1
|
Kim SH, Lee B, Lee SM, Kim Y. Restoring social deficits in IRSp53-deleted mice: chemogenetic inhibition of ventral dentate gyrus Emx1-expressing cells. Transl Psychiatry 2024; 14:425. [PMID: 39375329 PMCID: PMC11458854 DOI: 10.1038/s41398-024-03104-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024] Open
Abstract
IRSp53 is a synaptic scaffold protein reported to be involved in schizophrenia, autism spectrum disorders, and social deficits in knockout mice. Identifying critical brain regions and cells related to IRSp53 deletion is expected to be of great help in the treatment of psychiatric problems. In this study, we performed chemogenetic inhibition within the ventral dentate gyrus (vDG) of mice with IRSp53 deletion in Emx1-expressing cells (Emx1-Cre;IRSp53 flox/flox). We observed the recovery of social deficits after chemogenetic inhibition within vDG of Emx1-Cre;IRSp53 flox/flox mice. Additionally, chemogenetic activation induced social deficits in Emx1-Cre mice. CRHR1 expression increased in the hippocampus of Emx1-Cre;IRSp53 flox/flox mice, and CRHR1 was reduced by chemogenetic inhibition. Htd2, Ccn1, and Atp61l were decreased in bulk RNA sequencing, and Eya1 and Ecrg4 were decreased in single-cell RNA sequencing of the hippocampus in Emx1-Cre;IRSp53 flox/flox mice compared to control mice. This study determined that the vDG is a critical brain region for social deficits caused by IRSp53 deletion. Social deficits in Emx1-Cre;IRSp53 flox/flox mice were recovered through chemogenetic inhibition, providing clues for new treatment methods for psychiatric disorders accompanied by social deficits.
Collapse
Affiliation(s)
- Su Hyun Kim
- Mental Health Research Institute, National Center for Mental Health, Seoul, South Korea
| | - Bomee Lee
- Mental Health Research Institute, National Center for Mental Health, Seoul, South Korea
| | - Seong Mi Lee
- Mental Health Research Institute, National Center for Mental Health, Seoul, South Korea
| | - Yangsik Kim
- Department of Psychiatry, Inha University Hospital, College of Medicine, Inha University, Incheon, South Korea.
| |
Collapse
|
2
|
de Barcellos Filho PG, Dantzler HA, Hasser EM, Kline DD. Oxytocin and corticotropin-releasing hormone exaggerate nucleus tractus solitarii neuronal and synaptic activity following chronic intermittent hypoxia. J Physiol 2024; 602:3375-3400. [PMID: 38698722 PMCID: PMC11251298 DOI: 10.1113/jp286069] [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: 11/29/2023] [Accepted: 03/28/2024] [Indexed: 05/05/2024] Open
Abstract
Chronic intermittent hypoxia (CIH) in rodents mimics the hypoxia-induced elevation of blood pressure seen in individuals experiencing episodic breathing. The brainstem nucleus tractus solitarii (nTS) is the first site of visceral sensory afferent integration, and thus is critical for cardiorespiratory homeostasis and its adaptation during a variety of stressors. In addition, the paraventricular nucleus of the hypothalamus (PVN), in part through its nTS projections that contain oxytocin (OT) and/or corticotropin-releasing hormone (CRH), contributes to cardiorespiratory regulation. Within the nTS, these PVN-derived neuropeptides alter nTS activity and the cardiorespiratory response to hypoxia. Nevertheless, their contribution to nTS activity after CIH is not fully understood. We hypothesized that OT and CRH would increase nTS activity to a greater extent following CIH, and co-activation of OT+CRH receptors would further magnify nTS activity. Our data show that compared to their normoxic controls, 10 days' CIH exaggerated nTS discharge, excitatory synaptic currents and Ca2+ influx in response to CRH, which were further enhanced by the addition of OT. CIH increased the tonic functional contribution of CRH receptors, which occurred with elevation of mRNA and protein. Together, our data demonstrate that intermittent hypoxia exaggerates the expression and function of neuropeptides on nTS activity. KEY POINTS: Episodic breathing and chronic intermittent hypoxia (CIH) are associated with autonomic dysregulation, including elevated sympathetic nervous system activity. Altered nucleus tractus solitarii (nTS) activity contributes to this response. Neurons originating in the paraventricular nucleus (PVN), including those containing oxytocin (OT) and corticotropin-releasing hormone (CRH), project to the nTS, and modulate the cardiorespiratory system. Their role in CIH is unknown. In this study, we focused on OT and CRH individually and together on nTS activity from rats exposed to either CIH or normoxia control. We show that after CIH, CRH alone and with OT increased to a greater extent overall nTS discharge, neuronal calcium influx, synaptic transmission to second-order nTS neurons, and OT and CRH receptor expression. These results provide insights into the underlying circuits and mechanisms contributing to autonomic dysfunction during periods of episodic breathing.
Collapse
Affiliation(s)
- Procopio Gama de Barcellos Filho
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| | - Heather A. Dantzler
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| | - Eileen M. Hasser
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| | - David D. Kline
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| |
Collapse
|
3
|
Montero-Atalaya M, Expósito S, Muñoz-Arnaiz R, Makarova J, Bartolomé B, Martín E, Moreno-Arribas MV, Herreras O. A dietary polyphenol metabolite alters CA1 excitability ex vivo and mildly affects cortico-hippocampal field potential generators in anesthetized animals. Cereb Cortex 2023; 33:10411-10425. [PMID: 37550066 PMCID: PMC10545443 DOI: 10.1093/cercor/bhad292] [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: 02/16/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/09/2023] Open
Abstract
Dietary polyphenols have beneficial effects in situations of impaired cognition in acute models of neurodegeneration. The possibility that they may have a direct effect on the electrical activity of neuronal populations has not been tested. We explored the electrophysiological action of protocatechuic acid (PCA) on CA1 pyramidal cells ex vivo and network activity in anesthetized female rats using pathway-specific field potential (FP) generators obtained from laminar FPs in cortex and hippocampus. Whole-cell recordings from CA1 pyramidal cells revealed increased synaptic potentials, particularly in response to basal dendritic excitation, while the associated evoked firing was significantly reduced. This counterintuitive result was attributed to a marked increase of the rheobase and voltage threshold, indicating a decreased ability to generate spikes in response to depolarizing current. Systemic administration of PCA only slightly altered the ongoing activity of some FP generators, although it produced a striking disengagement of infraslow activities between the cortex and hippocampus on a scale of minutes. To our knowledge, this is the first report showing the direct action of a dietary polyphenol on electrical activity, performing neuromodulatory roles at both the cellular and network levels.
