1
|
Sanson A, Demarchi L, Rocaboy E, Bosch OJ. Increased CRF-R1 transmission in the nucleus accumbens shell facilitates maternal neglect in lactating rats and mediates anxiety-like behaviour in a sex-specific manner. Neuropharmacology 2025; 265:110256. [PMID: 39647775 DOI: 10.1016/j.neuropharm.2024.110256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
During the transition to motherhood, complex brain adaptations occur to ensure adequate maternal responses to offspring' needs accompanied by reduced anxiety. Among others, the corticotropin-releasing factor (CRF) and oxytocin (OXT) systems have emerged as crucial regulators of these essential postpartum adaptations. Here, we investigated their roles within the nucleus accumbens shell (NAcSh), a central region of the reward and maternal circuits, in maternal neglect of lactating rats. Furthermore, we assessed the contribution of the local CRF system to anxiety-like behaviour, comparing lactating female, virgin female and male rats to evaluate potential sex-differences. Increasing CRF receptor (CRF-R) 1 transmission via local CRF infusion in the NAcSh led to maternal neglect, reducing nursing and increasing self-directed behaviours. In turn, local CRF-R1 inhibition impaired maternal motivation. Intra-NAcSh Urocortin3 infusion did not promote maternal neglect but increased anxiety-like behaviour in lactating and virgin female rats, whereas CRF infusion had anxiogenic effects only in male rats. Crh-r1 mRNA expression was higher in male and lactating rats compared to virgin females; furthermore, male rats had increased Crh-bp mRNA expression compared to virgin female rats, only. Lastly, pharmacological manipulations of the OXT system did not affect maternal responses. In conclusion, finely balanced CRF-R1 signalling in the NAcSh is required for the proper expression of maternal behaviours. Dampened CRF-R2 signalling prevents the onset of anxiety-like behaviour in female rats, whereas CRF-R1 plays a more prominent role in males, highlighting complex sex-differences of the CRF system's regulation of anxiety within the NAcSh.
Collapse
Affiliation(s)
- Alice Sanson
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Luisa Demarchi
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Emma Rocaboy
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Oliver J Bosch
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
2
|
Eckenwiler EA, Ingebretson AE, Stolley JJ, Fusaro MA, Romportl AM, Ross JM, Petersen CL, Kale EM, Clark MS, Schattauer SS, Zweifel LS, Lemos JC. Corticotropin-Releasing Factor Release From a Unique Subpopulation of Accumbal Neurons Constrains Action-Outcome Acquisition in Reward Learning. Biol Psychiatry 2024:S0006-3223(24)01534-8. [PMID: 39181385 DOI: 10.1016/j.biopsych.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The nucleus accumbens (NAc) mediates reward learning and motivation. Despite an abundance of neuropeptides, peptidergic neurotransmission from the NAc has not been integrated into current models of reward learning. The existence of a sparse population of neurons containing corticotropin-releasing factor (CRF) has been previously documented. Here, we provide a comprehensive analysis of their identity and functional role in shaping reward learning. METHODS Our multidisciplinary approach included fluorescent in situ hybridization (n = ≥3 mice), tract tracing (n = 5 mice), ex vivo electrophysiology (n = ≥30 cells), in vivo calcium imaging with fiber photometry (n = ≥4 mice), and use of viral strategies in transgenic lines to selectively delete CRF peptide from NAc neurons (n = ≥4 mice). Behaviors used were instrumental learning, sucrose preference, and spontaneous exploration in an open field. RESULTS We showed that the vast majority of NAc CRF-containing neurons are spiny projection neurons (SPNs) comprising dopamine D1-, D2-, or D1/D2-containing SPNs that primarily project and connect to the ventral pallidum and to a lesser extent the ventral midbrain. As a population, they display mature and immature SPN firing properties. We demonstrated that NAc CRF-containing neurons track reward outcomes during operant reward learning and that CRF release from these neurons acts to constrain initial acquisition of action-outcome learning and at the same time facilitates flexibility in the face of changing contingencies. CONCLUSIONS CRF release from this sparse population of SPNs is critical for reward learning under normal conditions.
Collapse
Affiliation(s)
- Elizabeth A Eckenwiler
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Anna E Ingebretson
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey J Stolley
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Maxine A Fusaro
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Alyssa M Romportl
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Jack M Ross
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Christopher L Petersen
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Eera M Kale
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota
| | - Michael S Clark
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington
| | - Selena S Schattauer
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington; Department of Pharmacology, University of Washington, Seattle, Washington
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington; Department of Pharmacology, University of Washington, Seattle, Washington
| | - Julia C Lemos
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, Minnesota; Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
3
|
Domin H, Śmiałowska M. The diverse role of corticotropin-releasing factor (CRF) and its CRF1 and CRF2 receptors under pathophysiological conditions: Insights into stress/anxiety, depression, and brain injury processes. Neurosci Biobehav Rev 2024; 163:105748. [PMID: 38857667 DOI: 10.1016/j.neubiorev.2024.105748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/12/2024]
Abstract
Corticotropin-releasing factor (CRF, corticoliberin) is a neuromodulatory peptide activating the hypothalamic-pituitary-adrenal (HPA) axis, widely distributed in the central nervous system (CNS) in mammals. In addition to its neuroendocrine effects, CRF is essential in regulating many functions under physiological and pathophysiological conditions through CRF1 and CRF2 receptors (CRF1R, CRF2R). This review aims to present selected examples of the diverse and sometimes opposite effects of CRF and its receptor ligands in various pathophysiological states, including stress/anxiety, depression, and processes associated with brain injury. It seems interesting to draw particular attention to the fact that CRF and its receptor ligands exert different effects depending on the brain structures or subregions, likely stemming from the varied distribution of CRFRs in these regions and interactions with other neurotransmitters. CRFR-mediated region-specific effects might also be related to brain site-specific ligand binding and the associated activated signaling pathways. Intriguingly, different types of CRF molecules can also influence the diverse actions of CRF in the CNS.
Collapse
Affiliation(s)
- Helena Domin
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Neurobiology, 12 Smętna Street, Kraków 31-343, Poland.
| | - Maria Śmiałowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Neurobiology, 12 Smętna Street, Kraków 31-343, Poland
| |
Collapse
|
4
|
Ingebretson AE, Alonso-Caraballo Y, Razidlo JA, Lemos JC. Corticotropin releasing factor alters the functional diversity of accumbal cholinergic interneurons. J Neurophysiol 2024; 132:403-417. [PMID: 39106208 PMCID: PMC11427051 DOI: 10.1152/jn.00348.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 08/09/2024] Open
Abstract
Cholinergic interneurons (ChIs) provide the main source of acetylcholine in the striatum and have emerged as a critical modulator of behavioral flexibility, motivation, and associative learning. In the dorsal striatum (DS), ChIs display heterogeneous firing patterns. Here, we investigated the spontaneous firing patterns of ChIs in the nucleus accumbens (NAc) shell, a region of the ventral striatum. We identified four distinct ChI firing signatures: regular single-spiking, irregular single-spiking, rhythmic bursting, and a mixed-mode pattern composed of bursting activity and regular single spiking. ChIs from females had lower firing rates compared with males and had both a higher proportion of mixed-mode firing patterns and a lower proportion of regular single-spiking neurons compared with males. We further observed that across the estrous cycle, the diestrus phase was characterized by higher proportions of irregular ChI firing patterns compared with other phases. Using pooled data from males and females, we examined how the stress-associated neuropeptide corticotropin releasing factor (CRF) impacts these firing patterns. ChI firing patterns showed differential sensitivity to CRF. This translated into differential ChI sensitivity to CRF across the estrous cycle. Furthermore, CRF shifted the proportion of ChI firing patterns toward more regular spiking activity over bursting patterns. Finally, we found that repeated stressor exposure altered ChI firing patterns and sensitivity to CRF in the NAc core, but not the NAc shell. These findings highlight the heterogeneous nature of ChI firing patterns, which may have implications for accumbal-dependent motivated behaviors.NEW & NOTEWORTHY Cholinergic interneurons (ChIs) within the dorsal and ventral striatum can exert a major influence on network output and motivated behaviors. However, the firing patterns and neuromodulation of ChIs within the ventral striatum, specifically the nucleus accumbens (NAc) shell, are understudied. Here, we report that NAc shell ChIs have heterogeneous ChI firing patterns that are labile and can be modulated by the stress-linked neuropeptide corticotropin releasing factor (CRF) and by the estrous cycle.
Collapse
Affiliation(s)
- Anna E Ingebretson
- Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, United States
| | - Yanaira Alonso-Caraballo
- Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, United States
| | - John A Razidlo
- Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, United States
| | - Julia C Lemos
- Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
5
|
Eckenwiler EA, Ingebretson AE, Stolley JJ, Fusaro MA, Romportl AM, Ross JM, Petersen CL, Kale EM, Clark MS, Schattauer SS, Zweifel LS, Lemos JC. CRF release from a unique subpopulation of accumbal neurons constrains action-outcome acquisition in reward learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.16.567495. [PMID: 39005420 PMCID: PMC11244858 DOI: 10.1101/2023.11.16.567495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background The nucleus accumbens (NAc) mediates reward learning and motivation. Despite an abundance of neuropeptides, peptidergic neurotransmission from the NAc has not been integrated into current models of reward learning. The existence of a sparse population of neurons containing corticotropin releasing factor (CRF) has been previously documented. Here we provide a comprehensive analysis of their identity and functional role in shaping reward learning. Methods To do this, we took a multidisciplinary approach that included florescent in situ hybridization (N mice ≥ 3), tract tracing (N mice = 5), ex vivo electrophysiology (N cells ≥ 30), in vivo calcium imaging with fiber photometry (N mice ≥ 4) and use of viral strategies in transgenic lines to selectively delete CRF peptide from NAc neurons (N mice ≥ 4). Behaviors used were instrumental learning, sucrose preference and spontaneous exploration in an open field. Results Here we show that the vast majority of NAc CRF-containing (NAc CRF ) neurons are spiny projection neurons (SPNs) comprised of dopamine D1-, D2- or D1/D2-containing SPNs that primarily project and connect to the ventral pallidum and to a lesser extent the ventral midbrain. As a population, they display mature and immature SPN firing properties. We demonstrate that NAc CRF neurons track reward outcomes during operant reward learning and that CRF release from these neurons acts to constrain initial acquisition of action-outcome learning, and at the same time facilitates flexibility in the face of changing contingencies. Conclusion We conclude that CRF release from this sparse population of SPNs is critical for reward learning under normal conditions.
Collapse
|
6
|
Zhao W, Yu YM, Wang XY, Xia SH, Ma Y, Tang H, Tao M, Li H, Xu Z, Yang JX, Wu P, Zhang H, Ding HL, Cao JL. CRF regulates pain sensation by enhancement of corticoaccumbal excitatory synaptic transmission. Mol Psychiatry 2024; 29:2170-2184. [PMID: 38454083 DOI: 10.1038/s41380-024-02488-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 02/08/2024] [Accepted: 02/14/2024] [Indexed: 03/09/2024]
Abstract
Both peripheral and central corticotropin-releasing factor (CRF) systems have been implicated in regulating pain sensation. However, compared with the peripheral, the mechanisms underlying central CRF system in pain modulation have not yet been elucidated, especially at the neural circuit level. The corticoaccumbal circuit, a structure rich in CRF receptors and CRF-positive neurons, plays an important role in behavioral responses to stressors including nociceptive stimuli. The present study was designed to investigate whether and how CRF signaling in this circuit regulated pain sensation under physiological and pathological pain conditions. Our studies employed the viral tracing and circuit-, and cell-specific electrophysiological methods to label the CRF-containing circuit from the medial prefrontal cortex to the nucleus accumbens shell (mPFCCRF-NAcS) and record its neuronal propriety. Combining optogenetic and chemogenetic manipulation, neuropharmacological methods, and behavioral tests, we were able to precisely manipulate this circuit and depict its role in regulation of pain sensation. The current study found that the CRF signaling in the NAc shell (NAcS), but not NAc core, was necessary and sufficient for the regulation of pain sensation under physiological and pathological pain conditions. This process was involved in the CRF-mediated enhancement of excitatory synaptic transmission in the NAcS. Furthermore, we demonstrated that the mPFCCRF neurons monosynaptically connected with the NAcS neurons. Chronic pain increased the protein level of CRF in NAcS, and then maintained the persistent NAcS neuronal hyperactivity through enhancement of this monosynaptic excitatory connection, and thus sustained chronic pain behavior. These findings reveal a novel cell- and circuit-based mechanistic link between chronic pain and the mPFCCRF → NAcS circuit and provide a potential new therapeutic target for chronic pain.
