1
|
Fuller PJ, Yang J, Young MJ, Cole TJ. Mechanisms of ligand-mediated modulation of mineralocorticoid receptor signaling. Mol Cell Endocrinol 2025; 600:112504. [PMID: 39983891 DOI: 10.1016/j.mce.2025.112504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
The mineralocorticoid receptor plays a central role in homeostasis, mediating the regulation by aldosterone of epithelial sodium transport. In addition, it regulates a range of responses in other tissues where it is likely responding to both mineralocorticoids and glucocorticoids. Structural, functional and evolutionary studies have provided insights into the mechanisms of receptor activation by agonist ligands and how interactions within the domains of the mineralocorticoid receptor may modulate the response to individual ligands including the mechanisms of antagonism. This review will discuss the current understanding, including recent insights into these interactions, with implications for an emerging array of novel non-steroidal compounds targeting the mineralocorticoid receptor; and highlight their relevance to ligand- or tissue-specificity as well as their suitability as therapeutic agents.
Collapse
Affiliation(s)
- Peter J Fuller
- Centre of Endocrinology and Metabolism, Hudson Institute of Medical Research and Department of Molecular Translational Science, Monash University Clayton, Victoria, Australia.
| | - Jun Yang
- Centre of Endocrinology and Metabolism, Hudson Institute of Medical Research and Department of Molecular Translational Science, Monash University Clayton, Victoria, Australia
| | - Morag J Young
- Cardiovascular Endocrinology Laboratory, Discovery & Preclinical Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Timothy J Cole
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
2
|
Li Y, Xie H, Liu S, Ruan Z, Wang B. Resveratrol Alleviates Ischemia-Reperfusion-Induced Neuronal Damage by Inhibiting NR3C2-Mediated TRIM28 Expression. J Integr Neurosci 2025; 24:31375. [PMID: 40302259 DOI: 10.31083/jin31375] [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: 11/26/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (CIRI) exacerbates neuronal damage through mechanisms including apoptosis and autophagy dysregulation. Resveratrol (Res), a natural polyphenol with neuroprotective properties, may alleviate CIRI-induced damage by modulating key signaling pathways. This study investigates the therapeutic effects of Res on CIRI, focusing on its role in balancing apoptosis and autophagy via regulation of nuclear receptor subfamily 3 group C member 2 (NR3C2) and tripartite motif containing 28 (TRIM28). METHOD In vivo, cognitive impairment, neurological dysfunction, cerebral infarction, neuronal damage, and inflammatory response were assessed in Sprague Dawley (SD) rats subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) using the morris water maze, Longa and Bederson scores, 2,3,5-tripheny ltetrazolium chloride (TTC) staining, hematoxylin and eosin staining, Nissl staining, and enzyme-linked immunosorbent assay (ELISA). The expression of NR3C2 and TRIM28 were analyzed by real time quantitative polymerase chain reaction (RT-qPCR) and western blot (WB). In vitro, Res effects on oxygen-glucose deprivation/reperfusion (OGD/R)-treated PC12 cells were evaluated using cell counting kit-8 (CCK-8), ELISA, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) staining, and WB. The relationship between NR3C2 and TRIM28 was validated using dual luciferase and chromatin immunoprecipitation followed by quantitative polymerase chain reaction (ChIP-qPCR). RESULT Res treatment significantly improved cognitive performance in the morris water maze test, and the infarct area was reduced by 16.736%. It was accompanied by downregulation of NR3C2 and TRIM28 expression. In vitro, Res enhanced cell viability, reduced inflammatory responses and apoptosis (with a 17.70% decrease in cell apoptosis rate), and restored autophagy balance. Mechanistically, NR3C2 was shown to directly regulate TRIM28 transcription, mediating the observed neuroprotective effects. CONCLUSION Res inhibits NR3C2 expression, which in turn directly regulates the transcription of TRIM28 through NR3C2, alleviating apoptosis and autophagy dysregulation induced by CIRI. This mechanism clearly demonstrates the important role of NR3C2 in CIRI and reveals its regulatory relationship with TRIM28. By uncovering the neuroprotective effects of Res, we provide new insights for the treatment of CIRI and lay the foundation for future targeted therapeutic strategies involving NR3C2 and TRIM28.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, 421002 Hengyang, Hunan, China
| | - Haiwei Xie
- Department of Anesthesiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, 421002 Hengyang, Hunan, China
| | - Shuang Liu
- Department of Anesthesiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, 421002 Hengyang, Hunan, China
| | - Zhongfan Ruan
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 421001 Hengyang, Hunan, China
| | - Baiyun Wang
- Department of Anesthesiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, 421002 Hengyang, Hunan, China
| |
Collapse
|
3
|
Gasparini S, Peltekian L, McDonough MC, Mitchell CJ, Hefti M, Resch JM, Geerling JC. Aldosterone-induced salt appetite requires HSD2 neurons. JCI Insight 2024; 9:e175087. [PMID: 39446486 PMCID: PMC11623954 DOI: 10.1172/jci.insight.175087] [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] [Accepted: 10/22/2024] [Indexed: 10/26/2024] Open
Abstract
Excessive aldosterone production increases the risk of heart disease, stroke, dementia, and death. Aldosterone increases both sodium retention and sodium consumption, and increased sodium consumption may worsen end-organ damage in patients with aldosteronism. Preventing this increase could improve outcomes, but the behavioral mechanisms of aldosterone-induced sodium appetite remain unclear. In rodents, we previously identified aldosterone-sensitive neurons, which express the mineralocorticoid receptor and its prereceptor regulator, 11-β-hydroxysteroid dehydrogenase 2 (HSD2). In the present study, we identified HSD2 neurons in the human brain and then used a mouse model to evaluate their role in aldosterone-induced salt intake. First, we confirmed that dietary sodium deprivation increases aldosterone production, salt intake, and HSD2 neuron activity. Next, we showed that continuous chemogenetic stimulation of HSD2 neurons causes a large and specific increase in salt intake. Finally, we used dose-response studies and genetically targeted ablation of HSD2 neurons to show that these neurons are necessary for aldosterone-induced salt intake. Identifying HSD2 neurons in the human brain and establishing their necessity for aldosterone-induced salt intake in mice improves our understanding of appetitive circuits and highlights this small cell population as a therapeutic target for moderating dietary sodium.
Collapse
Affiliation(s)
| | | | | | | | - Marco Hefti
- Department of Pathology, and
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Jon M. Resch
- Department of Neuroscience and Pharmacology
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Joel C. Geerling
- Department of Neurology
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
4
|
Gasparini S, Almeida‐Pereira G, Munuzuri ASP, Resch JM, Geerling JC. Molecular Ontology of the Nucleus of Solitary Tract. J Comp Neurol 2024; 532:e70004. [PMID: 39629676 PMCID: PMC11615840 DOI: 10.1002/cne.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/03/2024] [Accepted: 11/15/2024] [Indexed: 12/08/2024]
Abstract
The nucleus of the solitary tract (NTS) receives visceral information and regulates appetitive, digestive, and cardiorespiratory systems. Within the NTS, diverse processes operate in parallel to sustain life, but our understanding of their cellular composition is incomplete. Here, we integrate histologic and transcriptomic analysis to identify and compare molecular features that distinguish neurons in this brain region. Most glutamatergic neurons in the NTS and area postrema co-express the transcription factors Lmx1b and Phox2b, except for a ventral band of neurons in the far-caudal NTS, which include the Gcg-expressing neurons that produce glucagon-like peptide 1 (GLP-1). GABAergic interneurons intermingle through the Lmx1b+Phox2b macropopulation, and dense clusters of GABAergic neurons surround the NTS. The Lmx1b+Phox2b macropopulation includes subpopulations with distinct distributions expressing Grp, Hsd11b2, Npff, Pdyn, Pou3f1, Sctr, Th, and other markers. These findings highlight Lmx1b-Phox2b co-expression as a common feature of glutamatergic neurons in the NTS and improve our understanding of the organization and distribution of neurons in this critical brain region.
Collapse
Affiliation(s)
| | | | | | - Jon M. Resch
- Department of Neuroscience and PharmacologyUniversity of IowaIowa CityIowaUSA
- Iowa Neuroscience InstituteUniversity of IowaIowa CityIowaUSA
| | - Joel C. Geerling
- Department of NeurologyUniversity of IowaIowa CityIowaUSA
- Iowa Neuroscience InstituteUniversity of IowaIowa CityIowaUSA
| |
Collapse
|
5
|
Nieckarz A, Graff B, Burnier M, Marcinkowska AB, Narkiewicz K. Aldosterone in the brain and cognition: knowns and unknowns. Front Endocrinol (Lausanne) 2024; 15:1456211. [PMID: 39553314 PMCID: PMC11563778 DOI: 10.3389/fendo.2024.1456211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
Mineralocorticoid receptors are expressed in several structures of the central nervous system, and aldosterone levels can be measured in the brain, although in smaller amounts than in plasma. Nevertheless, these amounts appear to be sufficient to elicit substantial clinical effects. Primary aldosteronism, characterized by high levels of plasma aldosterone, is one of the most common causes of secondary hypertension. In this context, high aldosterone levels may have both indirect and direct effects on the brain with a negative impact on several cerebral functions. Thus, chronic aldosterone excess has been associated with symptoms of anxiety and depression - two clinical entities themselves associated with cognitive deficits. Today, there is an increasing number of reports on the influence of aldosterone on the brain, but there is also a significant amount of uncertainty, such as the role of high aldosterone levels on cognitive functions and decline independently of blood pressure. In this mini review, we discuss the known and unknowns of the impact of aldosterone on the brain putting emphasis on cognitive functions.