Collapse
Affiliation(s)
- Marta Montero-Atalaya
- Dept Biotecnología y Microbiología de Alimentos, Institute of Food Science Research (CIAL), CSIC-UAM, c/Nicolás Cabrera, 9, 28049 Madrid, Spain
| | - Sara Expósito
- Dept Neurociencia Translacional, Cajal Institute, CSIC, Av Doctor Arce 37, 28002 Madrid, Spain
| | - Ricardo Muñoz-Arnaiz
- Dept Neurociencia Translacional, Cajal Institute, CSIC, Av Doctor Arce 37, 28002 Madrid, Spain
| | - Julia Makarova
- Dept Neurociencia Translacional, Cajal Institute, CSIC, Av Doctor Arce 37, 28002 Madrid, Spain
| | - Begoña Bartolomé
- Dept Biotecnología y Microbiología de Alimentos, Institute of Food Science Research (CIAL), CSIC-UAM, c/Nicolás Cabrera, 9, 28049 Madrid, Spain
| | - Eduardo Martín
- Dept Neurociencia Translacional, Cajal Institute, CSIC, Av Doctor Arce 37, 28002 Madrid, Spain
| | - María Victoria Moreno-Arribas
- Dept Biotecnología y Microbiología de Alimentos, Institute of Food Science Research (CIAL), CSIC-UAM, c/Nicolás Cabrera, 9, 28049 Madrid, Spain
| | - Oscar Herreras
- Dept Neurociencia Translacional, Cajal Institute, CSIC, Av Doctor Arce 37, 28002 Madrid, Spain
| |
Collapse
|
4
|
Colquitt BM, Li K, Green F, Veline R, Brainard MS. Neural circuit-wide analysis of changes to gene expression during deafening-induced birdsong destabilization. eLife 2023; 12:e85970. [PMID: 37284822 PMCID: PMC10259477 DOI: 10.7554/elife.85970] [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: 01/05/2023] [Accepted: 04/17/2023] [Indexed: 06/08/2023] Open
Abstract
Sensory feedback is required for the stable execution of learned motor skills, and its loss can severely disrupt motor performance. The neural mechanisms that mediate sensorimotor stability have been extensively studied at systems and physiological levels, yet relatively little is known about how disruptions to sensory input alter the molecular properties of associated motor systems. Songbird courtship song, a model for skilled behavior, is a learned and highly structured vocalization that is destabilized following deafening. Here, we sought to determine how the loss of auditory feedback modifies gene expression and its coordination across the birdsong sensorimotor circuit. To facilitate this system-wide analysis of transcriptional responses, we developed a gene expression profiling approach that enables the construction of hundreds of spatially-defined RNA-sequencing libraries. Using this method, we found that deafening preferentially alters gene expression across birdsong neural circuitry relative to surrounding areas, particularly in premotor and striatal regions. Genes with altered expression are associated with synaptic transmission, neuronal spines, and neuromodulation and show a bias toward expression in glutamatergic neurons and Pvalb/Sst-class GABAergic interneurons. We also found that connected song regions exhibit correlations in gene expression that were reduced in deafened birds relative to hearing birds, suggesting that song destabilization alters the inter-region coordination of transcriptional states. Finally, lesioning LMAN, a forebrain afferent of RA required for deafening-induced song plasticity, had the largest effect on groups of genes that were also most affected by deafening. Combined, this integrated transcriptomics analysis demonstrates that the loss of peripheral sensory input drives a distributed gene expression response throughout associated sensorimotor neural circuitry and identifies specific candidate molecular and cellular mechanisms that support the stability and plasticity of learned motor skills.
Collapse
Affiliation(s)
- Bradley M Colquitt
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Kelly Li
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Foad Green
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Robert Veline
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Michael S Brainard
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
5
|
Local production of corticotropin-releasing hormone in prefrontal cortex modulates male-specific novelty exploration. Proc Natl Acad Sci U S A 2022; 119:e2211454119. [PMID: 36442105 PMCID: PMC9894189 DOI: 10.1073/pnas.2211454119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Neuromodulatory substances can be released from distal afferents for communication between brain structures or produced locally to modulate neighboring circuit elements. Corticotropin-releasing hormone (CRH) from long-range neurons in the hypothalamus projecting to the medial prefrontal cortex (mPFC) has been shown to induce anxiety-like behaviors. However, the role of CRH produced in the mPFC has not been investigated. Here we demonstrate that a specific class of mPFC interneurons that express CRH (CrhINs) releases CRH upon high-frequency stimulation to enhance excitability of layer 2/3 pyramidal cells (L2/3 PCs) expressing the CRH receptors. When stimulated at low frequency, CrhINs release GABA resulting in the inhibition of oxytocin receptor-expressing interneurons (OxtrINs) and L2/3 PCs. Conditional deletion of CRH in mPFC CrhINs and chemogenetic activation of CrhINs have opposite effects on novelty exploration in male but not in female mice, and do not affect anxiety-related behaviors in either males or females. Our data reveal that CRH produced by local interneurons in the mPFC is required for sex-specific novelty exploration and suggest that our understanding of complex behaviors may require knowledge of local and remote neuromodulatory action.
Collapse
|
6
|
Rieger NS, Varela JA, Ng AJ, Granata L, Djerdjaj A, Brenhouse HC, Christianson JP. Insular cortex corticotropin-releasing factor integrates stress signaling with social affective behavior. Neuropsychopharmacology 2022; 47:1156-1168. [PMID: 35220413 PMCID: PMC9018766 DOI: 10.1038/s41386-022-01292-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/10/2022] [Accepted: 02/02/2022] [Indexed: 02/02/2023]
Abstract
Impairments in identifying and responding to the emotions of others manifest in a variety of psychopathologies. Therefore, elaborating the neurobiological mechanisms that underpin social responses to social emotions, or social affective behavior, is a translationally important goal. The insular cortex is consistently implicated in stress-related social and anxiety disorders, which are associated with diminished ability to make and use inferences about the emotions of others to guide behavior. We investigated how corticotropin-releasing factor (CRF), a neuromodulator evoked upon exposure to stressed conspecifics, influenced the insula. We hypothesized that social affective behavior requires CRF signaling in the insular cortex in order to detect stress in social interactions. In acute slices from male and female rats, CRF depolarized insular pyramidal neurons. In males, but not females, CRF suppressed presynaptic GABAergic inhibition leading to greater excitatory synaptic efficacy in a CRF receptor 1 (CRF1)- and cannabinoid receptor 1 (CB1)-dependent fashion. In males only, insular CRF increased social investigation, and CRF1 and CB1 antagonists interfered with social interactions with stressed conspecifics. To investigate the molecular and cellular basis for the effect of CRF we examined insular CRF1 and CB1 mRNAs and found greater total insula CRF1 mRNA in females but greater CRF1 and CB1 mRNA colocalization in male insular cortex glutamatergic neurons that suggest complex, sex-specific organization of CRF and endocannabinoid systems. Together these results reveal a new mechanism by which stress and affect contribute to social affective behavior.
