D1R expression by doxycycline treatment decreased spontaneous motor activity and impaired motor ability in the mice. Neuronal activity in the entopeduncular nucleus (EPN), one of the output nuclei of the rodent BG, was recorded in awake conditions to examine the mechanism of motor deficits. Cortically evoked inhibition in the EPN mediated by the cortico-striato-EPN direct pathway was mostly lost during suppression of D1R expression, whereas spontaneous firing rates and patterns remained unchanged. On the other hand, GPe activity changed little. These results suggest that D1R-mediated dopaminergic transmission maintains the information flow through the direct pathway to appropriately release motor actions.
Collapse
Affiliation(s)
- Satomi Chiken
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8585, Japan
| | - Asako Sato
- Department of Laboratory Animal Science, Kitasato University School of Medicine, Sagamihara 252-0374, Japan National Institute for Basic Biology, Okazaki 444-8585, Japan
| | - Chikara Ohta
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Makoto Kurokawa
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Satoshi Arai
- Department of Laboratory Animal Science, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Jun Maeshima
- Department of Laboratory Animal Science, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Tomoko Sunayama-Morita
- National Institute for Basic Biology, Okazaki 444-8585, Japan Department of Life Sciences, Graduate School of Arts and Science, The University of Tokyo, Tokyo 153-8902, Japan
| | - Toshikuni Sasaoka
- School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8585, Japan Department of Laboratory Animal Science, Kitasato University School of Medicine, Sagamihara 252-0374, Japan National Institute for Basic Biology, Okazaki 444-8585, Japan Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8585, Japan
| |
Collapse
|
25
|
Kim YC, Alberico SL, Emmons E, Narayanan NS. New therapeutic strategies targeting D1-type dopamine receptors for neuropsychiatric disease. ACTA ACUST UNITED AC 2015; 10:230-238. [PMID: 28280503 DOI: 10.1007/s11515-015-1360-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The neurotransmitter dopamine acts via two major classes of receptors, D1-type and D2-type. D1 receptors are highly expressed in the striatum and can also be found in the cerebral cortex. Here we review the role of D1 dopamine signaling in two major domains: L-DOPA-induced dyskinesias in Parkinson's disease and cognition in neuropsychiatric disorders. While there are many drugs targeting D2-type receptors, there are no drugs that specifically target D1 receptors. It has been difficult to use selective D1-receptor agonists for clinical applications due to issues with bioavailability, binding affinity, pharmacological kinetics, and side effects. We propose potential therapies that selectively modulate D1 dopamine signaling by targeting second messengers downstream of D1 receptors, allosteric modulators, or by making targeted modifications to D1-receptor machinery. The development of therapies specific to D1-receptor signaling could be a new frontier in the treatment of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Young-Cho Kim
- Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Eric Emmons
- Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Nandakumar S Narayanan
- Department of Neurology, University of Iowa, Iowa City, IA 52242, USA; Aging Mind and Brain Initiative, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
26
|
Nomura S, Bouhadana M, Morel C, Faure P, Cauli B, Lambolez B, Hepp R. Noradrenalin and dopamine receptors both control cAMP-PKA signaling throughout the cerebral cortex. Front Cell Neurosci 2014; 8:247. [PMID: 25191229 PMCID: PMC4140213 DOI: 10.3389/fncel.2014.00247] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/05/2014] [Indexed: 01/11/2023] Open
Abstract
Noradrenergic fibers innervate the entire cerebral cortex, whereas the cortical distribution of dopaminergic fibers is more restricted. However, the relative functional impact of noradrenalin and dopamine receptors in various cortical regions is largely unknown. Using a specific genetic label, we first confirmed that noradrenergic fibers innervate the entire cortex whereas dopaminergic fibers were present in all layers of restricted medial and lateral areas but only in deep layers of other areas. Imaging of a genetically encoded sensor revealed that noradrenalin and dopamine widely activate PKA in cortical pyramidal neurons of frontal, parietal and occipital regions with scarce dopaminergic fibers. Responses to noradrenalin had higher amplitude, velocity and occurred at more than 10-fold lower dose than those elicited by dopamine, whose amplitude and velocity increased along the antero-posterior axis. The pharmacology of these responses was consistent with the involvement of Gs-coupled beta1 adrenergic and D1/D5 dopaminergic receptors, but the inhibition of both noradrenalin and dopamine responses by beta adrenergic antagonists was suggestive of the existence of beta1-D1/D5 heteromeric receptors. Responses also involved Gi-coupled alpha2 adrenergic and D2-like dopaminergic receptors that markedly reduced their amplitude and velocity and contributed to their cell-to-cell heterogeneity. Our results reveal that noradrenalin and dopamine receptors both control cAMP-PKA signaling throughout the cerebral cortex with moderate regional and laminar differences. These receptors can thus mediate widespread effects of both catecholamines, which are reportedly co-released by cortical noradrenergic fibers beyond the territory of dopaminergic fibers.