Collapse
Affiliation(s)
- Weinan Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yu-Mei Yu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xiao-Yi Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Department of Anesthesiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Sun-Hui Xia
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yu Ma
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Huimei Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Mingshu Tao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - He Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Zheng Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Peng Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
7
|
Ingebretson AE, Alonso-Caraballo Y, Razidlo JA, Lemos JC. Corticotropin releasing factor alters the functional diversity of accumbal cholinergic interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.17.558116. [PMID: 37745598 PMCID: PMC10516029 DOI: 10.1101/2023.09.17.558116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Cholinergic interneurons (ChIs) provide the main source of acetylcholine in the striatum and have emerged as a critical modulator of behavioral flexibility, motivation, and associative learning. In the dorsal striatum, ChIs display heterogeneous firing patterns. Here, we investigated the spontaneous firing patterns of ChIs in the nucleus accumbens (NAc) shell, a region of the ventral striatum. We identified four distinct ChI firing signatures: regular single-spiking, irregular single-spiking, rhythmic bursting, and a mixed-mode pattern composed of bursting activity and regular single spiking. ChIs from females had lower firing rates compared to males and had both a higher proportion of mixed-mode firing patterns and a lower proportion of regular single-spiking neurons compared to males. We further observed that across the estrous cycle, the diestrus phase was characterized by higher proportions of irregular ChI firing patterns compared to other phases. Using pooled data from males and females, we examined how the stress-associated neuropeptide corticotropin releasing factor (CRF) impacts these firing patterns. ChI firing patterns showed differential sensitivity to CRF. This translated into differential ChI sensitivity to CRF across the estrous cycle. Furthermore, CRF shifted the proportion of ChI firing patterns toward more regular spiking activity over bursting patterns. Finally, we found that repeated stressor exposure altered ChI firing patterns and sensitivity to CRF in the NAc core, but not the NAc shell. These findings highlight the heterogeneous nature of ChI firing patterns, which may have implications for accumbal-dependent motivated behaviors.
Collapse
|
8
|
Gozen O, Aypar B, Ozturk Bintepe M, Tuzcu F, Balkan B, Koylu EO, Kanit L, Keser A. Chronic Nicotine Consumption and Withdrawal Regulate Melanocortin Receptor, CRF, and CRF Receptor mRNA Levels in the Rat Brain. Brain Sci 2024; 14:63. [PMID: 38248278 PMCID: PMC10813117 DOI: 10.3390/brainsci14010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
Alterations in the various neuropeptide systems in the mesocorticolimbic circuitry have been implicated in negative effects associated with drug withdrawal. The corticotropin-releasing factor (CRF) and α-melanocyte-stimulating hormone are two peptides that may be involved. This study investigated the regulatory effects of chronic nicotine exposure and withdrawal on the mRNA levels of melanocortin receptors (MC3R, MC4R), CRF, and CRF receptors (CRFR1 and CRFR2) expressed in the mesocorticolimbic system. Rats were given drinking water with nicotine or without nicotine (control group) for 12 weeks, after which they continued receiving nicotine (chronic exposure) or were withdrawn from nicotine for 24 or 48 h. The animals were decapitated following behavioral testing for withdrawal signs. Quantitative real-time PCR analysis demonstrated that nicotine exposure (with or without withdrawal) increased levels of CRF and CRFR1 mRNA in the amygdala, CRF mRNA in the medial prefrontal cortex, and CRFR1 mRNA in the septum. Nicotine withdrawal also enhanced MC3R and MC4R mRNA levels in different brain regions, while chronic nicotine exposure was associated with increased MC4R mRNA levels in the nucleus accumbens. These results suggest that chronic nicotine exposure and withdrawal regulate CRF and melanocortin signaling in the mesocorticolimbic system, possibly contributing to negative affective state and nicotine addiction.
Collapse
Affiliation(s)
- Oguz Gozen
- Department of Physiology, School of Medicine, Ege University, 35100 Izmir, Turkey; (O.G.)
- Center for Brain Research, Ege University, 35100 Izmir, Turkey
| | - Buket Aypar
- Department of Physiology, School of Medicine, Ege University, 35100 Izmir, Turkey; (O.G.)
| | - Meliha Ozturk Bintepe
- Department of Physiology, School of Medicine, Ege University, 35100 Izmir, Turkey; (O.G.)
| | - Fulya Tuzcu
- Department of Physiology, School of Medicine, Ege University, 35100 Izmir, Turkey; (O.G.)
| | - Burcu Balkan
- Department of Physiology, School of Medicine, Ege University, 35100 Izmir, Turkey; (O.G.)
- Center for Brain Research, Ege University, 35100 Izmir, Turkey
| | - Ersin O. Koylu
- Department of Physiology, School of Medicine, Ege University, 35100 Izmir, Turkey; (O.G.)
- Center for Brain Research, Ege University, 35100 Izmir, Turkey
| | - Lutfiye Kanit
- Department of Physiology, School of Medicine, Ege University, 35100 Izmir, Turkey; (O.G.)
- Center for Brain Research, Ege University, 35100 Izmir, Turkey
| | - Aysegul Keser
- Department of Physiology, School of Medicine, Ege University, 35100 Izmir, Turkey; (O.G.)
- Center for Brain Research, Ege University, 35100 Izmir, Turkey
| |
Collapse
|
9
|
Castro EM, Lotfipour S, Leslie FM. Nicotine on the developing brain. Pharmacol Res 2023; 190:106716. [PMID: 36868366 PMCID: PMC10392865 DOI: 10.1016/j.phrs.2023.106716] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Developmental periods such as gestation and adolescence have enhanced plasticity leaving the brain vulnerable to harmful effects from nicotine use. Proper brain maturation and circuit organization is critical for normal physiological and behavioral outcomes. Although cigarette smoking has declined in popularity, noncombustible nicotine products are readily used. The misperceived safety of these alternatives lead to widespread use among vulnerable populations such as pregnant women and adolescents. Nicotine exposure during these sensitive developmental windows is detrimental to cardiorespiratory function, learning and memory, executive function, and reward related circuitry. In this review, we will discuss clinical and preclinical evidence of the adverse alterations in the brain and behavior following nicotine exposure. Time-dependent nicotine-induced changes in reward related brain regions and drug reward behaviors will be discussed and highlight unique sensitivities within a developmental period. We will also review long lasting effects of developmental exposure persisting into adulthood, along with permanent epigenetic changes in the genome which can be passed to future generations. Taken together, it is critical to evaluate the consequences of nicotine exposure during these vulnerable developmental windows due to its direct impact on cognition, potential trajectories for other substance use, and implicated mechanisms for the neurobiology of substance use disorders.
Collapse
Affiliation(s)
- Emily M Castro
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Emergency Medicine, School of Medicine, University of California, Irvine, Irvine, CA, USA; Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Frances M Leslie
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
10
|
Zegers-Delgado J, Aguilera-Soza A, Calderón F, Davidson H, Verbel-Vergara D, Yarur HE, Novoa J, Blanlot C, Bastias CP, Andrés ME, Gysling K. Type 1 Corticotropin-Releasing Factor Receptor Differentially Modulates Neurotransmitter Levels in the Nucleus Accumbens of Juvenile versus Adult Rats. Int J Mol Sci 2022; 23:ijms231810800. [PMID: 36142716 PMCID: PMC9505341 DOI: 10.3390/ijms231810800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Adversity is particularly pernicious in early life, increasing the likelihood of developing psychiatric disorders in adulthood. Juvenile and adult rats exposed to social isolation show differences in anxiety-like behaviors and significant changes in dopamine (DA) neurotransmission in the nucleus accumbens (NAc). Brain response to stress is partly mediated by the corticotropin-releasing factor (CRF) system, composed of CRF and its two main receptors, CRF-R1 and CRF-R2. In the NAc shell of adult rats, CRF induces anxiety-like behavior and changes local DA balance. However, the role of CRF receptors in the control of neurotransmission in the NAc is not fully understood, nor is it known whether there are differences between life stages. Our previous data showed that infusion of a CRF-R1 antagonist into the NAc of juvenile rats increased DA levels in response to a depolarizing stimulus and decreased basal glutamate levels. To extend this analysis, we now evaluated the effect of a CRF-R1 antagonist infusion in the NAc of adult rats. Here, we describe that the opposite occurred in the NAc of adult compared to juvenile rats. Infusion of a CRF-R1 antagonist decreased DA and increased glutamate levels in response to a depolarizing stimulus. Furthermore, basal levels of DA, glutamate, and γ-Aminobutyric acid (GABA) were similar in juvenile animals compared to adults. CRF-R1 protein levels and localization were not different in juvenile compared to adult rats. Interestingly, we observed differences in the signaling pathways of CRF-R1 in the NAc of juveniles compared to adult rats. We propose that the function of CRF-R1 receptors is differentially modulated in the NAc according to life stage.
Collapse
|
11
|
Mazaheri S, Zendehdel M, Haghparast A. Role of orexinergic receptors within the ventral tegmental area in the development of morphine sensitization induced by forced swim stress in the rat. Prog Neuropsychopharmacol Biol Psychiatry 2022; 116:110539. [PMID: 35217126 DOI: 10.1016/j.pnpbp.2022.110539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/05/2023]
Abstract
The ventral tegmental area (VTA) has been suggested as part of a common system for reward, stress, and morphine sensitization. Repeated exposure to stress enhances sensitivity to drugs such as morphine. The role of orexin receptor type 1 (OX1R) and type 2 (OX2R) within the VTA in cross-sensitization of morphine with stress was assessed in this study. Various doses of OX1R antagonist (SB334867) and OX2R antagonist (TCS OX2 29) were microinjected into the VTA of 134 adult male albino Wistar rats through cannulae, which had been bilaterally implanted above this region. Five min after microinjection, animals were forced to swim for 6 min, and 10 min after forced swim stress (FSS) termination, a low dose of morphine (i.e., ineffective dose for sensitization) was subcutaneously injected (1 mg/kg; sc). This procedure was repeated for three consecutive days as a sensitization period followed by a 5-day drug/stress-free period. On the 9th day, sensitivity to morphine was examined by measuring antinociceptive responses to the ineffective dose of morphine via tail-flick test. The obtained findings revealed that while concurrent administration of FSS and an ineffective dose of morphine (1 mg/kg; sc) for three consecutive days induced sensitivity to morphine, intra-VTA administration of OX1R- and OX2R antagonists, dose-dependently blocked this sensitization. These results suggested that both orexin receptors located in the VTA have a considerable role in morphine sensitization induced by concurrent administration of FSS and a low dose of morphine. So, there is a contribution of the orexin system partly to stress-induced sensitization to morphine.