Collapse
Affiliation(s)
- Nathaniel S Rieger
- Department of Psychology & Neuroscience, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA
| | - Juan A Varela
- Department of Psychology & Neuroscience, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA
| | - Alexandra J Ng
- Department of Psychology & Neuroscience, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA
| | - Lauren Granata
- Psychology Department, Northeastern University, 360 Huntington Avenue, 115 Richards Hall, Boston, MA, 02115, USA
| | - Anthony Djerdjaj
- Department of Psychology & Neuroscience, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA
| | - Heather C Brenhouse
- Psychology Department, Northeastern University, 360 Huntington Avenue, 115 Richards Hall, Boston, MA, 02115, USA
| | - John P Christianson
- Department of Psychology & Neuroscience, Boston College, 140 Commonwealth Ave, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
7
|
Shimoura CG, Andrade MA, Toney GM. Central AT1 receptor signaling by circulating angiotensin II is permissive to acute intermittent hypoxia-induced sympathetic neuroplasticity. J Appl Physiol (1985) 2020; 128:1329-1337. [PMID: 32240022 DOI: 10.1152/japplphysiol.00094.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Acute intermittent hypoxia (AIH) triggers sympathetic long-term facilitation (sLTF), a progressive increase in sympathetic nerve activity (SNA) linked to central AT1 receptor (AT1R) activation by circulating angiotensin II (ANG II). Here, we investigated AIH activation of the peripheral renin-angiotensin system (RAS) and the extent to which the magnitude of RAS activation predicts the magnitude of AIH-induced sLTF. In anesthetized male Sprague-Dawley rats, plasma renin activity (PRA) increased in a linear fashion in response to 5 (P = 0.0342) and 10 (P < 0.0001) cycles of AIH, with PRA remaining at the 10th cycle level 1 h later, a period over which SNA progressively increased. On average, SNA ramping began at the AIH cycle 4.6 ± 0.9 (n = 12) and was similar in magnitude 1 h later whether AIH consisted of 5 or 10 cycles (n = 6/group). Necessity of central AT1R in post-AIH sLTF was affirmed by intracerebroventricular (icv) losartan (40 nmol, 2 µL; n = 5), which strongly attenuated both splanchnic (P = 0.0469) and renal (P = 0.0018) sLTF compared with vehicle [artificial cerebrospinal fluid (aCSF), 2 µL; n = 5]. Bilateral nephrectomy largely prevented sLTF, affirming the necessity of peripheral RAS activation. Sufficiency of central ANG II signaling was assessed in nephrectomized rats. Whereas ICV ANG II (0.5 ng/0.5 µL, 30 min) in nephrectomized rats exposed to sham AIH (n = 4) failed to cause SNA ramping, it rescued sLTF in nephrectomized rats exposed to five cycles of AIH [splanchnic SNA (SSNA), P = 0.0227; renal SNA (RSNA), P = 0.0390; n = 5]. Findings indicate that AIH causes progressive peripheral RAS activation, which stimulates an apparent threshold level of central AT1R signaling that plays a permissive role in triggering sLTF.NEW & NOTEWORTHY Acute intermittent hypoxia (AIH) triggers sympathetic long-term facilitation (sLTF) that relies on peripheral renin-angiotensin system (RAS) activation. Here, increasing AIH cycles from 5 to 10 proportionally increased RAS activity, but not the magnitude of post-AIH sLTF. Brain angiotensin II (ANG II) receptor blockade and nephrectomy each largely prevented sLTF, whereas central ANG II rescued it following nephrectomy. Peripheral RAS activation by AIH induces time-dependent neuroplasticity at an apparent central ANG II signaling threshold, triggering a stereotyped sLTF response.
Collapse
Affiliation(s)
- Caroline G Shimoura
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Mary Ann Andrade
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Glenn M Toney
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas.,Center for Biomedical Neuroscience, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
8
|
Repeated Exposure to Multiple Concurrent Stresses Induce Circuit Specific Loss of Inputs to the Posterior Parietal Cortex. J Neurosci 2020; 40:1849-1861. [PMID: 31949108 DOI: 10.1523/jneurosci.1838-19.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 11/21/2022] Open
Abstract
Severe loss of excitatory synapses in key brain regions is thought to be one of the major mechanisms underlying stress-induced cognitive impairment. To date, however, the identity of the affected circuits remains elusive. Here we examined the effect of exposure to repeated multiple concurrent stressors (RMS) on the connectivity of the posterior parietal cortex (PPC) in adolescent male mice. We found that RMS led to layer-specific elimination of excitatory synapses with the most pronounced loss observed in deeper cortical layers. Quantitative analysis of cortical projections to the PPC revealed a significant loss of sensory and retrosplenial inputs to the PPC while contralateral and frontal projections were preserved. These results were confirmed by decreased synaptic strength from sensory, but not from contralateral, projections in stress-exposed animals. Functionally, RMS disrupted visuospatial working memory performance, implicating disrupted higher-order visual processing. These effects were not observed in mice subjected to restraint-only stress for an identical period of time. The PPC is considered to be a cortical hub for multisensory integration, working memory, and perceptual decision-making. Our data suggest that sensory information streams targeting the PPC may be impacted by recurring stress, likely contributing to stress-induced cognitive impairment.SIGNIFICANCE STATEMENT Repeated exposure to stress profoundly impairs cognitive functions like memory, attention, or decision-making. There is emerging evidence that stress not only impacts high-order regions of the brain, but may affect earlier stages of cognitive processing. Our work focuses on the posterior parietal cortex, a brain region supporting short-term memory, multisensory integration, and decision-making. We show evidence that repeated stress specifically damages sensory inputs to this region. This disruption of synaptic connectivity is linked to working memory impairment and is specific to repeated exposure to multiple stressors. Altogether, our data provide a potential alternative explanation to ailments previously attributed to downstream, cognitive brain structures.
Collapse
|
9
|
Dedic N, Kühne C, Gomes KS, Hartmann J, Ressler KJ, Schmidt MV, Deussing JM. Deletion of CRH From GABAergic Forebrain Neurons Promotes Stress Resilience and Dampens Stress-Induced Changes in Neuronal Activity. Front Neurosci 2019; 13:986. [PMID: 31619956 PMCID: PMC6763571 DOI: 10.3389/fnins.2019.00986] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of the corticotropin-releasing hormone (CRH) system has been implicated in stress-related psychopathologies such as depression and anxiety. Although most studies have linked CRH/CRH receptor 1 signaling to aversive, stress-like behavior, recent work has revealed a crucial role for distinct CRH circuits in maintaining positive emotional valence and appetitive responses under baseline conditions. Here we addressed whether deletion of CRH, specifically from GABAergic forebrain neurons (Crh CKO-GABA mice) differentially affects general behavior under baseline and chronic stress conditions. Expression mapping in Crh CK O-GABA mice revealed absence of Crh in GABAergic neurons of the cortex and limbic regions including the hippocampus, central nucleus of the amygdala and the bed nucleus of the stria terminals, but not in the paraventricular nucleus of hypothalamus. Consequently, conditional CRH knockout animals exhibited no alterations in circadian and stress-induced corticosterone release compared to controls. Under baseline conditions, absence of Crh from forebrain GABAergic neurons resulted in social interaction deficits but had no effect on other behavioral measures including locomotion, anxiety, immobility in the forced swim test, acoustic startle response and fear conditioning. Interestingly, following exposure to chronic social defeat stress, Crh CKO-GABA mice displayed a resilient phenotype, which was accompanied by a dampened, stress-induced expression of immediate early genes c-fos and zif268 in several brain regions. Collectively our data reveals the requirement of GABAergic CRH circuits in maintaining appropriate social behavior in naïve animals and further supports the ability of CRH to promote divergent behavioral states under baseline and severe stress conditions.
Collapse
Affiliation(s)
- Nina Dedic
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, United States
| | - Claudia Kühne
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Karina S Gomes
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Laboratory of Neuropsychopharmacology, Paulista State University, Araraquara, Brazil
| | - Jakob Hartmann
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, United States.,Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, United States
| | - Mathias V Schmidt
- Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jan M Deussing
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
10
|
Gururajan A, van de Wouw M, Boehme M, Becker T, O'Connor R, Bastiaanssen TFS, Moloney GM, Lyte JM, Ventura Silva AP, Merckx B, Dinan TG, Cryan JF. Resilience to chronic stress is associated with specific neurobiological, neuroendocrine and immune responses. Brain Behav Immun 2019; 80:583-594. [PMID: 31059807 DOI: 10.1016/j.bbi.2019.05.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 04/23/2019] [Accepted: 05/02/2019] [Indexed: 12/12/2022] Open
Abstract
Research into the molecular basis of stress resilience is a novel strategy to identify potential therapeutic strategies to treat stress-induced psychopathologies such as anxiety and depression. Stress resilience is a phenomenon which is not solely driven by effects within the central nervous system (CNS) but involves multiple systems, central and peripheral, which interact with and influence each other. Accordingly, we used the chronic social defeat stress paradigm and investigated specific CNS, endocrine and immune responses to identify signatures of stress-resilience and stress susceptibility in mice. Our results showed that mice behaviourally susceptible to stress (indexed by a reduction in social interaction behaviour) had higher plasma corticosterone levels and adrenal hypertrophy. An increase in inflammatory circulating monocytes was another hallmark of stress susceptibility. Furthermore, prefrontal cortex mRNA expression of corticotrophin-releasing factor (Crf) was increased in susceptible mice relative to resilient mice. We also report differences in hippocampal synaptic plasticity between resilient and susceptible mice. Ongoing studies will interpret the functional relevance of these signatures which could potentially inform the development of novel psychotherapeutic strategies.
