1
|
Bramlett SN, Fitzmaurice SM, Harbin NH, Yan W, Bandlamudi C, Van Doorn GE, Smith Y, Hepler JR. Regulator of G protein signalling 14 (RGS14) protein expression profile in the adult mouse brain. Eur J Neurosci 2024; 60:7058-7085. [PMID: 39557622 DOI: 10.1111/ejn.16592] [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: 06/25/2024] [Revised: 10/02/2024] [Accepted: 10/20/2024] [Indexed: 11/20/2024]
Abstract
Regulator of G protein signalling 14 (RGS14) is a multifunctional signalling protein that serves as a natural suppressor of synaptic plasticity in the mouse brain. Our previous studies showed that RGS14 is highly expressed in postsynaptic dendrites and spines of pyramidal neurons in hippocampal area CA2 of the developing mouse brain. However, our more recent work with monkey brain shows that RGS14 is found in multiple neuron populations throughout hippocampal area CA1 and CA2, caudate nucleus, putamen, globus pallidus, substantia nigra and amygdala. In the mouse brain, we also have observed RGS14 protein in discrete limbic regions linked to reward behaviour and addiction, including the central amygdala and the nucleus accumbens, but a comprehensive mapping of RGS14 protein expression in the adult mouse brain is lacking. Here, we report that RGS14 is more broadly expressed in mouse brain than previously known. Intense RGS14 staining is observed in specific neuron populations of the hippocampal formation, amygdala, septum, bed nucleus of stria terminalis and ventral striatum/nucleus accumbens. RGS14 is also observed in axon fibre tracts including the dorsal fornix, fimbria, stria terminalis and the ventrohippocampal commissure. Moderate RGS14 staining is observed in various other adjacent regions not previously reported. These findings show that RGS14 is expressed in brain regions that govern aspects of core cognitive functions such as sensory perception, emotion, memory, motivation and execution of actions and suggest that RGS14 may serve to suppress plasticity and filter inputs in these brain regions to set the overall tone on experience-to-action processes.
Collapse
Affiliation(s)
- Sara N Bramlett
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shana M Fitzmaurice
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nicholas H Harbin
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Wuji Yan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Charan Bandlamudi
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - G Emme Van Doorn
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yoland Smith
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Neurology, Emory University School of Medcine, Atlanta, Georgia, USA
| | - John R Hepler
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
2
|
Hulsman AM, Klaassen FH, de Voogd LD, Roelofs K, Klumpers F. How Distributed Subcortical Integration of Reward and Threat May Inform Subsequent Approach-Avoidance Decisions. J Neurosci 2024; 44:e0794242024. [PMID: 39379152 PMCID: PMC11604143 DOI: 10.1523/jneurosci.0794-24.2024] [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: 04/12/2024] [Revised: 08/19/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
Healthy and successful living involves carefully navigating rewarding and threatening situations by balancing approach and avoidance behaviors. Excessive avoidance to evade potential threats often leads to forfeiting potential rewards. However, little is known about how reward and threat information is integrated neurally to inform approach or avoidance. In this preregistered study, participants (N behavior = 31, 17F; N MRI = 28, 15F) made approach-avoidance decisions under varying reward (monetary gains) and threat (electrical stimulations) prospects during functional MRI scanning. In contrast to theorized parallel subcortical processing of reward and threat information before cortical integration, Bayesian multivariate multilevel analyses revealed subcortical reward and threat integration prior to indicating approach-avoidance decisions. This integration occurred in the ventral striatum, thalamus, and bed nucleus of the stria terminalis (BNST). When reward was low, risk-diminishing avoidance decisions dominated, which was linked to more positive tracking of threat magnitude prior to indicating avoidance than approach decisions. In contrast, the amygdala exhibited dual sensitivity to reward and threat. While anticipating outcomes of risky approach decisions, we observed positive tracking of threat magnitude within the salience network (dorsal anterior cingulate cortex, thalamus, periaqueductal gray, BNST). Conversely, after risk-diminishing avoidance, characterized by reduced reward prospects, we observed more negative tracking of reward magnitude in the ventromedial prefrontal cortex and ventral striatum. These findings shed light on the temporal dynamics of approach-avoidance decision-making. Importantly, they demonstrate the role of subcortical integration of reward and threat information in balancing approach and avoidance, challenging theories positing predominantly separate subcortical processing prior to cortical integration.
Collapse
Affiliation(s)
- Anneloes M Hulsman
- Behavioural Science Institute, Radboud University, 6525 GD Nijmegen, The Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, 6525 EN Nijmegen, The Netherlands
| | - Felix H Klaassen
- Behavioural Science Institute, Radboud University, 6525 GD Nijmegen, The Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, 6525 EN Nijmegen, The Netherlands
| | - Lycia D de Voogd
- Behavioural Science Institute, Radboud University, 6525 GD Nijmegen, The Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, 6525 EN Nijmegen, The Netherlands
- Institute of Psychology and Leiden Institute for Brain and Cognition, Leiden University, 2333 AK Leiden, The Netherlands
| | - Karin Roelofs
- Behavioural Science Institute, Radboud University, 6525 GD Nijmegen, The Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, 6525 EN Nijmegen, The Netherlands
| | - Floris Klumpers
- Behavioural Science Institute, Radboud University, 6525 GD Nijmegen, The Netherlands
- Donders Centre for Cognitive Neuroimaging, Radboud University, 6525 EN Nijmegen, The Netherlands
| |
Collapse
|
3
|
Florido A, Curtis VR, Pégard NC, Rodriguez-Romaguera J. Disentangling the Neural Circuits of Arousal and Anxiety-Like Behavior. Curr Top Behav Neurosci 2024. [PMID: 39579325 DOI: 10.1007/7854_2024_539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Anxiety disorders are prevalent and debilitating conditions characterized by excessive concern and fear, affecting thoughts, behaviors, and sensations. A critical component of anxiety is arousal, a complex process involving alertness regulation and stimulus salience modulation. While arousal is adaptive in normal circumstances, dysregulation can lead to hypoarousal or hyperarousal, affecting response selection and threat perception. This chapter reviews challenges in studying arousal in preclinical anxiety models, emphasizing the need for multicomponent measurement and analysis. Novel methodologies integrating physiological measurement with activity tracking of neurons with single-cell resolution in awake animals are discussed, with emphasis in current challenges. Understanding these mechanisms is crucial for developing effective treatments for anxiety disorders.