Collapse
Affiliation(s)
- Anna Nieckarz
- Department of Hypertension and Diabetology, Medical University of Gdańsk, Gdańsk, Poland
| | - Beata Graff
- Department of Hypertension and Diabetology, Medical University of Gdańsk, Gdańsk, Poland
| | - Michel Burnier
- Department of Hypertension and Diabetology, Medical University of Gdańsk, Gdańsk, Poland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Anna B. Marcinkowska
- Applied Cognitive Neuroscience Lab, Department of Neurophysiology, Neuropsychology and Neuroinformatics, Medical University of Gdańsk, Gdańsk, Poland
| | - Krzysztof Narkiewicz
- Department of Hypertension and Diabetology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
6
|
Kuralay A, McDonough MC, Resch JM. Control of sodium appetite by hindbrain aldosterone-sensitive neurons. Mol Cell Endocrinol 2024; 592:112323. [PMID: 38936597 PMCID: PMC11381173 DOI: 10.1016/j.mce.2024.112323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/25/2024] [Indexed: 06/29/2024]
Abstract
Mineralocorticoids play a key role in hydromineral balance by regulating sodium retention and potassium wasting. Through favoring sodium, mineralocorticoids can cause hypertension from fluid overload under conditions of hyperaldosteronism, such as aldosterone-secreting tumors. An often-overlooked mechanism by which aldosterone functions to increase sodium is through stimulation of salt appetite. To drive sodium intake, aldosterone targets neurons in the hindbrain which uniquely express 11β-hydroxysteroid dehydrogenase type 2 (HSD2). This enzyme is a necessary precondition for aldosterone-sensing cells as it metabolizes glucocorticoids - preventing their activation of the mineralocorticoid receptor. In this review, we will consider the role of hindbrain HSD2 neurons in regulating sodium appetite by discussing HSD2 expression in the brain, regulation of hindbrain HSD2 neuron activity, and the circuitry mediating the effects of these aldosterone-sensitive neurons. Reducing the activity of hindbrain HSD2 neurons may be a viable strategy to reduce sodium intake and cardiovascular risk, particularly for conditions of hyperaldosteronism.
Collapse
Affiliation(s)
- Ahmet Kuralay
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA
| | - Miriam C McDonough
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA; Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
7
|
Ali MSS, Parastooei G, Raman S, Mack J, Kim YS, Chung MK. Genetic labeling of the nucleus of tractus solitarius neurons associated with electrical stimulation of the cervical or auricular vagus nerve in mice. Brain Stimul 2024; 17:987-1000. [PMID: 39173736 PMCID: PMC11555405 DOI: 10.1016/j.brs.2024.08.007] [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: 05/28/2024] [Revised: 07/31/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024] Open
Abstract
INTRODUCTION Vagus nerve stimulation (VNS) is clinically useful for treating epilepsy, depression, and chronic pain. Currently, cervical VNS (cVNS) treatment is well-established, while auricular VNS (aVNS) is under development. Vagal stimulation regulates functions in diverse brain regions; therefore, it is critical to better understand how electrically-evoked vagal inputs following cVNS and aVNS engage with different brain regions. OBJECTIVE As vagus inputs are predominantly transmitted to the nucleus of tractus solitarius (NTS), we directly compared the activation of NTS neurons by cVNS or aVNS and the brain regions directly projected by the activated NTS neurons in mice. METHODS We adopted the targeted recombination in active populations method, which allows for the activity-dependent, tamoxifen-inducible expression of mCherry-a reporter protein-in neurons specifically associated with cVNS or aVNS. RESULTS cVNS and aVNS induced comparable bilateral mCherry expressions in neurons within the NTS, especially in its caudal section (cNTS). However, the numbers of mCherry-expressing neurons within different subdivisions of cNTS was distinctive. In both cVNS and aVNS, anterogradely labeled mCherry-expressing axonal terminals were similarly observed across different areas of the forebrain, midbrain, and hindbrain. These terminals were enriched in the rostral ventromedial medulla, parabrachial nucleus, periaqueductal gray, thalamic nuclei, central amygdala, and the hypothalamus. Sex difference of cVNS- and aVNS-induced labeling of NTS neurons was modest. CONCLUSION The central projections of mCherry-expressing cNTS terminals are comparable between aVNS and cVNS, suggesting that cVNS and aVNS activate distinct but largely overlapping projections into the brain through the cNTS.
Collapse
Affiliation(s)
- Md Sams Sazzad Ali
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Ghazaal Parastooei
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Swarnalakshmi Raman
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Jalen Mack
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Yu Shin Kim
- Department of Oral & Maxillofacial Surgery, School of Dentistry, Programs in Integrated Biomedical Sciences, Translational Sciences, Biomedical Engineering, Radiological Sciences, University of Texas Health Science Center at San Antonio, USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, 21201, USA.
| |
Collapse
|
8
|
Lalonde R, Strazielle C. Neurochemical Anatomy of Cushing's Syndrome. Neurochem Res 2024; 49:1945-1964. [PMID: 38833089 DOI: 10.1007/s11064-024-04172-2] [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: 11/17/2023] [Revised: 03/05/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
The neurochemical anatomy underlying Cushing's syndrome is examined for regional brain metabolism as well as neurotransmitter levels and receptor binding of biogenic amines and amino acids. Preliminary studies generally indicate that glucose uptake, blood flow, and activation on fMRI scans decreased in neocortical areas and increased in subcortical areas of patients with Cushing's syndrome or disease. Glucocorticoid-mediated increases in hippocampal metabolism occurred despite in vitro evidence of glucocorticoid-induced decreases in glucose uptake or consumption, indicating that in vivo increases are the result of indirect, compensatory, or preliminary responses. In animal studies, glucocorticoid administration decreased 5HT levels and 5HT1A receptor binding in several brain regions while adrenalectomy increased such binding. Region-specific effects were also obtained in regard to the dopaminergic system, with predominant actions of glucocorticoid-induced potentiation of reuptake blockers and releasing agents. More in-depth neuroanatomical analyses are warranted of these and amino acid-related neurotransmission.
Collapse
Affiliation(s)
- Robert Lalonde
- Laboratory of Stress, Immunity, Pathogens (UR SIMPA), University of Lorraine, Campus Santé, Bât A/B 9, avenue de la Forêt de Haye, Vandoeuvre-les-Nancy, 54500, France.
| | - Catherine Strazielle
- Laboratory of Stress, Immunity, Pathogens (UR SIMPA), University of Lorraine, Campus Santé, Bât A/B 9, avenue de la Forêt de Haye, Vandoeuvre-les-Nancy, 54500, France
- CHRU Nancy, Vandoeuvre-les-Nancy, France
| |
Collapse
|
9
|
Shao L, Kong F, Tian X, Deng T, Wang Y, Ji Y, Wang X, Yu H, Yuan F, Fu C, Wang S. Whole-brain inputs and outputs of Phox2b and GABAergic neurons in the nucleus tractus solitarii. Front Neurosci 2024; 18:1427384. [PMID: 38948926 PMCID: PMC11211284 DOI: 10.3389/fnins.2024.1427384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024] Open
Abstract
The nucleus tractus solitarii (NTS) plays a critical role in the homeostatic regulation of respiration, blood pressure, sodium consumption and metabolic processes. Despite their significance, the circuitry mechanisms facilitating these diverse physiological functions remain incompletely understood. In this study, we present a whole-brain mapping of both the afferent and efferent connections of Phox2b-expressing and GABAergic neurons within the NTS. Our findings reveal that these neuronal populations not only receive monosynaptic inputs primarily from the medulla oblongata, pons, midbrain, supra-midbrain and cortical areas, but also mutually project their axons to these same locales. Moreover, intense monosynaptic inputs are received from the central amygdala, the paraventricular nucleus of the hypothalamus, the parasubthalamic nucleus and the intermediate reticular nucleus, along with brainstem nuclei explicitly engaged in respiratory regulation. In contrast, both neuronal groups extensively innervate brainstem nuclei associated with respiratory functions, although their projections to regions above the midbrain are comparatively limited. These anatomical findings provide a foundational platform for delineating an anatomical framework essential for dissecting the specific functional mechanisms of these circuits.