Collapse
Affiliation(s)
- Anand Gururajan
- Department of Anatomy & Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| | - Marcel van de Wouw
- Department of Anatomy & Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Marcus Boehme
- Department of Anatomy & Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Thorsten Becker
- Department of Anatomy & Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Rory O'Connor
- Department of Anatomy & Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Thomaz F S Bastiaanssen
- Department of Anatomy & Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Gerard M Moloney
- Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Joshua M Lyte
- APC Microbiome Ireland, University College Cork, Ireland
| | | | - Barbara Merckx
- Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Psychiatry & Neurobehavioural Science, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy & Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|
11
|
Ahmadi F, Zendehdel M, Babapour V, Panahi N. CRF1/CRF2 and MC3/MC4 Receptors Affect Glutamate- Induced Food Intake in Neonatal Meat-Type Chicken. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2019. [DOI: 10.1590/1806-9061-2018-0821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
12
|
Maruyama NO, Mitchell NC, Truong TT, Toney GM. Activation of the hypothalamic paraventricular nucleus by acute intermittent hypoxia: Implications for sympathetic long-term facilitation neuroplasticity. Exp Neurol 2018; 314:1-8. [PMID: 30605624 DOI: 10.1016/j.expneurol.2018.12.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/03/2018] [Accepted: 12/30/2018] [Indexed: 02/07/2023]
Abstract
Exposure to acute intermittent hypoxia (AIH) induces a progressive increase of sympathetic nerve activity (SNA) that reflects a form of neuroplasticity known as sympathetic long-term facilitation (sLTF). Our recent findings indicate that activity of neurons in the hypothalamic paraventricular nucleus (PVN) contributes to AIH-induced sLTF, but neither the intra-PVN distribution nor the neurochemical identity of AIH responsive neurons has been determined. Here, awake rats were exposed to 10 cycles of AIH and c-Fos immunohistochemistry was performed to identify transcriptionally activated neurons in rostral, middle and caudal planes of the PVN. Effects of graded intensities of AIH were investigated in separate groups of rats (n = 6/group) in which inspired oxygen (O2) was reduced every 6 min from 21% to nadirs of 10%, 8% or 6%. All intensities of AIH failed to increase c-Fos counts in the caudally located lateral parvocellular region of the PVN. c-Fos counts increased in the dorsal parvocellular and central magnocellular regions, but significance was achieved only with AIH to 6% O2 (P < 0.002). By contrast, graded intensities of AIH induced graded c-Fos activation in the stress-related medial parvocellular (MP) region. Focusing on AIH exposure to 8% O2, experiments next investigated the stress-regulatory neuropeptide content of AIH-activated MP neurons. Tissue sections immunostained for corticotropin-releasing hormone (CRH) or arginine vasopressin (AVP) revealed a significantly greater number of neurons stained for CRH than AVP (P < 0.0001), though AIH induced expression of c-Fos in a similar fraction (~14%) of each neurochemical class. Amongst AIH-activated MP neurons, ~30% stained for CRH while only ~2% stained for AVP. Most AIH-activated CRH neurons (~82%) were distributed in the rostral one-half of the PVN. Results indicate that AIH recruits CRH, but not AVP, neurons in rostral to middle levels of the MP region of PVN, and raise the possibility that these CRH neurons may be a substrate for AIH-induced sLTF neuroplasticity.
Collapse
Affiliation(s)
- Nadia Oliveira Maruyama
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Nathan C Mitchell
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Tamara T Truong
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Glenn M Toney
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
13
|
Dedic N, Chen A, Deussing JM. The CRF Family of Neuropeptides and their Receptors - Mediators of the Central Stress Response. Curr Mol Pharmacol 2018; 11:4-31. [PMID: 28260504 PMCID: PMC5930453 DOI: 10.2174/1874467210666170302104053] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 11/26/2015] [Accepted: 08/03/2016] [Indexed: 12/12/2022]
Abstract
Background: Dysregulated stress neurocircuits, caused by genetic and/or environmental changes, underlie the development of many neuropsychiatric disorders. Corticotropin-releasing factor (CRF) is the major physiological activator of the hypothalamic-pituitary-adrenal (HPA) axis and conse-quently a primary regulator of the mammalian stress response. Together with its three family members, urocortins (UCNs) 1, 2, and 3, CRF integrates the neuroendocrine, autonomic, metabolic and behavioral responses to stress by activating its cognate receptors CRFR1 and CRFR2. Objective: Here we review the past and current state of the CRF/CRFR field, ranging from pharmacologi-cal studies to genetic mouse models and virus-mediated manipulations. Results: Although it is well established that CRF/CRFR1 signaling mediates aversive responses, includ-ing anxiety and depression-like behaviors, a number of recent studies have challenged this viewpoint by revealing anxiolytic and appetitive properties of specific CRF/CRFR1 circuits. In contrast, the UCN/CRFR2 system is less well understood and may possibly also exert divergent functions on physiol-ogy and behavior depending on the brain region, underlying circuit, and/or experienced stress conditions. Conclusion: A plethora of available genetic tools, including conventional and conditional mouse mutants targeting CRF system components, has greatly advanced our understanding about the endogenous mecha-nisms underlying HPA system regulation and CRF/UCN-related neuronal circuits involved in stress-related behaviors. Yet, the detailed pathways and molecular mechanisms by which the CRF/UCN-system translates negative or positive stimuli into the final, integrated biological response are not completely un-derstood. The utilization of future complementary methodologies, such as cell-type specific Cre-driver lines, viral and optogenetic tools will help to further dissect the function of genetically defined CRF/UCN neurocircuits in the context of adaptive and maladaptive stress responses.
Collapse
Affiliation(s)
- Nina Dedic
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstr, 2-10, 80804 Munich. Germany
| |
Collapse
|
14
|
Hyper-diversity of CRH interneurons in mouse hippocampus. Brain Struct Funct 2018; 224:583-598. [PMID: 30456559 DOI: 10.1007/s00429-018-1793-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/09/2018] [Indexed: 12/20/2022]
Abstract
Hippocampal inhibitory interneurons comprise an anatomically, neurochemically and electrophysiologically diverse population of cells that are essential for the generation of the oscillatory activity underlying hippocampal spatial and episodic memory processes. Here, we aimed to characterize a population of interneurons that express the stress-related neuropeptide corticotropin-releasing hormone (CRH) within existing interneuronal categories through the use of combined electrophysiological and immunocytochemical approaches. Focusing on CA1 strata pyramidale and radiatum of mouse hippocampus, CRH interneurons were found to exhibit a heterogeneous neurochemical phenotype with parvalbumin, cholecystokinin and calretinin co-expression observed to varying degrees. In contrast, CRH and somatostatin were never co-expressed. Electrophysiological categorization identified heterogeneous firing pattern of CRH neurons, with two distinct subtypes within stratum pyramidale and stratum radiatum. Together, these findings indicate that CRH-expressing interneurons do not segregate into any single distinct subtype of interneuron using conventional criteria. Rather our findings suggest that CRH is likely co-expressed in subpopulations of previously described hippocampal interneurons. In addition, the observed heterogeneity suggests that distinct CRH interneuron subtypes may have specific functional roles in the both physiological and pathophysiological hippocampal processes.