Collapse
Affiliation(s)
- Shinobu Nomura
- Sorbonne Universités, UPMC Université Paris 06, UM CR 18, Neuroscience Paris Seine Paris, France ; Centre National de la Recherche Scientifique (CNRS), UMR 8246 Paris, France ; Institut National de la Santé et de la Recherche Médicale (INSERM), U 1130 Paris, France
| | - Maud Bouhadana
- Sorbonne Universités, UPMC Université Paris 06, UM CR 18, Neuroscience Paris Seine Paris, France ; Centre National de la Recherche Scientifique (CNRS), UMR 8246 Paris, France ; Institut National de la Santé et de la Recherche Médicale (INSERM), U 1130 Paris, France
| | - Carole Morel
- Sorbonne Universités, UPMC Université Paris 06, UM CR 18, Neuroscience Paris Seine Paris, France ; Centre National de la Recherche Scientifique (CNRS), UMR 8246 Paris, France ; Institut National de la Santé et de la Recherche Médicale (INSERM), U 1130 Paris, France
| | - Philippe Faure
- Sorbonne Universités, UPMC Université Paris 06, UM CR 18, Neuroscience Paris Seine Paris, France ; Centre National de la Recherche Scientifique (CNRS), UMR 8246 Paris, France ; Institut National de la Santé et de la Recherche Médicale (INSERM), U 1130 Paris, France
| | - Bruno Cauli
- Sorbonne Universités, UPMC Université Paris 06, UM CR 18, Neuroscience Paris Seine Paris, France ; Centre National de la Recherche Scientifique (CNRS), UMR 8246 Paris, France ; Institut National de la Santé et de la Recherche Médicale (INSERM), U 1130 Paris, France
| | - Bertrand Lambolez
- Sorbonne Universités, UPMC Université Paris 06, UM CR 18, Neuroscience Paris Seine Paris, France ; Centre National de la Recherche Scientifique (CNRS), UMR 8246 Paris, France ; Institut National de la Santé et de la Recherche Médicale (INSERM), U 1130 Paris, France
| | - Régine Hepp
- Sorbonne Universités, UPMC Université Paris 06, UM CR 18, Neuroscience Paris Seine Paris, France ; Centre National de la Recherche Scientifique (CNRS), UMR 8246 Paris, France ; Institut National de la Santé et de la Recherche Médicale (INSERM), U 1130 Paris, France
| |
Collapse
|
27
|
Vandeputte C, Reumers V, Aelvoet SA, Thiry I, De Swaef S, Van den Haute C, Pascual-Brazo J, Farr TD, Vande Velde G, Hoehn M, Himmelreich U, Van Laere K, Debyser Z, Gijsbers R, Baekelandt V. Bioluminescence imaging of stroke-induced endogenous neural stem cell response. Neurobiol Dis 2014; 69:144-55. [PMID: 24878507 DOI: 10.1016/j.nbd.2014.05.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 03/15/2014] [Accepted: 05/17/2014] [Indexed: 02/07/2023] Open
Abstract
Brain injury following stroke affects neurogenesis in the adult mammalian brain. However, a complete understanding of the origin and fate of the endogenous neural stem cells (eNSCs) in vivo is missing. Tools and technology that allow non-invasive imaging and tracking of eNSCs in living animals will help to overcome this hurdle. In this study, we aimed to monitor eNSCs in a photothrombotic (PT) stroke model using in vivo bioluminescence imaging (BLI). In a first strategy, inducible transgenic mice expressing firefly luciferase (Fluc) in the eNSCs were generated. In animals that received stroke, an increased BLI signal originating from the infarct region was observed. However, due to histological limitations, the identity and exact origin of cells contributing to the increased BLI signal could not be revealed. To overcome this limitation, we developed an alternative strategy employing stereotactic injection of conditional lentiviral vectors (Cre-Flex LVs) encoding Fluc and eGFP in the subventricular zone (SVZ) of Nestin-Cre transgenic mice, thereby specifically labeling the eNSCs. Upon induction of stroke, increased eNSC proliferation resulted in a significant increase in BLI signal between 2days and 2weeks after stroke, decreasing after 3months. Additionally, the BLI signal relocalized from the SVZ towards the infarct region during the 2weeks following stroke. Histological analysis at 90days post stroke showed that in the peri-infarct area, 36% of labeled eNSC progeny differentiated into astrocytes, while 21% differentiated into mature neurons. In conclusion, we developed and validated a novel imaging technique that unequivocally demonstrates that nestin(+) eNSCs originating from the SVZ respond to stroke injury by increased proliferation, migration towards the infarct region and differentiation into both astrocytes and neurons. In addition, this new approach allows non-invasive and specific monitoring of eNSCs over time, opening perspectives for preclinical evaluation of candidate stroke therapeutics.
Collapse
Affiliation(s)
- Caroline Vandeputte
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, 3000 Leuven, Flanders, Belgium; KU Leuven, Molecular Small Animal Imaging Center, MOSAIC, KU Leuven, 3000 Leuven, Flanders, Belgium; Division of Nuclear Medicine, University Hospital and KU Leuven, 3000 Leuven, Flanders, Belgium
| | - Veerle Reumers
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, 3000 Leuven, Flanders, Belgium
| | - Sarah-Ann Aelvoet
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, 3000 Leuven, Flanders, Belgium
| | - Irina Thiry
- KU Leuven, Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium
| | - Sylvie De Swaef
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, 3000 Leuven, Flanders, Belgium
| | - Chris Van den Haute
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, 3000 Leuven, Flanders, Belgium; KU Leuven, Leuven Viral Vector Core, 3000 Leuven, Flanders, Belgium
| | - Jesus Pascual-Brazo
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, 3000 Leuven, Flanders, Belgium
| | - Tracy D Farr
- In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, 50931 Cologne, Germany
| | - Greetje Vande Velde
- KU Leuven, Molecular Small Animal Imaging Center, MOSAIC, KU Leuven, 3000 Leuven, Flanders, Belgium; KU Leuven, Biomedical MRI, Department of Imaging and Pathology, 3000 Leuven, Flanders, Belgium
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, 50931 Cologne, Germany
| | - Uwe Himmelreich
- KU Leuven, Molecular Small Animal Imaging Center, MOSAIC, KU Leuven, 3000 Leuven, Flanders, Belgium; KU Leuven, Biomedical MRI, Department of Imaging and Pathology, 3000 Leuven, Flanders, Belgium
| | - Koen Van Laere
- KU Leuven, Molecular Small Animal Imaging Center, MOSAIC, KU Leuven, 3000 Leuven, Flanders, Belgium; Division of Nuclear Medicine, University Hospital and KU Leuven, 3000 Leuven, Flanders, Belgium
| | - Zeger Debyser
- KU Leuven, Molecular Small Animal Imaging Center, MOSAIC, KU Leuven, 3000 Leuven, Flanders, Belgium; KU Leuven, Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium
| | - Rik Gijsbers
- KU Leuven, Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, Flanders, Belgium; KU Leuven, Leuven Viral Vector Core, 3000 Leuven, Flanders, Belgium.
| | - Veerle Baekelandt
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, 3000 Leuven, Flanders, Belgium; KU Leuven, Molecular Small Animal Imaging Center, MOSAIC, KU Leuven, 3000 Leuven, Flanders, Belgium.