Collapse
Affiliation(s)
- Sajad Mazaheri
- Department of Physiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Morteza Zendehdel
- Department of Physiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Morin A, Poitras M, Plamondon H. Global Cerebral Ischemia in Male Long Evans Rats Impairs Dopaminergic/ΔFosB Signalling in the Mesocorticolimbic Pathway Without Altering Delay Discounting Rates. Front Behav Neurosci 2022; 15:770374. [PMID: 35058756 PMCID: PMC8763703 DOI: 10.3389/fnbeh.2021.770374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/18/2021] [Indexed: 11/24/2022] Open
Abstract
Global cerebral ischemia (GCI) in rats has been shown to promote exploration of anxiogenic zones of the Elevated-Plus Maze (EPM) and Open Field Test (OFT). This study investigated changes in impulsive choice and/or defensive responses as possible contributors of heightened anxiogenic exploration observed after ischemia. Impulsivity was assessed using delay discounting (DD) paradigms, while the Predator Odour Test (PO) served to assess changes in defensive responses towards a naturally aversive stimulus. Male Long Evans rats underwent 9 days of autoshaping training and 24 days of DD training prior to GCI or sham surgery (n = 9/group). Post-surgery, rats completed the OFT, EPM, and PO, followed by 6 days of DD sessions. Blood droplets served to evaluate corticosterone secretion associated with PO exposure. With impulsivity being regulated through mesocorticolimbic monoaminergic pathways, we also characterised post-ischemic changes in the expression of dopamine D2 receptors (DRD2), dopamine transporters (DAT), and 1FosB in the basolateral amygdala (BLA), nucleus accumbens core (NAcC) and shell (NAcS), and ventromedial prefrontal cortex (vmPFC) using immunohistofluorescence. Our findings revealed no impact of GCI on delay discounting rates, while PO approach behaviours were minimally affected. Nonetheless, GCI significantly reduced DRD2 and ΔFosB-ir in the NAcS and NAcC, respectively, while DAT-ir was diminished in both NAc subregions. Collectively, our findings refine the understanding of cognitive-behavioural and biochemical responses following stroke or cardiac arrest. They support significant alterations to the dopaminergic mesocorticolimbic pathway after ischemia, which are not associated with altered impulsive choice in a DD task but may influence locomotor exploration of the OFT and EPM.
Collapse
|
13
|
Dissociable roles of the nucleus accumbens core and shell subregions in the expression and extinction of conditioned fear. Neurobiol Stress 2021; 15:100365. [PMID: 34355048 PMCID: PMC8319794 DOI: 10.1016/j.ynstr.2021.100365] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/23/2021] [Accepted: 07/09/2021] [Indexed: 12/25/2022] Open
Abstract
The nucleus accumbens (NAc), consisting of core (NAcC) and shell (NAcS) sub-regions, has primarily been studied as a locus mediating the effects of drug reward and addiction. However, there is ample evidence that this region is also involved in regulating aversive responses, but the exact role of the NAc and its subregions in regulating associative fear processing remains unclear. Here, we investigated the specific contribution of the NAcC and NAcS in regulating both fear expression and fear extinction in C57BL/6J mice. Using Arc expression as an indicator of neuronal activity, we first show that the NAcC is specifically active only in response to an associative fear cue during an expression test. In contrast, the NAcS is specifically active during fear extinction. We next inactivated each subregion using lidocaine and demonstrated that the NAcC is necessary for fear expression, but not for extinction learning or consolidation of extinction. In contrast, we demonstrate that the NAcS is necessary for the consolidation of extinction, but not fear expression or extinction learning. Further, inactivation of mGluR1 or ERK signaling specifically in the NAcS disrupted the consolidation of extinction but had no effect on fear expression or extinction learning itself. Our data provide the first evidence for the importance of the ERK/MAPK pathway as the underlying neural mechanism facilitating extinction consolidation within the NAcS. These findings suggest that the NAc subregions play dissociable roles in regulating fear recall and the consolidation of fear extinction, and potentially implicate them as critical regions within the canonical fear circuit.
Collapse
|
14
|
McCorkle TA, Barson JR, Raghupathi R. A Role for the Amygdala in Impairments of Affective Behaviors Following Mild Traumatic Brain Injury. Front Behav Neurosci 2021; 15:601275. [PMID: 33746719 PMCID: PMC7969709 DOI: 10.3389/fnbeh.2021.601275] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/29/2021] [Indexed: 11/30/2022] Open
Abstract
Mild traumatic brain injury (TBI) results in chronic affective disorders such as depression, anxiety, and fear that persist up to years following injury and significantly impair the quality of life for patients. Although a great deal of research has contributed to defining symptoms of mild TBI, there are no adequate drug therapies for brain-injured individuals. Preclinical studies have modeled these deficits in affective behaviors post-injury to understand the underlying mechanisms with a view to developing appropriate treatment strategies. These studies have also unveiled sex differences that contribute to the varying phenotypes associated with each behavior. Although clinical and preclinical studies have viewed these behavioral deficits as separate entities with unique neurobiological mechanisms, mechanistic similarities suggest that a novel approach is needed to advance research on drug therapy. This review will discuss the circuitry involved in the expression of deficits in affective behaviors following mild TBI in humans and animals and provide evidence that the manifestation of impairment in these behaviors stems from an amygdala-dependent emotional processing deficit. It will highlight mechanistic similarities between these different types of affective behaviors that can potentially advance mild TBI drug therapy by investigating treatments for the deficits in affective behaviors as one entity, requiring the same treatment.
Collapse
Affiliation(s)
- Taylor A. McCorkle
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jessica R. Barson
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA, United States
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ramesh Raghupathi
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA, United States
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
15
|
Hudson R, Green M, Wright DJ, Renard J, Jobson CEL, Jung T, Rushlow W, Laviolette SR. Adolescent nicotine induces depressive and anxiogenic effects through ERK 1-2 and Akt-GSK-3 pathways and neuronal dysregulation in the nucleus accumbens. Addict Biol 2021; 26:e12891. [PMID: 32135573 DOI: 10.1111/adb.12891] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 02/11/2020] [Accepted: 02/15/2020] [Indexed: 12/16/2022]
Abstract
Long-term tobacco dependence typically develops during adolescence and neurodevelopmental nicotine exposure is associated with affective disturbances that manifest as a variety of neuropsychiatric comorbidities in clinical and preclinical studies, including mood and anxiety-related disorders. The nucleus accumbens shell (NASh) is critically involved in regulating emotional processing, and both molecular and neuronal disturbances in this structure are associated with mood and anxiety-related pathologies. In the present study, we used a rodent model of adolescent neurodevelopmental nicotine exposure to examine the expression of several molecular biomarkers associated with mood/anxiety-related phenotypes. We report that nicotine exposure during adolescence (but not adulthood) induces profound upregulation of the ERK 1-2 and Akt-GSK-3 signalling pathways directly within the NASh, as well as downregulation of local D1R expression that persists into adulthood. These adaptations were accompanied by decreases in τ, α, β, and γ-band oscillatory states, hyperactive medium spiny neuron activity with depressed bursting rates, and anxiety and depressive-like behavioural abnormalities. Pharmacologically targeting these molecular and neuronal adaptations revealed that selective inhibition of local ERK 1-2 and Akt-GSK-3 signalling cascades rescued nicotine-induced high-γ-band oscillatory signatures and phasic bursting rates in the NASh, suggesting that they are involved in mediating adolescent nicotine-induced depressive and anxiety-like neuropathological trajectories.
Collapse
Affiliation(s)
- Roger Hudson
- Addiction Research Group, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario London, London, Ontario, Canada
| | - Matthew Green
- Addiction Research Group, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario London, London, Ontario, Canada
| | - Daniel J Wright
- Addiction Research Group, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario London, London, Ontario, Canada
| | - Justine Renard
- Addiction Research Group, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario London, London, Ontario, Canada
| | - Christina E L Jobson
- Addiction Research Group, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario London, London, Ontario, Canada
| | - Tony Jung
- Addiction Research Group, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario London, London, Ontario, Canada
| | - Walter Rushlow
- Addiction Research Group, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario London, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario London, London, Ontario, Canada
| | - Steven R Laviolette
- Addiction Research Group, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario London, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario London, London, Ontario, Canada
| |
Collapse
|
16
|
Curtis GR, Oakes K, Barson JR. Expression and Distribution of Neuropeptide-Expressing Cells Throughout the Rodent Paraventricular Nucleus of the Thalamus. Front Behav Neurosci 2021; 14:634163. [PMID: 33584216 PMCID: PMC7873951 DOI: 10.3389/fnbeh.2020.634163] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022] Open
Abstract
The paraventricular nucleus of the thalamus (PVT) has been shown to make significant contributions to affective and motivated behavior, but a comprehensive description of the neurochemicals expressed in the cells of this brain region has never been presented. While the PVT is believed to be composed of projection neurons that primarily use as their neurotransmitter the excitatory amino acid, glutamate, several neuropeptides have also been described in this brain region. In this review article, we combine published literature with our observations from the Allen Brain Atlas to describe in detail the expression and distribution of neuropeptides in cells throughout the mouse and rat PVT, with a special focus on neuropeptides known to be involved in behavior. Several themes emerge from this investigation. First, while the majority of neuropeptides are expressed across the antero-posterior axis of the PVT, they generally exist in a gradient, in which expression is most dense but not exclusive in either the anterior or posterior PVT, although other neuropeptides display somewhat more equal expression in the anterior and posterior PVT but have reduced expression in the middle PVT. Second, we find overall that neuropeptides involved in arousal are more highly expressed in the anterior PVT, those involved in depression-like behavior are more highly expressed in the posterior PVT, and those involved in reward are more highly expressed in the medial PVT, while those involved in the intake of food and drugs of abuse are distributed throughout the PVT. Third, the pattern and content of neuropeptide expression in mice and rats appear not to be identical, and many neuropeptides found in the mouse PVT have not yet been demonstrated in the rat. Thus, while significantly more work is required to uncover the expression patterns and specific roles of individual neuropeptides in the PVT, the evidence thus far supports the existence of a diverse yet highly organized system of neuropeptides in this nucleus. Determined in part by their location within the PVT and their network of projections, the function of the neuropeptides in this system likely involves intricate coordination to influence both affective and motivated behavior.
Collapse
Affiliation(s)
- Genevieve R Curtis
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Kathleen Oakes
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jessica R Barson
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
17
|
Plasticity of the Reward Circuitry After Early-Life Adversity: Mechanisms and Significance. Biol Psychiatry 2020; 87:875-884. [PMID: 32081365 PMCID: PMC7211119 DOI: 10.1016/j.biopsych.2019.12.018] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/25/2019] [Accepted: 12/11/2019] [Indexed: 12/24/2022]
Abstract
Disrupted operation of the reward circuitry underlies many aspects of affective disorders. Such disruption may manifest as aberrant behavior including risk taking, depression, anhedonia, and addiction. Early-life adversity is a common antecedent of adolescent and adult affective disorders involving the reward circuitry. However, whether early-life adversity influences the maturation and operations of the reward circuitry, and the potential underlying mechanisms, remain unclear. Here, we present novel information using cutting-edge technologies in animal models to dissect out the mechanisms by which early-life adversity provokes dysregulation of the complex interactions of stress and reward circuitries. We propose that certain molecularly defined pathways within the reward circuitry are particularly susceptible to early-life adversity. We examine regions and pathways expressing the stress-sensitive peptide corticotropin-releasing factor (CRF), which has been identified in critical components of the reward circuitry and interacting stress circuits. Notably, CRF is strongly modulated by early-life adversity in several of these brain regions. Focusing on amygdala nuclei and their projections, we provide evidence suggesting that aberrant CRF expression and function may underlie augmented connectivity of the nucleus accumbens with fear/anxiety regions, disrupting the function of this critical locus of pleasure and reward.