Collapse
|
15
|
Deussing JM, Chen A. The Corticotropin-Releasing Factor Family: Physiology of the Stress Response. Physiol Rev 2018; 98:2225-2286. [DOI: 10.1152/physrev.00042.2017] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The physiological stress response is responsible for the maintenance of homeostasis in the presence of real or perceived challenges. In this function, the brain activates adaptive responses that involve numerous neural circuits and effector molecules to adapt to the current and future demands. A maladaptive stress response has been linked to the etiology of a variety of disorders, such as anxiety and mood disorders, eating disorders, and the metabolic syndrome. The neuropeptide corticotropin-releasing factor (CRF) and its relatives, the urocortins 1–3, in concert with their receptors (CRFR1, CRFR2), have emerged as central components of the physiological stress response. This central peptidergic system impinges on a broad spectrum of physiological processes that are the basis for successful adaptation and concomitantly integrate autonomic, neuroendocrine, and behavioral stress responses. This review focuses on the physiology of CRF-related peptides and their cognate receptors with the aim of providing a comprehensive up-to-date overview of the field. We describe the major molecular features covering aspects of gene expression and regulation, structural properties, and molecular interactions, as well as mechanisms of signal transduction and their surveillance. In addition, we discuss the large body of published experimental studies focusing on state-of-the-art genetic approaches with high temporal and spatial precision, which collectively aimed to dissect the contribution of CRF-related ligands and receptors to different levels of the stress response. We discuss the controversies in the field and unravel knowledge gaps that might pave the way for future research directions and open up novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jan M. Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; and Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany; and Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
16
|
Raftogianni A, Roth LC, García-González D, Bus T, Kühne C, Monyer H, Spergel DJ, Deussing JM, Grinevich V. Deciphering the Contributions of CRH Receptors in the Brain and Pituitary to Stress-Induced Inhibition of the Reproductive Axis. Front Mol Neurosci 2018; 11:305. [PMID: 30214395 PMCID: PMC6125327 DOI: 10.3389/fnmol.2018.00305] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/09/2018] [Indexed: 01/13/2023] Open
Abstract
Based on pharmacological studies, corticotropin-releasing hormone (CRH) and its receptors play a leading role in the inhibition of the hypothalamic–pituitary–gonadal (HPG) axis during acute stress. To further study the effects of CRH receptor signaling on the HPG axis, we generated and/or employed male mice lacking CRH receptor type 1 (CRHR1) or type 2 (CRHR2) in gonadotropin-releasing hormone neurons, GABAergic neurons, or in all central neurons and glia. The deletion of CRHRs revealed a preserved decrease of plasma luteinizing hormone (LH) in response to either psychophysical or immunological stress. However, under basal conditions, central infusion of CRH into mice lacking CRHR1 in all central neurons and glia, or application of CRH to pituitary cultures from mice lacking CRHR2, failed to suppress LH release, unlike in controls. Our results, taken together with those of the earlier pharmacological studies, suggest that inhibition of the male HPG axis during acute stress is mediated by other factors along with CRH, and that CRH suppresses the HPG axis at the central and pituitary levels via CRHR1 and CRHR2, respectively.
Collapse
Affiliation(s)
- Androniki Raftogianni
- Schaller Group on Neuropeptides, German Cancer Research Center, Heidelberg - Central Institute of Mental Health, Mannheim, Germany.,Department of Molecular Neurobiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Lena C Roth
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Diego García-González
- Department of Clinical Neurobiology, Medical Faculty of Heidelberg, University of Heidelberg - German Cancer Research Center, Heidelberg, Germany
| | - Thorsten Bus
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research, Heidelberg, Germany.,Max Planck Research Group at the Institute for Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Claudia Kühne
- Molecular Neurogenetics Research Group, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology, Medical Faculty of Heidelberg, University of Heidelberg - German Cancer Research Center, Heidelberg, Germany
| | - Daniel J Spergel
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
| | - Jan M Deussing
- Molecular Neurogenetics Research Group, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Valery Grinevich
- Schaller Group on Neuropeptides, German Cancer Research Center, Heidelberg - Central Institute of Mental Health, Mannheim, Germany.,Department of Molecular Neurobiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| |
Collapse
|
17
|
Persistent Stress-Induced Neuroplastic Changes in the Locus Coeruleus/Norepinephrine System. Neural Plast 2018; 2018:1892570. [PMID: 30008741 PMCID: PMC6020552 DOI: 10.1155/2018/1892570] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/09/2018] [Accepted: 05/27/2018] [Indexed: 11/25/2022] Open
Abstract
Neural plasticity plays a critical role in mediating short- and long-term brain responses to environmental stimuli. A major effector of plasticity throughout many regions of the brain is stress. Activation of the locus coeruleus (LC) is a critical step in mediating the neuroendocrine and behavioral limbs of the stress response. During stressor exposure, activation of the hypothalamic-pituitary-adrenal axis promotes release of corticotropin-releasing factor in LC, where its signaling promotes a number of physiological and cellular changes. While the acute effects of stress on LC physiology have been described, its long-term effects are less clear. This review will describe how stress changes LC neuronal physiology, function, and morphology from a genetic, cellular, and neuronal circuitry/transmission perspective. Specifically, we describe morphological changes of LC neurons in response to stressful stimuli and signal transduction pathways underlying them. Also, we will review changes in excitatory glutamatergic synaptic transmission in LC neurons and possible stress-induced modifications of AMPA receptors. This review will also address stress-related behavioral adaptations and specific noradrenergic receptors responsible for them. Finally, we summarize the results of several human studies which suggest a link between stress, altered LC function, and pathogenesis of posttraumatic stress disorder.
Collapse
|
18
|
Gunn BG, Cox CD, Chen Y, Frotscher M, Gall CM, Baram TZ, Lynch G. The Endogenous Stress Hormone CRH Modulates Excitatory Transmission and Network Physiology in Hippocampus. Cereb Cortex 2018; 27:4182-4198. [PMID: 28460009 PMCID: PMC6248689 DOI: 10.1093/cercor/bhx103] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Indexed: 01/06/2023] Open
Abstract
Memory is strongly influenced by stress but underlying mechanisms are unknown. Here, we
used electrophysiology, neuroanatomy, and network simulations to probe the role of the
endogenous, stress-related neuropeptide corticotropin-releasing hormone (CRH) in
modulating hippocampal function. We focused on neuronal excitability and the incidence of
sharp waves (SPWs), a form of intrinsic network activity associated with memory
consolidation. Specifically, we blocked endogenous CRH using 2 chemically distinct
antagonists of the principal hippocampal CRH receptor, CRHR1. The antagonists caused a
modest reduction of spontaneous excitatory transmission onto CA3 pyramidal cells,
mediated, in part by effects on IAHP. This was accompanied by a decrease in the
incidence but not amplitude of SPWs, indicating that the synaptic actions of CRH are
sufficient to alter the output of a complex hippocampal network. A biophysical model of
CA3 described how local actions of CRH produce macroscopic consequences including the
observed changes in SPWs. Collectively, the results provide a first demonstration of the
manner in which subtle synaptic effects of an endogenously released neuropeptide influence
hippocampal network level operations and, in the case of CRH, may contribute to the
effects of acute stress on memory.