Collapse
Affiliation(s)
- Antonio Florido
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Vincent R Curtis
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Nicolas C Pégard
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA.
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA.
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Stress Initiative, University of North Carolina, Chapel Hill, NC, USA.
| | - Jose Rodriguez-Romaguera
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA.
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Stress Initiative, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Institute for Developmental Disorders, University of North Carolina, Chapel Hill, NC, USA.
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Tamura Y, Maeda S, Takahashi H, Aoto Y, Matsuki T, Seki K. GABAergic circuit interaction between central amygdala and bed nucleus of the stria terminalis in lipopolysaccharide-induced despair-like behavior. Physiol Behav 2024; 288:114753. [PMID: 39551417 DOI: 10.1016/j.physbeh.2024.114753] [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: 10/05/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Hyperexcitability of central amygdala (CeA) induces depressive symptoms. The bed nucleus of the stria terminalis (BNST) receives GABAergic input from the CeA. However, it remains unclear whether the GABAergic neurons in the CeA projecting to BNST contribute to major depression. Here, we investigated the roles of GABAergic neurons in CeA and BNST in lipopolysaccharide (LPS)-induced despair-like behavior. We generated adeno-associated virus vectors (AAV) carrying shRNA against Gad67 to knock down GAD67 expression in CeA (Gad67-KD-CeA) or BNST (Gad67-KD-BNST) in C57BL/6J male mice. Despair-like behavior was assessed by tail suspension test (TST) 24 h after LPS administration. Saline-treated Gad67-KD-CeA mice exhibited longer immobility during TST than saline-treated AAV-injected control (AAV-Cont) mice. Although LPS increased immobility time in AAV-Cont mice, it did not affect immobility time in Gad67-KD-CeA mice. While LPS did not affect the immobility time in Gad67-KD-BNST mice, it increased immobility time in AAV-Cont mice. We injected GFP-expressing AAV with a Dlx promoter, specifically expressed in GABAergic neurons, into CeA, and FluoroGold, a retrograde neuronal tracer, into the BNST. GFP signals associated with CeA GABAergic neurons were detected in the BNST, contacting c-fos and GAD67-expressed cells following LPS. We detected the FluoroGold signals in GAD67- and c-fos-expressed neurons in the CeA after LPS administration. Bilateral intra-BNST injection of muscimol (2 pmol), a GABAA receptor agonist, increased immobility time during TST. These findings suggest that LPS-decreased GABAergic activity in the CeA may lead to disinhibition of GABAergic interneurons in the BNST, resulting in GABAA receptor activation and subsequently induces despair-like behavior.
Collapse
Affiliation(s)
- Yuka Tamura
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Sakura Maeda
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Haruna Takahashi
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Yuta Aoto
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Tohru Matsuki
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Aichi 480-0392, Japan.
| | - Kenjiro Seki
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan.
| |
Collapse
|
5
|
Horváth K, Vági P, Juhász B, Kuti D, Ferenczi S, Kovács KJ. Sex Differences in the Neuroendocrine Stress Response: A View from a CRH-Reporting Mouse Line. Int J Mol Sci 2024; 25:12004. [PMID: 39596070 PMCID: PMC11593550 DOI: 10.3390/ijms252212004] [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: 10/18/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Corticotropin-releasing hormone (CRH) neurons within the paraventricular hypothalamic nucleus (PVH) play a crucial role in initiating the neuroendocrine response to stress and are also pivotal in coordination of autonomic, metabolic, and behavioral stress reactions. Although the role of parvocellular CRHPVH neurons in activation of the hypothalamic-pituitary-adrenal (HPA) axis is well established, the distribution and function of CRH-expressing neurons across the whole central nervous system are less understood. Stress responses activate complex neural networks, which differ depending on the type of stressor and on the sex of the individual. Because of the technical difficulties of localizing CRH neurons throughout the rodent brain, several CRH reporter mouse lines have recently been developed. In this study, we used Crh-IRES-Cre;Ai9 reporter mice to examine whether CRH neurons are recruited in a stressor- or sex-specific manner, both within and outside the hypothalamus. In contrast to the clear sexual dimorphism of CRH-mRNA-expressing neurons, quantification of CRH-reporting, tdTomato-positive neurons in different stress-related brain areas revealed only subtle differences between male and female subjects. These results strongly imply that sex differences in CRH mRNA expression occur later in development under the influence of sex steroids and reflects the limitations of using genetic reporter constructs to reveal the current physiological/transcriptional status of a specific neuron population. Next, we compared the recruitment of stress-related, tdTomato-expressing (putative CRH) neurons in male and female Crh-IRES-Cre;Ai9 reporter mice that had been exposed to predator odor. In male mice, fox odor triggered more c-Fos in the CRH neurons of the paraventricular hypothalamic nucleus, central amygdala, and anterolateral bed nucleus of the stria terminalis compared to females. These results indicate that male mice are more sensitive to predator exposure due to a combination of hormonal, environmental, and behavioral factors.
Collapse
Affiliation(s)
- Krisztina Horváth
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Hungarian Research Network, 1083 Budapest, Hungary; (K.H.); (B.J.); (D.K.); (S.F.)
| | - Pál Vági
- Nikon Center of Excellence, Institute of Experimental Medicine, Hungarian Research Network, 1083 Budapest, Hungary;
| | - Balázs Juhász
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Hungarian Research Network, 1083 Budapest, Hungary; (K.H.); (B.J.); (D.K.); (S.F.)