Collapse
|
18
|
Bryce CA, Floresco SB. Alterations in effort-related decision-making induced by stimulation of dopamine D 1, D 2, D 3, and corticotropin-releasing factor receptors in nucleus accumbens subregions. Psychopharmacology (Berl) 2019; 236:2699-2712. [PMID: 30972447 DOI: 10.1007/s00213-019-05244-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/01/2019] [Indexed: 10/27/2022]
Abstract
RATIONALE Nucleus accumbens (NAc) dopamine (DA) plays an integral role in overcoming effort costs, as blockade of D1 and D2 receptors reduces the choice of larger, more-costly rewards. Similarly, the stress neuropeptide corticotropin-releasing factor (CRF) modulates DA transmission and mediates stress-induced alterations in effort-related choice. OBJECTIVES The current study explored how excessive stimulation of different DA receptors within the NAc core and shell alters effort-related decision-making and compared these effects to those induced by CRF stimulation. METHODS Male Long Evans rats were well-trained on an effort-discounting task wherein they choose between a low-effort/low-reward and a high-effort/high-reward lever where the effort requirement increased over blocks (2-20 presses). Dopamine D1 (SKF 81297, 0.2-2 μg), D2/3 (quinpirole, 1-10 μg), or D3 (PD 128,907, 1.5-3 μg) receptor agonists, or CRF (0.5 μg), were infused into the NAc core or shell prior to testing. RESULTS Stimulation of D2/3 receptors with quinpirole in the NAc core or shell markedly reduced the choice of high-effort option and increase choice latencies, without altering preference for larger vs smaller rewards. Stimulation of D1 or D3 receptors did not alter choice, although SKF 81297 infusions into the shell reduced response vigor. In comparison, core infusions of CRF flattened the discounting curve, reducing effortful choice when costs were low and increasing it when costs were high. CONCLUSIONS Excessive stimulation of NAc D2 receptors has detrimental effects on effort-related decision-making. Furthermore, CRF stimulation induces dissociable effects on decision-making compared with those induced the effects of stimulation of different DA receptors.
Collapse
Affiliation(s)
- Courtney A Bryce
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2136 West Mall, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2136 West Mall, Vancouver, British Columbia, V6T 1Z4, Canada.
| |
Collapse
|
19
|
Collins AL, Aitken TJ, Huang IW, Shieh C, Greenfield VY, Monbouquette HG, Ostlund SB, Wassum KM. Nucleus Accumbens Cholinergic Interneurons Oppose Cue-Motivated Behavior. Biol Psychiatry 2019; 86:388-396. [PMID: 30955842 PMCID: PMC7003647 DOI: 10.1016/j.biopsych.2019.02.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Environmental reward-predictive stimuli provide a major source of motivation for adaptive reward pursuit behavior. This cue-motivated behavior is known to be mediated by the nucleus accumbens (NAc) core. The cholinergic interneurons in the NAc are tonically active and densely arborized and thus well suited to modulate NAc function. However, their causal contribution to adaptive behavior remains unknown. Here we investigated the function of NAc cholinergic interneurons in cue-motivated behavior. METHODS We used chemogenetics, optogenetics, pharmacology, and a translationally analogous Pavlovian-to-instrumental transfer behavioral task designed to assess the motivating influence of a reward-predictive cue over reward-seeking actions in male and female rats. RESULTS The data show that NAc cholinergic interneuron activity critically opposes the motivating influence of appetitive cues. Chemogenetic inhibition of NAc cholinergic interneurons augmented cue-motivated behavior. Optical stimulation of acetylcholine release from NAc cholinergic interneurons prevented cues from invigorating reward-seeking behavior, an effect that was mediated by activation of β2-containing nicotinic acetylcholine receptors. CONCLUSIONS NAc cholinergic interneurons provide a critical regulatory influence over adaptive cue-motivated behavior and therefore are a potential therapeutic target for the maladaptive cue-motivated behavior that marks many psychiatric conditions, including addiction and depression.
Collapse
Affiliation(s)
- Anne L Collins
- Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Tara J Aitken
- Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - I-Wen Huang
- Department of Chemical Engineering, University of California, Los Angeles, Los Angeles, California
| | - Christine Shieh
- Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Venuz Y Greenfield
- Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Harold G Monbouquette
- Department of Chemical Engineering, University of California, Los Angeles, Los Angeles, California
| | - Sean B Ostlund
- Department of Anesthesiology and Perioperative Care, University of California, Irvine, Irvine, California
| | - Kate M Wassum
- Department of Psychology, University of California, Los Angeles, Los Angeles, California; Brain Research Institute, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
20
|
Striatal Cholinergic Interneurons Are a Novel Target of Corticotropin Releasing Factor. J Neurosci 2019; 39:5647-5661. [PMID: 31109960 DOI: 10.1523/jneurosci.0479-19.2019] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/03/2019] [Accepted: 05/11/2019] [Indexed: 12/21/2022] Open
Abstract
Cholinergic interneurons (CINs) are critical regulators of striatal network activity and output. Changes in CIN activity are thought to encode salient changes in the environment and stimulus-response-outcome associations. Here we report that the stress-associated neuropeptide corticotropin releasing factor (CRF) produces a profound and reliable increase in the spontaneous firing of CINs in both dorsal striatum and nucleus accumbens (NAc) through activation of CRF type 1 receptors, production of cAMP and reduction in spike accommodation in male mice. The increase of CIN firing by CRF results in the activation muscarinic acetylcholine receptors type 5, which mediate potentiation of dopamine transmission in the striatum. This study provides critical mechanistic insight into how CRF modulates striatal activity and dopamine transmission in the NAc to likely account for CRF facilitation of appetitive behaviors.SIGNIFICANCE STATEMENT Although the presence of CRF receptors in the dorsal and ventral striatum has been acknowledged, the cellular identity and the functional consequences of receptor activation is unknown. Here we report that striatal cholinergic interneurons express CRF-R1 receptors and are acutely activated by the neuropeptide CRF that is released in response to salient environmental stimuli. Cholinergic interneurons make <1% of the cells in the striatum but are critical regulators of the striatal circuitry and its output. CRF's fast and potent activation of cholinergic interneurons could have far reaching behavioral implications across motivated behaviors controlled by the striatum.
Collapse
|
21
|
Castro DC, Bruchas MR. A Motivational and Neuropeptidergic Hub: Anatomical and Functional Diversity within the Nucleus Accumbens Shell. Neuron 2019; 102:529-552. [PMID: 31071288 PMCID: PMC6528838 DOI: 10.1016/j.neuron.2019.03.003] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/22/2019] [Accepted: 03/01/2019] [Indexed: 01/14/2023]
Abstract
The mesocorticolimbic pathway is canonically known as the "reward pathway." Embedded within the center of this circuit is the striatum, a massive and complex network hub that synthesizes motivation, affect, learning, cognition, stress, and sensorimotor information. Although striatal subregions collectively share many anatomical and functional similarities, it has become increasingly clear that it is an extraordinarily heterogeneous region. In particular, the nucleus accumbens (NAc) medial shell has repeatedly demonstrated that the rules dictated by more dorsal aspects of the striatum do not apply or are even reversed in functional logic. These discrepancies are perhaps most easily captured when isolating the functions of various neuromodulatory peptide systems within the striatum. Endogenous peptides are thought to play a critical role in modulating striatal signals to either amplify or dampen evoked behaviors. Here we describe the anatomical-functional backdrop upon which several neuropeptides act within the NAc to modulate behavior, with a specific emphasis on nucleus accumbens medial shell and stress responsivity. Additionally, we propose that, as the field continues to dissect fast neurotransmitter systems within the NAc, we must also provide considerable contextual weight to the roles local peptides play in modulating these circuits to more comprehensively understand how this important subregion gates motivated behaviors.
Collapse
Affiliation(s)
- Daniel C Castro
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Michael R Bruchas
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
22
|
Itoga CA, Chen Y, Fateri C, Echeverry PA, Lai JM, Delgado J, Badhon S, Short A, Baram TZ, Xu X. New viral-genetic mapping uncovers an enrichment of corticotropin-releasing hormone-expressing neuronal inputs to the nucleus accumbens from stress-related brain regions. J Comp Neurol 2019; 527:2474-2487. [PMID: 30861133 DOI: 10.1002/cne.24676] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/14/2019] [Accepted: 03/04/2019] [Indexed: 12/18/2022]
Abstract
Corticotropin-releasing hormone (CRH) is an essential, evolutionarily-conserved stress neuropeptide. In addition to hypothalamus, CRH is expressed in brain regions including amygdala and hippocampus where it plays crucial roles in modulating the function of circuits underlying emotion and cognition. CRH+ fibers are found in nucleus accumbens (NAc), where CRH modulates reward/motivation behaviors. CRH actions in NAc may vary by the individual's stress history, suggesting roles for CRH in neuroplasticity and adaptation of the reward circuitry. However, the origin and extent of CRH+ inputs to NAc are incompletely understood. We employed viral genetic approaches to map both global and CRH+ projection sources to NAc in mice. We injected into NAc variants of a new designer adeno-associated virus that permits robust retrograde access to NAc-afferent projection neurons. Cre-dependent viruses injected into CRH-Cre mice enabled selective mapping of CRH+ afferents. We employed anterograde AAV1-directed axonal tracing to verify NAc CRH+ fiber projections and established the identity of genetic reporter-labeled cells via validated antisera against native CRH. We quantified the relative contribution of CRH+ neurons to total NAc-directed projections. Combined retrograde and anterograde tracing identified the paraventricular nucleus of the thalamus, bed nucleus of stria terminalis, basolateral amygdala, and medial prefrontal cortex as principal sources of CRH+ projections to NAc. CRH+ NAc afferents were selectively enriched in NAc-projecting brain regions involved in diverse aspects of the sensing, processing and memory of emotionally salient events. These findings suggest multiple, complex potential roles for the molecularly-defined, CRH-dependent circuit in modulation of reward and motivation behaviors.
Collapse
Affiliation(s)
- Christy A Itoga
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Yuncai Chen
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California.,Department of Pediatrics, School of Medicine, University of California-Irvine, Irvine, California
| | - Cameron Fateri
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Paula A Echeverry
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Jennifer M Lai
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Jasmine Delgado
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Shapatur Badhon
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Annabel Short
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California.,Department of Pediatrics, School of Medicine, University of California-Irvine, Irvine, California
| | - Tallie Z Baram
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California.,Department of Pediatrics, School of Medicine, University of California-Irvine, Irvine, California
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California.,Department of Biomedical Engineering, University of California, Irvine, California.,Department of Microbiology and Molecular Genetics, University of California, Irvine, California
| |
Collapse
|
23
|
Ztaou S, Lhost J, Watabe I, Torromino G, Amalric M. Striatal cholinergic interneurons regulate cognitive and affective dysfunction in partially dopamine-depleted mice. Eur J Neurosci 2018; 48:2988-3004. [PMID: 30230645 DOI: 10.1111/ejn.14153] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 08/20/2018] [Accepted: 08/31/2018] [Indexed: 02/02/2023]
Abstract
Early non-motor symptoms such as mood disorders and cognitive deficits are increasingly recognised in Parkinson's disease (PD). They may precede the characteristic motor symptomatology caused by dopamine (DA) neuronal loss in the substantia nigra pars compacta (SNc). It is well known that striatal cholinergic interneurons (ChIs) are emerging as key regulators of PD motor symptom, however, their involvement in the cognitive and affective alterations occurring in the premotor phase of PD is poorly understood. We used optogenetic photoinhibition of striatal ChIs in mice with mild nigrostriatal 6-hydroxydopamine (6-OHDA) lesions and assessed their role in anxiety-like behaviour in the elevated plus maze, social memory recognition of a congener and visuospatial object recognition. In transgenic mice specifically expressing halorhodopsin (eNpHR) in cholinergic neurons, striatal ChIs photoinhibition reduced the anxiety-like behaviour and reversed social and spatial short-term memory impairment induced by moderate DA depletion (e.g., 50% loss of tyrosine hydroxylase TH-positive neurons in the SNc). Systemic injection of telenzepine (0.3 mg/kg), a preferential M1 muscarinic cholinergic receptors antagonist, improved anxiety-like behaviour, social memory recognition but not spatial memory deficits. Our results suggest that dysfunction of the striatal cholinergic system may play a role in the short-term cognitive and emotional deficits of partially DA-depleted mice. Blocking cholinergic activity with M1 muscarinic receptor antagonists may represent a possible therapeutic target, although not exclusive, to modulate these early non-motor deficits.
