1
|
Medrano M, Allaoui W, Haddad RES, Makrini-Maleville L, Valjent E, Smolders I, Kormos V, Gaszner B, De Bundel D. Neuromedin U Neurons in the Edinger-Westphal Nucleus Respond to Alcohol Without Interfering with the Urocortin 1 Response. Neurochem Res 2024; 49:3277-3296. [PMID: 39266897 PMCID: PMC11502588 DOI: 10.1007/s11064-024-04238-1] [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: 07/06/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024]
Abstract
The Edinger-Westphal nucleus (EW) is a midbrain nucleus composed of a preganglionic, cholinergic subpopulation and a densely clustered peptidergic subpopulation (EWcp). The EWcp is one of the few brain regions that show consistent induction of FOS following voluntary alcohol intake. Previous results in rodents point to urocortin 1 (UCN1) as one of the peptides most involved in the control of ethanol intake and preference. Notably, the functions described for UCN1, such as reward processing, stress coping or the regulation of feeding behavior are similar to those described for the neuropeptide neuromedin U (NMU). Interestingly, NMU has been recently associated with the modulation of alcohol-related behaviors. However, little is known about the expression and functionality of NMU neurons in alcohol-responsive areas. In this study, we used the recently developed Nmu-Cre knock-in mouse model to examine the expression of NMU in the subaqueductal paramedian zone comprising the EWcp. We delved into the characterization and co-expression of NMU with other markers already described in the EWcp. Moreover, using FOS as a marker of neuronal activity, we tested whether NMU neurons were sensitive to acute alcohol administration. Overall, we provided novel insights on NMU expression and functionality in the EW region. We showed the presence of NMU within a subpopulation of UCN1 neurons in the EWcp and demonstrated that this partial co-expression does not interfere with the responsivity of UCN1-containing cells to alcohol. Moreover, we proposed that the UCN1 content in these neurons may be influenced by sex.
Collapse
Affiliation(s)
- Mireia Medrano
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Wissal Allaoui
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Ra'fat Ehab Salim Haddad
- Medical School, Research Group for Mood Disorders, Department of Anatomy and Centre for Neuroscience, University of Pécs, Szigeti út 12, 7624, Pécs, Hungary
| | | | - Emmanuel Valjent
- IGF, Université de Montpellier, CNRS, Inserm, Montpellier, France
| | - Ilse Smolders
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Viktória Kormos
- Medical School, Department of Pharmacology and Pharmacotherapy, University of Pécs, Szigeti út 12, 7624, Pécs, Hungary
| | - Balázs Gaszner
- Medical School, Research Group for Mood Disorders, Department of Anatomy and Centre for Neuroscience, University of Pécs, Szigeti út 12, 7624, Pécs, Hungary.
| | - Dimitri De Bundel
- Center for Neurosciences, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| |
Collapse
|
2
|
Bagosi Z, Megyesi K, Ayman J, Rudersdorf H, Ayaz MK, Csabafi K. The Role of Corticotropin-Releasing Factor (CRF) and CRF-Related Peptides in the Social Behavior of Rodents. Biomedicines 2023; 11:2217. [PMID: 37626714 PMCID: PMC10452353 DOI: 10.3390/biomedicines11082217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/02/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Since the corticotropin-releasing factor (CRF) was isolated from an ovine brain, a growing family of CRF-related peptides has been discovered. Today, the mammalian CRF system consists of four ligands (CRF, urocortin 1 (Ucn1), urocortin 2 (Ucn2), and urocortin 3 (Ucn3)); two receptors (CRF receptor type 1 (CRF1) and CRF receptor type 2 (CRF2)); and a CRF-binding protein (CRF-BP). Besides the regulation of the neuroendocrine, autonomic, and behavioral responses to stress, CRF and CRF-related peptides are also involved in different aspects of social behavior. In the present study, we review the experiments that investigated the role of CRF and the urocortins involved in the social behavior of rats, mice, and voles, with a special focus on sociability and preference for social novelty, as well as the ability for social recognition, discrimination, and memory. In general, these experiments demonstrate that CRF, Ucn1, Ucn2, and Ucn3 play important, but distinct roles in the social behavior of rodents, and that they are mediated by CRF1 and/or CRF2. In addition, we suggest the possible brain regions and pathways that express CRF and CRF-related peptides and that might be involved in social interactions. Furthermore, we also emphasize the differences between the species, strains, and sexes that make translation of these roles from rodents to humans difficult.
Collapse
Affiliation(s)
- Zsolt Bagosi
- Department of Pathophysiology, Albert Szent-Györgyi School of Medicine, University of Szeged, 6720 Szeged, Hungary; (H.R.); (M.K.A.); (K.C.)
| | - Kíra Megyesi
- Interdisciplinary Center for Excellence, Clinical Research Competence Center, Albert Szent-Györgyi School of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Jázmin Ayman
- Department of Obstetrics and Gynecology, Albert Szent-Györgyi Albert School of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Hanna Rudersdorf
- Department of Pathophysiology, Albert Szent-Györgyi School of Medicine, University of Szeged, 6720 Szeged, Hungary; (H.R.); (M.K.A.); (K.C.)
| | - Maieda Khan Ayaz
- Department of Pathophysiology, Albert Szent-Györgyi School of Medicine, University of Szeged, 6720 Szeged, Hungary; (H.R.); (M.K.A.); (K.C.)
| | - Krisztina Csabafi
- Department of Pathophysiology, Albert Szent-Györgyi School of Medicine, University of Szeged, 6720 Szeged, Hungary; (H.R.); (M.K.A.); (K.C.)
| |
Collapse
|
3
|
Bhargava A. Unraveling corticotropin-releasing factor family-orchestrated signaling and function in both sexes. VITAMINS AND HORMONES 2023; 123:27-65. [PMID: 37717988 DOI: 10.1016/bs.vh.2023.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Stress responses to physical, psychological, environmental, or cellular stressors, has two arms: initiation and recovery. Corticotropin-releasing factor (CRF) is primarily responsible for regulating and/or initiating stress responses via, whereas urocortins (UCNs) are involved in the recovery response to stress via feedback inhibition. Stress is a loaded, polysemous word and is experienced in a myriad of ways. Some stressors are good for an individual, in fact essential, whereas other stressors are associated with bad outcomes. Perceived stress, like beauty, lies in the eye of the beholder, and hence the same stressor can result in individual-specific outcomes. In mammals, there are two main biological sexes with reproduction as primary function. Reproduction and nutrition can also be viewed as stressors; based on a body of work from my laboratory, we propose that the functions of all other organs have co-evolved to optimize and facilitate an individual's nutritional and reproductive functions. Hence, sex differences in physiologically relevant outcomes are innate and occur at all levels- molecular, endocrine, immune, and (patho)physiological. CRF and three UCNs are peptide hormones that mediate their physiological effects by binding to two known G protein-coupled receptors (GPCRs), CRF1 and CRF2. Expression and function of CRF family of hormones and their receptors is likely to be sexually dimorphic in all organs. In this chapter, based on the large body of work from others and my laboratory, an overview of the CRF family with special emphasis on sex-specific actions of peripherally expressed CRF2 receptor in health and disease is provided.