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Dániel Kuti
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Hungarian Research Network, 1083 Budapest, Hungary; (K.H.); (B.J.); (D.K.); (S.F.)
| | - Szilamér Ferenczi
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Hungarian Research Network, 1083 Budapest, Hungary; (K.H.); (B.J.); (D.K.); (S.F.)
| | - Krisztina J. Kovács
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Hungarian Research Network, 1083 Budapest, Hungary; (K.H.); (B.J.); (D.K.); (S.F.)
| |
Collapse
|
6
|
Bramlett SN, Foster SL, Weinshenker D, Hepler JR. Endogenous Regulator of G protein Signaling 14 (RGS14) suppresses cocaine-induced emotionally motivated behaviors in female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612719. [PMID: 39314405 PMCID: PMC11419016 DOI: 10.1101/2024.09.12.612719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Addictive drugs hijack the neuronal mechanisms of learning and memory in motivation and emotion processing circuits to reinforce their own use. Regulator of G-protein Signaling 14 (RGS14) is a natural suppressor of post-synaptic plasticity underlying learning and memory in the hippocampus. The present study used immunofluorescence and RGS14 knockout mice to assess the role of RGS14 in behavioral plasticity and reward learning induced by chronic cocaine in emotional-motivational circuits. We report that RGS14 is strongly expressed in discrete regions of the ventral striatum and extended amygdala in wild-type mice, and is co-expressed with D1 and D2 dopamine receptors in neurons of the nucleus accumbens (NAc). Of note, we found that RGS14 is upregulated in the NAc in mice with chronic cocaine history following acute cocaine treatment. We found significantly increased cocaine-induced locomotor sensitization, as well as enhanced conditioned place preference and conditioned locomotor activity in RGS14-deficient mice compared to wild-type littermates. Together, these findings suggest that endogenous RGS14 suppresses cocaine-induced plasticity in emotional-motivational circuits, implicating RGS14 as a protective agent against the maladaptive neuroplastic changes that occur during addiction.
Collapse
Affiliation(s)
- Sara N. Bramlett
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Stephanie L. Foster
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - John R. Hepler
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Bramlett SN, Fitzmaurice SM, Harbin NH, Yan W, Bandlamudi C, Van Doorn GE, Smith Y, Hepler JR. Regulator of G Protein Signaling 14 protein expression profile in the adult mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.22.600169. [PMID: 38979272 PMCID: PMC11230234 DOI: 10.1101/2024.06.22.600169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Regulator of G protein signaling 14 (RGS14) is a multifunctional signaling protein that serves as a natural suppressor of synaptic plasticity in the mouse brain. Our previous studies showed that RGS14 is highly expressed in postsynaptic dendrites and spines of pyramidal neurons in hippocampal area CA2 of the developing mouse brain. However, our more recent work with adult rhesus macaque brain shows that RGS14 is found in multiple neuron populations throughout hippocampal area CA1 and CA2, caudate nucleus, putamen, globus pallidus, substantia nigra, and amygdala in the adult rhesus monkey brain. In the mouse brain, we also have observed RGS14 protein in discrete limbic regions linked to reward behavior and addiction, including the central amygdala and the nucleus accumbens, but a comprehensive mapping of RGS14 protein expression in the adult mouse brain is lacking. Here, we report that RGS14 is more broadly expressed in mouse brain than previously known. Intense RGS14 staining is observed in specific neuron populations of the hippocampal formation, amygdala, septum, bed nucleus of stria terminalis and ventral striatum/nucleus accumbens. RGS14 is also observed in axon fiber tracts including the dorsal fornix, fimbria, stria terminalis, and the ventrohippocampal commissure. Moderate RGS14 staining is observed in various other adjacent regions not previously reported. These findings show that RGS14 is expressed in brain regions that govern aspects of core cognitive functions such as sensory perception, emotion, memory, motivation, and execution of actions, and suggests that RGS14 may serve to suppress plasticity and filter inputs in these brain regions to set the overall tone on experience-to-action processes.
Collapse
|
8
|
Sotelo-Hitschfeld T, Minère M, Klemm P, Borgmann D, Wnuk-Lipinski D, Jais A, Jia X, Corneliussen S, Kloppenburg P, Fenselau H, Brüning JC. GABAergic disinhibition from the BNST to PNOC ARC neurons promotes HFD-induced hyperphagia. Cell Rep 2024; 43:114343. [PMID: 38865247 DOI: 10.1016/j.celrep.2024.114343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/01/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
Activation of prepronociceptin (PNOC)-expressing neurons in the arcuate nucleus (ARC) promotes high-fat-diet (HFD)-induced hyperphagia. In turn, PNOCARC neurons can inhibit the anorexic response of proopiomelanocortin (POMC) neurons. Here, we validate the necessity of PNOCARC activity for HFD-induced inhibition of POMC neurons in mice and find that PNOCARC-neuron-dependent inhibition of POMC neurons is mediated by gamma-aminobutyric acid (GABA) release. When monitoring individual PNOCARC neuron activity via Ca2+ imaging, we find a subpopulation of PNOCARC neurons that is inhibited upon gastrointestinal calorie sensing and disinhibited upon HFD feeding. Combining retrograde rabies tracing and circuit mapping, we find that PNOC neurons from the bed nucleus of the stria terminalis (PNOCBNST) provide inhibitory input to PNOCARC neurons, and this inhibitory input is blunted upon HFD feeding. This work sheds light on how an increase in caloric content of the diet can rewire a neuronal circuit, paving the way to overconsumption and obesity development.