Collapse
Affiliation(s)
- Samira Ztaou
- Aix Marseille Univ, CNRS, LNC, FR3C, Marseille, France
| | | | | | - Giulia Torromino
- Aix Marseille Univ, CNRS, LNC, FR3C, Marseille, France.,Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
24
|
Chemokine receptor CCR2 contributes to neuropathic pain and the associated depression via increasing NR2B-mediated currents in both D1 and D2 dopamine receptor-containing medium spiny neurons in the nucleus accumbens shell. Neuropsychopharmacology 2018; 43:2320-2330. [PMID: 29993042 PMCID: PMC6135748 DOI: 10.1038/s41386-018-0115-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/20/2018] [Accepted: 06/01/2018] [Indexed: 01/08/2023]
Abstract
Patients with neuropathic pain are usually accompanied by depression. Chemokine-mediated neuroinflammation is involved in a variety of diseases, including neurodegenerative diseases, depression, and chronic pain. The nucleus accumbens (NAc) is an important area in mediating pain sensation and depression. Here we report that spinal nerve ligation (SNL) induced upregulation of chemokine CCL2 and its major receptor CCR2 in both dopamine D1 and D2 receptor (D1R and D2R)-containing neurons in the NAc. Inhibition of CCR2 by shRNA lentivirus in the NAc shell attenuated SNL-induced pain hypersensitivity and depressive behaviors. Conversely, intra-NAc injection of CCL2-expressing lentivirus-induced nociceptive and depressive behaviors in naïve mice. Whole-cell patch clamp recording of D1R-positive or D2R-positive medium spiny neurons (MSNs) showed that SNL increased NMDA receptor (NMDAR)-mediated currents that are induced by stimulation of prefrontal cortical afferents to MSNs, which was inhibited by a CCR2 antagonist. Furthermore, Ccr2 shRNA also reduced NMDAR-mediated currents, and this reduction was mainly mediated via NR2B subunit. Consistently, NR2B, colocalized with CCR2 in the NAc, was phosphorylated after SNL and was inhibited by intra-NAc injection of Ccr2 shRNA. Furthermore, SNL or CCL2 induced ERK activation in the NAc. Inhibition of ERK by a MEK inhibitor reduced NR2B phosphorylation induced by SNL or CCL2. Finally, intra-NAc injection of NR2B antagonist or MEK inhibitor attenuated SNL-induced pain hypersensitivity and depressive behaviors. Collectively, these results suggest that CCL2/CCR2 signaling in the NAc shell is important in mediating neuropathic pain and depression via regulating NR2B-mediated NMDAR function in D1R- and D2R-containing neurons following peripheral nerve injury.
Collapse
|
25
|
Dimatelis JJ, Mtintsilana A, Naidoo V, Stein DJ, Russell VA. Chronic light exposure alters serotonergic and orexinergic systems in the rat brain and reverses maternal separation-induced increase in orexin receptors in the prefrontal cortex. Metab Brain Dis 2018; 33:433-441. [PMID: 29039077 DOI: 10.1007/s11011-017-0123-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 10/04/2017] [Indexed: 12/22/2022]
Abstract
Maternal separation (MS) is a well-established rodent model of depression. Chronic constant light (CCL) treatment during adolescence has been shown to reverse the depression-like behaviour induced by MS. We aimed to further delineate the antidepressant effect of light by investigating the involvement of the dopaminergic, serotonergic and orexinergic systems. MS was used to induce changes in adult male Sprague-Dawley rats, some of whom were also treated with CCL for 3 weeks during adolescence. At P80, rats were decapitated and brain tissue collected for analysis of glutamate- and potassium-stimulated dopamine release in the nucleus accumbens (NAc) using an in vitro superfusion technique. Enzyme-linked immunosorbent assays were employed to measure 5-hydroxytryptamine (5-HT) levels in the hypothalamus and prefrontal cortex (PFC). Western blotting was used to measure orexin receptor 1 (OXR-1) and 2 (OXR-2) in the PFC. MS did not affect 5-HT levels in these rats. However, CCL increased hypothalamic 5-HT and reduced 5-HT levels in the PFC. CCL had opposite effects on OXR levels in the PFC of maternally separated and non-separated rats. MS increased OXR-1 and OXR-2 levels in the PFC, an effect that was normalized by CCL treatment. MS reduced glutamate-stimulated dopamine release in the NAc, an effect that was not reversed by CCL. The present results suggest that CCL treatment affects 5-HT and orexinergic systems in the MS model while not affecting the MS-induced decrease in dopamine release in the NAc. The reversal of changes in the orexinergic system may be of particular relevance to the antidepressant effect of CCL in depression.
Collapse
Affiliation(s)
- J J Dimatelis
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa.
| | - A Mtintsilana
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - V Naidoo
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - D J Stein
- Department of Psychiatry and Mental Health and MRC Unit on Anxiety & Stress Disorders, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - V A Russell
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| |
Collapse
|
26
|
Lalonde R, Strazielle C. Neuroanatomical pathways underlying the effects of hypothalamo-hypophysial-adrenal hormones on exploratory activity. Rev Neurosci 2018; 28:617-648. [PMID: 28609296 DOI: 10.1515/revneuro-2016-0075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/16/2017] [Indexed: 12/25/2022]
Abstract
When injected via the intracerebroventricular route, corticosterone-releasing hormone (CRH) reduced exploration in the elevated plus-maze, the center region of the open-field, and the large chamber in the defensive withdrawal test. The anxiogenic action of CRH in the elevated plus-maze also occurred when infused in the basolateral amygdala, ventral hippocampus, lateral septum, bed nucleus of the stria terminalis, nucleus accumbens, periaqueductal grey, and medial frontal cortex. The anxiogenic action of CRH in the defensive withdrawal test was reproduced when injected in the locus coeruleus, while the amygdala, hippocampus, lateral septum, nucleus accumbens, and lateral globus pallidus contribute to center zone exploration in the open-field. In addition to elevated plus-maze and open-field tests, the amygdala appears as a target region for CRH-mediated anxiety in the elevated T-maze. Thus, the amygdala is the principal brain region identified with these three tests, and further research must identify the neural circuits underlying this form of anxiety.
Collapse
Affiliation(s)
| | - Catherine Strazielle
- , Laboratoire 'Stress, Immunité, Pathogènes' EA 7300 and Service de Microscopie Electronique, Faculté de Médecine
| |
Collapse
|
27
|
Yanovich C, Kirby ML, Michaelevski I, Yadid G, Pinhasov A. Social rank-associated stress vulnerability predisposes individuals to cocaine attraction. Sci Rep 2018; 8:1759. [PMID: 29379100 PMCID: PMC5789078 DOI: 10.1038/s41598-018-19816-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/03/2018] [Indexed: 12/15/2022] Open
Abstract
Studies of personality have suggested that dissimilarities in ability to cope with stressful situations results in differing tendency to develop addictive behaviors. The present study used selectively bred stress-resilient, socially-dominant (Dom) and stress-vulnerable, socially-submissive (Sub) mice to investigate the interaction between environmental stress and inbred predisposition to develop addictive behavior to cocaine. In a Conditioned Place Preference (CPP) paradigm using cocaine, Sub mice displayed an aversion to drug, whereas Dom mice displayed drug attraction. Following a 4-week regimen of Chronic Mild Stress (CMS), Sub mice in CPP displayed a marked increase (>400%) in cocaine attraction, whereas Dom mice did not differ in attraction from their non-stressed state. Examination of hippocampal gene expression revealed in Sub mice, exposure to external stimuli, stress or cocaine, increased CRH expression (>100%), which was evoked in Dom mice only by cocaine exposure. Further, stress-induced decreases in DRD1 (>60%) and DRD2 (>50%) expression in Sub mice differed markedly from a complete lack of change in Dom mice. From our findings, we propose that social stratification dictates vulnerability to stress-induced attraction that may lead to addiction via differential regulation of hippocampal response to dopaminergic input, which in turn may influence differing tendency to develop addictive behaviors.
Collapse
Affiliation(s)
- Chen Yanovich
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Michael L Kirby
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | | | - Gal Yadid
- Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center and the Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| | - Albert Pinhasov
- Department of Molecular Biology, Ariel University, Ariel, Israel.
| |
Collapse
|
28
|
Abstract
Most of the energy we get to spend is furnished by mitochondria, minuscule living structures sitting inside our cells or dispatched back and forth within them to where they are needed. Mitochondria produce energy by burning down what remains of our meal after we have digested it, but at the cost of constantly corroding themselves and us. Here we review how our mitochondria evolved from invading bacteria and have retained a small amount of independence from us; how we inherit them only from our mother; and how they are heavily implicated in learning, memory, cognition, and virtually every mental or neurological affliction. We discuss why counteracting mitochondrial corrosion with antioxidant supplements is often unwise, and why our mitochondria, and therefore we ourselves, benefit instead from exercise, meditation, sleep, sunshine, and particular eating habits. Finally, we describe how malfunctioning mitochondria force rats to become socially subordinate to others, how such disparity can be evened off by a vitamin, and why these findings are relevant to us.
Collapse
Affiliation(s)
- Peter Kramer
- Department of General Psychology, University of Padua, Italy
| | - Paola Bressan
- Department of General Psychology, University of Padua, Italy
| |
Collapse
|
29
|
Abstract
BACKGROUND AMPAkines augment the function of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the brain to increase excitatory outputs. These drugs are known to relieve persistent pain. However, their role in acute pain is unknown. Furthermore, a specific molecular and anatomic target for these novel analgesics remains elusive. METHODS The authors studied the analgesic role of an AMPAkine, CX546, in a rat paw incision (PI) model of acute postoperative pain. The authors measured the effect of AMPAkines on sensory and depressive symptoms of pain using mechanical hypersensitivity and forced swim tests. The authors asked whether AMPA receptors in the nucleus accumbens (NAc), a key node in the brain's reward and pain circuitry, can be a target for AMPAkine analgesia. RESULTS Systemic administration of CX546 (n = 13), compared with control (n = 13), reduced mechanical hypersensitivity (50% withdrawal threshold of 6.05 ± 1.30 g [mean ± SEM] vs. 0.62 ± 0.13 g), and it reduced depressive features of pain by decreasing immobility on the forced swim test in PI-treated rats (89.0 ± 15.5 vs. 156.7 ± 18.5 s). Meanwhile, CX546 delivered locally into the NAc provided pain-relieving effects in both PI (50% withdrawal threshold of 6.81 ± 1.91 vs. 0.50 ± 0.03 g; control, n = 6; CX546, n = 8) and persistent postoperative pain (spared nerve injury) models (50% withdrawal threshold of 3.85 ± 1.23 vs. 0.45 ± 0.00 g; control, n = 7; CX546, n = 11). Blocking AMPA receptors in the NAc with 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione inhibited these pain-relieving effects (50% withdrawal threshold of 7.18 ± 1.52 vs. 1.59 ± 0.66 g; n = 8 for PI groups; 10.70 ± 3.45 vs. 1.39 ± 0.88 g; n = 4 for spared nerve injury groups). CONCLUSIONS AMPAkines relieve postoperative pain by acting through AMPA receptors in the NAc.