| |
Collapse
|
28
|
Differential roles of the dopamine 1-class receptors, D1R and D5R, in hippocampal dependent memory. Proc Natl Acad Sci U S A 2014; 111:8245-50. [PMID: 24843151 DOI: 10.1073/pnas.1407395111] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Activation of the hippocampal dopamine 1-class receptors (D1R and D5R) are implicated in contextual fear conditioning (CFC). However, the specific role of the D1R versus D5R in hippocampal dependent CFC has not been investigated. Generation of D1R- and D5R-specific in situ hybridization probes showed that D1R and D5R mRNA expression was greatest in the dentate gyrus (DG) of the hippocampus. To identify the role of each receptor in CFC we generated spatially restricted KO mice that lack either the D1R or D5R in DG granule cells. DG D1R KOs displayed significant fear memory deficits, whereas DG D5R KOs did not. Furthermore, D1R KOs but not D5R KOs, exhibited generalized fear between two similar but different contexts. In the familiar home cage context, c-Fos expression was relatively low in the DG of control mice, and it increased upon exposure to a novel context. This level of c-Fos expression in the DG did not further increase when a footshock was delivered in the novel context. In DG D1R KOs, DG c-Fos levels in the home cage was higher than that of the control mice, but it did not further increase upon exposure to a novel context and remained at the same level upon a shock delivery. In contrast, the levels of DG c-Fos expression was unaffected by the deletion of DG D5R neither in the home cage nor upon a shock delivery. These results suggest that DG D1Rs, but not D5Rs, contribute to the formation of distinct contextual representations of novel environments.
Collapse
|
29
|
Parnaudeau S, Dongelmans ML, Turiault M, Ambroggi F, Delbes AS, Cansell C, Luquet S, Piazza PV, Tronche F, Barik J. Glucocorticoid receptor gene inactivation in dopamine-innervated areas selectively decreases behavioral responses to amphetamine. Front Behav Neurosci 2014; 8:35. [PMID: 24574986 PMCID: PMC3921555 DOI: 10.3389/fnbeh.2014.00035] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/23/2014] [Indexed: 11/13/2022] Open
Abstract
The meso-cortico-limbic system, via dopamine release, encodes the rewarding and reinforcing properties of natural rewards. It is also activated in response to abused substances and is believed to support drug-related behaviors. Dysfunctions of this system lead to several psychiatric conditions including feeding disorders and drug addiction. These disorders are also largely influenced by environmental factors and in particular stress exposure. Stressors activate the corticotrope axis ultimately leading to glucocorticoid hormone (GCs) release. GCs bind the glucocorticoid receptor (GR) a transcription factor ubiquitously expressed including within the meso-cortico-limbic tract. While GR within dopamine-innervated areas drives cocaine's behavioral responses, its implication in responses to other psychostimulants such as amphetamine has never been clearly established. Moreover, while extensive work has been made to uncover the role of this receptor in addicted behaviors, its contribution to the rewarding and reinforcing properties of food has yet to be investigated. Using mouse models carrying GR gene inactivation in either dopamine neurons or in dopamine-innervated areas, we found that GR in dopamine responsive neurons is essential to properly build amphetamine-induced conditioned place preference and locomotor sensitization. c-Fos quantification in the nucleus accumbens further confirmed defective neuronal activation following amphetamine injection. These diminished neuronal and behavioral responses to amphetamine may involve alterations in glutamate transmission as suggested by the decreased MK801-elicited hyperlocomotion and by the hyporeactivity to glutamate of a subpopulation of medium spiny neurons. In contrast, GR inactivation did not affect rewarding and reinforcing properties of food suggesting that responding for natural reward under basal conditions is preserved in these mice.
Collapse
Affiliation(s)
- Sébastien Parnaudeau
- UMR 7224 CNRS, Physiopathologie des Maladies du Système Nerveux Central, "Gene Regulation and Adaptive Behaviors" Group Paris, France ; INSERM, UMRs 952, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; Université Pierre et Marie Curie, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; Department of Psychiatry, Columbia University New York, NY, USA
| | - Marie-Louise Dongelmans
- UMR 7224 CNRS, Physiopathologie des Maladies du Système Nerveux Central, "Gene Regulation and Adaptive Behaviors" Group Paris, France ; INSERM, UMRs 952, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; Université Pierre et Marie Curie, Physiopathologie des Maladies du Système Nerveux Central Paris, France
| | - Marc Turiault
- UMR 7224 CNRS, Physiopathologie des Maladies du Système Nerveux Central, "Gene Regulation and Adaptive Behaviors" Group Paris, France ; INSERM, UMRs 952, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; Université Pierre et Marie Curie, Physiopathologie des Maladies du Système Nerveux Central Paris, France
| | - Frédéric Ambroggi
- Pathophysiology of Addiction, Institut National de la Santé et de la Recherche Médicale, U862, NeuroCentre Magendie Bordeaux Cedex, France ; Department of Neurology, Center for Integrative Neuroscience and the Ernest Gallo Clinic and Research Center, University of California at San Francisco San Francisco, CA, USA
| | - Anne-Sophie Delbes
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, UMR 8251 CNRS, Université Paris Diderot Paris, France
| | - Céline Cansell
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, UMR 8251 CNRS, Université Paris Diderot Paris, France
| | - Serge Luquet
- Unité de Biologie Fonctionnelle et Adaptative, Sorbonne Paris Cité, UMR 8251 CNRS, Université Paris Diderot Paris, France
| | - Pier-Vincenzo Piazza
- Pathophysiology of Addiction, Institut National de la Santé et de la Recherche Médicale, U862, NeuroCentre Magendie Bordeaux Cedex, France
| | - François Tronche
- UMR 7224 CNRS, Physiopathologie des Maladies du Système Nerveux Central, "Gene Regulation and Adaptive Behaviors" Group Paris, France ; INSERM, UMRs 952, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; Université Pierre et Marie Curie, Physiopathologie des Maladies du Système Nerveux Central Paris, France
| | - Jacques Barik
- UMR 7224 CNRS, Physiopathologie des Maladies du Système Nerveux Central, "Gene Regulation and Adaptive Behaviors" Group Paris, France ; INSERM, UMRs 952, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; Université Pierre et Marie Curie, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275 Valbonne, France
| |
Collapse
|
30
|
Kreiner G, Bierhoff H, Armentano M, Rodriguez-Parkitna J, Sowodniok K, Naranjo JR, Bonfanti L, Liss B, Schütz G, Grummt I, Parlato R. A neuroprotective phase precedes striatal degeneration upon nucleolar stress. Cell Death Differ 2013; 20:1455-64. [PMID: 23764776 PMCID: PMC3792439 DOI: 10.1038/cdd.2013.66] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 05/03/2013] [Accepted: 05/16/2013] [Indexed: 01/17/2023] Open
Abstract
The nucleolus is implicated in sensing and responding to cellular stress by stabilizing p53. The pro-apoptotic effect of p53 is associated with several neurodegenerative disorders, including Huntington's disease (HD), which is characterized by the progressive loss of medium spiny neurons (MSNs) in the striatum. Here we show that disruption of nucleolar integrity and function causes nucleolar stress and is an early event in MSNs of R6/2 mice, a transgenic model of HD. Targeted perturbation of nucleolar function in MSNs by conditional knockout of the RNA polymerase I-specific transcription initiation factor IA (TIF-IA) leads to late progressive striatal degeneration, HD-like motor abnormalities and molecular signatures. Significantly, p53 prolongs neuronal survival in TIF-IA-deficient MSNs by transient upregulation of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a tumor suppressor that inhibits mammalian target of rapamycin signaling and induces autophagy. The results emphasize the initial role of nucleolar stress in neurodegeneration and uncover a p53/PTEN-dependent neuroprotective response.