Collapse
Affiliation(s)
- Tamara Sotelo-Hitschfeld
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Marielle Minère
- Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Synaptic Transmission in Energy Homeostasis Research Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Diba Borgmann
- Synaptic Transmission in Energy Homeostasis Research Group, Max Planck Institute for Metabolism Research, Cologne, Germany; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Daria Wnuk-Lipinski
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Alexander Jais
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Xianglian Jia
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Svenja Corneliussen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Institute of Zoology, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Peter Kloppenburg
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Institute of Zoology, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Henning Fenselau
- Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Synaptic Transmission in Energy Homeostasis Research Group, Max Planck Institute for Metabolism Research, Cologne, Germany.
| | - Jens Claus Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
9
|
Kovács A, Szabó E, László K, Kertes E, Zagorácz O, Mintál K, Tóth A, Gálosi R, Berta B, Lénárd L, Hormay E, László B, Zelena D, Tóth ZE. Brain RFamide Neuropeptides in Stress-Related Psychopathologies. Cells 2024; 13:1097. [PMID: 38994950 PMCID: PMC11240450 DOI: 10.3390/cells13131097] [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: 04/29/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024] Open
Abstract
The RFamide peptide family is a group of proteins that share a common C-terminal arginine-phenylalanine-amide motif. To date, the family comprises five groups in mammals: neuropeptide FF, LPXRFamides/RFamide-related peptides, prolactin releasing peptide, QRFP, and kisspeptins. Different RFamide peptides have their own cognate receptors and are produced by different cell populations, although they all can also bind to neuropeptide FF receptors with different affinities. RFamide peptides function in the brain as neuropeptides regulating key aspects of homeostasis such as energy balance, reproduction, and cardiovascular function. Furthermore, they are involved in the organization of the stress response including modulation of pain. Considering the interaction between stress and various parameters of homeostasis, the role of RFamide peptides may be critical in the development of stress-related neuropathologies. This review will therefore focus on the role of RFamide peptides as possible key hubs in stress and stress-related psychopathologies. The neurotransmitter coexpression profile of RFamide-producing cells is also discussed, highlighting its potential functional significance. The development of novel pharmaceutical agents for the treatment of stress-related disorders is an ongoing need. Thus, the importance of RFamide research is underlined by the emergence of peptidergic and G-protein coupled receptor-based therapeutic targets in the pharmaceutical industry.
Collapse
Affiliation(s)
- Anita Kovács
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Evelin Szabó
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kristóf László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Erika Kertes
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Olga Zagorácz
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kitti Mintál
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Attila Tóth
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Rita Gálosi
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bea Berta
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - László Lénárd
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Edina Hormay
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bettina László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Dóra Zelena
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Zsuzsanna E. Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary
| |
Collapse
|
10
|
van Velthoven CTJ, Gao Y, Kunst M, Lee C, McMillen D, Chakka AB, Casper T, Clark M, Chakrabarty R, Daniel S, Dolbeare T, Ferrer R, Gloe J, Goldy J, Guzman J, Halterman C, Ho W, Huang M, James K, Nguy B, Pham T, Ronellenfitch K, Thomas ED, Torkelson A, Pagan CM, Kruse L, Dee N, Ng L, Waters J, Smith KA, Tasic B, Yao Z, Zeng H. The transcriptomic and spatial organization of telencephalic GABAergic neuronal types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599583. [PMID: 38948843 PMCID: PMC11212977 DOI: 10.1101/2024.06.18.599583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The telencephalon of the mammalian brain comprises multiple regions and circuit pathways that play adaptive and integrative roles in a variety of brain functions. There is a wide array of GABAergic neurons in the telencephalon; they play a multitude of circuit functions, and dysfunction of these neurons has been implicated in diverse brain disorders. In this study, we conducted a systematic and in-depth analysis of the transcriptomic and spatial organization of GABAergic neuronal types in all regions of the mouse telencephalon and their developmental origins. This was accomplished by utilizing 611,423 single-cell transcriptomes from the comprehensive and high-resolution transcriptomic and spatial cell type atlas for the adult whole mouse brain we have generated, supplemented with an additional single-cell RNA-sequencing dataset containing 99,438 high-quality single-cell transcriptomes collected from the pre- and postnatal developing mouse brain. We present a hierarchically organized adult telencephalic GABAergic neuronal cell type taxonomy of 7 classes, 52 subclasses, 284 supertypes, and 1,051 clusters, as well as a corresponding developmental taxonomy of 450 clusters across different ages. Detailed charting efforts reveal extraordinary complexity where relationships among cell types reflect both spatial locations and developmental origins. Transcriptomically and developmentally related cell types can often be found in distant and diverse brain regions indicating that long-distance migration and dispersion is a common characteristic of nearly all classes of telencephalic GABAergic neurons. Additionally, we find various spatial dimensions of both discrete and continuous variations among related cell types that are correlated with gene expression gradients. Lastly, we find that cortical, striatal and some pallidal GABAergic neurons undergo extensive postnatal diversification, whereas septal and most pallidal GABAergic neuronal types emerge simultaneously during the embryonic stage with limited postnatal diversification. Overall, the telencephalic GABAergic cell type taxonomy can serve as a foundational reference for molecular, structural and functional studies of cell types and circuits by the entire community.
Collapse
Affiliation(s)
| | - Yuan Gao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mike Huang
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Beagan Nguy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
11
|
Zheng X, Dingpeng L, Yan X, Yao X, Wang Y. The role and mechanism of 5-HTDRN-BNST neural circuit in anxiety and fear lesions. Front Neurosci 2024; 18:1362899. [PMID: 38784088 PMCID: PMC11111893 DOI: 10.3389/fnins.2024.1362899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/10/2024] [Indexed: 05/25/2024] Open
Abstract
Central 5-hydroxytryptaminergic dorsal raphe nucleus (5-HTDRN)-bed nucleus of stria terminalis (BNST) neural circuit dysfunction is one of the important neurobiological basis of anxiety and fear disorders. Under stress, 5-hydroxytryptamine (5-HT) neurons act on BNST receptors to attenuate anxiety and fear responses or enhance anxiety and fear. In BNST, corticotropin releasing factor neurons play a role in regulating emotions by reversely regulating excitatory or inhibitory 5-HT neurons. The composition of 5-HTDRN-BNST neural circuit, the pathological changes of 5-HTDRN-BNST neural circuit function damage under stress, and the effects of 5-HTDRN-BNST neural circuit on anxiety disorder, panic disorder and post-traumatic stress disorder were analyzed and are summarized in this paper. The characteristics of functional changes of the neural circuit and its effects on brain functional activities provide a basis and ideas for the treatment of anxiety and fear disorders through the regulation of 5-HTDRN-BNST neural circuit, and they also provide a new perspective for understanding the pathological mechanism of such diseases.