Collapse
|
30
|
Reichard RA, Subramanian S, Desta MT, Sura T, Becker ML, Ghobadi CW, Parsley KP, Zahm DS. Abundant collateralization of temporal lobe projections to the accumbens, bed nucleus of stria terminalis, central amygdala and lateral septum. Brain Struct Funct 2017; 222:1971-1988. [PMID: 27704219 PMCID: PMC5378696 DOI: 10.1007/s00429-016-1321-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022]
Abstract
Behavioral flexibility is subserved in part by outputs from the cerebral cortex to telencephalic subcortical structures. In our earlier evaluation of the organization of the cortical-subcortical output system (Reynolds and Zahm, J Neurosci 25:11757-11767, 2005), retrograde double-labeling was evaluated in the prefrontal cortex following tracer injections into pairs of the following subcortical telencephalic structures: caudate-putamen, core and shell of the accumbens (Acb), bed nucleus of stria terminalis (BST) and central nucleus of the amygdala (CeA). The present study was done to assess patterns of retrograde labeling in the temporal lobe after similar paired tracer injections into most of the same telencephalic structures plus the lateral septum (LS). In contrast to the modest double-labeling observed in the prefrontal cortex in the previous study, up to 60-80 % of neurons in the basal and accessory basal amygdaloid nuclei and amygdalopiriform transition area exhibited double-labeling in the present study. The most abundant double-labeling was generated by paired injections into structures affiliated with the extended amygdala, including the CeA, BST and Acb shell. Injections pairing the Acb core with the BST or CeA produced significantly fewer double-labeled neurons. The ventral subiculum exhibited modest amounts of double-labeling associated with paired injections into the Acb, BST, CeA and LS. The results raise the issue of how an extraordinarily collateralized output from the temporal lobe may contribute to behavioral flexibility.
Collapse
Affiliation(s)
- Rhett A Reichard
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Suriya Subramanian
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Mikiyas T Desta
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Tej Sura
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Mary L Becker
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Comeron W Ghobadi
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Kenneth P Parsley
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Daniel S Zahm
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA.
| |
Collapse
|
31
|
Hayase T. Putative Epigenetic Involvement of the Endocannabinoid System in Anxiety- and Depression-Related Behaviors Caused by Nicotine as a Stressor. PLoS One 2016; 11:e0158950. [PMID: 27404492 PMCID: PMC4942073 DOI: 10.1371/journal.pone.0158950] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 06/26/2016] [Indexed: 01/01/2023] Open
Abstract
Like various stressors, the addictive use of nicotine (NC) is associated with emotional symptoms such as anxiety and depression, although the underlying mechanisms have not yet been fully elucidated due to the complicated involvement of target neurotransmitter systems. In the elicitation of these emotional symptoms, the fundamental involvement of epigenetic mechanisms such as histone acetylation has recently been suggested. Furthermore, among the interacting neurotransmitter systems implicated in the effects of NC and stressors, the endocannabinoid (ECB) system is considered to contribute indispensably to anxiety and depression. In the present study, the epigenetic involvement of histone acetylation induced by histone deacetylase (HDAC) inhibitors was investigated in anxiety- and depression-related behavioral alterations caused by NC and/or immobilization stress (IM). Moreover, based on the contributing roles of the ECB system, the interacting influence of ECB ligands on the effects of HDAC inhibitors was evaluated in order to examine epigenetic therapeutic interventions. Anxiety-like (elevated plus-maze test) and depression-like (forced swimming test) behaviors, which were observed in mice treated with repeated (4 days) NC (subcutaneous 0.8 mg/kg) and/or IM (10 min), were blocked by the HDAC inhibitors sodium butyrate (SB) and valproic acid (VA). The cannabinoid type 1 (CB1) agonist ACPA (arachidonylcyclopropylamide; AC) also antagonized these behaviors. Conversely, the CB1 antagonist SR 141716A (SR), which counteracted the effects of AC, attenuated the anxiolytic-like effects of the HDAC inhibitors commonly in the NC and/or IM groups. SR also attenuated the antidepressant-like effects of the HDAC inhibitors, most notably in the IM group. From these results, the combined involvement of histone acetylation and ECB system was shown in anxiety- and depression-related behaviors. In the NC treatment groups, the limited influence of SR against the HDAC inhibitor-induced antidepressant-like effects may reflect the characteristic involvement of histone acetylation within the NC-related neurotransmitter systems other than the ECB system.
Collapse
Affiliation(s)
- Tamaki Hayase
- Department of Legal Medicine, Kyoto University, Kyoto 606–8501, Japan
- * E-mail:
| |
Collapse
|
32
|
Barra de la Tremblaye P, Plamondon H. Alterations in the corticotropin-releasing hormone (CRH) neurocircuitry: Insights into post stroke functional impairments. Front Neuroendocrinol 2016; 42:53-75. [PMID: 27455847 DOI: 10.1016/j.yfrne.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/04/2016] [Accepted: 07/06/2016] [Indexed: 10/21/2022]
Abstract
Although it is well accepted that changes in the regulation of the hypothalamic-pituitary adrenal (HPA) axis may increase susceptibility to affective disorders in the general population, this link has been less examined in stroke patients. Yet, the bidirectional association between depression and cardiovascular disease is strong, and stress increases vulnerability to stroke. Corticotropin-releasing hormone (CRH) is the central stress hormone of the HPA axis pathway and acts by binding to CRH receptors (CRHR) 1 and 2, which are located in several stress-related brain regions. Evidence from clinical and animal studies suggests a role for CRH in the neurobiological basis of depression and ischemic brain injury. Given its importance in the regulation of the neuroendocrine, autonomic, and behavioral correlates of adaptation and maladaptation to stress, CRH is likely associated in the pathophysiology of post stroke emotional impairments. The goals of this review article are to examine the clinical and experimental data describing (1) that CRH regulates the molecular signaling brain circuit underlying anxiety- and depression-like behaviors, (2) the influence of CRH and other stress markers in the pathophysiology of post stroke emotional and cognitive impairments, and (3) context and site specific interactions of CRH and BDNF as a basis for the development of novel therapeutic targets. This review addresses how the production and release of the neuropeptide CRH within the various regions of the mesocorticolimbic system influences emotional and cognitive behaviors with a look into its role in psychiatric disorders post stroke.
Collapse
Affiliation(s)
- P Barra de la Tremblaye
- School of Psychology, Behavioral Neuroscience Program, University of Ottawa, 136 Jean-Jacques Lussier, Vanier Building, Ottawa, Ontario K1N 6N5, Canada
| | - H Plamondon
- School of Psychology, Behavioral Neuroscience Program, University of Ottawa, 136 Jean-Jacques Lussier, Vanier Building, Ottawa, Ontario K1N 6N5, Canada.
| |
Collapse
|
33
|
Small KM, Nunes E, Hughley S, Addy NA. Ventral tegmental area muscarinic receptors modulate depression and anxiety-related behaviors in rats. Neurosci Lett 2016; 616:80-5. [PMID: 26828299 PMCID: PMC4798862 DOI: 10.1016/j.neulet.2016.01.057] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/16/2016] [Accepted: 01/26/2016] [Indexed: 01/16/2023]
Abstract
Cholinergic and dopaminergic mechanisms within the mesolimbic dopamine system are suggested to play a role in the manifestation of depression and anxiety-related disorders. However, despite the fact that cholinergic mechanisms in the ventral tegmental area (VTA) highly regulate dopamine activity, the role of VTA cholinergic mechanisms in depression-related behaviors is relatively unknown. Here we sought to determine whether enhancing cholinergic tone in the VTA would alter depression and anxiety-related behavior in the forced swim test (FST), elevated plus maze (EPM) and sucrose preference test (SPT). Adult Sprague Dawley male rats received VTA infusion of the acetylcholinesterase inhibitor, physostigmine (0, 1, 2μg/side), immediately prior to the FST, EPM, or SPT. Physostigmine administration increased immobility time in the FST, decreased time spent on open arms in the EPM, and decreased sucrose preference. We also examined whether activation of VTA muscarinic receptors was sufficient to alter behavior in the FST and EPM. Similar to physostigmine, VTA infusion of the muscarinic receptor agonist, pilocarpine (0, 3, 30μg/side), increased immobility time in the FST and decreased time spent on open arms in the EPM. These data suggest that enhanced VTA cholinergic tone promotes pro-depressive and anxiogenic-like effects and demonstrate that specific activation of VTA muscarinic receptors is also sufficient to induce pro-depressive and anxiogenic responses. Together, these findings reveal a novel role of VTA cholinergic, and specifically muscarinic receptor, mechanisms in mediating responses to stress and anxiety.
Collapse
Affiliation(s)
- Keri M Small
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511, United States
| | - Eric Nunes
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511, United States
| | - Shannon Hughley
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511, United States
| | - Nii A Addy
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511, United States; Department of Cellular and Molecular Physiology, New Haven, CT 06511, United States; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06511, United States.
| |
Collapse
|
34
|
Hoffman KL. New dimensions in the use of rodent behavioral tests for novel drug discovery and development. Expert Opin Drug Discov 2016; 11:343-53. [DOI: 10.1517/17460441.2016.1153624] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
35
|
Oxytocin in the nucleus accumbens shell reverses CRFR2-evoked passive stress-coping after partner loss in monogamous male prairie voles. Psychoneuroendocrinology 2016; 64:66-78. [PMID: 26615473 PMCID: PMC4698175 DOI: 10.1016/j.psyneuen.2015.11.011] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/12/2015] [Accepted: 11/12/2015] [Indexed: 12/21/2022]
Abstract
Loss of a partner can have severe effects on mental health. Here we explore the neural mechanisms underlying increased passive stress-coping, indicative of depressive-like behavior, following the loss of the female partner in the monogamous male prairie vole. We demonstrate that corticotropin-releasing factor receptor 2 (CRFR2) in the nucleus accumbens shell mediates social loss-induced passive coping. Further, we show that partner loss compromises the oxytocin system through multiple mechanisms. Finally, we provide evidence for an interaction of the CRFR2 and oxytocin systems in mediating the emotional consequences of partner loss. Our results suggest that chronic activation of CRFR2 and suppression of striatal oxytocin signaling following partner loss result in an aversive emotional state that may share underlying mechanisms with bereavement. We propose that the suppression of oxytocin signaling is likely adaptive during short separations to encourage reunion with the partner and may have evolved to maintain long-term partnerships. Additionally, therapeutic strategies targeting these systems should be considered for treatment of social loss-mediated depression.