Collapse
Affiliation(s)
- Liuqi Shao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fanrao Kong
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaochen Tian
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tianjiao Deng
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yakun Wang
- Department of Sleep Medicine, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yake Ji
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoyi Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hongxiao Yu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fang Yuan
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Congrui Fu
- Nursing School, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sheng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, Hebei, China
| |
Collapse
|
10
|
Buller S, Blouet C. Brain access of incretins and incretin receptor agonists to their central targets relevant for appetite suppression and weight loss. Am J Physiol Endocrinol Metab 2024; 326:E472-E480. [PMID: 38381398 PMCID: PMC11193531 DOI: 10.1152/ajpendo.00250.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/05/2024] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
New incretin-based pharmacotherapies provide efficient and safe therapeutic options to curb appetite and produce weight loss in patients with obesity. Delivered systemically, these molecules produce pleiotropic metabolic benefits, but the target sites mediating their weight-suppressive action are located within the brain. Recent research has increased our understanding of the neural circuits and behavioral mechanisms involved in the anorectic and metabolic consequences of glucagon-like peptide 1 (GLP-1)-based weight loss strategies, yet little is known about how these drugs access their functional targets in the brain to produce sustained weight loss. The majority of brain cells expressing incretin receptors are located behind the blood-brain barrier, shielded from the circulation and fluctuations in the availability of peripheral signals, which is a major challenge for the development of CNS-targeted therapeutic peptides. GLP-1 receptor (GLP-1R) agonists with increased half-life and enhanced therapeutic benefit do not cross the blood-brain barrier, yet they manage to access discrete brain sites relevant to the regulation of energy homeostasis. In this review, we give a brief overview of the different routes for peptide hormones to access the brain. We then examine the evidence informing the routes employed by incretins and incretin receptor agonists to access brain targets relevant for their appetite and weight-suppressive actions. We highlight existing controversies and suggest future directions to further establish the functionally relevant access routes for GLP-1-based weight loss compounds, which might guide the development and selection of the future generation of incretin receptor polypharmacologies.
Collapse
Affiliation(s)
- Sophie Buller
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Clemence Blouet
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
11
|
Adolf C, Murck H, Sarkis AL, Schneider H, Fischer I, Steiger A, Braun LT, Reincke M, Künzel H. Sleep disturbances in primary aldosteronism are associated to depressive symptoms - Could specific mineralocorticoidreceptors be a common pathway? J Psychiatr Res 2024; 172:66-70. [PMID: 38367319 DOI: 10.1016/j.jpsychires.2024.01.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/28/2023] [Accepted: 01/22/2024] [Indexed: 02/19/2024]
Abstract
Symptoms of depression and anxiety are frequent in patients with primary aldosteronism (PA) and are supposed to be independent risk factors for cardiovascular diseases (CVD). As patients with PA have an increased cardiovascular risk compared to patients with essential hypertension, sleep disturbances, which often accompany depressive and anxiety symptoms, may be an additional contributor to the cardiometabolic consequences of PA. To clarify this possible link we investigated 132 patients with PA at baseline and after one year after initiation of treatment either by adrenalectomy (ADX) or mineralocorticoid-receptor-antagonist (MRA). Sleep disturbances and daytime sleepiness were assessed with Pittsburg sleep Inventory (PSQI) and Epworth sleepiness scale (ESS). Patients with PA showed pathological scores for sleep disturbances at baseline according to PSQI, with females being more affected (8.1 vs. 5.7 p < 0.001), which was significantly improved after initiation of specific treatment (p = 0.002). For ESS we found scores within the normal range, but higher than the general population, which significantly improved at follow-up (p < 0.001). The intensity of sleep disturbances was highly correlated with scores of anxiety and depression at baseline and follow-up. However, clinical and biochemical markers of PA (e.g. aldosterone, blood pressure) and metabolic markers did not show a consistent association with sleep changes. The degree of improvement in PSQI was significantly associated with the improvement of brief patients health questionnaire (PHQD) (p = 0.0151). Sleep disturbances seem not to be an independent risk factor for cardiovascular and metabolic problems in PA. They are strongly associated to depressive symptoms and maybe mediated by the same mineralocorticoid receptor circuits.
Collapse
Affiliation(s)
- Christian Adolf
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Ziemssenstraße 1, 80336, Munich, Germany
| | - Harald Murck
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, Germany
| | - Anna-Lina Sarkis
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Ziemssenstraße 1, 80336, Munich, Germany
| | - Holger Schneider
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Ziemssenstraße 1, 80336, Munich, Germany
| | - Ina Fischer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Ziemssenstraße 1, 80336, Munich, Germany
| | - Axel Steiger
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Ziemssenstraße 1, 80336, Munich, Germany
| | - Leah T Braun
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Ziemssenstraße 1, 80336, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Ziemssenstraße 1, 80336, Munich, Germany
| | - Heike Künzel
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Ziemssenstraße 1, 80336, Munich, Germany.
| |
Collapse
|
12
|
Nardone S, De Luca R, Zito A, Klymko N, Nicoloutsopoulos D, Amsalem O, Brannigan C, Resch JM, Jacobs CL, Pant D, Veregge M, Srinivasan H, Grippo RM, Yang Z, Zeidel ML, Andermann ML, Harris KD, Tsai LT, Arrigoni E, Verstegen AMJ, Saper CB, Lowell BB. A spatially-resolved transcriptional atlas of the murine dorsal pons at single-cell resolution. Nat Commun 2024; 15:1966. [PMID: 38438345 PMCID: PMC10912765 DOI: 10.1038/s41467-024-45907-7] [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: 05/05/2023] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
The "dorsal pons", or "dorsal pontine tegmentum" (dPnTg), is part of the brainstem. It is a complex, densely packed region whose nuclei are involved in regulating many vital functions. Notable among them are the parabrachial nucleus, the Kölliker Fuse, the Barrington nucleus, the locus coeruleus, and the dorsal, laterodorsal, and ventral tegmental nuclei. In this study, we applied single-nucleus RNA-seq (snRNA-seq) to resolve neuronal subtypes based on their unique transcriptional profiles and then used multiplexed error robust fluorescence in situ hybridization (MERFISH) to map them spatially. We sampled ~1 million cells across the dPnTg and defined the spatial distribution of over 120 neuronal subtypes. Our analysis identified an unpredicted high transcriptional diversity in this region and pinpointed the unique marker genes of many neuronal subtypes. We also demonstrated that many neuronal subtypes are transcriptionally similar between humans and mice, enhancing this study's translational value. Finally, we developed a freely accessible, GPU and CPU-powered dashboard ( http://harvard.heavy.ai:6273/ ) that combines interactive visual analytics and hardware-accelerated SQL into a data science framework to allow the scientific community to query and gain insights into the data.
Collapse
Affiliation(s)
- Stefano Nardone
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Roberto De Luca
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Antonino Zito
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Nataliya Klymko
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA
| | | | - Oren Amsalem
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Cory Brannigan
- HEAVY.AI, 100 Montgomery St Fl 5, San Francisco, California, 94104, USA
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center. University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Christopher L Jacobs
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Deepti Pant
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Molly Veregge
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Harini Srinivasan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ryan M Grippo
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Zongfang Yang
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Mark L Zeidel
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA
| | - Mark L Andermann
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Kenneth D Harris
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Linus T Tsai
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elda Arrigoni
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Anne M J Verstegen
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA.
| | - Clifford B Saper
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA.
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Murck H, Karailiev P, Karailievova L, Puhova A, Jezova D. Treatment with Glycyrrhiza glabra Extract Induces Anxiolytic Effects Associated with Reduced Salt Preference and Changes in Barrier Protein Gene Expression. Nutrients 2024; 16:515. [PMID: 38398838 PMCID: PMC10893552 DOI: 10.3390/nu16040515] [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: 12/15/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
We have previously identified that low responsiveness to antidepressive therapy is associated with higher aldosterone/cortisol ratio, lower systolic blood pressure, and higher salt preference. Glycyrrhiza glabra (GG) contains glycyrrhizin, an inhibitor of 11β-hydroxysteroid-dehydrogenase type-2 and antagonist of toll-like receptor 4. The primary hypothesis of this study is that food enrichment with GG extract results in decreased anxiety behavior and reduced salt preference under stress and non-stress conditions. The secondary hypothesis is that the mentioned changes are associated with altered gene expression of barrier proteins in the prefrontal cortex. Male Sprague-Dawley rats were exposed to chronic mild stress for five weeks. Both stressed and unstressed rats were fed a diet with or without an extract of GG roots for the last two weeks. GG induced anxiolytic effects in animals independent of stress exposure, as measured in elevated plus maze test. Salt preference and intake were significantly reduced by GG under control, but not stress conditions. The gene expression of the barrier protein claudin-11 in the prefrontal cortex was increased in control rats exposed to GG, whereas stress-induced rise was prevented. Exposure to GG-enriched diet resulted in reduced ZO-1 expression irrespective of stress conditions. In conclusion, the observed effects of GG are in line with a reduction in the activity of central mineralocorticoid receptors. The treatment with GG extract or its active components may, therefore, be a useful adjunct therapy for patients with subtypes of depression and anxiety disorders with heightened renin-angiotensin-aldosterone system and/or inflammatory activity.