Collapse
Affiliation(s)
- Xianli Zheng
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Li Dingpeng
- Gansu Provincial Second People’s Hospital, Lanzhou, Gansu, China
| | - Xingke Yan
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Xiaoqiang Yao
- Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Yongrui Wang
- Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
12
|
Han RW, Zhang ZY, Jiao C, Hu ZY, Pan BX. Synergism between two BLA-to-BNST pathways for appropriate expression of anxiety-like behaviors in male mice. Nat Commun 2024; 15:3455. [PMID: 38658548 PMCID: PMC11043328 DOI: 10.1038/s41467-024-47966-2] [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: 08/22/2023] [Accepted: 04/15/2024] [Indexed: 04/26/2024] Open
Abstract
Understanding how distinct functional circuits are coordinated to fine-tune mood and behavior is of fundamental importance. Here, we observe that within the dense projections from basolateral amygdala (BLA) to bed nucleus of stria terminalis (BNST), there are two functionally opposing pathways orchestrated to enable contextually appropriate expression of anxiety-like behaviors in male mice. Specifically, the anterior BLA neurons predominantly innervate the anterodorsal BNST (adBNST), while their posterior counterparts send massive fibers to oval BNST (ovBNST) with moderate to adBNST. Optogenetic activation of the anterior and posterior BLA inputs oppositely regulated the activity of adBNST neurons and anxiety-like behaviors, via disengaging and engaging the inhibitory ovBNST-to-adBNST microcircuit, respectively. Importantly, the two pathways exhibited synchronized but opposite responses to both anxiolytic and anxiogenic stimuli, partially due to their mutual inhibition within BLA and the different inputs they receive. These findings reveal synergistic interactions between two BLA-to-BNST pathways for appropriate anxiety expression with ongoing environmental demands.
Collapse
Affiliation(s)
- Ren-Wen Han
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Zi-Yi Zhang
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Chen Jiao
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Ze-Yu Hu
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
13
|
Fulenwider HD, Zhang Y, Ryabinin AE. Characterization of social hierarchy formation and maintenance in same-sex, group-housed male and female C57BL/6 J mice. Horm Behav 2024; 157:105452. [PMID: 37977023 PMCID: PMC10841988 DOI: 10.1016/j.yhbeh.2023.105452] [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: 03/30/2023] [Revised: 10/31/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Social hierarchies are a prevalent feature of all animal groups, and an individual's rank within the group can significantly affect their overall health, typically at the greatest expense of the lowest-ranked individuals, or omegas. These subjects have been shown to exhibit various stress-related phenotypes, such as increased hypothalamic-pituitary axis activity and increased amygdalar corticotropin-releasing factor levels compared to higher-ranked subjects. However, these findings have been primarily characterized in males and in models requiring exhibition of severe aggression. The goals of the current study, therefore, were to characterize the formation and maintenance of social hierarchies using the tube test and palatable liquid competition in same-sex groups of male and female C57BL/6 J mice. We also aimed to examine the effects of tube test-determined social rank on plasma and hypothalamic oxytocin and vasopressin levels, peptides with established roles in social behaviors and the stress response. Lastly, we assessed the effects of environmental enrichment and length of testing on the measures outlined above. Overall, we demonstrated that males and females develop social hierarchies and that these hierarchies can be determined using the tube test. While we were unable to establish a consistent connection between peptide levels and social rank, we observed transient changes in these peptides reflecting complex interactions between social rank, sex, environment, and length of testing. We also found that many male and female omegas began to exhibit passive coping behavior after repeated tube test losses, demonstrating the potential of this assay to serve as a model of chronic, mild psychosocial stress.
Collapse
Affiliation(s)
- Hannah D Fulenwider
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Yangmiao Zhang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Andrey E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
14
|
Ding SL. A novel subdivision of the bed nucleus of stria terminalis in monkey, rat, and mouse brains. J Comp Neurol 2023; 531:2121-2145. [PMID: 36583448 PMCID: PMC11406555 DOI: 10.1002/cne.25446] [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: 10/19/2022] [Revised: 11/17/2022] [Accepted: 12/05/2022] [Indexed: 12/31/2022]
Abstract
The bed nucleus of stria terminalis (BST) is a critical structure that mediates sustained vigilant responses to contextual, diffuse, and unpredictable threats. Dysfunction of the BST could lead to excessive anxiety and hypervigilance, which are often observed in posttraumatic stress disorder and anxiety disorders. Vigilance of potential future threats from the external environment is a basic brain function and probably requires rapid and/or short neural circuits, which enable both quick detection of the potential threats and fast adaptive responses. However, the BST in literature does not appear to receive spatial information directly from earlier visual or spatial processing structures. In this study, a novel subdivision of the BST is uncovered in monkey, rat, and mouse brains based on the human equivalent and is found in mouse to receive direct inputs from the ventral lateral geniculate nucleus and pretectal nucleus as well as from the spatial processing structures such as subiculum, presubiculum, and medial entorhinal cortex. This new subdivision, termed spindle-shaped small cell subdivision (BSTsc), is located between the known BST and the anterior thalamus. In addition to the unique afferent connections and cell morphology, the BSTsc also displays unique molecular signature (e.g., positive for excitatory markers) compared with other BST subdivisions, which are mostly composed of inhibitory GABAergic neurons. The BSTsc appears to have largely overlapping efferent projections with other BST subdivisions such as the projections to the amygdala, hypothalamus, nucleus accumbens, septum, and brainstem. Together, the present study suggests that the BSTsc is poised to serve as a shortcut bridge directly linking spatial information from the environment to vigilant adaptive internal responses.