Collapse
Affiliation(s)
- G Kreiner
- 1] Department of Molecular Biology of the Cell I, DKFZ-ZMBH Alliance, German Cancer Research Center, Heidelberg, Germany [2] Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Cracow, Poland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lenz JD, Lobo MK. Optogenetic insights into striatal function and behavior. Behav Brain Res 2013; 255:44-54. [PMID: 23628212 DOI: 10.1016/j.bbr.2013.04.018] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 04/10/2013] [Accepted: 04/15/2013] [Indexed: 10/26/2022]
Abstract
Recent breakthroughs in optogenetic technologies to alter neuronal firing and function with light, combined with cell type-specific transgenic animal lines, has led to important insights into the function of distinct neuronal cell subtypes and afferent connections in the heterogeneously complex striatum. A vital part of the basal ganglia, the striatum is heavily implicated in both motor control and motivation-based behavior; as well as in neurological disorders and psychiatric diseases including Parkinson's Disease, Huntington's Disease, drug addiction, depression, and schizophrenia. Researchers are able to manipulate firing and cell signaling with temporal precision using optogenetics in the two striatal medium spiny neuron (MSN) subpopulations, the striatal interneurons, and striatal afferents. These studies confirmed the classical hypothesis of movement control and reward seeking behavior through direct versus indirect pathway MSNs; illuminated a selective role for TANs in cocaine reward; dissected the roles of glutamatergic and dopaminergic inputs to striatum in reward; and highlighted a role for striatal signaling molecules including an adrenergic G-protein coupled receptor in reward and the rho-GTPase Rac1 in cocaine reward and cocaine induced structural plasticity. This review focuses on how the evolving optogenetic toolbox provides insight into the distinct behavioral roles of striatal cell subpopulations and striatal afferents, which has clinically relevant implications into neurological disorders and psychiatric disease.
Collapse
Affiliation(s)
- Jeffrey D Lenz
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | |
Collapse
|
32
|
Barik J, Marti F, Morel C, Fernandez SP, Lanteri C, Godeheu G, Tassin JP, Mombereau C, Faure P, Tronche F. Chronic stress triggers social aversion via glucocorticoid receptor in dopaminoceptive neurons. Science 2013; 339:332-5. [PMID: 23329050 DOI: 10.1126/science.1226767] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Repeated traumatic events induce long-lasting behavioral changes that are key to organism adaptation and that affect cognitive, emotional, and social behaviors. Rodents subjected to repeated instances of aggression develop enduring social aversion and increased anxiety. Such repeated aggressions trigger a stress response, resulting in glucocorticoid release and activation of the ascending dopamine (DA) system. We bred mice with selective inactivation of the gene encoding the glucocorticoid receptor (GR) along the DA pathway, and exposed them to repeated aggressions. GR in dopaminoceptive but not DA-releasing neurons specifically promoted social aversion as well as dopaminergic neurochemical and electrophysiological neuroadaptations. Anxiety and fear memories remained unaffected. Acute inhibition of the activity of DA-releasing neurons fully restored social interaction in socially defeated wild-type mice. Our data suggest a GR-dependent neuronal dichotomy for the regulation of emotional and social behaviors, and clearly implicate GR as a link between stress resiliency and dopaminergic tone.
Collapse
Affiliation(s)
- Jacques Barik
- Molecular Genetics, Neurophysiology and Behavior Group, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7224, Paris, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Dual control of dopamine synthesis and release by presynaptic and postsynaptic dopamine D2 receptors. J Neurosci 2012; 32:9023-34. [PMID: 22745501 DOI: 10.1523/jneurosci.0918-12.2012] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Dysfunctions of dopaminergic homeostasis leading to either low or high dopamine (DA) levels are causally linked to Parkinson's disease, schizophrenia, and addiction. Major sites of DA synthesis are the mesencephalic neurons originating in the substantia nigra and ventral tegmental area; these structures send major projections to the dorsal striatum (DSt) and nucleus accumbens (NAcc), respectively. DA finely tunes its own synthesis and release by activating DA D2 receptors (D2R). To date, this critical D2R-dependent function was thought to be solely due to activation of D2Rs on dopaminergic neurons (D2 autoreceptors); instead, using site-specific D2R knock-out mice, we uncover that D2 heteroreceptors located on non-DAergic medium spiny neurons participate in the control of DA levels. This D2 heteroreceptor-mediated mechanism is more efficient in the DSt than in NAcc, indicating that D2R signaling differentially regulates mesolimbic- versus nigrostriatal-mediated functions. This study reveals previously unappreciated control of DA signaling, shedding new light on region-specific regulation of DA-mediated effects.
Collapse
|
34
|
Kühne C, Puk O, Graw J, Hrabě de Angelis M, Schütz G, Wurst W, Deussing JM. Visualizing corticotropin-releasing hormone receptor type 1 expression and neuronal connectivities in the mouse using a novel multifunctional allele. J Comp Neurol 2012; 520:3150-80. [DOI: 10.1002/cne.23082] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
35
|
Abstract
Muscarinic acetylcholine (ACh) receptors (mAChRs; M₁-M₅) regulate the activity of an extraordinarily large number of important physiological processes. During the past 10-15 years, studies with whole-body M₁-M₅ mAChR knockout mice have provided many new insights into the physiological and pathophysiological roles of the individual mAChR subtypes. This review will focus on the characterization of a novel generation of mAChR mutant mice, including mice in which distinct mAChR genes have been excised in a tissue- or cell type-specific fashion, various transgenic mouse lines that overexpress wild-type or different mutant M₃ mAChRs in certain tissues or cells only, as well as a novel M₃ mAChR knockin mouse strain deficient in agonist-induced M₃ mAChR phosphorylation. Phenotypic analysis of these new animal models has greatly advanced our understanding of the physiological roles of the various mAChR subtypes and has identified potential targets for the treatment of type 2 diabetes, schizophrenia, Parkinson's disease, drug addiction, cognitive disorders, and several other pathophysiological conditions.