Collapse
Affiliation(s)
- Aditi Bhargava
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
4
|
Ma Y, Giardino WJ. Neural circuit mechanisms of the cholecystokinin (CCK) neuropeptide system in addiction. ADDICTION NEUROSCIENCE 2022; 3:100024. [PMID: 35983578 PMCID: PMC9380858 DOI: 10.1016/j.addicn.2022.100024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Given historical focus on the roles for cholecystokinin (CCK) as a peripheral hormone controlling gastrointestinal processes and a brainstem peptide regulating food intake, the study of CCK as a limbic neuromodulator coordinating reward-seeking and emotional behavior remains underappreciated. Furthermore, localization of CCK to specialized interneurons throughout the hippocampus and cortex relegated CCK to being examined primarily as a static cell type marker rather than a dynamic functional neuromodulator. Yet, over three decades of literature have been generated by efforts to delineate the central mechanisms of addiction-related behaviors mediated by the CCK system across the striatum, amygdala, hypothalamus, and midbrain. Here, we cover fundamental findings that implicate CCK neuron activity and CCK receptor signaling in modulating drug intake and drug-seeking (focusing on psychostimulants, opioids, and alcohol). In doing so, we highlight the few studies that indicate sex differences in CCK expression and corresponding drug effects, emphasizing the importance of examining hormonal influences and sex as a biological variable in translating basic science discoveries to effective treatments for substance use disorders in human patients. Finally, we point toward understudied subcortical sources of endogenous CCK and describe how continued neurotechnology advancements can be leveraged to modernize understanding of the neural circuit mechanisms underlying CCK release and signaling in addiction-relevant behaviors.
Collapse
Affiliation(s)
- Yihe Ma
- Department of Psychiatry & Behavioral Sciences and Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - William J. Giardino
- Department of Psychiatry & Behavioral Sciences and Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
5
|
Zuniga A, Smith ML, Caruso M, Ryabinin AE. Vesicular glutamate transporter 2-containing neurons of the centrally-projecting Edinger-Westphal nucleus regulate alcohol drinking and body temperature. Neuropharmacology 2021; 200:108795. [PMID: 34555367 DOI: 10.1016/j.neuropharm.2021.108795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 08/28/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022]
Abstract
Previous studies in rodents have repeatedly demonstrated that the centrally-projecting Edinger-Westphal nucleus (EWcp) is highly sensitive to alcohol and is also involved in regulating alcohol intake and body temperature. Historically, the EWcp has been known as the main site of Urocortin 1 (Ucn1) expression, a corticotropin-releasing factor-related peptide, in the brain. However, the EWcp also contains other populations of neurons, including neurons that express the vesicular glutamate transporter 2 (Vglut2). Here we transduced the EWcp with adeno-associated viruses (AAVs) encoding Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) to test the role of the EWcp in alcohol drinking and in the regulation of body temperature. Activation of the EWcp with excitatory DREADDs inhibited alcohol intake in a 2-bottle choice procedure in male C57BL/6J mice, whereas inhibition of the EWcp with DREADDs had no effect. Surprisingly, analysis of DREADD expression indicated Ucn1-containing neurons of the EWcp did not express DREADDs. In contrast, AAVs transduced non-Ucn1-containing EWcp neurons. Subsequent experiments showed that the inhibitory effect of EWcp activation on alcohol intake was also present in male Ucn1 KO mice, suggesting that a Ucn1-devoid population of EWcp regulates alcohol intake. A final set of chemogenetic experiments showed that activation of Vglut2-expressing EWcp neurons inhibited alcohol intake and induced hypothermia in male and female mice. These studies expand on previous literature by indicating that a glutamatergic, Ucn1-devoid subpopulation of the EWcp regulates alcohol consumption and body temperature.
Collapse
Affiliation(s)
- Alfredo Zuniga
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA.
| | - Monique L Smith
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Maya Caruso
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| |
Collapse
|
6
|
Cano G, Hernan SL, Sved AF. Centrally Projecting Edinger-Westphal Nucleus in the Control of Sympathetic Outflow and Energy Homeostasis. Brain Sci 2021; 11:1005. [PMID: 34439626 PMCID: PMC8392615 DOI: 10.3390/brainsci11081005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
The centrally projecting Edinger-Westphal nucleus (EWcp) is a midbrain neuronal group, adjacent but segregated from the preganglionic Edinger-Westphal nucleus that projects to the ciliary ganglion. The EWcp plays a crucial role in stress responses and in maintaining energy homeostasis under conditions that require an adjustment of energy expenditure, by virtue of modulating heart rate and blood pressure, thermogenesis, food intake, and fat and glucose metabolism. This modulation is ultimately mediated by changes in the sympathetic outflow to several effector organs, including the adrenal gland, heart, kidneys, brown and white adipose tissues and pancreas, in response to environmental conditions and the animal's energy state, providing for appropriate energy utilization. Classic neuroanatomical studies have shown that the EWcp receives inputs from forebrain regions involved in these functions and projects to presympathetic neuronal populations in the brainstem. Transneuronal tracing with pseudorabies virus has demonstrated that the EWcp is connected polysynaptically with central circuits that provide sympathetic innervation to all these effector organs that are critical for stress responses and energy homeostasis. We propose that EWcp integrates multimodal signals (stress, thermal, metabolic, endocrine, etc.) and modulates the sympathetic output simultaneously to multiple effector organs to maintain energy homeostasis under different conditions that require adjustments of energy demands.
Collapse
Affiliation(s)
- Georgina Cano
- Department of Neuroscience, A210 Langley Hall, University of Pittsburgh, Pittsburgh, PA 15260, USA; (S.L.H.); (A.F.S.)
| | | | | |
Collapse
|
7
|
Zuniga A, Ryabinin AE, Cunningham CL. Effects of pharmacological inhibition of the centrally-projecting Edinger-Westphal nucleus on ethanol-induced conditioned place preference and body temperature. Alcohol 2020; 87:121-131. [PMID: 31926294 DOI: 10.1016/j.alcohol.2020.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/18/2019] [Accepted: 01/06/2020] [Indexed: 11/18/2022]
Abstract
Alcohol use disorder is a chronic disease characterized in part by repeated relapsing events. Exposure to environmental stimuli or cues that have previously been associated with the effects of alcohol can promote relapse through the triggering of craving for alcohol. Therefore, identifying and characterizing neuronal populations that may regulate these associations is of the upmost importance. Previous studies have implicated the centrally-projecting Edinger Westphal nucleus (EWcp) in this process, as the EWcp is both sensitive to, and can regulate alcohol intake. To date however, it is unclear if the EWcp is involved in the formation or expression of these alcohol-cue associations. As such, the present studies examined the involvement of the EWcp in male DBA/2J mice in the acquisition and expression of place preference for an alcohol-paired cue using the conditioned place preference (CPP) procedure. Pharmacological inhibition of the EWcp via the GABAA and GABAB receptor agonists muscimol and baclofen did not affect either the acquisition or the expression of CPP. Follow up studies did find however, that pharmacological inhibition of the EWcp increased body temperature and prevented alcohol-induced increases in c-Fos expression in the EWcp. When considered in light of previous studies, the present results indicate that the EWcp may be involved in the regulation of alcohol self-administration, and not conditioned alcohol-seeking. Additionally, the present studies provide further evidence for the involvement of the EWcp in thermoregulation and help elucidate the molecular mechanisms by which alcohol increases c-Fos in the EWcp.
Collapse
Affiliation(s)
- Alfredo Zuniga
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239-3098, USA.