Collapse
Affiliation(s)
- Song-Lin Ding
- Allen Institute for Brain Science, Seattle, Washington, USA
| |
Collapse
|
15
|
Takenawa S, Nagasawa Y, Go K, Chérasse Y, Mizuno S, Sano K, Ogawa S. Activity of estrogen receptor β expressing neurons in the medial amygdala regulates preference toward receptive females in male mice. Proc Natl Acad Sci U S A 2023; 120:e2305950120. [PMID: 37819977 PMCID: PMC10589649 DOI: 10.1073/pnas.2305950120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
The processing of information regarding the sex and reproductive state of conspecific individuals is critical for successful reproduction and survival in males. Generally, male mice exhibit a preference toward the odor of sexually receptive (RF) over nonreceptive females (XF) or gonadally intact males (IM). Previous studies suggested the involvement of estrogen receptor beta (ERβ) expressed in the medial amygdala (MeA) in male preference toward RF. To further delineate the role played by ERβ in the MeA in the neuronal network regulating male preference, we developed a new ERβ-iCre mouse line using the CRISPR-Cas9 system. Fiber photometry Ca2+ imaging revealed that ERβ-expressing neurons in the postero-dorsal part of the MeA (MeApd-ERβ+ neurons) were more active during social investigation toward RF compared to copresented XF or IM mice in a preference test. Chemogenetic inhibition of MeApd-ERβ+ neuronal activity abolished a preference to RF in "RF vs. XF," but not "RF vs. IM," tests. Analysis with cre-dependent retrograde tracing viral vectors identified the principal part of the bed nucleus of stria terminalis (BNSTp) as a primary projection site of MeApd-ERβ+ neurons. Fiber photometry recording in the BNSTp during a preference test revealed that chemogenetic inhibition of MeApd-ERβ+ neurons abolished differential neuronal activity of BNSTp cells as well as a preference to RF against XF but not against IM mice. Collectively, these findings demonstrate for the first time that MeApd-ERβ+ neuronal activity is required for expression of receptivity-based preference (i.e., RF vs. XF) but not sex-based preference (i.e., RF vs. IM) in male mice.
Collapse
Affiliation(s)
- Satoshi Takenawa
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba305-8577, Japan
| | - Yutaro Nagasawa
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba305-8577, Japan
| | - Kim Go
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba305-8577, Japan
| | - Yoan Chérasse
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center and Trans-border Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Kazuhiro Sano
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba305-8577, Japan
| | - Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba305-8577, Japan
| |
Collapse
|
16
|
van de Poll Y, Cras Y, Ellender TJ. The neurophysiological basis of stress and anxiety - comparing neuronal diversity in the bed nucleus of the stria terminalis (BNST) across species. Front Cell Neurosci 2023; 17:1225758. [PMID: 37711509 PMCID: PMC10499361 DOI: 10.3389/fncel.2023.1225758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/03/2023] [Indexed: 09/16/2023] Open
Abstract
The bed nucleus of the stria terminalis (BNST), as part of the extended amygdala, has become a region of increasing interest regarding its role in numerous human stress-related psychiatric diseases, including post-traumatic stress disorder and generalized anxiety disorder amongst others. The BNST is a sexually dimorphic and highly complex structure as already evident by its anatomy consisting of 11 to 18 distinct sub-nuclei in rodents. Located in the ventral forebrain, the BNST is anatomically and functionally connected to many other limbic structures, including the amygdala, hypothalamic nuclei, basal ganglia, and hippocampus. Given this extensive connectivity, the BNST is thought to play a central and critical role in the integration of information on hedonic-valence, mood, arousal states, processing emotional information, and in general shape motivated and stress/anxiety-related behavior. Regarding its role in regulating stress and anxiety behavior the anterolateral group of the BNST (BNSTALG) has been extensively studied and contains a wide variety of neurons that differ in their electrophysiological properties, morphology, spatial organization, neuropeptidergic content and input and output synaptic organization which shape their activity and function. In addition to this great diversity, further species-specific differences are evident on multiple levels. For example, classic studies performed in adult rat brain identified three distinct neuron types (Type I-III) based on their electrophysiological properties and ion channel expression. Whilst similar neurons have been identified in other animal species, such as mice and non-human primates such as macaques, cross-species comparisons have revealed intriguing differences such as their comparative prevalence in the BNSTALG as well as their electrophysiological and morphological properties, amongst other differences. Given this tremendous complexity on multiple levels, the comprehensive elucidation of the BNSTALG circuitry and its role in regulating stress/anxiety-related behavior is a major challenge. In the present Review we bring together and highlight the key differences in BNSTALG structure, functional connectivity, the electrophysiological and morphological properties, and neuropeptidergic profiles of BNSTALG neurons between species with the aim to facilitate future studies of this important nucleus in relation to human disease.
Collapse
Affiliation(s)
- Yana van de Poll
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Yasmin Cras
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tommas J. Ellender
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Slabe Z, Pechler GA, van Heerikhuize J, Samuels BA, Živin M, Zorović M, Swaab DF. Increased pituitary adenylate cyclase-activating polypeptide in the central bed nucleus of the stria terminalis in mood disorders in men. Neurobiol Dis 2023:106191. [PMID: 37290577 DOI: 10.1016/j.nbd.2023.106191] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/09/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023] Open
Abstract
The mood disorders major depressive disorder (MDD) and bipolar disorder (BD) are highly prevalent worldwide. Women are more vulnerable to these psychopathologies than men. The bed nucleus of the stria terminalis (BNST), the amygdala and the hypothalamus are the crucial interconnected structures involved in the stress response. In mood disorders, stress systems in the brain are put into a higher gear. The BNST is implicated in mood, anxiety, and depression. The stress-related neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is highly abundant in the central BNST (cBNST). In this study, we investigated alterations in PACAP in the cBNST of patients with mood disorders. Immunohistochemical (IHC) staining of PACAP and in situ hybridization (ISH) of PACAP mRNA were performed on the cBNST of post-mortem human brain samples. Quantitative IHC revealed elevated PACAP levels in the cBNST in both mood disorders, MDD and BD, but only in men, not in women. The PACAP ISH was negative, indicating that PACAP is not produced in the cBNST. The results support the possibility that PACAP innervation of the cBNST plays a role in mood disorder pathophysiology in men.