Collapse
Affiliation(s)
- B. G. Gunn
- Department of Pediatrics, University of
California-Irvine, Irvine, CA, USA
| | - C. D. Cox
- Department of Anatomy/Neurobiology, University of
California-Irvine, Irvine, CA, USA
| | - Y. Chen
- Department of Pediatrics, University of
California-Irvine, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of
California-Irvine, Irvine, CA, USA
| | - M. Frotscher
- ZMNH, Institute for Structural
Neurobiology, D-20251 Hamburg,
Germany
| | - C. M. Gall
- Department of Anatomy/Neurobiology, University of
California-Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of
California-Irvine, Irvine, CA, USA
| | - T. Z. Baram
- Department of Pediatrics, University of
California-Irvine, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of
California-Irvine, Irvine, CA, USA
- Department of Neurology, University of
California-Irvine, Irvine, CA, USA
- Address correspondence to Prof. T. Z. Baram, Departments of Pediatrics;
Anatomy & Neurobiology; Neurology, University of California-Irvine, Medical Sciences
I, ZOT: 4475, Irvine, CA 92697-4475, USA.
| | - G. Lynch
- Department of Anatomy/Neurobiology, University of
California-Irvine, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University
of California-Irvine, Irvine, CA, USA
| |
Collapse
|
19
|
Dedic N, Pöhlmann ML, Richter JS, Mehta D, Czamara D, Metzger MW, Dine J, Bedenk BT, Hartmann J, Wagner KV, Jurik A, Almli LM, Lori A, Moosmang S, Hofmann F, Wotjak CT, Rammes G, Eder M, Chen A, Ressler KJ, Wurst W, Schmidt MV, Binder EB, Deussing JM. Cross-disorder risk gene CACNA1C differentially modulates susceptibility to psychiatric disorders during development and adulthood. Mol Psychiatry 2018; 23:533-543. [PMID: 28696432 PMCID: PMC5822460 DOI: 10.1038/mp.2017.133] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/25/2017] [Accepted: 05/04/2017] [Indexed: 12/17/2022]
Abstract
Single-nucleotide polymorphisms (SNPs) in CACNA1C, the α1C subunit of the voltage-gated L-type calcium channel Cav1.2, rank among the most consistent and replicable genetics findings in psychiatry and have been associated with schizophrenia, bipolar disorder and major depression. However, genetic variants of complex diseases often only confer a marginal increase in disease risk, which is additionally influenced by the environment. Here we show that embryonic deletion of Cacna1c in forebrain glutamatergic neurons promotes the manifestation of endophenotypes related to psychiatric disorders including cognitive decline, impaired synaptic plasticity, reduced sociability, hyperactivity and increased anxiety. Additional analyses revealed that depletion of Cacna1c during embryonic development also increases the susceptibility to chronic stress, which suggest that Cav1.2 interacts with the environment to shape disease vulnerability. Remarkably, this was not observed when Cacna1c was deleted in glutamatergic neurons during adulthood, where the later deletion even improved cognitive flexibility, strengthened synaptic plasticity and induced stress resilience. In a parallel gene × environment design in humans, we additionally demonstrate that SNPs in CACNA1C significantly interact with adverse life events to alter the risk to develop symptoms of psychiatric disorders. Overall, our results further validate Cacna1c as a cross-disorder risk gene in mice and humans, and additionally suggest a differential role for Cav1.2 during development and adulthood in shaping cognition, sociability, emotional behavior and stress susceptibility. This may prompt the consideration for pharmacological manipulation of Cav1.2 in neuropsychiatric disorders with developmental and/or stress-related origins.
Collapse
Affiliation(s)
- N Dedic
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - M L Pöhlmann
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - J S Richter
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - D Mehta
- Queensland Brain Institute, University of Queensland, St. Lucia, QLD, Australia
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - D Czamara
- Queensland Brain Institute, University of Queensland, St. Lucia, QLD, Australia
| | - M W Metzger
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - J Dine
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - B T Bedenk
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - J Hartmann
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, USA
| | - K V Wagner
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - A Jurik
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
| | - L M Almli
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - A Lori
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - S Moosmang
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
| | - F Hofmann
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
| | - C T Wotjak
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - G Rammes
- Clinic of Anaesthesiology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - M Eder
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - A Chen
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- The Ruhman Family Laboratory for Research on the Neurobiology of Stress, Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - K J Ressler
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - W Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - M V Schmidt
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - E B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - J M Deussing
- Molecular Neurogenetics, Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
20
|
Hrvatin S, Hochbaum DR, Nagy MA, Cicconet M, Robertson K, Cheadle L, Zilionis R, Ratner A, Borges-Monroy R, Klein AM, Sabatini BL, Greenberg ME. Single-cell analysis of experience-dependent transcriptomic states in the mouse visual cortex. Nat Neurosci 2018; 21:120-129. [PMID: 29230054 PMCID: PMC5742025 DOI: 10.1038/s41593-017-0029-5] [Citation(s) in RCA: 314] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 10/17/2017] [Indexed: 12/17/2022]
Abstract
Activity-dependent transcriptional responses shape cortical function. However, a comprehensive understanding of the diversity of these responses across the full range of cortical cell types, and how these changes contribute to neuronal plasticity and disease, is lacking. To investigate the breadth of transcriptional changes that occur across cell types in the mouse visual cortex after exposure to light, we applied high-throughput single-cell RNA sequencing. We identified significant and divergent transcriptional responses to stimulation in each of the 30 cell types characterized, thus revealing 611 stimulus-responsive genes. Excitatory pyramidal neurons exhibited inter- and intralaminar heterogeneity in the induction of stimulus-responsive genes. Non-neuronal cells showed clear transcriptional responses that may regulate experience-dependent changes in neurovascular coupling and myelination. Together, these results reveal the dynamic landscape of the stimulus-dependent transcriptional changes occurring across cell types in the visual cortex; these changes are probably critical for cortical function and may be sites of deregulation in developmental brain disorders.
Collapse
Affiliation(s)
- Sinisa Hrvatin
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Daniel R Hochbaum
- Society of Fellows, Harvard University, Cambridge, MA, USA
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - M Aurel Nagy
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Marcelo Cicconet
- Image and Data Analysis Core, Harvard Medical School, Boston, MA, USA
| | - Keiramarie Robertson
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Lucas Cheadle
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Rapolas Zilionis
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Vilnius University Institute of Biotechnology, Vilnius, Lithuania
| | - Alex Ratner
- ICCB-L Single Cell Core, Harvard Medical School, Boston, MA, USA
| | - Rebeca Borges-Monroy
- Program for Bioinformatics and Integrative Genomics, Graduate School of Arts and Science, Division of Medical Sciences, Harvard University, Cambridge, MA, USA
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
21
|
Phumsatitpong C, Moenter SM. Estradiol-Dependent Stimulation and Suppression of Gonadotropin-Releasing Hormone Neuron Firing Activity by Corticotropin-Releasing Hormone in Female Mice. Endocrinology 2018; 159:414-425. [PMID: 29069304 PMCID: PMC5761586 DOI: 10.1210/en.2017-00747] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/17/2017] [Indexed: 11/19/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are the final central regulators of reproduction, integrating various inputs that modulate fertility. Stress typically inhibits reproduction but can be stimulatory; stress effects can also be modulated by steroid milieu. Corticotropin-releasing hormone (CRH) released during the stress response may suppress reproduction independent of downstream glucocorticoids. We hypothesized CRH suppresses fertility by decreasing GnRH neuron firing activity. To test this, mice were ovariectomized (OVX) and either implanted with an estradiol capsule (OVX+E) or not treated further to examine the influence of estradiol on GnRH neuron response to CRH. Targeted extracellular recordings were used to record firing activity from green fluorescent protein-identified GnRH neurons in brain slices before and during CRH treatment; recordings were done in the afternoon when estradiol has a positive feedback effect to increase GnRH neuron firing. In OVX mice, CRH did not affect the firing rate of GnRH neurons. In contrast, CRH exhibited dose-dependent stimulatory (30 nM) or inhibitory (100 nM) effects on GnRH neuron firing activity in OVX+E mice; both effects were reversible. The dose-dependent effects of CRH appear to result from activation of different receptor populations; a CRH receptor type-1 agonist increased firing activity in GnRH neurons, whereas a CRH receptor type-2 agonist decreased firing activity. CRH and specific agonists also differentially regulated short-term burst frequency and burst properties, including burst duration, spikes/burst, and/or intraburst interval. These results indicate that CRH alters GnRH neuron activity and that estradiol is required for CRH to exert both stimulatory and inhibitory effects on GnRH neurons.