Collapse
Affiliation(s)
- Harald Murck
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, 35039 Marburg, Germany
| | - Peter Karailiev
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Lucia Karailievova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Agnesa Puhova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| |
Collapse
|
14
|
de Kloet ER, Joëls M. The cortisol switch between vulnerability and resilience. Mol Psychiatry 2024; 29:20-34. [PMID: 36599967 DOI: 10.1038/s41380-022-01934-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023]
Abstract
In concert with neuropeptides and transmitters, the end products of the hypothalamus-pituitary-adrenal (HPA) axis, the glucocorticoid hormones cortisol and corticosterone (CORT), promote resilience: i.e., the ability to cope with threats, adversity, and trauma. To exert this protective action, CORT activates mineralocorticoid receptors (MR) and glucocorticoid receptors (GR) that operate in a complementary manner -as an on/off switch- to coordinate circadian events, stress-coping, and adaptation. The evolutionary older limbic MR facilitates contextual memory retrieval and supports an on-switch in the selection of stress-coping styles at a low cost. The rise in circulating CORT concentration after stress subsequently activates a GR-mediated off-switch underlying recovery of homeostasis by providing the energy for restraining the primary stress reactions and promoting cognitive control over emotional reactivity. GR activation facilitates contextual memory storage of the experience to enable future stress-coping. Such complementary MR-GR-mediated actions involve rapid non-genomic and slower gene-mediated mechanisms; they are time-dependent, conditional, and sexually dimorphic, and depend on genetic background and prior experience. If coping fails, GR activation impairs cognitive control and promotes emotional arousal which eventually may compromise resilience. Such breakdown of resilience involves a transition to a chronic stress construct, where information processing is crashed; it leads to an imbalanced MR-GR switch and hence increased vulnerability. Novel MR-GR modulators are becoming available that may reset a dysregulated stress response system to reinstate the cognitive flexibility required for resilience.
Collapse
Affiliation(s)
- E Ronald de Kloet
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, The Netherlands.
- Leiden/Amsterdam Center of Drug Research, Leiden University, Leiden, The Netherlands.
| | - Marian Joëls
- Dept. Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
15
|
Aitken CM, Jaramillo JCM, Davis W, Brennan‐Xie L, McDougall SJ, Lawrence AJ, Ryan PJ. Feeding signals inhibit fluid-satiation signals in the mouse lateral parabrachial nucleus to increase intake of highly palatable, caloric solutions. J Neurochem 2023; 167:648-667. [PMID: 37855271 PMCID: PMC10952698 DOI: 10.1111/jnc.15991] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/20/2023]
Abstract
Chemogenetic activation of oxytocin receptor-expressing neurons in the parabrachial nucleus (OxtrPBN neurons) acts as a satiation signal for water. In this research, we investigated the effect of activating OxtrPBN neurons on satiation for different types of fluids. Chemogenetic activation of OxtrPBN neurons in male and female transgenic OxtrCre mice robustly suppressed the rapid, initial (15-min) intake of several solutions after dehydration: water, sucrose, ethanol and saccharin, but only slightly decreased intake of Ensure®, a highly caloric solution (1 kcal/mL; containing 3.72 g protein, 3.27 g fat, 13.42 g carbohydrates, and 1.01 g dietary fibre per 100 mL). OxtrPBN neuron activation also suppressed cumulative, longer-term (2-h) intake of lower caloric, less palatable solutions, but not highly caloric, palatable solutions. These results suggest that OxtrPBN neurons predominantly control initial fluid-satiation responses after rehydration, but not longer-term intake of highly caloric, palatable solutions. The suppression of fluid intake was not because of anxiogenesis, but because OxtrPBN neuron activation decreased anxiety-like behaviour. To investigate the role of different PBN subdivisions on the intake of different solutions, we examined FOS as a proxy marker of PBN neuron activation. Different PBN subdivisions were activated by different solutions: the dorsolateral PBN similarly by all fluids; the external lateral PBN by caloric but not non-caloric solutions; and the central lateral PBN primarily by highly palatable solutions, suggesting PBN subdivisions regulate different aspects of fluid intake. To explore the possible mechanisms underlying the minimal suppression of Ensure® after OxtrPBN neuron activation, we demonstrated in in vitro slice recordings that the feeding-associated agouti-related peptide (AgRP) inhibited OxtrPBN neuron firing in a concentration-related manner, suggesting possible inhibition by feeding-related neurocircuitry of fluid satiation neurocircuitry. Overall, this research suggests that although palatable beverages like sucrose- and ethanol-containing beverages activate fluid satiation signals encoded by OxtrPBN neurons, these neurons can be inhibited by hunger-related signals (agouti-related peptide, AgRP), which may explain why these fluids are often consumed in excess of what is required for fluid satiation.
Collapse
Affiliation(s)
- Connor M. Aitken
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Janine C. M. Jaramillo
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Warren Davis
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Liam Brennan‐Xie
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Stuart J. McDougall
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Andrew J. Lawrence
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Philip J. Ryan
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
16
|
Nardone S, De Luca R, Zito A, Klymko N, Nicoloutsopoulos D, Amsalem O, Brannigan C, Resch JM, Jacobs CL, Pant D, Veregge M, Srinivasan H, Grippo RM, Yang Z, Zeidel ML, Andermann ML, Harris KD, Tsai LT, Arrigoni E, Verstegen AMJ, Saper CB, Lowell BB. A spatially-resolved transcriptional atlas of the murine dorsal pons at single-cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558047. [PMID: 38014113 PMCID: PMC10680649 DOI: 10.1101/2023.09.18.558047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The "dorsal pons", or "dorsal pontine tegmentum" (dPnTg), is part of the brainstem. It is a complex, densely packed region whose nuclei are involved in regulating many vital functions. Notable among them are the parabrachial nucleus, the Kölliker Fuse, the Barrington nucleus, the locus coeruleus, and the dorsal, laterodorsal, and ventral tegmental nuclei. In this study, we applied single-nucleus RNA-seq (snRNA-seq) to resolve neuronal subtypes based on their unique transcriptional profiles and then used multiplexed error robust fluorescence in situ hybridization (MERFISH) to map them spatially. We sampled ~1 million cells across the dPnTg and defined the spatial distribution of over 120 neuronal subtypes. Our analysis identified an unpredicted high transcriptional diversity in this region and pinpointed many neuronal subtypes' unique marker genes. We also demonstrated that many neuronal subtypes are transcriptionally similar between humans and mice, enhancing this study's translational value. Finally, we developed a freely accessible, GPU and CPU-powered dashboard (http://harvard.heavy.ai:6273/) that combines interactive visual analytics and hardware-accelerated SQL into a data science framework to allow the scientific community to query and gain insights into the data.
Collapse
Affiliation(s)
- Stefano Nardone
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Roberto De Luca
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Antonino Zito
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Nataliya Klymko
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA 02215, USA
| | | | - Oren Amsalem
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Cory Brannigan
- HEAVY.AI, 100 Montgomery St Fl 5, San Francisco, California, 94104, USA
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center. University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Christopher L Jacobs
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Deepti Pant
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Molly Veregge
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Harini Srinivasan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ryan M Grippo
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Zongfang Yang
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Mark L Zeidel
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA 02215, USA
| | - Mark L Andermann
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Kenneth D Harris
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Linus T Tsai
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elda Arrigoni
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Anne M J Verstegen
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA 02215, USA
| | - Clifford B Saper
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Pince CL, Whiting KE, Wang T, Lékó AH, Farinelli LA, Cooper D, Farokhnia M, Vendruscolo LF, Leggio L. Role of aldosterone and mineralocorticoid receptor (MR) in addiction: A scoping review. Neurosci Biobehav Rev 2023; 154:105427. [PMID: 37858908 PMCID: PMC10865927 DOI: 10.1016/j.neubiorev.2023.105427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/24/2023] [Accepted: 10/13/2023] [Indexed: 10/21/2023]
Abstract
Preclinical and human studies suggest a role of aldosterone and mineralocorticoid receptor (MR) in addiction. This scoping review aimed to summarize (1) the relationship between alcohol and other substance use disorders (ASUDs) and dysfunctions of the aldosterone and MR, and (2) how pharmacological manipulations of MR may affect ASUD-related outcomes. Our search in four databases (MEDLINE, Embase, Web of Science, and Cochrane Library) indicated that most studies focused on the relationship between aldosterone, MR, and alcohol (n = 30), with the rest focused on opioids (n = 5), nicotine (n = 9), and other addictive substances (n = 9). Despite some inconsistencies, the overall results suggest peripheral and central dysregulations of aldosterone and MR in several species and that these dysregulations depended on the pattern of drug exposure and genetic factors. We conclude that MR antagonism may be a promising target in ASUD, yet future studies are warranted.
Collapse
Affiliation(s)
- Claire L Pince
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA; Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA; Stress & Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA
| | - Kimberly E Whiting
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA; Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA
| | - Tammy Wang
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA
| | - András H Lékó
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA; Center on Compulsive Behaviors, Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lisa A Farinelli
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA
| | - Diane Cooper
- Office of Research Services, Division of Library Services, National Institutes of Health, Building 10, Bethesda, MD 20892, USA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA
| | - Leandro F Vendruscolo
- Stress & Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA.