Collapse
Affiliation(s)
- Zala Slabe
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; University of Ljubljana, Faculty of Medicine, Institute of Pharmacology and Experimental Toxicology, Korytkova ulica 2, Ljubljana, Slovenia
| | - Gwyneth A Pechler
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; University of Ljubljana, Faculty of Medicine, Institute of Pharmacology and Experimental Toxicology, Korytkova ulica 2, Ljubljana, Slovenia
| | - Joop van Heerikhuize
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | | | - Marko Živin
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Maja Zorović
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Dick F Swaab
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands; University of Ljubljana, Faculty of Medicine, Institute of Pharmacology and Experimental Toxicology, Korytkova ulica 2, Ljubljana, Slovenia.
| |
Collapse
|
18
|
Coolen RL, Cambier JC, van Asselt E, Blok BFM. Androgen receptors in the forebrain: A study in adult male cats. J Morphol 2023; 284:e21553. [PMID: 36601705 PMCID: PMC10107852 DOI: 10.1002/jmor.21553] [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: 09/21/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023]
Abstract
Androgens and their receptors are present throughout the body. Various structures such as muscles, genitals, and prostate express androgen receptors. The central nervous system also expresses androgen receptors. Androgens cross the blood-brain barrier to reach these central areas. In the central nervous system, androgens are involved in multiple functions. The current study investigated in which forebrain areas androgens are expressed in the male cat. Androgen receptor immunoreactive (AR-IR) nuclei were plotted and the results were quantified with a Heidelberg Topaz II + scanner and Linocolor 5.0 software. The density and intensity of the labeled cells were the main outcomes of interest. The analysis revealed a dense distribution of AR-IR nuclei in the preoptic area, periventricular complex of the hypothalamus, posterior hypothalamic area, ventromedial hypothalamic, parvocellular hypothalamic, infundibular, and supramammillary nucleus. Numerous AR-IR cells were also observed in the dorsal division of the anterior olfactory nucleus, lateral septal nucleus, medial and lateral divisions of the bed nucleus of the stria terminalis, lateral olfactory tract nucleus, anterior amygdaloid area, and the central and medial amygdaloid nuclei. AR-IR nuclei were predominantly observed in areas involved in autonomic and neuroendocrinergic responses which are important for many physiological processes and behaviors.
Collapse
Affiliation(s)
- Rosa L Coolen
- Department of Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Els van Asselt
- Department of Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bertil F M Blok
- Department of Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
19
|
Coexistence of chronic hyperalgesia and multilevel neuroinflammatory responses after experimental SCI: a systematic approach to profiling neuropathic pain. J Neuroinflammation 2022; 19:264. [PMID: 36309729 PMCID: PMC9617391 DOI: 10.1186/s12974-022-02628-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022] Open
Abstract
Background People with spinal cord injury (SCI) frequently develop neuropathic pain (NP) that worsens disability and diminishes rehabilitation efficacy. Chronic NP is presently incurable due to poor understanding of underlying mechanisms. We hypothesized that multilocus neuroinflammation (NIF) might be a driver of SCI NP, and tested it by investigating whether NP coexisted with central NIF, neurotransmission (NTM), neuromodulation (NML) and neuroplasticity (NPL) changes post-SCI. Methods Female Sprague–Dawley rats (230–250 g) with T10 compression or laminectomy were evaluated for physical conditions, coordinated hindlimb functions, neurological reflexes, and mechanical/thermal sensitivity thresholds at 1 day post-injury (p.i.) and weekly thereafter. Eight weeks p.i., central nervous system tissues were histochemically and immunohistochemically characterized for parameters/markers of histopathology and NIF/NTM/NML/NPL. Also analyzed was the correlative relationship between levels of selected biomarkers and thermosensitivity thresholds via statistical linear regression. Results SCI impaired sensorimotor functions, altered reflexes, and produced spontaneous pain signs and hypersensitivity to evoked nociceptive, mechanical, and thermal inputs. Only injured spinal cords exhibited neural lesion, microglia/astrocyte activation, and abnormal expression of proinflammatory cytokines, as well as NIF/NTM/NML/NPL markers. Brains of SCI animals displayed similar pathophysiological signs in the gracile and parabrachial nuclei (GrN and PBN: sensory relay), raphe magnus nucleus and periaqueduct gray (RMN and PAG: pain modulation), basolateral amygdala (BLA: emotional-affective dimension of pain), and hippocampus (HPC: memory/mood/neurogenesis). SCI augmented sensory NTM/NPL (GrN and PBN); increased GAD67 (PAG) level; reduced serotonin (RMN) and fear-off neuronal NTR2 (BLA) expressions; and perturbed neurogenesis (HPC). Conclusion T10 compression caused chronic hyperalgesia that coexisted with NIF/NTM/NML/NPL responses at multilevel neuroaxis centers. The data have provided multidimensional biomarkers as new mechanistic leads to profile SCI NP for therapeutic/therapy development. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02628-2.
Collapse
|
20
|
Divergent Neural and Endocrine Responses in Wild-Caught and Laboratory-Bred Rattus Norvegicus. Behav Brain Res 2022; 432:113978. [PMID: 35753530 DOI: 10.1016/j.bbr.2022.113978] [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: 02/14/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 11/23/2022]
Abstract
Although rodents have represented the most intensely studied animals in neurobiological investigations for more than a century, few studies have systematically compared neural and endocrine differences between wild rodents in their natural habitats and laboratory strains raised in traditional laboratory environments. In the current study, male and female Rattus norvegicus rats were trapped in an urban setting and compared to weight-and sex-matched conspecifics living in standard laboratory housing conditions. Brains were extracted for neural assessments and fecal boli were collected for endocrine [corticosterone and dehydroepiandrosterone (DHEA)] assays. Additionally, given their role in immune and stress functions, spleen and adrenal weights were recorded. A separate set of wild rats was trapped at a dairy farm and held in captivity for one month prior to assessments; in these animals, brains were processed but no hormone data were available. The results indicated that wild-trapped rats exhibited 31% heavier brains, including higher densities of cerebellar neurons and glial cells in the bed nucleus of the stria terminalis. The wild rats also had approximately 300% greater spleen and adrenal weights, and more than a six-fold increase in corticosterone levels than observed in laboratory rats. Further research on neurobiological variables in wild vs. lab animals will inform the extensive neurobiological knowledge base derived from laboratory investigations using selectively bred rodents in laboratory environments, knowledge that will enhance the translational value of preclinical laboratory rodent studies.