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239-3098, USA
| | - Christopher L Cunningham
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239-3098, USA; Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR, 97239-3098, USA
| |
Collapse
|
8
|
Song SY, Li Y, Zhai XM, Li YH, Bao CY, Shan CJ, Hong J, Cao JL, Zhang LC. Connection Input Mapping and 3D Reconstruction of the Brainstem and Spinal Cord Projections to the CSF-Contacting Nucleus. Front Neural Circuits 2020; 14:11. [PMID: 32296310 PMCID: PMC7136615 DOI: 10.3389/fncir.2020.00011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/10/2020] [Indexed: 01/04/2023] Open
Abstract
Objective To investigate whether the CSF-contacting nucleus receives brainstem and spinal cord projections and to understand the functional significance of these connections. Methods The retrograde tracer cholera toxin B subunit (CB) was injected into the CSF-contacting nucleus in Sprague-Dawley rats according the previously reported stereotaxic coordinates. After 7–10 days, these rats were perfused and their brainstem and spinal cord were sliced (thickness, 40 μm) using a freezing microtome. All the sections were subjected to CB immunofluorescence staining. The distribution of CB-positive neuron in different brainstem and spinal cord areas was observed under fluorescence microscope. Results The retrograde labeled CB-positive neurons were found in the midbrain, pons, medulla oblongata, and spinal cord. Four functional areas including one hundred and twelve sub-regions have projections to the CSF-contacting nucleus. However, the density of CB-positive neuron distribution ranged from sparse to dense. Conclusion Based on the connectivity patterns of the CSF-contacting nucleus receives anatomical inputs from the brainstem and spinal cord, we preliminarily conclude and summarize that the CSF-contacting nucleus participates in pain, visceral activity, sleep and arousal, emotion, and drug addiction. The present study firstly illustrates the broad projections of the CSF-contacting nucleus from the brainstem and spinal cord, which implies the complicated functions of the nucleus especially for the unique roles of coordination in neural and body fluids regulation.
Collapse
Affiliation(s)
- Si-Yuan Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Ying Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Xiao-Meng Zhai
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Yue-Hao Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Cheng-Yi Bao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Cheng-Jing Shan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Jia Hong
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Li-Cai Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
9
|
Zuniga A, Ryabinin AE. Involvement of Centrally Projecting Edinger-Westphal Nucleus Neuropeptides in Actions of Addictive Drugs. Brain Sci 2020; 10:brainsci10020067. [PMID: 31991932 PMCID: PMC7071833 DOI: 10.3390/brainsci10020067] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 11/16/2022] Open
Abstract
The centrally-projecting Edinger-Westphal nucleus (EWcp) is a brain region distinct from the preganglionic Edinger-Westphal nucleus (EWpg). In contrast to the EWpg, the EWcp does not send projections to the ciliary ganglion and appears not to regulate oculomotor function. Instead, evidence is accumulating that the EWcp is extremely sensitive to alcohol and several other drugs of abuse. Studies using surgical, genetic knockout, and shRNA approaches further implicate the EWcp in the regulation of alcohol sensitivity and self-administration. The EWcp is also known as the site of preferential expression of urocortin 1, a peptide of the corticotropin-releasing factor family. However, neuroanatomical data indicate that the EWcp is not a monotypic brain region and consists of several distinct subpopulations of neurons. It is most likely that these subpopulations of the EWcp are differentially involved in the regulation of actions of addictive drugs. This review summarizes and analyzes the current literature of the EWcp's involvement in actions of drugs of abuse in male and female subjects in light of the accumulating evidence of complexities of this brain region.
Collapse
|
10
|
Poznanski P, Lesniak A, Korostynski M, Sacharczuk M. Ethanol consumption following mild traumatic brain injury is related to blood-brain barrier permeability. Addict Biol 2020; 25:e12683. [PMID: 30334599 DOI: 10.1111/adb.12683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/12/2018] [Accepted: 09/14/2018] [Indexed: 12/21/2022]
Abstract
Several preclinical and clinical studies that deal with the neuropathological consequences of mild traumatic brain injury (mTBI) have focused on unraveling its effect on ethanol drinking behavior. Previous reports describe changes in ethanol consumption, both in animal models of mTBI as well as in patients, after concussive brain injury. However, the neurobiological mechanisms underlying this phenomenon are still poorly understood. In the present study, we used a unique model of mouse lines divergently selected for high (HA) or low (LA) swim stress-induced analgesia to examine the effect of mTBI on ethanol drinking behavior. In comparison with LA mice, their HA counterparts exhibited increased blood-brain barrier (BBB) permeability, lower basal alcohol preference, and lower level of stress-induced ethanol intake. Here, we showed that mTBI attenuates voluntary ethanol intake in LA, but not in HA mice. Interestingly, BBB disruption after mannitol infusion also decreases the level of ethanol drinking behavior in this line. We conclude that in alcohol-preferring LA mice, BBB disruption as a consequence of mTBI attenuates ethanol consumption. Our results suggest that the innate level of BBB integrity plays a pivotal role in regulation of ethanol consumption in mice showing differential endogenous opioid system activity.
Collapse
Affiliation(s)
- Piotr Poznanski
- Laboratory of NeurogenomicsInstitute of Genetics and Animal Breeding, Polish Academy of Sciences Magdalenka Poland
| | - Anna Lesniak
- Department of Pharmacodynamics, Centre for Preclinical Research and TechnologyMedical University of Warsaw Warsaw Poland
| | - Michal Korostynski
- Department of Molecular NeuropharmacologyInstitute of Pharmacology Krakow Poland
| | - Mariusz Sacharczuk
- Laboratory of NeurogenomicsInstitute of Genetics and Animal Breeding, Polish Academy of Sciences Magdalenka Poland
- Department of Pharmacodynamics, Centre for Preclinical Research and TechnologyMedical University of Warsaw Warsaw Poland
- Department of Internal Medicine, Hypertension and Vascular DiseasesMedical University of Warsaw Warsaw Poland
| |
Collapse
|
11
|
Neasta J, Darcq E, Jeanblanc J, Carnicella S, Ben Hamida S. GPCR and Alcohol-Related Behaviors in Genetically Modified Mice. Neurotherapeutics 2020; 17:17-42. [PMID: 31919661 PMCID: PMC7007453 DOI: 10.1007/s13311-019-00828-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest class of cell surface signaling receptors and regulate major neurobiological processes. Accordingly, GPCRs represent primary targets for the treatment of brain disorders. Several human genetic polymorphisms affecting GPCRs have been associated to different components of alcohol use disorder (AUD). Moreover, GPCRs have been reported to contribute to several features of alcohol-related behaviors in animal models. Besides traditional pharmacological tools, genetic-based approaches mostly aimed at deleting GPCR genes provided substantial information on how key GPCRs drive alcohol-related behaviors. In this review, we summarize the alcohol phenotypes that ensue from genetic manipulation, in particular gene deletion, of key GPCRs in rodents. We focused on GPCRs that belong to fundamental neuronal systems that have been shown as potential targets for the development of AUD treatment. Data are reviewed with particular emphasis on alcohol reward, seeking, and consumption which are behaviors that capture essential aspects of AUD. Literature survey indicates that in most cases, there is still a gap in defining the intracellular transducers and the functional crosstalk of GPCRs as well as the neuronal populations in which their signaling regulates alcohol actions. Further, the implication of only a few orphan GPCRs has been so far investigated in animal models. Combining advanced pharmacological technologies with more specific genetically modified animals and behavioral preclinical models is likely necessary to deepen our understanding in how GPCR signaling contributes to AUD and for drug discovery.