Collapse
|
36
|
Ena S, de Kerchove d'Exaerde A, Schiffmann SN. Unraveling the differential functions and regulation of striatal neuron sub-populations in motor control, reward, and motivational processes. Front Behav Neurosci 2011; 5:47. [PMID: 21847377 PMCID: PMC3148764 DOI: 10.3389/fnbeh.2011.00047] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 07/18/2011] [Indexed: 12/15/2022] Open
Abstract
The striatum, the major input structure of the basal ganglia, is critically involved in motor control and learning of habits and skills, and is also involved in motivational and reward processes. The dorsal striatum, caudate–putamen, is primarily implicated in motor functions whereas the ventral striatum, the nucleus accumbens, is essential for motivation and drug reinforcement. Severe basal ganglia dysfunction occurs in movement disorders as Parkinson's and Huntington's disease, and in psychiatric disorders such as schizophrenia and drug addiction. The striatum is essentially composed of GABAergic medium-sized spiny neurons (MSNs) that are output neurons giving rise to the so-called direct and indirect pathways and are targets of the cerebral cortex and mesencephalic dopaminergic neurons. Although the involvement of striatal sub-areas in motor control and motivation has been thoroughly characterized, major issues remained concerning the specific and respective functions of the two MSNs sub-populations, D2R-striatopallidal (dopamine D2 receptor-positive) and D1R-striatonigral (dopamine D1 receptor-positive) neurons, as well as their specific regulation. Here, we review recent advances that gave new insight in the understanding of the differential roles of striatopallidal and striatonigral neurons in the basal ganglia circuit. We discuss innovative techniques developed in the last decade which allowed a much precise evaluation of molecular pathways implicated in motivational processes and functional roles of striatopallidal and striatonigral neurons in motor control and in the establishment of reward-associated behavior.
Collapse
Affiliation(s)
- Sabrina Ena
- Laboratory of Neurophysiology, School of Medicine, Université Libre de Bruxelles Brussels, Belgium
| | | | | |
Collapse
|
37
|
Lobo MK, Nestler EJ. The striatal balancing act in drug addiction: distinct roles of direct and indirect pathway medium spiny neurons. Front Neuroanat 2011; 5:41. [PMID: 21811439 PMCID: PMC3140647 DOI: 10.3389/fnana.2011.00041] [Citation(s) in RCA: 256] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 07/05/2011] [Indexed: 12/16/2022] Open
Abstract
The striatum plays a key role in mediating the acute and chronic effects of addictive drugs, with drugs of abuse causing long-lasting molecular and cellular alterations in both dorsal striatum and nucleus accumbens (ventral striatum). Despite the wealth of research on the biological actions of abused drugs in striatum, until recently, the distinct roles of the striatum’s two major subtypes of medium spiny neurons (MSNs) in drug addiction remained elusive. Recent advances in cell-type-specific technologies, including fluorescent reporter mice, transgenic, or knockout mice, and viral-mediated gene transfer, have advanced the field toward a more comprehensive understanding of the two MSN subtypes in the long-term actions of drugs of abuse. Here we review progress in defining the distinct molecular and functional contributions of the two MSN subtypes in mediating addiction.
Collapse
Affiliation(s)
- Mary Kay Lobo
- Fishberg Department of Neuroscience, Friedman Brain Institute, Mount Sinai School of Medicine New York, NY, USA
| | | |
Collapse
|
38
|
Durieux PF, Schiffmann SN, de Kerchove d'Exaerde A. Targeting neuronal populations of the striatum. Front Neuroanat 2011; 5:40. [PMID: 21811438 PMCID: PMC3139926 DOI: 10.3389/fnana.2011.00040] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 07/03/2011] [Indexed: 12/30/2022] Open
Abstract
The striatum is critically involved in motor and motivational functions. The dorsal striatum, caudate–putamen, is primarily implicated in motor control and the learning of habits and skills, whereas the ventral striatum, the nucleus accumbens, is essential for motivation and drug reinforcement. The GABA medium-sized spiny neurons (MSNs, about 95% of striatal neurons), which are targets of the cerebral cortex and the midbrain dopaminergic neurons, form two pathways. The dopamine D1 receptor-positive (D1R) striatonigral MSNs project to the medial globus pallidus and substantia nigra pars reticulata (direct pathway) and co-express D1R and substance P, whereas dopamine D2 receptor-positive (D2R) striatopallidal MSNs project to the lateral globus pallidus (indirect pathway) and co-express D2R, adenosine A2A receptor (A2AR) and enkephalin (Enk). The specific role of the two efferent pathways in motor and motivational control remained poorly understood until recently. Indeed, D1R striatonigral and D2R striatopallidal neurons, are intermingled and morphologically indistinguishable, and, hence, cannot be functionally dissociated with techniques such as chemical lesions or surgery. In view of the still debated respective functions of projection D2R striatopallidal and D1R striatonigral neurons and striatal interneurons, both in motor control and learning but also in more cognitive processes such as motivation, the present review sum up the development of new models and techniques (bacterial artificial chromosome transgenesis, optogenetic, viral transgenesis) allowing the selective targeting of these striatal neuronal populations in adult animal brain to understand their specific roles.