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA.
| |
Collapse
|
18
|
de Kloet ER. Glucocorticoid feedback paradox: a homage to Mary Dallman. Stress 2023; 26:2247090. [PMID: 37589046 DOI: 10.1080/10253890.2023.2247090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/07/2023] [Indexed: 08/18/2023] Open
Abstract
As the end product of the hypothalamus-pituitary-adrenal (HPA) axis, the glucocorticoid hormones cortisol and corticosterone coordinate circadian activities, stress-coping, and adaptation to change. For this purpose, the hormone promotes energy metabolism and controls defense reactions in the body and brain. This life-sustaining action exerted by glucocorticoids occurs in concert with the autonomic nervous and immune systems, transmitters, growth factors/cytokines, and neuropeptides. The current contribution will focus on the glucocorticoid feedback paradox in the HPA-axis: the phenomenon that stress responsivity remains resilient if preceded by stress-induced secretion of glucocorticoid hormone, but not if this hormone is previously administered. Furthermore, in animal studies, the mixed progesterone/glucocorticoid antagonist RU486 or mifepristone switches to an apparent partial agonist upon repeated administration. To address these enigmas several interesting phenomena are highlighted. These include the conditional nature of the excitation/inhibition balance in feedback regulation, the role of glucose as a determinant of stress responsivity, and the potential of glucocorticoids in resetting the stress response system. The analysis of the feedback paradox provides also a golden opportunity to review the progress in understanding the role of glucocorticoid hormone in resilience and vulnerability during stress, the science that was burned deeply in Mary Dallman's emotions.
Collapse
Affiliation(s)
- Edo Ronald de Kloet
- Department of Clinical Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
19
|
Jeanneteau F. Stress and the risk of Alzheimer dementia: Can deconstructed engrams be rebuilt? J Neuroendocrinol 2023; 35:e13235. [PMID: 36775895 DOI: 10.1111/jne.13235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
The exact neuropathological mechanism by which the dementia process unfolds is under intense scrutiny. The disease affects about 38 million people worldwide, 70% of which are clinically diagnosed with Alzheimer's disease (AD). If the destruction of synapses essential for learning, planning and decision-making is part of the problem, must the restoration of previously lost synapses be part of the solution? It is plausible that neuronal capacity to restitute information corresponds with the adaptive capacity of its connectivity reserve. A challenge will be to promote the functional connectivity that can compensate for the lost one. This will require better clarification of the remodeling of functional connectivity during the progression of AD dementia and its reversal upon experimental treatment. A major difficulty is to promote the neural pathways that are atrophied in AD dementia while suppressing others that are bolstered. Therapeutic strategies should aim at scaling functional connectivity to a just balance between the atrophic and hypertrophic systems. However, the exact factors that can help reach this objective are still unclear. Similarities between the effects of chronic stress and some neuropathological mechanisms underlying AD dementia support the idea that common components deserve prime attention as therapeutic targets.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
20
|
Zhang R, Huang D, Gasparini S, Geerling JC. Efferent projections of Nps-expressing neurons in the parabrachial region. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.13.553140. [PMID: 37645772 PMCID: PMC10462015 DOI: 10.1101/2023.08.13.553140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
In the brain, connectivity determines function. Neurons in the parabrachial nucleus (PB) relay diverse information to widespread brain regions, but the connections and functions of PB neurons that express Nps (neuropeptide S) remain mysterious. Here, we use Cre-dependent anterograde tracing and whole-brain analysis to map their output connections. While many other PB neurons project ascending axons through the central tegmental tract, NPS axons reach the forebrain via distinct periventricular and ventral pathways. Along the periventricular pathway, NPS axons target the tectal longitudinal column and periaqueductal gray then continue rostrally to target the paraventricular nucleus of the thalamus. Along the ventral pathway, NPS axons blanket much of the hypothalamus but avoid the ventromedial and mammillary nuclei. They also project prominently to the ventral bed nucleus of the stria terminalis, A13 cell group, and magnocellular subparafasciular nucleus. In the hindbrain, NPS axons have fewer descending projections, targeting primarily the superior salivatory nucleus, nucleus of the lateral lemniscus, and periolivary region. Combined with what is known about NPS and its receptor, the output pattern of Nps-expressing neurons in the PB region predicts a role in threat response and circadian behavior.
Collapse
Affiliation(s)
- Richie Zhang
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa
| | - Dake Huang
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa
| | - Silvia Gasparini
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa
| | - Joel C. Geerling
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa
| |
Collapse
|
21
|
Peltekian L, Gasparini S, Fazan FS, Karthik S, Iverson G, Resch JM, Geerling JC. Sodium appetite and thirst do not require angiotensinogen production in astrocytes or hepatocytes. J Physiol 2023; 601:3499-3532. [PMID: 37291801 DOI: 10.1113/jp283169] [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: 12/28/2022] [Accepted: 06/02/2023] [Indexed: 06/10/2023] Open
Abstract
In addition to its renal and cardiovascular functions, angiotensin signalling is thought to be responsible for the increases in salt and water intake caused by hypovolaemia. However, it remains unclear whether these behaviours require angiotensin production in the brain or liver. Here, we use in situ hybridization to identify tissue-specific expression of the genes required for producing angiotensin peptides, and then use conditional genetic deletion of the angiotensinogen gene (Agt) to test whether production in the brain or liver is necessary for sodium appetite and thirst. In the mouse brain, we identified expression of Agt (the precursor for all angiotensin peptides) in a large subset of astrocytes. We also identified Ren1 and Ace (encoding enzymes required to produce angiotensin II) expression in the choroid plexus, and Ren1 expression in neurons within the nucleus ambiguus compact formation. In the liver, we confirmed that Agt is widely expressed in hepatocytes. We next tested whether thirst and sodium appetite require angiotensinogen production in astrocytes or hepatocytes. Despite virtually eliminating expression in the brain, deleting astrocytic Agt did not reduce thirst or sodium appetite. Despite markedly reducing angiotensinogen in the blood, eliminating Agt from hepatocytes did not reduce thirst or sodium appetite, and in fact, these mice consumed the largest amounts of salt and water after sodium deprivation. Deleting Agt from both astrocytes and hepatocytes also did not prevent thirst or sodium appetite. Our findings suggest that angiotensin signalling is not required for sodium appetite or thirst and highlight the need to identify alternative signalling mechanisms. KEY POINTS: Angiotensin signalling is thought to be responsible for the increased thirst and sodium appetite caused by hypovolaemia, producing elevated water and sodium intake. Specific cells in separate brain regions express the three genes needed to produce angiotensin peptides, but brain-specific deletion of the angiotensinogen gene (Agt), which encodes the lone precursor for all angiotensin peptides, did not reduce thirst or sodium appetite. Double-deletion of Agt from brain and liver also did not reduce thirst or sodium appetite. Liver-specific deletion of Agt reduced circulating angiotensinogen levels without reducing thirst or sodium appetite. Instead, these angiotensin-deficient mice exhibited an enhanced sodium appetite. Because the physiological mechanisms controlling thirst and sodium appetite continued functioning without angiotensin production in the brain and liver, understanding these mechanisms requires a renewed search for the hypovolaemic signals necessary for activating each behaviour.
Collapse
Affiliation(s)
- Lila Peltekian
- Department of Neurology, University of Iowa, Iowa City, IA, USA
| | | | | | | | | | - Jon M Resch
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Joel C Geerling
- Department of Neurology, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
22
|
Jun S, Ou X, Shi L, Yu H, Deng T, Chen J, Nie X, Hao Y, Shi Y, Liu W, Tian Y, Wang S, Yuan F. Circuit-Specific Control of Blood Pressure by PNMT-Expressing Nucleus Tractus Solitarii Neurons. Neurosci Bull 2023; 39:1193-1209. [PMID: 36588135 PMCID: PMC10387028 DOI: 10.1007/s12264-022-01008-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/18/2022] [Indexed: 01/03/2023] Open
Abstract
The nucleus tractus solitarii (NTS) is one of the morphologically and functionally defined centers that engage in the autonomic regulation of cardiovascular activity. Phenotypically-characterized NTS neurons have been implicated in the differential regulation of blood pressure (BP). Here, we investigated whether phenylethanolamine N-methyltransferase (PNMT)-expressing NTS (NTSPNMT) neurons contribute to the control of BP. We demonstrate that photostimulation of NTSPNMT neurons has variable effects on BP. A depressor response was produced during optogenetic stimulation of NTSPNMT neurons projecting to the paraventricular nucleus of the hypothalamus, lateral parabrachial nucleus, and caudal ventrolateral medulla. Conversely, photostimulation of NTSPNMT neurons projecting to the rostral ventrolateral medulla produced a robust pressor response and bradycardia. In addition, genetic ablation of both NTSPNMT neurons and those projecting to the rostral ventrolateral medulla impaired the arterial baroreflex. Overall, we revealed the neuronal phenotype- and circuit-specific mechanisms underlying the contribution of NTSPNMT neurons to the regulation of BP.