Collapse
|
21
|
Sakharkar M, Rockland KS, Duque A. Complex Neurochemical Microstructure of the Stria Terminalis in Infant and Adult Macaque Monkey. Front Neuroanat 2022; 16:891608. [PMID: 35692258 PMCID: PMC9174467 DOI: 10.3389/fnana.2022.891608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/20/2022] [Indexed: 11/29/2022] Open
Abstract
The stria terminalis (ST) is a major bidirectional fiber tract anchored in the amygdala and bed nucleus (BNST). Extensive investigations in rodents report a complex arrangement of neurochemically diverse neurons within the ST, but fewer data are available for non-human primates. Given the functional importance of the ST, we investigated its microarchitecture in one newborn, four infant, and two adult macaque brains, by parallel immunocytochemical series for cells or fibers. Main results are as follows: (1) The pan-neuronal marker NeuN shows scattered neurons and small neuronal clusters in both the dorsal and ventral ST, but more numerous dorsally; (2) smaller neuronal subpopulations are labeled by calretinin (CR), neuropeptide Y (NPY), calbindin (CB), and somatostatin (SOM), of which the CR + neurons are the most numerous; (3) the infant brains on average have more neurons in the ST than the adult brains, but across our sample, there is notable individual variability; and (4) fiber architectonics have a complex organization, which can be referenced to myelin-poor or myelin-dense zones. Myelin-poor zones coincide with concentrations of fibers positive for CB, CR, or tyrosine hydroxylase (TH). Neurons have been reported in other white matter domains (e.g., anterior commissure, corpus callosum, cingulum bundle, and subcortical white matter). Like these, at least some neurons within the ST may give rise to long-distance connections, and/or participate in more local functions, such as vascular regulation or axon guidance/maintenance.
Collapse
Affiliation(s)
| | - Kathleen S. Rockland
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Alvaro Duque
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
- *Correspondence: Alvaro Duque,
| |
Collapse
|
22
|
Boucher MN, Aktar M, Braas KM, May V, Hammack SE. Activation of Lateral Parabrachial Nucleus (LPBn) PACAP-Expressing Projection Neurons to the Bed Nucleus of the Stria Terminalis (BNST) Enhances Anxiety-like Behavior. J Mol Neurosci 2022; 72:451-458. [PMID: 34811712 PMCID: PMC8930475 DOI: 10.1007/s12031-021-01946-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/08/2021] [Indexed: 10/19/2022]
Abstract
Anxiety disorders are among the most common psychiatric disorders, and understanding the underlying neurocircuitry of anxiety- and stress-related behaviors may be important for treatment. The bed nucleus of the stria terminalis (BNST) has been studied for its role in many stress-related pathologies, such as anxiety, pain, depression, and addiction. Our prior work has demonstrated that pituitary adenylate cyclase-activating polypeptide (PACAP) receptor activation in the BNST mediates many of the behavioral consequences of chronic stress. While the BNST contains local PACAP-expressing neurons, a major source of afferent PACAP is the lateral parabrachial nucleus (LPBn), and excitotoxic lesions of the LPBn substantially decreasess PACAP immunostaining in the BNST. Here, we first assessed Cre-dependent reporter expression by injecting AAV2-hSyn-DIO-mCherry into the LPBn of PACAP-IRES-Cre mice for circuit mapping studies and identified PACAP projections to the BNST, lateral capsular central nucleus of the amygdala (CeLC), and ventromedial hypothalamus (VMH). In a second study, we assessed the effects of chemogenetically activating LPBn PACAP afferents in the BNST by injecting AAV2-hSyn-DIO-hM3D(Gq)-mCherry into the LPBn of PACAP-IRES-Cre mice for Cre-dependent expression of excitatory designer receptors exclusively activated by designer drugs (DREADDs). Before behavioral testing, clozapine-N-oxide (CNO), the selective agonist of our DREADD, was infused directly into the BNST. We found that after specific activation of LPBn PACAP afferents in the BNST, mice had increased anxiety-like behavior compared with controls, while total locomotor activity was unaffected. These results indicate that activation of PACAPergic LPBn projections to the BNST may play an important role in producing anxiety-like behavior.
Collapse
Affiliation(s)
- Melissa N. Boucher
- Department of Psychological Science, University of Vermont, 2 Colchester Avenue, Burlington, VT 05405, USA,Department of Neurological Sciences, College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT 05405, USA
| | - Mahafuza Aktar
- Department of Psychological Science, University of Vermont, 2 Colchester Avenue, Burlington, VT 05405, USA,Department of Neurological Sciences, College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT 05405, USA
| | - Karen M. Braas
- Department of Psychological Science, University of Vermont, 2 Colchester Avenue, Burlington, VT 05405, USA,Department of Neurological Sciences, College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT 05405, USA
| | - Victor May
- Department of Psychological Science, University of Vermont, 2 Colchester Avenue, Burlington, VT 05405, USA,Department of Neurological Sciences, College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT 05405, USA
| | - Sayamwong E. Hammack
- Department of Psychological Science, University of Vermont, 2 Colchester Avenue, Burlington, VT 05405, USA,Department of Neurological Sciences, College of Medicine, University of Vermont, 149 Beaumont Avenue, Burlington, VT 05405, USA,Corresponding author:
| |
Collapse
|