Collapse
|
36
|
Sand E, Linninge C, Lozinska L, Egecioglu E, Roth B, Molin G, Weström B, Ekblad E, Ohlsson B. Buserelin treatment to rats causes enteric neurodegeneration with moderate effects on CRF-immunoreactive neurons and Enterobacteriaceae in colon, and in acetylcholine-mediated permeability in ileum. BMC Res Notes 2015; 8:824. [PMID: 26710832 PMCID: PMC4693429 DOI: 10.1186/s13104-015-1800-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 12/14/2015] [Indexed: 02/07/2023] Open
Abstract
Background The gonadotropin-releasing hormone (GnRH) analog buserelin causes enteric neuronal loss. Acute stress or injection of corticotropin-releasing factor (CRF) affects motility, secretion, and barrier function of the gastrointestinal tract. The aim of the study was to characterize the CRF immunoreactivity in enteric neurons after buserelin treatment, and to evaluate possible effects of enteric neuropathy on gut microbiota, intestinal permeability, and stress response behavior. Results Sixty rats were given buserelin (20 μg) or saline subcutaneously for 5 days, repeated four times with 3 weeks in-between. At the study end, enteric neuronal density, enteric expression of CRF, gut microbial composition, and plasma levels of adrenocorticotropic hormone (ACTH) and CRF were analyzed. Intestinal permeability was examined in Ussing chambers and the reaction to stressful events was measured by behavior tests. Buserelin treatment reduced the number of neurons along the entire gastrointestinal tract, with increased relative numbers of CRF-immunoreactive submucosal and myenteric neurons in colon (p < 0.05 and p < 0.01, respectively). The overall microbial diversity and relative abundance did not differ between groups, but Enterobacteriaceae was decreased in colon in buserelin-treated rats (p = 0.020). Basal intestinal permeability did not differ between groups, whereas carbachol stimulation increased ileum permeability in controls (p < 0.05), but not in buserelin-treated rats. Buserelin did not affect stress behavior. Conclusions Although buserelin treatment leads to enteric neuronal loss along the gastrointestinal tract with an increased percentage of CRF-immunoreactive neurons in colon, the physiology is well preserved, with modest effects on colon microbiota and absence of carbachol-induced permeability in ileum as the only observed changes. Electronic supplementary material The online version of this article (doi:10.1186/s13104-015-1800-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elin Sand
- Division of Internal Medicine, Department of Clinical Sciences, Skåne University Hospital, Lund University, Inga Marie Nilssons street 32, 205 02, Malmö, Sweden. .,Neurogastroenterology Unit, Department of Experimental Medical Science, BMC B11, Lund University, 221 84, Lund, Sweden.
| | - Caroline Linninge
- Department of Food Technology, Engineering and Nutrition, Lund University, 22100, Lund, Sweden.
| | - Liudmyla Lozinska
- Department of Biology, Functional Biology, Lund University, 221 84, Lund, Sweden.
| | - Emil Egecioglu
- Department of Clinical Neuroscience and Rehabilitation, University of Gothenburg, 405 30, Gothenburg, Sweden.
| | - Bodil Roth
- Division of Internal Medicine, Department of Clinical Sciences, Skåne University Hospital, Lund University, Inga Marie Nilssons street 32, 205 02, Malmö, Sweden.
| | - Göran Molin
- Department of Food Technology, Engineering and Nutrition, Lund University, 22100, Lund, Sweden.
| | - Björn Weström
- Department of Biology, Functional Biology, Lund University, 221 84, Lund, Sweden.
| | - Eva Ekblad
- Neurogastroenterology Unit, Department of Experimental Medical Science, BMC B11, Lund University, 221 84, Lund, Sweden.
| | - Bodil Ohlsson
- Division of Internal Medicine, Department of Clinical Sciences, Skåne University Hospital, Lund University, Inga Marie Nilssons street 32, 205 02, Malmö, Sweden.
| |
Collapse
|
37
|
Xu D, Su C, Lin HY, Manders T, Wang J. Persistent neuropathic pain increases synaptic GluA1 subunit levels in core and shell subregions of the nucleus accumbens. Neurosci Lett 2015; 609:176-81. [PMID: 26477778 DOI: 10.1016/j.neulet.2015.10.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/09/2015] [Accepted: 10/10/2015] [Indexed: 11/18/2022]
Abstract
The nucleus accumbens (NAc) is a key component of the brain reward system, and it is composed of core and shell subregions. Glutamate transmission through AMPA-type receptors in both core and shell of the NAc has been shown to regulate reward- and aversion-type behaviors. Previous studies have additionally demonstrated a role for AMPA receptor signaling in the NAc in chronic pain states. Here, we show that persistent neuropathic pain, modeled by spared nerve injury (SNI), selectively increases the numbers of GluA1 subunits of AMPA receptors at the synapse of both core and shell subregions. Such increases are not observed, however, for the GluA2 subunits. Furthermore, we find that phosphorylation at Ser845-GluA1 is increased by SNI at both core and shell subregions. These results demonstrate that persistent neuropathic pain increases AMPA receptor delivery to the synapse in both NAc core and shell, implying a role for AMPA receptor signaling in these regions in pain states.
Collapse
Affiliation(s)
- Duo Xu
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York, NY 10016, United States
| | - Chen Su
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York, NY 10016, United States
| | - Hau-Yueh Lin
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York, NY 10016, United States
| | - Toby Manders
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York, NY 10016, United States
| | - Jing Wang
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York, NY 10016, United States; Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, United States.
| |
Collapse
|
38
|
Urstadt KR, Stanley BG. Direct hypothalamic and indirect trans-pallidal, trans-thalamic, or trans-septal control of accumbens signaling and their roles in food intake. Front Syst Neurosci 2015; 9:8. [PMID: 25741246 PMCID: PMC4327307 DOI: 10.3389/fnsys.2015.00008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 01/15/2015] [Indexed: 01/01/2023] Open
Abstract
Due in part to the increasing incidence of obesity in developed nations, recent research aims to elucidate neural circuits that motivate humans to overeat. Earlier research has described how the nucleus accumbens shell (AcbSh) motivates organisms to feed by activating neuronal populations in the lateral hypothalamus (LH). However, more recent research suggests that the LH may in turn communicate with the AcbSh, both directly and indirectly, to re-tune the motivation to consume foods with homeostatic and food-related sensory signals. Here, we discuss the functional and anatomical evidence for an LH to AcbSh connection and its role in eating behaviors. The LH appears to modulate Acb activity directly, using neurotransmitters such as hypocretin/orexin or melanin concentrating hormone (MCH). The LH also indirectly regulates AcbSh activity through certain subcortical "relay" regions, such as the lateral septum (LS), ventral pallidum (VP), and paraventricular thalamus, using a variety of neurotransmitters. This review aims to summarize studies on these topics and outline a model by which LH circuits processing energy balance can modulate AcbSh neural activity to regulate feeding behavior.
Collapse
Affiliation(s)
- Kevin R Urstadt
- Department of Psychology, University of Michigan Ann Arbor, MI, USA
| | - B Glenn Stanley
- Departments of Psychology and Cell Biology and Neuroscience, University of California - Riverside Riverside, CA, USA
| |
Collapse
|
39
|
Blockade of mGluR5 in the nucleus accumbens shell but not core attenuates heroin seeking behavior in rats. Acta Pharmacol Sin 2014; 35:1485-92. [PMID: 25399651 DOI: 10.1038/aps.2014.93] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 08/20/2014] [Indexed: 11/08/2022] Open
Abstract
AIM Glutamatergic neurotransmission in the nucleus accumbens (NAc) is crucial for the relapse to heroin seeking. The aim of this study was to determine whether mGluR5 in the NAc core or shell involved in heroin seeking behavior in rats. METHODS Male SD rats were self-administered heroin under a fixed-ratio 1 (FR1) reinforcement schedule for 14 d, and subsequently withdrawn for 2 weeks. The selective mGluR5 antagonist 2-methyl-6-phenylethynyl-pyridine (MPEP, 5, 15 and 50 nmol per side) was then microinjected into the NAc core or shell 10 min before a heroin-seeking test induced by context, cues or heroin priming. RESULTS Microinjection of MPEP into the NAc shell dose-dependently decreased the heroin seeking induced by context, cues or heroin priming. In contrast, microinjection of MPEP into the NAc core did not alter the heroin seeking induced by cues or heroin priming. In addition, microinjection with MPEP (15 nmol per side) in the NAc shell reversed both the percentage of open arms entries (OE%) and the percentage of time spent in open arms (OT%) after heroin withdrawal. Microinjection of MPEP (50 nmol per side) in the striatum as a control location did not affect the heroin seeking behavior. Microinjection of MPEP in the 3 locations did not change the locomotion activities. CONCLUSION Blockade of mGluR5 in NAc shell in rats specifically suppresses the relapse to heroin-seeking and anxiety-like behavior, suggesting that mGluR5 antagonists may be a potential candidate for the therapy of heroin addiction.
Collapse
|
40
|
CRF1 receptor-deficiency induces anxiety-like vulnerability to cocaine. Psychopharmacology (Berl) 2014; 231:3965-72. [PMID: 24687410 DOI: 10.1007/s00213-014-3534-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 03/03/2014] [Indexed: 01/23/2023]
Abstract
RATIONALE The intake of psychostimulant drugs may induce cognitive dysfunction and negative affective-like states, and is associated with increased activity of stress-responsive systems. The corticotropin-releasing factor (CRF) system mediates neuroendocrine, behavioural and autonomic responses to stressors, and might be implicated in substance-related disorders. CRF signalling is mediated by two receptor types, named CRF1 and CRF2. OBJECTIVES The present study aims to elucidate the role for the CRF1 receptor in cognitive dysfunction and anxiety-like states induced by cocaine. RESULTS The genetic inactivation of the CRF1 receptor (CRF1+/- and CRF1-/-) does not influence recognition memory in drug-naïve mice, as assessed by the novel object recognition (NOR) test. Moreover, the chronic administration of escalating doses of cocaine (5-20 mg/kg, i.p.) induces NOR deficits, which are unaffected by CRF1 receptor-deficiency. However, the same drug regimen reveals an anxiety-like vulnerability to cocaine in CRF1-/- but not in wild-type or CRF1+/- mice, as assessed by the elevated plus maze test. CONCLUSIONS The present findings indicate dissociation of cognitive dysfunction and anxiety-like states induced by cocaine. Moreover, they unravel a novel mechanism of vulnerability to psychostimulant drugs.
Collapse
|
41
|
Wang T, Zhou C, Yuan D, Lin F, Chen H, Wu H, Wei R, Xin Z, Liu J, Gao Y, Li Z. Schizothorax prenanti corticotropin-releasing hormone (CRH): molecular cloning, tissue expression, and the function of feeding regulation. FISH PHYSIOLOGY AND BIOCHEMISTRY 2014; 40:1407-1415. [PMID: 24696302 DOI: 10.1007/s10695-014-9935-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/26/2014] [Indexed: 06/03/2023]
Abstract
Corticotropin-releasing hormone (CRH) is a potent mediator of endocrine, autonomic, behavioral, and immune responses to stress. For a better understanding of the structure and function of the CRH gene and to study its effect on feeding regulation in cyprinid fish, the cDNA of the CRH gene from the brain of Schizothorax prenanti was cloned and sequenced. The full-length CRH cDNA consisted of 1,046 bp with an open reading frame of 489 bp encoding a protein of 162 amino acids. Real-time quantitative PCR analyses revealed that CRH was widely expressed in central and peripheral tissues. In particular, high expression level of CRH was detected in brain. Furthermore, CRH mRNA expression was examined in different brain regions, especially high in hypothalamus. In addition, there was no significant change in CRH mRNA expression in fed group compared with the fasted group in the S. prenanti hypothalamus during short-term fasting. However, CRH gene expression presented significant decrease in the hypothalamus in fasted group compared with the fed group (P < 0.05) on day 7; thereafter, re-feeding could lead to a significant increase in CRH mRNA expression in fasted group on day 9. The results suggest that the CRH may play a critical role in feeding regulation in S. prenanti.
Collapse
Affiliation(s)
- Tao Wang
- Department of Aquaculture, Sichuan Agricultural University, 46# Xinkang Road, Ya'an, 625014, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Rivier JE, Rivier CL. Corticotropin-releasing factor peptide antagonists: design, characterization and potential clinical relevance. Front Neuroendocrinol 2014; 35:161-70. [PMID: 24269930 PMCID: PMC3965584 DOI: 10.1016/j.yfrne.2013.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 10/18/2013] [Accepted: 10/20/2013] [Indexed: 12/19/2022]
Abstract
Elusive for more than half a century, corticotropin-releasing factor (CRF) was finally isolated and characterized in 1981 from ovine hypothalami and shortly thereafter, from rat brains. Thirty years later, much has been learned about the function and localization of CRF and related family members (Urocortins 1, 2 and 3) and their 2 receptors, CRF receptor type 1 (CRFR1) and CRF receptor type 2 (CRFR2). Here, we report the stepwise development of peptide CRF agonists and antagonists, which led to the CRFR1 agonist Stressin1; the long-acting antagonists Astressin2-B which is specific for CRFR2; and Astressin B, which binds to both CRFR1 and CRFR2.This analog has potential for the treatment of CRF-dependent diseases in the periphery, such as irritable bowel syndrome.