Collapse
Affiliation(s)
- Shirui Jun
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xianhong Ou
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Luo Shi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hongxiao Yu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Tianjiao Deng
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jinting Chen
- Core Facilities and Centers, Institute of Medicine and Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiaojun Nie
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yinchao Hao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yishuo Shi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Wei Liu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yanming Tian
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Sheng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China.
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, 050017, China.
| | - Fang Yuan
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China.
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, 050017, China.
| |
Collapse
|
23
|
Yang H, Narayan S, Schmidt MV. From Ligands to Behavioral Outcomes: Understanding the Role of Mineralocorticoid Receptors in Brain Function. Stress 2023; 26:2204366. [PMID: 37067948 DOI: 10.1080/10253890.2023.2204366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/18/2023] Open
Abstract
Stress is a normal response to situational pressures or demands. Exposure to stress activates the hypothalamic-pituitary-adrenal (HPA) axis and leads to the release of corticosteroids, which act in the brain via two distinct receptors: mineralocorticoid receptors (MR) and glucocorticoid receptors (GR). Persistent HPA axis overactivation or dysregulation can disrupt an individual's homeostasis, thereby contributing to an increased risk for mental illness. On the other hand, successful coping with stressful events involves adaptive and cognitive processes in the brain that render individuals more resilient to similar stressors in the future. Here we review the role of the MR in these processes, starting with an overview of the physiological structure, ligand binding, and expression of MR, and further summarizing its role in the brain, its relevance to psychiatric disorders, and related rodent studies. Given the central role of MR in cognitive and emotional functioning, and its importance as a target for promoting resilience, future research should investigate how MR modulation can be used to alleviate disturbances in emotion and behavior, as well as cognitive impairment, in patients with stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Huanqing Yang
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Sowmya Narayan
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Department Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
24
|
Dos Santos NL, Lenert ME, Castillo ZW, Mody PH, Thompson LT, Burton MD. Age and sex drive differential behavioral and neuroimmune phenotypes during postoperative pain. Neurobiol Aging 2023; 123:129-144. [PMID: 36577640 PMCID: PMC9892227 DOI: 10.1016/j.neurobiolaging.2022.09.008] [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: 03/12/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022]
Abstract
Surgical procedures in the geriatric population are steadily increasing, driven by improved healthcare technologies and longer lifespans. However, effective postoperative pain treatments are lacking, and this diminishes quality of life and recovery. Here we present one of the first preclinical studies to pursue sex- and age-specific differences in postoperative neuroimmune phenotypes and pain. We found that aged males, but not females, had a delayed onset of mechanical hypersensitivity post-surgery and faster resolution than young counterparts. This sex-specific age effect was accompanied by decreased paw innervation and increased local inflammation. Additionally, we find evidence of an age-dependent decrease in hyperalgesic priming and perioperative changes in nociceptor populations and spinal microglia in the aged. These findings suggest that impaired neuronal function and maladaptive inflammatory mechanisms influence postoperative pain development in advanced age. Elucidation of these neuroimmune phenotypes across age and sex enables the development of novel therapies that can be tailored for improved pain relief.
Collapse
Affiliation(s)
- Natalia L Dos Santos
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX, USA
| | - Melissa E Lenert
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX, USA
| | - Zachary W Castillo
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX, USA
| | - Prapti H Mody
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX, USA
| | - Lucien T Thompson
- Aging and Memory Research Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson TX, USA
| | - Michael D Burton
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX, USA.
| |
Collapse
|
25
|
Deng Y, Dickey JE, Saito K, Deng G, Singh U, Jiang J, Toth BA, Zhu Z, Zingman LV, Resch JM, Grobe JL, Cui H. Elucidating the role of Rgs2 expression in the PVN for metabolic homeostasis in mice. Mol Metab 2022; 66:101622. [PMID: 36307046 PMCID: PMC9638802 DOI: 10.1016/j.molmet.2022.101622] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/09/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE RGS2 is a GTPase activating protein that modulates GPCR-Gα signaling and mice lacking RGS2 globally exhibit metabolic alterations. While RGS2 is known to be broadly expressed throughout the body including the brain, the relative contribution of brain RGS2 to metabolic homeostasis remains unknown. The purpose of this study was to characterize RGS2 expression in the paraventricular nucleus of hypothalamus (PVN) and test its role in metabolic homeostasis. METHODS We used a combination of RNAscope in situ hybridization (ISH), immunohistochemistry, and bioinformatic analyses to characterize the pattern of Rgs2 expression in the PVN. We then created mice lacking Rgs2 either prenatally or postnatally in the PVN and evaluated their metabolic consequences. RESULTS RNAscope ISH analysis revealed a broad but regionally enriched Rgs2 mRNA expression throughout the mouse brain, with the highest expression being observed in the PVN along with several other brain regions, such as the arcuate nucleus of hypothalamus and the dorsal raphe nucleus. Within the PVN, we found that Rgs2 is specifically enriched in CRH+ endocrine neurons and is further increased by calorie restriction. Functionally, although Sim1-Cre-mediated prenatal deletion of Rgs2 in PVN neurons had no major effects on metabolic homeostasis, AAV-mediated adult deletion of Rgs2 in the PVN led to significantly increased food intake, body weight (both fat and fat-free masses), body length, and blood glucose levels in both male and female mice. Strikingly, we found that prolonged postnatal loss of Rgs2 leads to neuronal cell death in the PVN, while rapid body weight gain in the early phase of viral-mediated PVN Rgs2 deletion is independent of PVN neuronal loss. CONCLUSIONS Our results provide the first evidence to show that PVN Rgs2 expression is not only sensitive to metabolic challenge but also critically required for PVN endocrine neurons to function and maintain metabolic homeostasis.
Collapse
Affiliation(s)
- Yue Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jacob E Dickey
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Kenji Saito
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Uday Singh
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jingwei Jiang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Brandon A Toth
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Zhiyong Zhu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Leonid V Zingman
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States; F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States; F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States.
| |
Collapse
|
26
|
Arias A, Manubens-Gil L, Dierssen M. Fluorescent transgenic mouse models for whole-brain imaging in health and disease. Front Mol Neurosci 2022; 15:958222. [PMID: 36211979 PMCID: PMC9538927 DOI: 10.3389/fnmol.2022.958222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022] Open
Abstract
A paradigm shift is occurring in neuroscience and in general in life sciences converting biomedical research from a descriptive discipline into a quantitative, predictive, actionable science. Living systems are becoming amenable to quantitative description, with profound consequences for our ability to predict biological phenomena. New experimental tools such as tissue clearing, whole-brain imaging, and genetic engineering technologies have opened the opportunity to embrace this new paradigm, allowing to extract anatomical features such as cell number, their full morphology, and even their structural connectivity. These tools will also allow the exploration of new features such as their geometrical arrangement, within and across brain regions. This would be especially important to better characterize brain function and pathological alterations in neurological, neurodevelopmental, and neurodegenerative disorders. New animal models for mapping fluorescent protein-expressing neurons and axon pathways in adult mice are key to this aim. As a result of both developments, relevant cell populations with endogenous fluorescence signals can be comprehensively and quantitatively mapped to whole-brain images acquired at submicron resolution. However, they present intrinsic limitations: weak fluorescent signals, unequal signal strength across the same cell type, lack of specificity of fluorescent labels, overlapping signals in cell types with dense labeling, or undetectable signal at distal parts of the neurons, among others. In this review, we discuss the recent advances in the development of fluorescent transgenic mouse models that overcome to some extent the technical and conceptual limitations and tradeoffs between different strategies. We also discuss the potential use of these strains for understanding disease.
Collapse
Affiliation(s)
- Adrian Arias
- Department of System Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Linus Manubens-Gil
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Mara Dierssen
- Department of System Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| |
Collapse
|
27
|
Chemogenetic inhibition of Phox2-expressing neurons in the commissural NTS decreases blood pressure in anesthetized spontaneously hypertensive rats. Neurosci Lett 2022; 787:136817. [PMID: 35905886 DOI: 10.1016/j.neulet.2022.136817] [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: 03/27/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 11/21/2022]
Abstract
Interruption of the activity of neurons in the commissural portion of the nucleus of the solitary tract (cNTS) decreases blood pressure (BP) in experimental models of hypertension, such as the spontaneously hypertensive (SH) rat. To examine whether PHOX2B expressing cNTS neurons are involved in maintaining the elevated BP, we used replication-deficient viruses with a modified Phox2 binding site promoter to express the inhibitory chemogenetic allatostatin receptor or green fluorescent protein in the cNTS. Following administration of allatostatin, we observed a depressor and bradycardic response in anesthetized SH rats that expressed the allatostatin receptor. Injection of allatostatin did not affect BP or heart rate (HR) in control SH rats expressing green fluorescent protein in the cNTS. Immunohistochemistry showed that the majority of transduced cNTS neurons were PHOX2B-immunoreactive and some also expressed tyrosine hydroxylase. We conclude that in anesthetized SH rat, the Phox2B expressing cNTS neurons maintain elevated BP.