Collapse
Affiliation(s)
- Pierre F Durieux
- Laboratory of Neurophysiology, School of Medicine, Université Libre de Bruxelles Brussels, Belgium
| | | | | |
Collapse
|
39
|
Glucocorticoid receptors in dopaminoceptive neurons, key for cocaine, are dispensable for molecular and behavioral morphine responses. Biol Psychiatry 2010; 68:231-9. [PMID: 20554270 DOI: 10.1016/j.biopsych.2010.03.037] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 03/29/2010] [Accepted: 03/31/2010] [Indexed: 11/21/2022]
Abstract
BACKGROUND Psychostimulants and opiates trigger similar enduring neuroadaptations within the reward circuitry thought to underlie addiction. Transcription factors are key to mediating these enduring behavioral alterations. The facilitation of these maladaptive changes by glucocorticoid hormones suggests that the glucocorticoid receptor (GR), a transcription factor involved in the stress response, could be a common mediator of responses to pharmacologically distinct classes of abused drugs. METHODS We employed mouse models carrying GR gene inactivation in either dopamine or dopaminoceptive neurons to determine the involvement of this transcription factor in behavioral responses to cocaine and morphine. We then combined microarray analysis, drug-elicited immediate early gene induction, and in vivo microdialysis to elucidate the molecular underpinnings of these responses. RESULTS Inactivating GR within dopaminoceptive neurons markedly reduces cocaine-induced conditioned place preference and the expression of locomotor sensitization. In striking contrast, GR had no effect on behavioral morphine responses in either dopaminoceptive or dopamine neurons. The dopaminoceptive mutation engenders alterations in the expression of striatal genes that are implicated in glutamatergic transmission and plasticity. Within the nucleus accumbens, impaired cellular responses to cocaine are conspicuous; a pronounced deficit in cocaine-elicited extracellular dopamine release, expression of the key IEGs c-Fos and Zif268, and phosphorylation of extracellular signal-regulated kinases 1/2 in mutants were observed. In contrast, these molecular and neurochemical changes were not observed in response to morphine, mirroring the lack of effect on behavioral responses to morphine. CONCLUSION Combined behavioral and molecular approaches have identified a subset of neurons in which GR differentially influences cocaine- and morphine-induced responses.
Collapse
|
40
|
Development of a BAC vector for integration-independent and tight regulation of transgenes in rodents via the Tet system. Transgenic Res 2010; 20:709-20. [PMID: 20640885 DOI: 10.1007/s11248-010-9427-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 07/03/2010] [Indexed: 01/02/2023]
Abstract
The establishment of functional transgenic mouse lines is often limited by problems caused by integration site effects on the expression construct. Similarly, tetracycline (Tet) controlled transcription units most commonly used for conditional transgene expression in mice are strongly influenced by their genomic surrounding. Using bacterial artificial chromosome (BAC) technology in constitutive expression systems, it has been shown that integration site effects resulting in unwanted expression patterns can be largely eliminated. Here we describe a strategy to minimize unfavourable integration effects on conditional expression constructs based on a 75 kb genomic BAC fragment. This fragment was derived from a transgenic mouse line, termed LC-1, which carries the Tet-inducible genes luciferase and cre (Schönig et al. 2002). Animals of this mouse line have previously been shown to exhibit optimal expression properties in terms of tightness in the off state and the absolute level of induction, when mated to appropriate transactivator expressing mice. Here we report the cloning and identification of the transgenic LC-1 integration site which was subsequently inserted into a bacterial artificial chromosome. We demonstrate that this vector facilitates the efficient generation of transgenic mouse and rat lines, where the Tet-controlled expression unit is shielded from perturbations caused by the integration site.
Collapse
|
41
|
Parkitna JR, Bilbao A, Rieker C, Engblom D, Piechota M, Nordheim A, Spanagel R, Schütz G. Loss of the serum response factor in the dopamine system leads to hyperactivity. FASEB J 2010; 24:2427-35. [PMID: 20223941 DOI: 10.1096/fj.09-151423] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The serum response factor (SRF) is a key regulator of neural development and cellular plasticity, which enables it to act as a regulator of long-term adaptations in neurons. Here we performed a comprehensive analysis of SRF function in the murine dopamine system. We found that loss of SRF in dopaminoceptive, but not dopaminergic, neurons is responsible for the development of a hyperactivity syndrome, characterized by reduced body weight into adulthood, enhanced motor activity, and deficits in habituation processes. Most important, the hyperactivity also develops when the ablation of SRF is induced in adult animals. On the molecular level, the loss of SRF in dopaminoceptive cells is associated with altered expression of neuronal plasticity-related genes, in particular transcripts involved in calcium ion binding, formation of the cytoskeleton, and transcripts encoding neuropeptide precursors. Furthermore, abrogation of SRF causes specific deficits in activity-dependent transcription, especially a complete lack of psychostimulant-induced expression of the Egr genes. We inferred that alterations in SRF-dependent gene expression underlie the observed hyperactive behavior. Thus, SRF depletion in dopaminoceptive neurons might trigger molecular mechanisms responsible for development of psychopathological conditions involving hyperactivity.
Collapse
Affiliation(s)
- Jan Rodriguez Parkitna
- Molecular Biology of the Cell I, German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
A subpopulation of neuronal M4 muscarinic acetylcholine receptors plays a critical role in modulating dopamine-dependent behaviors. J Neurosci 2010; 30:2396-405. [PMID: 20147565 DOI: 10.1523/jneurosci.3843-09.2010] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Acetylcholine (ACh) regulates many key functions of the CNS by activating cell surface receptors referred to as muscarinic ACh receptors (M(1)-M(5) mAChRs). Like other mAChR subtypes, the M(4) mAChR is widely expressed in different regions of the forebrain. Interestingly, M(4) mAChRs are coexpressed with D(1) dopamine receptors in a specific subset of striatal projection neurons. To investigate the physiological relevance of this M(4) mAChR subpopulation in modulating dopamine-dependent behaviors, we used Cre/loxP technology to generate mutant mice that lack M(4) mAChRs only in D(1) dopamine receptor-expressing cells. The newly generated mutant mice displayed several striking behavioral phenotypes, including enhanced hyperlocomotor activity and increased behavioral sensitization following treatment with psychostimulants. These behavioral changes were accompanied by a lack of muscarinic inhibition of D(1) dopamine receptor-mediated cAMP stimulation in the striatum and an increase in dopamine efflux in the nucleus accumbens. These novel findings demonstrate that a distinct subpopulation of neuronal M(4) mAChRs plays a critical role in modulating several important dopamine-dependent behaviors. Since enhanced central dopaminergic neurotransmission is a hallmark of several severe disorders of the CNS, including schizophrenia and drug addiction, our findings have substantial clinical relevance.