Collapse
Affiliation(s)
- Jérémie Neasta
- Laboratoire de Pharmacologie, Faculté de Pharmacie, University of Montpellier, 34093, Montpellier, France
| | - Emmanuel Darcq
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada
| | - Jérôme Jeanblanc
- Research Group on Alcohol and Pharmacodependences-INSERM U1247, University of Picardie Jules Verne, 80025, Amiens, France
| | - Sebastien Carnicella
- INSERM U1216, Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, 38000, Grenoble, France
| | - Sami Ben Hamida
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada.
| |
Collapse
|
12
|
CRF modulation of central monoaminergic function: Implications for sex differences in alcohol drinking and anxiety. Alcohol 2018; 72:33-47. [PMID: 30217435 DOI: 10.1016/j.alcohol.2018.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/03/2018] [Accepted: 01/19/2018] [Indexed: 01/06/2023]
Abstract
Decades of research have described the importance of corticotropin-releasing factor (CRF) signaling in alcohol addiction, as well as in commonly co-expressed neuropsychiatric diseases, including anxiety and mood disorders. However, CRF signaling can also acutely regulate binge alcohol consumption, anxiety, and affect in non-dependent animals, possibly via modulation of central monoaminergic signaling. We hypothesize that basal CRF tone is particularly high in animals and humans with an inherent propensity for high anxiety and alcohol consumption, and thus these individuals are at increased risk for the development of alcohol use disorder and comorbid neuropsychiatric diseases. The current review focuses on extrahypothalamic CRF circuits, particularly those stemming from the bed nucleus of the stria terminalis (BNST), found to play a role in basal phenotypes, and examines whether the intrinsic hyperactivity of these circuits is sufficient to escalate the expression of these behaviors and steepen the trajectory of development of disease states. We focus our efforts on describing CRF modulation of biogenic amine neuron populations that have widespread projections to the forebrain to modulate behaviors, including alcohol and drug intake, stress reactivity, and anxiety. Further, we review the known sex differences and estradiol modulation of these neuron populations and CRF signaling at their synapses to address the question of whether females are more susceptible to the development of comorbid addiction and stress-related neuropsychiatric diseases because of hyperactive extrahypothalamic CRF circuits compared to males.
Collapse
|
13
|
Cunningham CL, Shields CN. Effects of sex on ethanol conditioned place preference, activity and variability in C57BL/6J and DBA/2J mice. Pharmacol Biochem Behav 2018; 173:84-89. [PMID: 30036544 DOI: 10.1016/j.pbb.2018.07.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/20/2018] [Accepted: 07/20/2018] [Indexed: 01/25/2023]
Abstract
Previous studies of ethanol drinking in rodents have shown greater intake in females than in males, but the reasons behind this difference are unknown. To address one possible interpretation of the drinking difference, these studies tested the hypothesis that female and male mice differ in sensitivity to the rewarding effects of ethanol using the conditioned place preference (CPP) procedure. To increase the generalizability of the results, sex differences were examined in two inbred mouse strains known to differ in their sensitivity to ethanol reward: C57BL/6J (B6) and DBA/2J (D2). Mice were conditioned in an unbiased CPP procedure using either 1 or 2 g/kg ethanol. To detect possible differences in learning rate, they were tested once at the midpoint of conditioning and again after conditioning ended. As expected, CPP was stronger with 2 g/kg than with 1 g/kg, and D2 mice generally showed stronger CPP than B6 mice. However, there were no sex differences in the rate of CPP acquisition or in CPP magnitude, suggesting no sex difference in ethanol reward sensitivity as indexed by CPP. Nevertheless, there were sex differences in locomotor activity. B6 females were generally more active than B6 males during CPP acquisition whereas D2 females were slightly less active than D2 males during both CPP acquisition and preference testing. Unexpectedly, female mice showed more variability than males in the behavioral measures recorded in these studies, encouraging greater attention to variability in the design, analysis and interpretation of future studies of sex differences in mice.
Collapse
Affiliation(s)
- Christopher L Cunningham
- Department of Behavioral Neuroscience, Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239-3098, USA.
| | - Chloe N Shields
- Department of Behavioral Neuroscience, Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239-3098, USA
| |
Collapse
|
14
|
Corticotropin-Releasing Factor (CRF) Neurocircuitry and Neuropharmacology in Alcohol Drinking. Handb Exp Pharmacol 2018; 248:435-471. [PMID: 29374836 DOI: 10.1007/164_2017_86] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alcohol use is pervasive in the United States. In the transition from nonhazardous drinking to hazardous drinking and alcohol use disorder, neuroadaptations occur within brain reward and brain stress systems. One brain signaling system that has received much attention in animal models of excessive alcohol drinking and alcohol dependence is corticotropin-releasing factor (CRF). The CRF system is composed of CRF, the urocortins, CRF-binding protein, and two receptors - CRF type 1 and CRF type 2. This review summarizes how acute, binge, and chronic alcohol dysregulates CRF signaling in hypothalamic and extra-hypothalamic brain regions and how this dysregulation may contribute to changes in alcohol reinforcement, excessive alcohol consumption, symptoms of negative affect during withdrawal, and alcohol relapse. In addition, it summarizes clinical work examining CRF type 1 receptor antagonists in humans and discusses why the brain CRF system is still relevant in alcohol research.
Collapse
|
15
|
Karelina K, Gaier KR, Weil ZM. Traumatic brain injuries during development disrupt dopaminergic signaling. Exp Neurol 2017; 297:110-117. [PMID: 28802560 DOI: 10.1016/j.expneurol.2017.08.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/19/2017] [Accepted: 08/08/2017] [Indexed: 12/16/2022]
Abstract
Traumatic brain injuries (TBI) sustained during peri-adolescent development produce lasting neuro-behavioral changes that render individuals at an increased risk for developing substance abuse disorders. Experimental and clinical evidence of a prolonged period of hypodopaminergia after TBI have been well documented, but the effect of juvenile TBI on dopaminergic dysfunction and its relationship with substance abuse have not been investigated. In order to determine the effect of juvenile brain injury on dopaminergic signaling, female mice were injured at 21days of age and then beginning seven weeks later were assessed for behavioral sensitization to amphetamine, a drug that increases synaptic dopamine availability. Together with a histological analysis of tyrosine hydroxylase, dopamine transporter, and dopamine D2 receptor expression, our data are indicative of a persistent state of hypodopaminergia well into adulthood after a juvenile TBI. Further, mice that sustained a juvenile TBI exhibited a significantly reduced activation of cFos in the urocortin-positive cells of the Edinger-Westphal nucleus in response to ethanol administration. Taken together, these data provide strong evidence for the vulnerability of juveniles to the development of lasting neuro-behavioral problems following TBI, and indicate a role of injury-induced hypodopaminergia as a risk factor for substance abuse later in life.
Collapse
Affiliation(s)
- Kate Karelina
- Department of Neuroscience, Group in Behavioral Neuroendocrinology, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Kristopher R Gaier
- Department of Neuroscience, Group in Behavioral Neuroendocrinology, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Zachary M Weil
- Department of Neuroscience, Group in Behavioral Neuroendocrinology, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
16
|
Contribution of Urocortin to the Development of Excessive Drinking. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:275-291. [PMID: 29056154 DOI: 10.1016/bs.irn.2017.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The corticotropin-releasing factor (CRF) system plays a role in alcohol consumption, and its dysregulation can contribute to alcohol use disorder. This system includes four peptide ligands: CRF, urocortin (Ucn)1, Ucn2, and Ucn3. Historically, attention focused on CRF, however, Ucn1 also plays a critical role in excessive alcohol use. This review covers evidence for this contribution and contrasts the role of Ucn1 with CRF. While CRF can promote binge consumption, this regulation occurs through generalized mechanisms that are not specific for alcohol. In contrast, inhibition of Ucn1 action specifically blunts escalation of alcohol drinking. Lesions, genetic knockout, and RNA interference experiments indicate that the centrally projecting Edinger-Westphal nucleus is the neuroanatomical source of Ucn1 critical for alcohol drinking. We propose that the contributions of Ucn1 to excessive drinking likely occur through enhancing rewarding properties of alcohol and symptoms of alcohol withdrawal, whereas CRF drives dependence-induced drinking at later stages of alcohol use. The transition from occasional binge drinking to dependence intricately depends on CRF system plasticity and coordination of CRF and Ucn1.