Collapse
|
28
|
de Kloet ER. Brain mineralocorticoid and glucocorticoid receptor balance in neuroendocrine regulation and stress-related psychiatric etiopathologies. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 24:100352. [PMID: 38037568 PMCID: PMC10687720 DOI: 10.1016/j.coemr.2022.100352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Cortisol and corticosterone (CORT) coordinate circadian events and manage the stress response by differential activation of two complementary brain receptor systems, i.e., the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR), which mediate rapid non-genomic and slow genomic actions. Several recent discoveries are highlighted from molecular fine-tuning of the MR/GR balance by FKBP5 to CORTs role in neural network regulation underlying stress adaptation in emotional, cognitive, and social domains of behavior. The data suggest that MR mediates CORT action on risk assessment, social interaction, and response selection, while GR activation promotes memory consolidation and behavioral adaptation; there are also sex differences in CORT action. New evidence suggests that targeting the MR/GR balance resets a dysregulated stress response system and promotes resilience.
Collapse
Affiliation(s)
- Edo Ronald de Kloet
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, University of Leiden, Leiden, the Netherlands
| |
Collapse
|
29
|
Daskalakis NP, Meijer OC, de Kloet ER. Mineralocorticoid receptor and glucocorticoid receptor work alone and together in cell-type-specific manner: Implications for resilience prediction and targeted therapy. Neurobiol Stress 2022; 18:100455. [PMID: 35601687 PMCID: PMC9118500 DOI: 10.1016/j.ynstr.2022.100455] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/30/2022] [Accepted: 04/19/2022] [Indexed: 12/24/2022] Open
Abstract
'You can't roll the clock back and reverse the effects of experiences' Bruce McEwen used to say when explaining how allostasis labels the adaptive process. Here we will for once roll the clock back to the times that the science of the glucocorticoid hormone was honored with a Nobel prize and highlight the discovery of their receptors in the hippocampus as inroad to its current status as master regulator in control of stress coping and adaptation. Glucocorticoids operate in concert with numerous neurotransmitters, neuropeptides, and other hormones with the aim to facilitate processing of information in the neurocircuitry of stress, from anticipation and perception of a novel experience to behavioral adaptation and memory storage. This action, exerted by the glucocorticoids, is guided by two complementary receptor systems, mineralocorticoid receptors (MR) and glucocorticoid receptors (GR), that need to be balanced for a healthy stress response pattern. Here we discuss the cellular, neuroendocrine, and behavioral studies underlying the MR:GR balance concept, highlight the relevance of hypothalamic-pituitary-adrenal (HPA) -axis patterns and note the limited understanding yet of sexual dimorphism in glucocorticoid actions. We conclude with the prospect that (i) genetically and epigenetically regulated receptor variants dictate cell-type-specific transcriptome signatures of stress-related neuropsychiatric symptoms and (ii) selective receptor modulators are becoming available for more targeted treatment. These two new developments may help to 'restart the clock' with the prospect to support resilience.
Collapse
Affiliation(s)
| | - Onno C. Meijer
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - E. Ron de Kloet
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
30
|
Karthik S, Huang D, Delgado Y, Laing JJ, Peltekian L, Iverson GN, Grady F, Miller RL, McCann CM, Fritzsch B, Iskusnykh IY, Chizhikov VV, Geerling JC. Molecular ontology of the parabrachial nucleus. J Comp Neurol 2022; 530:1658-1699. [PMID: 35134251 PMCID: PMC9119955 DOI: 10.1002/cne.25307] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 11/07/2022]
Abstract
This article has been removed because of a technical problem in the rendering of the PDF. 11 February 2022.
Collapse
Affiliation(s)
| | - Dake Huang
- Department of NeurologyUniversity of IowaIowa CityIowaUSA
| | | | | | - Lila Peltekian
- Department of NeurologyUniversity of IowaIowa CityIowaUSA
| | | | - Fillan Grady
- Department of NeurologyUniversity of IowaIowa CityIowaUSA
| | - Rebecca L. Miller
- Department of Anatomy and NeurobiologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Corey M. McCann
- Department of Anatomy and NeurobiologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Bernd Fritzsch
- Iowa Neuroscience InstituteIowa CityIowaUSA
- Department of BiologyUniversity of IowaIowa CityIowaUSA
| | - Igor Y. Iskusnykh
- Department of Anatomy and NeurobiologyUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Victor V. Chizhikov
- Department of Anatomy and NeurobiologyUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Joel C. Geerling
- Department of NeurologyUniversity of IowaIowa CityIowaUSA
- Iowa Neuroscience InstituteIowa CityIowaUSA
| |
Collapse
|
31
|
Hirsch D, Kohl A, Wang Y, Sela-Donenfeld D. Axonal Projection Patterns of the Dorsal Interneuron Populations in the Embryonic Hindbrain. Front Neuroanat 2022; 15:793161. [PMID: 35002640 PMCID: PMC8738170 DOI: 10.3389/fnana.2021.793161] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Unraveling the inner workings of neural circuits entails understanding the cellular origin and axonal pathfinding of various neuronal groups during development. In the embryonic hindbrain, different subtypes of dorsal interneurons (dINs) evolve along the dorsal-ventral (DV) axis of rhombomeres and are imperative for the assembly of central brainstem circuits. dINs are divided into two classes, class A and class B, each containing four neuronal subgroups (dA1-4 and dB1-4) that are born in well-defined DV positions. While all interneurons belonging to class A express the transcription factor Olig3 and become excitatory, all class B interneurons express the transcription factor Lbx1 but are diverse in their excitatory or inhibitory fate. Moreover, within every class, each interneuron subtype displays its own specification genes and axonal projection patterns which are required to govern the stage-by-stage assembly of their connectivity toward their target sites. Remarkably, despite the similar genetic landmark of each dINs subgroup along the anterior-posterior (AP) axis of the hindbrain, genetic fate maps of some dA/dB neuronal subtypes uncovered their contribution to different nuclei centers in relation to their rhombomeric origin. Thus, DV and AP positional information has to be orchestrated in each dA/dB subpopulation to form distinct neuronal circuits in the hindbrain. Over the span of several decades, different axonal routes have been well-documented to dynamically emerge and grow throughout the hindbrain DV and AP positions. Yet, the genetic link between these distinct axonal bundles and their neuronal origin is not fully clear. In this study, we reviewed the available data regarding the association between the specification of early-born dorsal interneuron subpopulations in the hindbrain and their axonal circuitry development and fate, as well as the present existing knowledge on molecular effectors underlying the process of axonal growth.
Collapse
Affiliation(s)
- Dana Hirsch
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.,Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, College of Medicine, Florida State University, Tallahassee, FL, United States
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
32
|
Signal Transduction of Mineralocorticoid and Angiotensin II Receptors in the Central Control of Sodium Appetite: A Narrative Review. Int J Mol Sci 2021; 22:ijms222111735. [PMID: 34769164 PMCID: PMC8584094 DOI: 10.3390/ijms222111735] [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] [Received: 09/29/2021] [Revised: 10/16/2021] [Accepted: 10/25/2021] [Indexed: 11/17/2022] Open
Abstract
Sodium appetite is an innate behavior occurring in response to sodium depletion that induces homeostatic responses such as the secretion of the mineralocorticoid hormone aldosterone from the zona glomerulosa of the adrenal cortex and the stimulation of the peptide hormone angiotensin II (ANG II). The synergistic action of these hormones signals to the brain the sodium appetite that represents the increased palatability for salt intake. This narrative review summarizes the main data dealing with the role of mineralocorticoid and ANG II receptors in the central control of sodium appetite. Appropriate keywords and MeSH terms were identified and searched in PubMed. References to original articles and reviews were examined, selected, and discussed. Several brain areas control sodium appetite, including the nucleus of the solitary tract, which contains aldosterone-sensitive HSD2 neurons, and the organum vasculosum lamina terminalis (OVLT) that contains ANG II-sensitive neurons. Furthermore, sodium appetite is under the control of signaling proteins such as mitogen-activated protein kinase (MAPK) and inositol 1,4,5-thriphosphate (IP3). ANG II stimulates salt intake via MAPK, while combined ANG II and aldosterone action induce sodium intake via the IP3 signaling pathway. Finally, aldosterone and ANG II stimulate OVLT neurons and suppress oxytocin secretion inhibiting the neuronal activity of the paraventricular nucleus, thus disinhibiting the OVLT activity to aldosterone and ANG II stimulation.