Collapse
Affiliation(s)
- Chayarndorn Phumsatitpong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Suzanne M. Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
22
|
Gunn BG, Baram TZ. Stress and Seizures: Space, Time and Hippocampal Circuits. Trends Neurosci 2017; 40:667-679. [PMID: 28916130 DOI: 10.1016/j.tins.2017.08.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/11/2017] [Accepted: 08/23/2017] [Indexed: 10/18/2022]
Abstract
Stress is a major trigger of seizures in people with epilepsy. Exposure to stress results in the release of several stress mediators throughout the brain, including the hippocampus, a region sensitive to stress and prone to seizures. Stress mediators interact with their respective receptors to produce distinct effects on the excitability of hippocampal neurons and networks. Crucially, these stress mediators and their actions exhibit unique spatiotemporal profiles, generating a complex combinatorial output with time- and space-dependent effects on hippocampal network excitability and seizure generation.
Collapse
Affiliation(s)
- B G Gunn
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - T Z Baram
- Department of Pediatrics, University of California, Irvine, CA, USA; Department of Anatomy & Neurobiology, University of California, Irvine, CA, USA; Department of Neurology, University of California, Irvine, CA, USA.
| |
Collapse
|
23
|
Inda C, Armando NG, Dos Santos Claro PA, Silberstein S. Endocrinology and the brain: corticotropin-releasing hormone signaling. Endocr Connect 2017; 6:R99-R120. [PMID: 28710078 PMCID: PMC5551434 DOI: 10.1530/ec-17-0111] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/14/2017] [Indexed: 01/01/2023]
Abstract
Corticotropin-releasing hormone (CRH) is a key player of basal and stress-activated responses in the hypothalamic-pituitary-adrenal axis (HPA) and in extrahypothalamic circuits, where it functions as a neuromodulator to orchestrate humoral and behavioral adaptive responses to stress. This review describes molecular components and cellular mechanisms involved in CRH signaling downstream of its G protein-coupled receptors (GPCRs) CRHR1 and CRHR2 and summarizes recent findings that challenge the classical view of GPCR signaling and impact on our understanding of CRHRs function. Special emphasis is placed on recent studies of CRH signaling that revealed new mechanistic aspects of cAMP generation and ERK1/2 activation in physiologically relevant contexts of the neurohormone action. In addition, we present an overview of the pathophysiological role of the CRH system, which highlights the need for a precise definition of CRHRs signaling at molecular level to identify novel targets for pharmacological intervention in neuroendocrine tissues and specific brain areas involved in CRH-related disorders.
Collapse
Affiliation(s)
- Carolina Inda
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
- DFBMCFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia G Armando
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - Paula A Dos Santos Claro
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - Susana Silberstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
- DFBMCFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
24
|
Abulseoud OA, Ho MC, Choi DS, Stanojević A, Čupić Ž, Kolar-Anić L, Vukojević V. Corticosterone oscillations during mania induction in the lateral hypothalamic kindled rat-Experimental observations and mathematical modeling. PLoS One 2017; 12:e0177551. [PMID: 28542167 PMCID: PMC5436765 DOI: 10.1371/journal.pone.0177551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 04/28/2017] [Indexed: 01/03/2023] Open
Abstract
Changes in the hypothalamic-pituitary-adrenal (HPA) axis activity constitute a key component of bipolar mania, but the extent and nature of these alterations are not fully understood. We use here the lateral hypothalamic-kindled (LHK) rat model to deliberately induce an acute manic-like episode and measure serum corticosterone concentrations to assess changes in HPA axis activity. A mathematical model is developed to succinctly describe the entwined biochemical transformations that underlay the HPA axis and emulate by numerical simulations the considerable increase in serum corticosterone concentration induced by LHK. Synergistic combination of the LHK rat model and dynamical systems theory allows us to quantitatively characterize changes in HPA axis activity under controlled induction of acute manic-like states and provides a framework to study in silico how the dynamic integration of neurochemical transformations underlying the HPA axis is disrupted in these states.
Collapse
Affiliation(s)
- Osama A. Abulseoud
- Department of Psychiatry and Psychology; Mayo Clinic, Rochester, Minnesota, United States of America
- Chemistry and Drug Metabolism, IRP, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Man Choi Ho
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Doo-Sup Choi
- Department of Psychiatry and Psychology; Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ana Stanojević
- University of Belgrade, Faculty of Physical Chemistry, Studentski trg 12–16, Belgrade, Serbia
| | - Željko Čupić
- University of Belgrade, Institute of Chemistry, Technology and Metallurgy, Department of Catalysis and Chemical Engineering, Njegoševa 12, Belgrade, Serbia
| | - Ljiljana Kolar-Anić
- University of Belgrade, Faculty of Physical Chemistry, Studentski trg 12–16, Belgrade, Serbia
- University of Belgrade, Institute of Chemistry, Technology and Metallurgy, Department of Catalysis and Chemical Engineering, Njegoševa 12, Belgrade, Serbia
| | - Vladana Vukojević
- Karolinska Institute, Department of Clinical Neuroscience, Center for Molecular Medicine CMM L8:01, Stockholm, Sweden
| |
Collapse
|
25
|
Li WL, Fu C, Xuan A, Shi DP, Gao YJ, Zhang J, Xu JL. Preliminary study of brain glucose metabolism changes in patients with lung cancer of different histological types. Chin Med J (Engl) 2015; 128:301-4. [PMID: 25635423 PMCID: PMC4837858 DOI: 10.4103/0366-6999.150089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Cerebral glucose metabolism changes are always observed in patients suffering from malignant tumors. This preliminary study aimed to investigate the brain glucose metabolism changes in patients with lung cancer of different histological types. METHODS One hundred and twenty patients with primary untreated lung cancer, who visited People's Hospital of Zhengzhou University from February 2012 to July 2013, were divided into three groups based on histological types confirmed by biopsy or surgical pathology, which included adenocarcinoma (52 cases), squamous cell carcinoma (43 cases), and small-cell carcinoma (25 cases). The whole body 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT) of these cases was retrospectively studied. The brain PET data of three groups were analyzed individually using statistical parametric maps (SPM) software, with 50 age-matched and gender-matched healthy controls for comparison. RESULTS The brain resting glucose metabolism in all three lung cancer groups showed regional cerebral metabolic reduction. The hypo-metabolic cerebral regions were mainly distributed at the left superior and middle frontal, bilateral superior and middle temporal and inferior and middle temporal gyrus. Besides, the hypo-metabolic regions were also found in the right inferior parietal lobule and hippocampus in the small-cell carcinoma group. The area of the total hypo-metabolic cerebral regions in the small-cell carcinoma group (total voxel value 3255) was larger than those in the adenocarcinoma group (total voxel value 1217) and squamous cell carcinoma group (total voxel value 1292). CONCLUSIONS The brain resting glucose metabolism in patients with lung cancer shows regional cerebral metabolic reduction and the brain hypo-metabolic changes are related to the histological types of lung cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jun-Ling Xu