Collapse
Affiliation(s)
- Jean E Rivier
- The Salk Institute, The Clayton Foundation Laboratories for Peptide Biology, La Jolla, CA 92037, USA.
| | - Catherine L Rivier
- The Salk Institute, The Clayton Foundation Laboratories for Peptide Biology, La Jolla, CA 92037, USA
| |
Collapse
|
43
|
Sequeira-Cordero A, Mora-Gallegos A, Cuenca-Berger P, Fornaguera-Trías J. Individual differences in the forced swimming test and the effect of environmental enrichment: searching for an interaction. Neuroscience 2014; 265:95-107. [PMID: 24508814 DOI: 10.1016/j.neuroscience.2014.01.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 01/22/2014] [Accepted: 01/24/2014] [Indexed: 01/28/2023]
Abstract
Animals with low and high immobility in the forced swimming test (FST) differ in a number of neurobehavioral factors. A growing body of evidence suggests that the exposure to enriched environments mediates a number of changes in the brain. Therefore, we studied if animals' individuality can somehow modulate the response to environmental stimuli. Male rats were classified according to their immobility time scores in the FST test session as animals with low, medium or high immobility. Then, rats from groups with low and high immobility were randomly distributed in two groups to be reared in different housing conditions (i.e., enriched and standard conditions) during 8weeks. Animals were subjected to the open field test (OFT) before and 6weeks after the start of housing protocol. Rats with high immobility in the FST also showed high ambulation and high rearing time in the first OFT. Such findings were not observed in the second OFT. Conversely, an effect of environmental enrichment was found in the second OFT where enriched animals showed lower ambulation and higher grooming time than the standard control group. Rats were sacrificed after the housing protocol and neurochemical content and/or gene expression were studied in three different brain regions: the prefrontal cortex, the hippocampus and the nucleus accumbens. Rats with low immobility showed significantly higher accumbal 5-HT levels than animals with high immobility, whereas no neurochemical differences were observed between enriched and standard animals. Regarding expression data, however, an effect of enrichment on accumbal corticotropin-releasing factor (CRF) and its receptor 1 (CRFR1) levels was observed, and such effect depended on immobility levels. Thus, our results not only allowed us to identify a number of differences between animals with low and high immobility or animals housed in standard and enriched conditions, but also suggested that animals' individuality modulated in some way the response to environmental stimuli.
Collapse
Affiliation(s)
- A Sequeira-Cordero
- Instituto de Investigaciones en Salud, Universidad de Costa Rica, ZIP code 11501-2060, San Pedro, San José, Costa Rica; Centro de Investigación en Neurociencias, Universidad de Costa Rica, ZIP code 11501-2060, San Pedro, San José, Costa Rica.
| | - A Mora-Gallegos
- Centro de Investigación en Neurociencias, Universidad de Costa Rica, ZIP code 11501-2060, San Pedro, San José, Costa Rica
| | - P Cuenca-Berger
- Instituto de Investigaciones en Salud, Universidad de Costa Rica, ZIP code 11501-2060, San Pedro, San José, Costa Rica; Centro de Investigación en Neurociencias, Universidad de Costa Rica, ZIP code 11501-2060, San Pedro, San José, Costa Rica; Depto. De Bioquímica, Escuela de Medicina, Universidad de Costa Rica, ZIP code 11501-2060, San Pedro, San José, Costa Rica
| | - J Fornaguera-Trías
- Centro de Investigación en Neurociencias, Universidad de Costa Rica, ZIP code 11501-2060, San Pedro, San José, Costa Rica; Depto. De Bioquímica, Escuela de Medicina, Universidad de Costa Rica, ZIP code 11501-2060, San Pedro, San José, Costa Rica
| |
Collapse
|
44
|
Calcium-permeable AMPA receptors in the nucleus accumbens regulate depression-like behaviors in the chronic neuropathic pain state. J Neurosci 2014; 33:19034-44. [PMID: 24285907 DOI: 10.1523/jneurosci.2454-13.2013] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Depression is a salient emotional feature of chronic pain. Depression alters the pain threshold and impairs functional recovery. To date, however, there has been limited understanding of synaptic or circuit mechanisms that regulate depression in the pain state. Here, we demonstrate that depression-like behaviors are induced in a rat model of chronic neuropathic pain. Using this model, we show that chronic pain selectively increases the level of GluA1 subunits of AMPA-type glutamate receptors at the synapses of the nucleus accumbens (NAc), a key component of the brain reward system. We find, in addition, that this increase in GluA1 levels leads to the formation of calcium-permeable AMPA receptors (CPARs). Surprisingly, pharmacologic blockade of these CPARs in the NAc increases depression-like behaviors associated with pain. Consistent with these findings, an AMPA receptor potentiator delivered into the NAc decreases pain-induced depression. These results show that transmission through CPARs in the NAc represents a novel molecular mechanism modulating the depressive symptoms of pain, and thus CPARs may be a promising therapeutic target for the treatment of pain-induced depression. More generally, these findings highlight the role of central glutamate signaling in pain states and define the brain reward system as an important region for the regulation of depressive symptoms of pain.
Collapse
|
45
|
AChE and RACK1 promote the anti-inflammatory properties of fluoxetine. J Mol Neurosci 2013; 53:306-15. [PMID: 24258317 DOI: 10.1007/s12031-013-0174-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 11/05/2013] [Indexed: 10/26/2022]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) show anti-inflammatory effects, suggesting a possible interaction with both Toll-like-receptor 4 (TLR4) responses and cholinergic signaling through as yet unclear molecular mechanism(s). Our results of structural modeling support the concept that the antidepressant fluoxetine physically interacts with the TLR4-myeloid differentiation factor-2 complex at the same site as bacterial lipopolysaccharide (LPS). We also demonstrate reduced LPS-induced pro-inflammatory interleukin-6 and tumor necrosis factor alpha in human peripheral blood mononuclear cells preincubated with fluoxetine. Furthermore, we show that fluoxetine intercepts the LPS-induced decreases in intracellular acetylcholinesterase (AChE-S) and that AChE-S interacts with the nuclear factor kappa B (NFκB)-activating intracellular receptor for activated C kinase 1 (RACK1). This interaction may prevent NFκB activation by residual RACK1 and its interacting protein kinase PKCβII. Our findings attribute the anti-inflammatory properties of SSRI to surface membrane interference with leukocyte TLR4 activation accompanied by intracellular limitation of pathogen-inducible changes in AChE-S, RACK1, and PKCβII.
Collapse
|
46
|
Abstract
This study examined the involvement of the brain stress system in the reinforcing effects of morphine. One group of mice was conditioned to morphine using the conditioned place preference (CPP) paradigm and the other group received morphine in a home-cage (non-conditioned). Adrenocorticotropic hormone and corticosterone levels were measured by radioimmunoassay; phospho (p) CREB expression and the number of corticotropin-releasing factor (CRF) neurons and fibres were measured by immunohistochemistry in different brain areas. We observed that the number of CRF neurons in the paraventricular nucleus (PVN) was increased after morphine-induced CPP, which was paralleled with enhanced CRF-immunoreactivity fibres in the nucleus tractus solitarius (NTS) and ventral tegmental area (VTA) vs. home-cage group injected with morphine. Morphine exposure induced an increase in CREB phosphorylated at Ser133 in the PVN and central amygdale (CeA), whereas mice exhibiting morphine CPP had higher levels of pCREB in the PVN, CeA and bed nucleus of the stria terminalis (BNST). We also found that most of the CRF-positive neurons in the PVN, CeA and BNST co-express pCREB after morphine CPP expression, suggesting that the drug-associated environmental contexts can elicit neuronal activity in the brain stress system. From the present results it is clear that exposure to a drug-associated context remains a potent activator of signalling pathways leading to CRF activation in the brain stress system.
Collapse
|
47
|
Clinical doses of citalopram or reboxetine differentially modulate passive and active behaviors of female Wistar rats with high or low immobility time in the forced swimming test. Pharmacol Biochem Behav 2013; 110:89-97. [DOI: 10.1016/j.pbb.2013.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 05/29/2013] [Accepted: 06/04/2013] [Indexed: 11/24/2022]
|
48
|
Sex differences in the antidepressant-like effects of ketamine. Neuropharmacology 2013; 70:27-34. [DOI: 10.1016/j.neuropharm.2012.12.009] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 11/27/2012] [Accepted: 12/10/2012] [Indexed: 11/21/2022]
|
49
|
Matsuda K, Hagiwara Y, Shibata H, Sakashita A, Wada K. Ovine corticotropin-releasing hormone (oCRH) exerts an anxiogenic-like action in the goldfish, Carassius auratus. Gen Comp Endocrinol 2013; 188:118-22. [PMID: 23321398 DOI: 10.1016/j.ygcen.2013.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 12/25/2012] [Accepted: 01/03/2013] [Indexed: 01/30/2023]
Abstract
Corticotropin-releasing hormone (CRH) is a member of the hypothalamic neuropeptide family that includes urocortins, urotensin I and sauvagine in vertebrates. CRH and urocortin act as anorexigenic factors for satiety regulation in rodents. In a goldfish model, intracerebroventricular (ICV) administration of ovine CRH (oCRH) affects not only food intake, but also locomotor activity. However, there is no information regarding the psychophysiological roles of CRH in goldfish. Therefore, we investigated the effect of oCRH on psychomotor activity in this species. ICV administration of synthetic oCRH at 20 pmol/g body weight (BW) enhanced locomotor activity. Since intact goldfish prefer the lower to the upper area of a tank, we developed a method for measuring the time taken for fish to move from the lower to the upper area. ICV administration of oCRH at 20 pmol/g BW and the central-type benzodiazepine receptor inverse agonist FG-7142 (an anxiogenic agent) at 1-4 pmol/g BW both increased the time taken to move from the lower to the upper area. This anxiogenic-like effect of oCRH was abolished by the CRH receptor antagonist α-helical CRH(9-41) (100 pmol/g BW). These results indicate that CRH can potently affect locomotor and psychomotor activities in goldfish.
Collapse
Affiliation(s)
- Kouhei Matsuda
- Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan.
| | | | | | | | | |
Collapse
|
50
|
Matsuda K. Regulation of feeding behavior and psychomotor activity by corticotropin-releasing hormone (CRH) in fish. Front Neurosci 2013; 7:91. [PMID: 23754974 PMCID: PMC3667241 DOI: 10.3389/fnins.2013.00091] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 05/14/2013] [Indexed: 11/21/2022] Open
Abstract
Corticotropin-releasing hormone (CRH) is a hypothalamic neuropeptide belonging to a family of neuropeptides that includes urocortins, urotensin I, and sauvagine in vertebrates. CRH and urocortin act as anorexigenic factors for satiety regulation in fish. In a goldfish model, intracerebroventricular (ICV) administration of CRH has been shown to affect not only food intake, but also locomotor and psychomotor activities. In particular, CRH elicits anxiety-like behavior as an anxiogenic neuropeptide in goldfish, as is the case in rodents. This paper reviews current knowledge of CRH and its related peptides derived from studies of teleost fish, as representative non-mammals, focusing particularly on the role of the CRH system, and examines its significance from a comparative viewpoint.
Collapse
Affiliation(s)
- Kouhei Matsuda
- Laboratory of Regulatory Biology, Graduate School of Science and Engineering, Graduate School of Innovative Life Science, University of Toyama Toyama, Japan
| |
Collapse
|