Collapse
|
17
|
Giardino WJ, Rodriguez ED, Smith ML, Ford MM, Galili D, Mitchell SH, Chen A, Ryabinin AE. Control of chronic excessive alcohol drinking by genetic manipulation of the Edinger-Westphal nucleus urocortin-1 neuropeptide system. Transl Psychiatry 2017; 7:e1021. [PMID: 28140406 PMCID: PMC5299395 DOI: 10.1038/tp.2016.293] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/25/2016] [Accepted: 12/15/2016] [Indexed: 11/23/2022] Open
Abstract
Midbrain neurons of the centrally projecting Edinger-Westphal nucleus (EWcp) are activated by alcohol, and enriched with stress-responsive neuropeptide modulators (including the paralog of corticotropin-releasing factor, urocortin-1). Evidence suggests that EWcp neurons promote behavioral processes for alcohol-seeking and consumption, but a definitive role for these cells remains elusive. Here we combined targeted viral manipulations and gene array profiling of EWcp neurons with mass behavioral phenotyping in C57BL/6 J mice to directly define the links between EWcp-specific urocortin-1 expression and voluntary binge alcohol intake, demonstrating a specific importance for EWcp urocortin-1 activity in escalation of alcohol intake.
Collapse
Affiliation(s)
- W J Giardino
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - E D Rodriguez
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - M L Smith
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - M M Ford
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - D Galili
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - S H Mitchell
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - A Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - A E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA,Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA. E-mail:
| |
Collapse
|
18
|
Quadros IMH, Macedo GC, Domingues LP, Favoretto CA. An Update on CRF Mechanisms Underlying Alcohol Use Disorders and Dependence. Front Endocrinol (Lausanne) 2016; 7:134. [PMID: 27818644 PMCID: PMC5073134 DOI: 10.3389/fendo.2016.00134] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/09/2016] [Indexed: 12/31/2022] Open
Abstract
Alcohol is the most commonly used and abused substance worldwide. The emergence of alcohol use disorders, and alcohol dependence in particular, is accompanied by functional changes in brain reward and stress systems, which contribute to escalated alcohol drinking and seeking. Corticotropin-releasing factor (CRF) systems have been critically implied in the transition toward problematic alcohol drinking and alcohol dependence. This review will discuss how dysregulation of CRF function contributes to the vulnerability for escalated alcohol drinking and other consequences of alcohol consumption, based on preclinical evidence. CRF signaling, mostly via CRF1 receptors, seems to be particularly important in conditions of excessive alcohol taking and seeking, including during early and protracted withdrawal, relapse, as well as during withdrawal-induced anxiety and escalated aggression promoted by alcohol. Modulation of CRF1 function seems to exert a less prominent role over low to moderate alcohol intake, or to species-typical behaviors. While CRF mechanisms in the hypothalamic-pituitary-adrenal axis have some contribution to the neurobiology of alcohol abuse and dependence, a pivotal role for extra-hypothalamic CRF pathways, particularly in the extended amygdala, is well characterized. More recent studies further suggest a direct modulation of brain reward function by CRF signaling in the ventral tegmental area, nucleus accumbens, and the prefrontal cortex, among other structures. This review will further discuss a putative role for other components of the CRF system that contribute for the overall balance of CRF function in reward and stress pathways, including CRF2 receptors, CRF-binding protein, and urocortins, a family of CRF-related peptides.
Collapse
Affiliation(s)
- Isabel Marian Hartmann Quadros
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Giovana Camila Macedo
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Liz Paola Domingues
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Cristiane Aparecida Favoretto
- Department of Psychobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Smith ML, Hostetler CM, Heinricher MM, Ryabinin AE. Social transfer of pain in mice. SCIENCE ADVANCES 2016; 2:e1600855. [PMID: 27774512 PMCID: PMC5072181 DOI: 10.1126/sciadv.1600855] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/19/2016] [Indexed: 06/06/2023]
Abstract
A complex relationship exists between the psychosocial environment and the perception and experience of pain, and the mechanisms of the social communication of pain have yet to be elucidated. The present study examined the social communication of pain and demonstrates that "bystander" mice housed and tested in the same room as mice subjected to inflammatory pain or withdrawal from morphine or alcohol develop corresponding hyperalgesia. Olfactory cues mediate the transfer of hyperalgesia to the bystander mice, which can be measured using mechanical, thermal, and chemical tests. Hyperalgesia in bystanders does not co-occur with anxiety or changes in corticosterone and cannot be explained by visually dependent emotional contagion or stress-induced hyperalgesia. These experiments reveal the multifaceted relationship between the social environment and pain behavior and support the use of mice as a model system for investigating these factors. In addition, these experiments highlight the need for proper consideration of how experimental animals are housed and tested.
Collapse
Affiliation(s)
- Monique L. Smith
- Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L470, Portland, OR 97239, USA
| | - Caroline M. Hostetler
- Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L470, Portland, OR 97239, USA
| | - Mary M. Heinricher
- Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L470, Portland, OR 97239, USA
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR 97239, USA
| | - Andrey E. Ryabinin
- Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail Code L470, Portland, OR 97239, USA
| |
Collapse
|
20
|
Mayfield J, Arends MA, Harris RA, Blednov YA. Genes and Alcohol Consumption: Studies with Mutant Mice. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:293-355. [PMID: 27055617 PMCID: PMC5302130 DOI: 10.1016/bs.irn.2016.02.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this chapter, we review the effects of global null mutant and overexpressing transgenic mouse lines on voluntary self-administration of alcohol. We examine approximately 200 publications pertaining to the effects of 155 mouse genes on alcohol consumption in different drinking models. The targeted genes vary in function and include neurotransmitter, ion channel, neuroimmune, and neuropeptide signaling systems. The alcohol self-administration models include operant conditioning, two- and four-bottle choice continuous and intermittent access, drinking in the dark limited access, chronic intermittent ethanol, and scheduled high alcohol consumption tests. Comparisons of different drinking models using the same mutant mice are potentially the most informative, and we will highlight those examples. More mutants have been tested for continuous two-bottle choice consumption than any other test; of the 137 mouse genes examined using this model, 97 (72%) altered drinking in at least one sex. Overall, the effects of genetic manipulations on alcohol drinking often depend on the sex of the mice, alcohol concentration and time of access, genetic background, as well as the drinking test.
Collapse
Affiliation(s)
- J Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - M A Arends
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, United States
| | - R A Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States.
| | - Y A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
21
|
Tang X, Büttner-Ennever JA, Mustari MJ, Horn AKE. Internal organization of medial rectus and inferior rectus muscle neurons in the C group of the oculomotor nucleus in monkey. J Comp Neurol 2015; 523:1809-23. [PMID: 25684641 DOI: 10.1002/cne.23760] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 02/08/2015] [Accepted: 02/09/2015] [Indexed: 11/11/2022]
Abstract
Mammalian extraocular muscles contain singly innervated twitch muscle fibers (SIF) and multiply innervated nontwitch muscle fibers (MIF). In monkey, MIF motoneurons lie around the periphery of oculomotor nuclei and have premotor inputs different from those of the motoneurons inside the nuclei. The most prominent MIF motoneuron group is the C group, which innervates the medial rectus (MR) and inferior rectus (IR) muscle. To explore the organization of both cell groups within the C group, we performed small injections of choleratoxin subunit B into the myotendinous junction of MR or IR in monkeys. In three animals the IR and MR myotendinous junction of one eye was injected simultaneously with different tracers (choleratoxin subunit B and wheat germ agglutinin). This revealed that both muscles were supplied by two different, nonoverlapping populations in the C group. The IR neurons lie adjacent to the dorsomedial border of the oculomotor nucleus, whereas MR neurons are located farther medially. A striking feature was the differing pattern of dendrite distribution of both cell groups. Whereas the dendrites of IR neurons spread into the supraoculomotor area bilaterally, those of the MR neurons were restricted to the ipsilateral side and sent a focused bundle dorsally to the preganglionic neurons of the Edinger-Westphal nucleus, which are involved in the "near response." In conclusion, MR and IR are innervated by independent neuron populations from the C group. Their dendritic branching pattern within the supraoculomotor area indicates a participation in the near response providing vergence but also reflects their differing functional roles.