Collapse
|
43
|
Stress and addiction: glucocorticoid receptor in dopaminoceptive neurons facilitates cocaine seeking. Nat Neurosci 2009; 12:247-9. [PMID: 19234455 DOI: 10.1038/nn.2282] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2008] [Accepted: 01/22/2009] [Indexed: 11/08/2022]
Abstract
The glucocorticoid receptor is a ubiquitous transcription factor mediating adaptation to environmental challenges and stress. Selective Nr3c1 (the glucocorticoid receptor gene) ablation in mouse dopaminoceptive neurons expressing dopamine receptor 1a, but not in dopamine-releasing neurons, markedly decreased the motivation of mice to self-administer cocaine, dopamine cell firing and the control exerted by dopaminoceptive neurons on dopamine cell firing activity. In contrast, anxiety was unaffected, indicating that glucocorticoid receptors modify a number of behavioral disorders through different neuronal populations.
Collapse
|
44
|
Abstract
The endocannabinoid (ECB) system comprises cannabinoid receptors, ECBs and the whole machinery for the synthesis and degradation of ECBs. It has emerged as an important signalling system in the nervous system, controlling numerous physiological processes, including synaptic transmission, learning and memory, reward, feeding, neuroprotection, neuroinflammation, and neural development. This system is also implicated in various diseases of the nervous system, and thus has become a promising therapeutic target. The use of genetically modified mice has contributed crucially to our rapidly expanding knowledge of the ECB system. In this chapter, the existing mouse mutants targeting the ECB system will be discussed in detail. The use of conditional mutants has given an additional dimension to the analysis of the system, and, it is hoped, will finally enable us to understand this widespread and complex system in the context of intricate networks where different brain regions and neurotransmitter systems interact tightly with each other.
Collapse
Affiliation(s)
- Krisztina Monory
- Department of Physiological Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 6, D-55099, Mainz, Germany.
| | | |
Collapse
|
45
|
Molecular profiling of striatonigral and striatopallidal medium spiny neurons past, present, and future. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 89:1-35. [PMID: 19900613 DOI: 10.1016/s0074-7742(09)89001-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Defining distinct molecular properties of the two striatal medium spiny neurons (MSNs) has been a challenging task for basal ganglia (BG) neuroscientists. Identifying differential molecular components in each MSN subtype is crucial for BG researchers to understand functional properties of these two neurons. The two MSN populations are morphologically identical except in their projections through the direct verses indirect BG pathways and they are heterogeneously dispersed throughout the dorsal striatum (dStr) and nucleus accumbens (NAc). These characteristics have made it difficult for researchers to distinguish and isolate these two neuronal populations thereby hindering progress toward a more comprehensive understanding of their differential molecular properties. Researchers began to investigate molecular differences in the striatonigral and striatopallidal neurons using in situ hybridization (ISH) techniques and single cell reverse transcription-polymerase chain reaction (scRT-PCR). Currently the field is utilizing more advanced techniques for large-scale gene expression studies including fluorescence activated cell sorting (FACS) of MSNs, from which RNA is purified, from fluorescent reporter transgenic mice or use of transgenic mice in which ribosomes from each MSN are tagged and can be immunoprecipitated followed by RNA isolation, a technique termed translating ribosomal affinity purification (TRAP). Additionally, the availability of fluorescent reporter mice for each MSN subtype is allowing, scientists to perform more accurate histology studies evaluating differential protein expression and signaling changes in each cell subtype. Finally, researchers are able to evaluate the role of specific genes in vivo by utilizing cell type-specific mouse models including Cre driver lines that can be crossed with conditional overexpression or knockout systems. This is a very exciting time in the BG field because researchers are well equipped with the most progressive tools to comprehensively evaluate molecular components in the two MSNs and their consequence on BG functional output in the normal, diseased, and developing brain.
Collapse
|
46
|
Loss of the Ca2+/calmodulin-dependent protein kinase type IV in dopaminoceptive neurons enhances behavioral effects of cocaine. Proc Natl Acad Sci U S A 2008; 105:17549-54. [PMID: 19001277 DOI: 10.1073/pnas.0803959105] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The persistent nature of addiction has been associated with activity-induced plasticity of neurons within the striatum and nucleus accumbens (NAc). To identify the molecular processes leading to these adaptations, we performed Cre/loxP-mediated genetic ablations of two key regulators of gene expression in response to activity, the Ca(2+)/calmodulin-dependent protein kinase IV (CaMKIV) and its postulated main target, the cAMP-responsive element binding protein (CREB). We found that acute cocaine-induced gene expression in the striatum was largely unaffected by the loss of CaMKIV. On the behavioral level, mice lacking CaMKIV in dopaminoceptive neurons displayed increased sensitivity to cocaine as evidenced by augmented expression of locomotor sensitization and enhanced conditioned place preference and reinstatement after extinction. However, the loss of CREB in the forebrain had no effect on either of these behaviors, even though it robustly blunted acute cocaine-induced transcription. To test the relevance of these observations for addiction in humans, we performed an association study of CAMK4 and CREB promoter polymorphisms with cocaine addiction in a large sample of addicts. We found that a single nucleotide polymorphism in the CAMK4 promoter was significantly associated with cocaine addiction, whereas variations in the CREB promoter regions did not correlate with drug abuse. These findings reveal a critical role for CaMKIV in the development and persistence of cocaine-induced behaviors, through mechanisms dissociated from acute effects on gene expression and CREB-dependent transcription.
Collapse
|
47
|
Mann K, Kiefer F, Spanagel R, Littleton J. Acamprosate: recent findings and future research directions. Alcohol Clin Exp Res 2008; 32:1105-10. [PMID: 18540918 DOI: 10.1111/j.1530-0277.2008.00690.x] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This article explores the mechanisms of action and the potential responder profile of acamprosate, a compound efficacious in relapse prevention of alcoholism. New evidence at the molecular and cellular level suggests that acamprosate attenuates hyper-glutamatergic states that occur during early abstinence and involves iono (NMDA)- and metabotrotropic (mGluR5) glutamate receptors along with augmented intracellular calcium release and electrophysiological changes. Thus mutant mice with enhanced glutamate levels exhibit higher alcohol consumption than wild type mice and respond better to acamprosate, demonstrating that acamprosate acts mainly on a hyper-glutamatergic system. This mode of action further suggests that acamprosate exhibits neuroprotective properties. In rats, cue-induced reinstatement behavior is significantly reduced by acamprosate treatment whereas cue-induced craving responses in alcohol-dependent patients seem not to be affected by this treatment. An ongoing study ("Project Predict") defines specific responder profiles for an individualized use of acamprosate and naltrexone. Neurophysiological as well as psychometric data are used to define 2 groups of patients: "reward cravers" and "relief cravers". While naltrexone should work better in the first group, acamprosate is hypothesized to be efficacious in the latter where withdrawal associated and/or cue induced hyper-glutamatergic states are thought to trigger relapse. Further research should target the definition of subgroups applying endophenotypic approaches, e.g. by detecting a hyperglutamatergic syndrome using MR spectroscopy.