- Department of Nuclear Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| |
Collapse
|
26
|
Narla C, Dunn HA, Ferguson SSG, Poulter MO. Suppression of piriform cortex activity in rat by corticotropin-releasing factor 1 and serotonin 2A/C receptors. Front Cell Neurosci 2015; 9:200. [PMID: 26074770 PMCID: PMC4446537 DOI: 10.3389/fncel.2015.00200] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/10/2015] [Indexed: 11/29/2022] Open
Abstract
The piriform cortex (PC) is richly innervated by corticotropin-releasing factor (CRF) and serotonin (5-HT) containing axons arising from central amygdala and Raphe nucleus. CRFR1 and 5-HT2A/2CRs have been shown to interact in manner where CRFR activation subsequently potentiates the activity of 5-HT2A/2CRs. The purpose of this study was to determine how the activation of CRFR1 and/or 5-HT2Rs modulates PC activity at both the circuit and cellular level. Voltage sensitive dye imaging showed that CRF acting through CRFR1 dampened activation of the Layer II of PC and interneurons of endopiriform nucleus. Application of the selective 5-HT2A/CR agonist 2,5-dimethoxy-4-iodoamphetamine (DOI) following CRFR1 activation potentiated this effect. Blocking the interaction between CRFR1 and 5-HT2R with a Tat-CRFR1-CT peptide abolished this potentiation. Application of forskolin did not mimic CRFR1 activity but instead blocked it, while a protein kinase A antagonist had no effect. However, activation and antagonism of protein kinase C (PKC) either mimicked or blocked CRF modulation, respectively. DOI had no effect when applied alone indicating that the prior activation of CRFR1 receptors was critical for DOI to show significant effects similar to CRF. Patch clamp recordings showed that both CRF and DOI reduced the synaptic responsiveness of Layer II pyramidal neurons. CRF had highly variable effects on interneurons within Layer III, both increasing and decreasing their excitability, but DOI had no effect on the excitability of this group of neurons. These data show that CRF and 5-HT, acting through both CRFR1 and 5-HT2A/CRs, reduce the activation of the PC. This modulation may be an important blunting mechanism of stressor behaviors mediated through the olfactory cortex.
Collapse
Affiliation(s)
- Chakravarthi Narla
- Molecular Medicine Research Group, Department of Physiology and Pharmacology, Robarts Research Institute, Faculty of Medicine, Schulich School of Medicine, University of Western Ontario London, ON, Canada
| | - Henry A Dunn
- Molecular Medicine Research Group, Department of Physiology and Pharmacology, Robarts Research Institute, Faculty of Medicine, Schulich School of Medicine, University of Western Ontario London, ON, Canada
| | - Stephen S G Ferguson
- Molecular Medicine Research Group, Department of Physiology and Pharmacology, Robarts Research Institute, Faculty of Medicine, Schulich School of Medicine, University of Western Ontario London, ON, Canada
| | - Michael O Poulter
- Molecular Medicine Research Group, Department of Physiology and Pharmacology, Robarts Research Institute, Faculty of Medicine, Schulich School of Medicine, University of Western Ontario London, ON, Canada
| |
Collapse
|
27
|
Balaraman Y, Lahiri DK, Nurnberger JI. Variants in Ion Channel Genes Link Phenotypic Features of Bipolar Illness to Specific Neurobiological Process Domains. MOLECULAR NEUROPSYCHIATRY 2015; 1:23-35. [PMID: 27602355 DOI: 10.1159/000371886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/05/2015] [Indexed: 11/19/2022]
Abstract
Recent advances in genome-wide association studies are pointing towards a major role for voltage-gated ion channels in neuropsychiatric disorders and, in particular, bipolar disorder (BD). The phenotype of BD is complex, with symptoms during mood episodes and deficits persisting between episodes. We have tried to elucidate the common neurobiological mechanisms associated with ion channel signaling in order to provide a new perspective on the clinical symptoms and possible endophenotypes seen in BD patients. We propose a model in which the multiple variants in genes coding for ion channel proteins would perturb motivational circuits, synaptic plasticity, myelination, hypothalamic-pituitary-adrenal axis function, circadian neuronal rhythms, and energy regulation. These changes in neurobiological mechanisms would manifest in endophenotypes of aberrant reward processing, white matter hyperintensities, deficits in executive function, altered frontolimbic connectivity, increased amygdala activity, increased melatonin suppression, decreased REM latency, and aberrant myo-inositol/ATP shuttling. The endophenotypes result in behaviors of poor impulse control, motivational changes, cognitive deficits, abnormal stress response, sleep disturbances, and energy changes involving different neurobiological process domains. The hypothesis is that these disturbances start with altered neural circuitry during development, following which multiple environmental triggers may disrupt the neuronal excitability balance through an activity-dependent molecular process, resulting in clinical mood episodes.
Collapse
Affiliation(s)
- Yokesh Balaraman
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| | - Debomoy K Lahiri
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| | - John I Nurnberger
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| |
Collapse
|
28
|
Fiorenza A, Lopez-Atalaya JP, Rovira V, Scandaglia M, Geijo-Barrientos E, Barco A. Blocking miRNA Biogenesis in Adult Forebrain Neurons Enhances Seizure Susceptibility, Fear Memory, and Food Intake by Increasing Neuronal Responsiveness. Cereb Cortex 2015; 26:1619-1633. [PMID: 25595182 DOI: 10.1093/cercor/bhu332] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The RNase Dicer is essential for the maturation of most microRNAs, a molecular system that plays an essential role in fine-tuning gene expression. To gain molecular insight into the role of Dicer and the microRNA system in brain function, we conducted 2 complementary RNA-seq screens in the hippocampus of inducible forebrain-restricted Dicer1 mutants aimed at identifying the microRNAs primarily affected by Dicer loss and their targets, respectively. Functional genomics analyses predicted the main biological processes and phenotypes associated with impaired microRNA maturation, including categories related to microRNA biology, signal transduction, seizures, and synaptic transmission and plasticity. Consistent with these predictions, we found that, soon after recombination, Dicer-deficient mice exhibited an exaggerated seizure response, enhanced induction of immediate early genes in response to different stimuli, stronger and more stable fear memory, hyperphagia, and increased excitability of CA1 pyramidal neurons. In the long term, we also observed slow and progressive excitotoxic neurodegeneration. Overall, our results indicate that interfering with microRNA biogenesis causes an increase in neuronal responsiveness and disrupts homeostatic mechanisms that protect the neuron against overactivation, which may explain both the initial and late phenotypes associated with the loss of Dicer in excitatory neurons.
Collapse
Affiliation(s)
- Anna Fiorenza
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Jose P Lopez-Atalaya
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Victor Rovira
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Marilyn Scandaglia
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Emilio Geijo-Barrientos
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Angel Barco
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| |
Collapse
|