Collapse
Affiliation(s)
- Xiaofang Tang
- Institute of Anatomy and Cell Biology, Department I, Ludwig-Maximilians-University of Munich, D-80336, Munich, Germany
| | - Jean A Büttner-Ennever
- Institute of Anatomy and Cell Biology, Department I, Ludwig-Maximilians-University of Munich, D-80336, Munich, Germany
| | - Michael J Mustari
- Washington National Primate Research Center and Department of Ophthalmology, University of Washington, Seattle, Washington, 98195
| | - Anja K E Horn
- Institute of Anatomy and Cell Biology, Department I, Ludwig-Maximilians-University of Munich, D-80336, Munich, Germany
| |
Collapse
|
22
|
Phillips TJ, Reed C, Pastor R. Preclinical evidence implicating corticotropin-releasing factor signaling in ethanol consumption and neuroadaptation. GENES, BRAIN, AND BEHAVIOR 2015; 14:98-135. [PMID: 25565358 PMCID: PMC4851463 DOI: 10.1111/gbb.12189] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/22/2014] [Accepted: 11/25/2014] [Indexed: 12/15/2022]
Abstract
The results of many studies support the influence of the corticotropin-releasing factor (CRF) system on ethanol (EtOH) consumption and EtOH-induced neuroadaptations that are critical in the addiction process. This review summarizes the preclinical data in this area after first providing an overview of the components of the CRF system. This complex system involves hypothalamic and extra-hypothalamic mechanisms that play a role in the central and peripheral consequences of stressors, including EtOH and other drugs of abuse. In addition, several endogenous ligands and targets make up this system and show differences in their involvement in EtOH drinking and in the effects of chronic or repeated EtOH treatment. In general, genetic and pharmacological approaches paint a consistent picture of the importance of CRF signaling via type 1 CRF receptors (CRF(1)) in EtOH-induced neuroadaptations that result in higher levels of intake, encourage alcohol seeking during abstinence and alter EtOH sensitivity. Furthermore, genetic findings in rodents, non-human primates and humans have provided some evidence of associations of genetic polymorphisms in CRF-related genes with EtOH drinking, although additional data are needed. These results suggest that CRF(1) antagonists have potential as pharmacotherapeutics for alcohol use disorders. However, given the broad and important role of these receptors in adaptation to environmental and other challenges, full antagonist effects may be too profound and consideration should be given to treatments with modulatory effects.
Collapse
Affiliation(s)
- T. J. Phillips
- VA Portland Health Care System, Portland Alcohol Research Center, Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Portland Alcohol Research Center, Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - C. Reed
- Department of Behavioral Neuroscience, Portland Alcohol Research Center, Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - R. Pastor
- Department of Behavioral Neuroscience, Portland Alcohol Research Center, Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
- Area de Psicobiología, Universitat Jaume I, 12071 Castellón, Spain
- Department of Psychology, Reed College, Portland, OR, USA
| |
Collapse
|
23
|
Smith ML, Li J, Ryabinin AE. Increased alcohol consumption in urocortin 3 knockout mice is unaffected by chronic inflammatory pain. Alcohol Alcohol 2014; 50:132-9. [PMID: 25451237 DOI: 10.1093/alcalc/agu084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
AIMS Stress neurocircuitry may modulate the relationship between alcohol drinking and chronic pain. The corticotropin-releasing factor (CRF) system is crucial for regulation of stress responses. The current study aimed to elucidate the role of the endogenous CRF ligand Urocortin 3 (Ucn3) in the relationship between alcohol drinking behavior and chronic pain using a genetic approach. METHODS Ucn3 (KO) and wildtype (WT) littermates were subjected to a 24-h access drinking procedure prior to and following induction of chronic inflammatory pain. RESULTS Ucn3 KO mice displayed significantly increased ethanol intake and preference compared with WT across the time course. There were no long-term effects of chronic pain on alcohol drinking behavior, regardless of genotype, nor any evidence for alcohol-induced analgesia. CONCLUSION The increased drinking in Ucn3 KO supports a role for this peptide in alcohol-related behavior. These data suggest the necessity for more research exploring the relationship between alcohol drinking, chronic pain and the CRF system in rodent models.
Collapse
Affiliation(s)
- Monique L Smith
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road L470, Portland, OR 97239-3098, USA
| | - Ju Li
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road L470, Portland, OR 97239-3098, USA
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road L470, Portland, OR 97239-3098, USA
| |
Collapse
|
24
|
Corticotropin releasing factor: a key role in the neurobiology of addiction. Front Neuroendocrinol 2014; 35:234-44. [PMID: 24456850 PMCID: PMC4213066 DOI: 10.1016/j.yfrne.2014.01.001] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 01/03/2014] [Accepted: 01/06/2014] [Indexed: 11/20/2022]
Abstract
Drug addiction is a chronically relapsing disorder characterized by loss of control over intake and dysregulation of stress-related brain emotional systems. Since the discovery by Wylie Vale and his colleagues of corticotropin-releasing factor (CRF) and the structurally-related urocortins, CRF systems have emerged as mediators of the body's response to stress. Relatedly, CRF systems have a prominent role in driving addiction via actions in the central extended amygdala, producing anxiety-like behavior, reward deficits, excessive, compulsive-like drug self-administration and stress-induced reinstatement of drug seeking. CRF neuron activation in the medial prefrontal cortex may also contribute to the loss of control. Polymorphisms in CRF system molecules are associated with drug use phenotypes in humans, often in interaction with stress history. Drug discovery efforts have yielded brain-penetrant CRF1 antagonists with activity in preclinical models of addiction. The results support the hypothesis that brain CRF-CRF1 systems contribute to the etiology and maintenance of addiction.
Collapse
|
25
|
Yong W, Spence JP, Eskay R, Fitz SD, Damadzic R, Lai D, Foroud T, Carr LG, Shekhar A, Chester JA, Heilig M, Liang T. Alcohol-preferring rats show decreased corticotropin-releasing hormone-2 receptor expression and differences in HPA activation compared to alcohol-nonpreferring rats. Alcohol Clin Exp Res 2014; 38:1275-83. [PMID: 24611993 DOI: 10.1111/acer.12379] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 01/07/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND Corticotropin-releasing hormone (CRH) and urocortins (UCNs) bind to corticotropin-releasing hormone type 2 receptor (CRF2 receptor ), a Gs protein-coupled receptor that plays an important role in modulation of anxiety and stress responses. The Crhr2 gene maps to a quantitative trait locus (QTL) for alcohol preference on chromosome 4 previously identified in inbred alcohol-preferring (iP) and-nonpreferring (iNP) F2 rats. METHODS Real-time polymerase chain reaction was utilized to screen for differences in Crhr2 mRNA expression in the central nervous system (CNS) of male iP and iNP rats. DNA sequence analysis was then performed to screen for polymorphism in Crhr2 in order to identify genetic variation, and luciferase reporter assays were then applied to test their functional significance. Next, binding assays were used to determine whether this polymorphism affected CRF2 receptor binding affinity as well as CRF2 receptor density in the CNS. Finally, social interaction and corticosterone levels were measured in the P and NP rats before and after 30-minute restraint stress. RESULTS Crhr2 mRNA expression studies found lower levels of Crhr2 mRNA in iP rats compared to iNP rats. In addition, DNA sequencing identified polymorphisms in the promoter region, coding region, and 3'-untranslated region between the iP and iNP rats. A 7 bp insertion in the Crhr2 promoter of iP rats altered expression in vitro as measured by reporter assays, and we found that CRF2 receptor density was lower in the amygdala of iP as compared to iNP rats. Male P rats displayed decreased social interaction and significantly higher corticosterone levels directly following 30-minute restraint when compared to male NP rats. CONCLUSIONS This study identified Crhr2 as a candidate gene of interest underlying the chromosome 4 QTL for alcohol consumption that was previously identified in the P and NP model. Crhr2 promoter polymorphism is associated with reduced mRNA expression in certain brain regions, particularly the amygdala, and lowered the density of CRF2 receptor in the amygdala of iP compared to iNP rats. Together, these differences between the animals may contribute to the drinking disparity as well as the anxiety differences of the P and NP rats.