Collapse
Affiliation(s)
- Karl Mann
- The Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health University of Heidelberg, Mannheim, Germany
| | | | | | | |
Collapse
|
48
|
Heusner CL, Beutler LR, Houser CR, Palmiter RD. Deletion of GAD67 in dopamine receptor-1 expressing cells causes specific motor deficits. Genesis 2008; 46:357-67. [PMID: 18615733 PMCID: PMC3360952 DOI: 10.1002/dvg.20405] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The medium spiny neurons (MSNs), which comprise the direct and indirect output pathways from the striatum, use gamma-aminobutyric acid (GABA) as their major fact-acting neurotransmitter. We generated mice carrying a conditional allele of the Gad1 gene, which encodes GAD67, one of the two enzymes responsible for GABA biosynthesis, and bred them to mice expressing Cre recombinase at the dopamine D1 receptor locus (Drd1a) to selectively reduce GABA synthesis in the direct output pathway from the striatum. We show that these mice are deficient in some types of motor skills, but normal for others, suggesting a differential role for GABA release from D1 receptor-containing neurons.
Collapse
Affiliation(s)
- Carrie L Heusner
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, USA
| | | | | | | |
Collapse
|
49
|
Kolber BJ, Wieczorek L, Muglia LJ. Hypothalamic-pituitary-adrenal axis dysregulation and behavioral analysis of mouse mutants with altered glucocorticoid or mineralocorticoid receptor function. Stress 2008; 11:321-38. [PMID: 18609295 PMCID: PMC2744095 DOI: 10.1080/10253890701821081] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Corticosteroid receptors are critical for the maintenance of homeostasis after both psychological and physiological stress. To understand the different roles and interactions of the glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) during stress, it is necessary to dissect the role of corticosteroid signaling at both the system and sub-system level. A variety of GR transgenic mouse lines have recently been used to characterize the role of GR in the CNS as a whole and particularly in the forebrain. We will describe both the behavioral and cellular/molecular implications of disrupting GR function in these animal models and describe the implications of this data for our understanding of normal endocrine function and stress adaptation. MRs in tight epithelia have a long established role in sodium homeostasis. Recently however, evidence has suggested that MRs in the limbic brain also play an important role in psychological stress. Just as with GR, targeted mutations in MR induce a variety of behavioral changes associated with stress adaptation. In this review, we will discuss the implications of this work on MR. Finally, we will discuss the possible interaction between MR and GR and how future work using double mutants (through conventional means or virus based gene alteration) will be needed to more fully understand how signaling through these two steroid receptors provides the adaptive mechanisms to deal with a variety of stressors.
Collapse
Affiliation(s)
- Benedict J. Kolber
- Departments of Pediatrics and Molecular Biology and Pharmacology and Program in Neuroscience, Washington University in St. Louis, St. Louis, MO 63110
| | - Lindsay Wieczorek
- Departments of Pediatrics and Molecular Biology and Pharmacology and Program in Neuroscience, Washington University in St. Louis, St. Louis, MO 63110
| | - Louis J. Muglia
- Departments of Pediatrics and Molecular Biology and Pharmacology and Program in Neuroscience, Washington University in St. Louis, St. Louis, MO 63110
- Corresponding Author: Louis J. Muglia, , Washington University in St. Louis, 660 S. Euclid, Box 8208, St. Louis, MO 63110
| |
Collapse
|
50
|
Chen S, Owens GC, Edelman DB. Dopamine inhibits mitochondrial motility in hippocampal neurons. PLoS One 2008; 3:e2804. [PMID: 18665222 PMCID: PMC2467486 DOI: 10.1371/journal.pone.0002804] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 07/05/2008] [Indexed: 01/24/2023] Open
Abstract
Background The trafficking of mitochondria within neurons is a highly regulated process. In an earlier study, we found that serotonin (5-HT), acting through the 5-HT1A receptor subtype, promotes axonal transport of mitochondria in cultured hippocampal neurons by increasing Akt activity, and consequently decreasing glycogen synthase kinase (GSK3β) activity. This finding suggests a critical role for neuromodulators in the regulation of mitochondrial trafficking in neurons. In the present study, we investigate the effects of a second important neuromodulator, dopamine, on mitochondrial transport in hippocampal neurons. Methodology/Principal Findings Here, we show that dopamine, like 5-HT, regulates mitochondrial motility in cultured hippocampal neurons through the Akt-GSK3β signaling cascade. But, in contrast to the stimulatory effect of 5-HT, administration of exogenous dopamine or bromocriptine, a dopamine 2 receptor (D2R) agonist, caused an inhibition of mitochondrial movement. Moreover, pretreatment with bromocriptine blocked the stimulatory effect of 5-HT on mitochondrial movement. Conversely, in cells pretreated with 5-HT, no further increases in movement were observed after administration of haloperidol, a D2R antagonist. In contrast to the effect of the D2R agonist, addition of SKF38393, a dopamine 1 receptor (D1R) agonist, promoted mitochondrial transport, indicating that the inhibitory effect of dopamine was actually the net summation of opposing influences of the two receptor subtypes. The most pronounced effect of dopamine signals was on mitochondria that were already moving directionally. Western blot analysis revealed that treatment with either a D2R agonist or a D1R antagonist decreased Akt activity, and conversely, treatment with either a D2R antagonist or a D1R agonist increased Akt activity. Conclusions/Significance Our observations strongly suggest a role for both dopamine and 5-HT in regulating mitochondrial movement, and indicate that the integrated effects of these two neuromodulators may be important in determining the distribution of energy sources in neurons.
Collapse
Affiliation(s)
- Sigeng Chen
- The Neurosciences Institute, San Diego, California, United States of America
| | - Geoffrey C. Owens
- The Neurosciences Institute, San Diego, California, United States of America
| | - David B. Edelman
- The Neurosciences Institute, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|