Collapse
Affiliation(s)
- Weidong Yong
- Institute of Laboratory Animal Science , Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China; Department of Pediatrics , Indiana University School of Medicine, Indianapolis, Indiana
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Cocaine- and amphetamine-regulated transcript (CART) peptide immunoreactivity in feeding- and reward-related brain areas of young OLETF rats. J Chem Neuroanat 2013; 50-51:75-84. [PMID: 23545074 DOI: 10.1016/j.jchemneu.2013.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 03/21/2013] [Accepted: 03/21/2013] [Indexed: 02/06/2023]
Abstract
Cocaine- and amphetamine-regulated transcript (CART) peptide is expressed in brain areas involved in the control of appetite, drug reward and homeostatic regulation and it has an overall anorexigenic effect. Recently, we have shown that CART peptide immunoreactivity was significantly reduced in the rostral part of the nucleus accumbens and in the rostro-medial part of the nucleus of the solitary tract in adult CCK-1 receptor deficient obese diabetic Otsuka Long Evans Tokushima Fatty (OLETF) rats compared to Long Evans Tokushima Otsuka (LETO) lean controls. It is not clear, however, whether altered CART expression is caused primarily by the deficiency in CCK-1 signaling or whether is related to the obese and diabetic phenotype of the OLETF strain which develops at a later age. Therefore, in the present study, CART-immunoreaction in feeding-related areas of the brain was compared in young, age-matched (6-7 weeks old) non-obese, non-diabetic OLETF rats and in LETO controls. We found that, young, non-diabetic OLETF rats revealed unaltered distribution of CART-peptide expressing neurons and axons throughout the brain when compared to age-matched LETO rats. In contrast to previous results observed in the obese diabetic adult rats, intensity of CART immunoreaction did not differ in the areas related to control of food-intake and reward in the young OLETFs compared to young LETO rats. Our findings suggest that factors secondary to obesity and/or diabetes rather than impaired CCK-1 receptor signaling may contribute to altered CART expression in the OLETF strain.
Collapse
|
27
|
Giardino WJ, Ryabinin AE. CRF1 receptor signaling regulates food and fluid intake in the drinking-in-the-dark model of binge alcohol consumption. Alcohol Clin Exp Res 2013; 37:1161-70. [PMID: 23398267 DOI: 10.1111/acer.12076] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/30/2012] [Indexed: 11/29/2022]
Abstract
BACKGROUND Several recent studies implementing the standard "drinking-in-the-dark" (DID) model of short-term binge-like ethanol (EtOH) intake in C57BL/6J mice highlighted a role for the stress-related neuropeptide corticotropin-releasing factor (CRF) and its primary binding partner, the CRF type-1 (CRF1) receptor. METHODS We evaluated the selectivity of CRF1 involvement in binge-like EtOH intake by interrupting CRF1 function via pharmacological and genetic methods in a slightly modified 2-bottle choice DID model that allowed calculation of an EtOH preference ratio. In addition to determining EtOH intake and preference, we also measured consumption of food and H2 O during the DID period, both in the presence and absence of EtOH and sweet tastant solutions. RESULTS Treatment with either of the CRF1-selective antagonists CP-376,395 (CP; 10 to 20 mg/kg, i.p.) or NBI-27914 (10 to 30 mg/kg, i.p.) decreased intake of 15% EtOH in male C57BL/6J mice, but did so in the absence of a concomitant decrease in EtOH preference. These findings were replicated genetically in a CRF1 knockout (KO) mouse model (also on a C57BL/6J background). In contrast to effects on EtOH intake, pharmacological blockade of CRF1 with CP increased intake of 10% sucrose, consistent with previous findings in CRF1 KO mice. Finally, pharmacological and genetic disruption of CRF1 activity significantly reduced feeding and/or total caloric intake in all experiments, confirming the existence of nonspecific effects. CONCLUSIONS Our findings indicate that blockade of CRF1 receptors does not exert specific effects on EtOH intake in the DID paradigm, and that slight modifications to this procedure, as well as additional consummatory control experiments, may be useful when evaluating the selectivity of pharmacological and genetic manipulations on binge-like EtOH intake.
Collapse
Affiliation(s)
- William J Giardino
- Department of Behavioral Neuroscience and Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | | |
Collapse
|
28
|
Alcohol in excess: CRF₁ receptors in the rat and mouse VTA and DRN. Psychopharmacology (Berl) 2013; 225:313-27. [PMID: 22885872 PMCID: PMC3518642 DOI: 10.1007/s00213-012-2820-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/16/2012] [Indexed: 12/29/2022]
Abstract
RATIONALE Manipulation of the stress neuropeptide corticotropin-releasing factor (CRF), specifically central antagonism of the type 1 receptors (CRF-R1), effectively reduces alcoholic-like ethanol drinking in rodents. Escalated consumption is largely controlled by neurocircuitry that is important for reward and affect, such as the ventral tegmental area (VTA) and the dorsal raphé nucleus (DRN). OBJECTIVE The current studies investigated the role of CRF-R1 within the VTA and DRN and their relation to escalated ethanol drinking in two species. An additional goal was to explore whether high alcohol-drinking individuals would be more affected by CRF-R1 antagonism than low alcohol-drinking individuals. METHODS With a two-bottle choice drinking procedure, adult male C57BL/6J mice and Long-Evans rats were given 24-h access to 20 % ethanol and water on an intermittent schedule. Rats and mice were implanted with cannulae targeting the VTA or DRN. Doses of the CRF-R1 antagonist CP-154,526 (butyl-[2,4,6-trimethylphenyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]ethylamine)) were microinfused to modulate drinking of ethanol and water over the course of 24 h. RESULTS In both mice and rats, intra-VTA CP-154,526 selectively decreased ethanol intake, while identical doses (0.3 and 0.6 μg) infused intra-DRN reduced both ethanol and water drinking. Long-Evans rats displayed a range of individual differences for ethanol preference, and CP-154,526 suppressed ethanol drinking in the high-preferring animals regardless of brain site manipulation. CONCLUSIONS The current findings confirm previous studies that blockade of CRF-R1 efficaciously reduces escalated drinking while also suggesting that the effects of intermittent access on alcohol consumption may require CRF interaction with dopamine in the VTA.
Collapse
|
29
|
Schank JR, Ryabinin AE, Giardino WJ, Ciccocioppo R, Heilig M. Stress-related neuropeptides and addictive behaviors: beyond the usual suspects. Neuron 2012; 76:192-208. [PMID: 23040815 PMCID: PMC3495179 DOI: 10.1016/j.neuron.2012.09.026] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Addictive disorders are chronic, relapsing conditions that cause extensive disease burden. Genetic factors partly account for susceptibility to addiction, but environmental factors such as stressful experiences and prolonged exposure of the brain to addictive drugs promote its development. Progression to addiction involves neuroadaptations within neurocircuitry that mediates stress responses and is influenced by several peptidergic neuromodulators. While corticotrophin releasing factor is the prototypic member of this class, recent work has identified several additional stress-related neuropeptides that play an important role in regulation of drug intake and relapse, including the urocortins, nociceptin, substance P, and neuropeptide S. Here, we review this emerging literature, discussing to what extent the properties of these neuromodulators are shared or distinct and considering their potential as drug targets.
Collapse
Affiliation(s)
- Jesse R. Schank
- Laboratory of Clinical and Translational Studies, National Inst. on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Andrey E. Ryabinin
- Dept. of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239-3098
| | - William J. Giardino
- Dept. of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239-3098
| | - Roberto Ciccocioppo
- Dept. of Experimental Medicine and Public Health, Camerino University, Italy
| | - Markus Heilig
- Laboratory of Clinical and Translational Studies, National Inst. on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
30
|
Laryea G, Arnett MG, Muglia LJ. Behavioral Studies and Genetic Alterations in Corticotropin-Releasing Hormone (CRH) Neurocircuitry: Insights into Human Psychiatric Disorders. Behav Sci (Basel) 2012; 2:135-71. [PMID: 23077729 PMCID: PMC3471213 DOI: 10.3390/bs2020135] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Revised: 05/23/2012] [Accepted: 06/15/2012] [Indexed: 12/20/2022] Open
Abstract
To maintain well-being, all organisms require the ability to re-establish homeostasis in the presence of adverse physiological or psychological experiences. The regulation of the hypothalamic-pituitary adrenal (HPA) axis during stress is important in preventing maladaptive responses that may increase susceptibility to affective disorders. Corticotropin-releasing hormone (CRH) is a central stress hormone in the HPA axis pathway and has been implicated in stress-induced psychiatric disorders, reproductive and cardiac function, as well as energy metabolism. In the context of psychiatric disorders, CRH dysfunction is associated with the occurrence of post-traumatic stress disorder, major depression, anorexia nervosa, and anxiety disorders. Here, we review the synthesis, molecular signaling and regulation, as well as synaptic activity of CRH. We go on to summarize studies of altered CRH signaling in mutant animal models. This assembled data demonstrate an important role for CRH in neuroendocrine, autonomic, and behavioral correlates of adaptation and maladaptation. Next, we present findings regarding human genetic polymorphisms in CRH pathway genes that are associated with stress and psychiatric disorders. Finally, we discuss a role for regulators of CRH activity as potential sites for therapeutic intervention aimed at treating maladaptive behaviors associated with stress.
Collapse
Affiliation(s)
- Gloria Laryea
- Neuroscience Graduate Program, School of Medicine, Vanderbilt University, 465 21st. Avenue South, Nashville, TN 37232, USA; E-Mail:
- Center for Preterm Birth Research, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; E-Mail:
| | - Melinda G. Arnett
- Center for Preterm Birth Research, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; E-Mail:
| | - Louis J. Muglia
- Center for Preterm Birth Research, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; E-Mail:
| |
Collapse
|
31
|
Ryabinin AE, Tsoory MM, Kozicz T, Thiele TE, Neufeld-Cohen A, Chen A, Lowery-Gionta EG, Giardino WJ, Kaur S. Urocortins: CRF's siblings and their potential role in anxiety, depression and alcohol drinking behavior. Alcohol 2012; 46:349-57. [PMID: 22444954 PMCID: PMC3358480 DOI: 10.1016/j.alcohol.2011.10.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 10/10/2011] [Accepted: 10/10/2011] [Indexed: 12/30/2022]
Abstract
It is widely accepted that stress, anxiety, depression and alcohol abuse-related disorders are in large part controlled by corticotropin-releasing factor (CRF) receptors. However, evidence is accumulating that some of the actions on these receptors are mediated not by CRF, but by a family of related Urocortin (Ucn) peptides Ucn1, Ucn2 and Ucn3. The initial narrow focus on CRF as the potential main player acting on CRF receptors appears outdated. Instead it is suggested that CRF and the individual Ucns act in a complementary and brain region-specific fashion to regulate anxiety-related behaviors and alcohol consumption. This review, based on a symposium held in 2011 at the research meeting on "Alcoholism and Stress" in Volterra, Italy, highlights recent evidence for regulation of these behaviors by Ucns. In studies on stress and anxiety, the roles of Ucns, and in particular Ucn1, appear more visible in experiments analyzing adaptation to stressors rather than testing basal anxiety states. Based on these studies, we propose that the contribution of Ucn1 to regulating mood follows a U-like pattern with both high and low activity of Ucn1 contributing to high anxiety states. In studies on alcohol use disorders, the CRF system appears to regulate not only dependence-induced drinking, but also binge drinking and even basal consumption of alcohol. While dependence-induced and binge drinking rely on the actions of CRF on CRFR1 receptors, alcohol consumption in models of these behaviors is inhibited by actions of Ucns on CRFR2. In contrast, alcohol preference is positively influenced by actions of Ucn1, which is capable of acting on both CRFR1 and CRFR2. Because of complex distribution of Ucns in the nervous system, advances in this field will critically depend on development of new tools allowing site-specific analyses of the roles of Ucns and CRF.
Collapse
Affiliation(s)
- Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, L470, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Giardino WJ, Cote DM, Li J, Ryabinin AE. Characterization of Genetic Differences within the Centrally Projecting Edinger-Westphal Nucleus of C57BL/6J and DBA/2J Mice by Expression Profiling. Front Neuroanat 2012; 6:22. [PMID: 22347848 PMCID: PMC3278674 DOI: 10.3389/fnana.2012.00005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 06/05/2012] [Indexed: 11/13/2022] Open
Abstract
Detailed examination of the midbrain Edinger–Westphal (EW) nucleus revealed the existence of two distinct nuclei. One population of EW preganglionic (EWpg) neurons was found to control oculomotor functions, and a separate population of EW centrally projecting (EWcp) neurons was found to contain stress- and feeding-related neuropeptides. Although it has been shown that EWcp neurons are highly responsive to drugs of abuse and behavioral stress, a genetic characterization of the EWcp was needed. To identify genetic differences in the EWcp of inbred mouse strains that differ in behaviors relevant to EWcp function, we used publicly available tools from the Allen Brain Atlas to identify 68 transcripts that were selectively expressed in the EWcp, and examined their expression within tissue punch microdissection samples containing the EWcp of adult male C57BL/6J (B6) and DBA/2J (D2) mice. Using 96-well quantitative real-time PCR (qPCR) arrays that included the EWcp-specific genes, several other genes of interest, and five housekeeping genes, we identified strain differences in expression of 11 EWcp-specific genes (BC023892, Btg3, Bves, Cart, Cck, Ghsr, Neto1, Postn, Ptprn, Rcn1, and Ucn), two immediate early genes (Egr1 and Fos), and one dopamine-related gene (Drd5). All significant expression differences were greater in B6 vs. D2 mice, and several of these were verified either at the protein level using immunohistochemistry (IHC) or in silico using microarray data sets from whole brain and other brain areas. These results demonstrate a significant advance in our understanding of the EWcp on three levels. First, we generated a list of EWcp-specific genes (most of which had not yet been reported within the EWcp in the literature) that will be informative for future studies of EWcp function. Second, due to similarity in results from qPCR and IHC, we revealed that strain differences in basal EWcp neuropeptide content are accounted for by differential transcription and number of peptidergic neurons, rather than by differential rates of peptide release. And third, our identification of differentially expressed EWcp-specific genes between B6 and D2 mice may hold powerful insight into the neurogenetic contributions of the EWcp to stress- and addiction-related behaviors.
Collapse
Affiliation(s)
- William J Giardino
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, OR, USA
| | | | | | | |
Collapse
|
33
|
Corticotropin-releasing factor: innocent until proven guilty. Nat Rev Neurosci 2011; 13:70; author reply 70. [PMID: 22183439 DOI: 10.1038/nrn3110-c1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|