Collapse
|
33
|
Mietlicki-Baase EG, Santollo J, Daniels D. Fluid intake, what's dopamine got to do with it? Physiol Behav 2021; 236:113418. [PMID: 33838203 DOI: 10.1016/j.physbeh.2021.113418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/12/2021] [Accepted: 04/05/2021] [Indexed: 12/27/2022]
Abstract
Maintaining fluid balance is critical for life. The central components that control fluid intake are only partly understood. This contribution to the collection of papers highlighting work by members of the Society for the Study of Ingestive Behavior focuses on the role that dopamine has on fluid intake and describes the roles that various bioregulators can have on thirst and sodium appetite by influencing dopamine systems in the brain. The goal of the review is to highlight areas in need of more research and to propose a framework to guide that research. We hope that this framework will inspire researchers in the field to investigate these interesting questions in order to form a more complete understanding of how fluid intake is controlled.
Collapse
Affiliation(s)
- Elizabeth G Mietlicki-Baase
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, United States; Center for Ingestive Behavior Research, University at Buffalo, State University of New York, Buffalo, NY 14260, United States
| | - Jessica Santollo
- Department of Biology, University of Kentucky, Lexington, KY 40506, United States
| | - Derek Daniels
- Center for Ingestive Behavior Research, University at Buffalo, State University of New York, Buffalo, NY 14260, United States; Department of Psychology, University at Buffalo, State University of New York, Buffalo, NY 14260, United States
| |
Collapse
|
34
|
Lucera GM, Menani JV, Colombari E, Colombari DSA. ANG II and Aldosterone Acting Centrally Participate in the Enhanced Sodium Intake in Water-Deprived Renovascular Hypertensive Rats. Front Pharmacol 2021; 12:679985. [PMID: 34113255 PMCID: PMC8186501 DOI: 10.3389/fphar.2021.679985] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/04/2021] [Indexed: 11/29/2022] Open
Abstract
Renovascular hypertension is a type of secondary hypertension caused by renal artery stenosis, leading to an increase in the renin–angiotensin–aldosterone system (RAAS). Two-kidney, 1-clip (2K1C) is a model of renovascular hypertension in which rats have an increased sodium intake induced by water deprivation (WD), a common situation found in the nature. In addition, a high-sodium diet in 2K1C rats induces glomerular lesion. Therefore, the purpose of this study was to investigate whether angiotensin II (ANG II) and/or aldosterone participates in the increased sodium intake in 2K1C rats under WD. In addition, we also verified if central AT1 and mineralocorticoid receptor blockade would change the high levels of arterial pressure in water-replete (WR) and WD 2K1C rats, because blood pressure changes can facilitate or inhibit water and sodium intake. Finally, possible central areas activated during WD or WD followed by partial rehydration (PR) in 2K1C rats were also investigated. Male Holtzman rats (150–180 g) received a silver clip around the left renal artery to induce renovascular hypertension. Six weeks after renal surgery, a stainless-steel cannula was implanted in the lateral ventricle, followed by 5–7 days of recovery before starting tests. Losartan (AT1 receptor antagonist) injected intracerebroventricularly attenuated water intake during the thirst test. Either icv losartan or RU28318 (mineralocorticoid receptor antagonist) reduced 0.3 M NaCl intake, whereas the combination of losartan and RU28318 icv totally blocked 0.3 M NaCl intake induced by WD in 2K1C rats. Losartan and RU28318 icv did not change hypertension levels of normohydrated 2K1C rats, but reduced the increase in mean arterial pressure (MAP) produced by WD. c-Fos expression increased in the lamina terminalis and in the NTS in WD condition, and increased even more after WD-PR. These results suggest the participation of ANG II and aldosterone acting centrally in the enhanced sodium intake in WD 2K1C rats, and not in the maintenance of hypertension in satiated and fluid-replete 2K1C rats.
Collapse
Affiliation(s)
- Gabriela Maria Lucera
- Department of Physiology and Pathology, School of Dentistry, Sao Paulo State University, Araraquara, Brazil
| | - José Vanderlei Menani
- Department of Physiology and Pathology, School of Dentistry, Sao Paulo State University, Araraquara, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, Sao Paulo State University, Araraquara, Brazil
| | | |
Collapse
|
35
|
Gasparini S, Resch JM, Gore AM, Peltekian L, Geerling JC. Pre-locus coeruleus neurons in rat and mouse. Am J Physiol Regul Integr Comp Physiol 2021; 320:R342-R361. [PMID: 33296280 PMCID: PMC7988775 DOI: 10.1152/ajpregu.00261.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022]
Abstract
Previously, we identified a population of neurons in the hindbrain tegmentum, bordering the locus coeruleus (LC). We named this population the pre-locus coeruleus (pre-LC) because in rats its neurons lie immediately rostral to the LC. In mice, however, pre-LC and LC neurons intermingle, making them difficult to distinguish. Here, we use molecular markers and anterograde tracing to clarify the location and distribution of pre-LC neurons in mice, relative to rats. First, we colocalized the transcription factor FoxP2 with the activity marker Fos to identify pre-LC neurons in sodium-deprived rats and show their distribution relative to surrounding catecholaminergic and cholinergic neurons. Next, we used sodium depletion and chemogenetic activation of the aldosterone-sensitive HSD2 neurons in the nucleus of the solitary tract (NTS) to identify the homologous population of pre-LC neurons in mice, along with a related population in the central lateral parabrachial nucleus. Using Cre-reporter mice for Pdyn, we confirmed that most of these sodium-depletion-activated neurons are dynorphinergic. Finally, after confirming that these neurons receive excitatory input from the NTS and paraventricular hypothalamic nucleus, plus convergent input from the inhibitory AgRP neurons in the arcuate hypothalamic nucleus, we identify a major, direct input projection from the medial prefrontal cortex. This new information on the location, distribution, and input to pre-LC neurons provides a neuroanatomical foundation for cell-type-specific investigation of their properties and functions in mice. Pre-LC neurons likely integrate homeostatic information from the brainstem and hypothalamus with limbic, contextual information from the cerebral cortex to influence ingestive behavior.
Collapse
Affiliation(s)
- Silvia Gasparini
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Jon M Resch
- Department of Medicine, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | - Anuradha M Gore
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Lila Peltekian
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Joel C Geerling
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| |
Collapse
|
36
|
Fazan FS, Colombari E, Loewy AD, Geerling JC. Despite increasing aldosterone, elevated potassium is not necessary for activating aldosterone-sensitive HSD2 neurons or sodium appetite. Physiol Rep 2021; 9:e14714. [PMID: 33463885 PMCID: PMC7814482 DOI: 10.14814/phy2.14714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 12/15/2020] [Indexed: 11/24/2022] Open
Abstract
Restricting dietary sodium promotes sodium appetite in rats. Prolonged sodium restriction increases plasma potassium (pK), and elevated pK is largely responsible for a concurrent increase in aldosterone, which helps promote sodium appetite. In addition to increasing aldosterone, we hypothesized that elevated potassium directly influences the brain to promote sodium appetite. To test this, we restricted dietary potassium in sodium‐deprived rats. Potassium restriction reduced pK and blunted the increase in aldosterone caused by sodium deprivation, but did not prevent sodium appetite or the activation of aldosterone‐sensitive HSD2 neurons. Conversely, supplementing potassium in sodium‐deprived rats increased pK and aldosterone, but did not increase sodium appetite or the activation of HSD2 neurons relative to potassium restriction. Supplementing potassium without sodium deprivation did not significantly increase aldosterone and HSD2 neuronal activation and only modestly increased saline intake. Overall, restricting dietary sodium activated the HSD2 neurons and promoted sodium appetite across a wide range of pK and aldosterone, and saline consumption inactivated the HSD2 neurons despite persistent hyperaldosteronism. In conclusion, elevated potassium is important for increasing aldosterone, but it is neither necessary nor sufficient for activating HSD2 neurons and increasing sodium appetite.
Collapse
Affiliation(s)
- Frederico S Fazan
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA.,Department of Physiology and Pathology, São Paulo State University, Araraquara, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, São Paulo State University, Araraquara, Brazil
| | - Arthur D Loewy
- Department of Neuroscience, Washington University School of Medicine in Saint Louis, St Louis, Missouri, USA
| | - Joel C Geerling
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa Hospital and Clinics, Iowa City, Iowa, USA
| |
Collapse
|
37
|
Huang D, Grady FS, Peltekian L, Geerling JC. Efferent projections of Vglut2, Foxp2, and Pdyn parabrachial neurons in mice. J Comp Neurol 2020; 529:657-693. [PMID: 32621762 DOI: 10.1002/cne.24975] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/16/2022]
Abstract
The parabrachial nucleus (PB) is a complex structure located at the junction of the midbrain and hindbrain. Its neurons have diverse genetic profiles and influence a variety of homeostatic functions. While its cytoarchitecture and overall efferent projections are known, we lack comprehensive information on the projection patterns of specific neuronal subtypes in the PB. In this study, we compared the projection patterns of glutamatergic neurons here with a subpopulation expressing the transcription factor Foxp2 and a further subpopulation expressing the neuropeptide Pdyn. To do this, we injected an AAV into the PB region to deliver a Cre-dependent anterograde tracer (synaptophysin-mCherry) in three different strains of Cre-driver mice. We then analyzed 147 neuroanatomical regions for labeled boutons in every brain (n = 11). Overall, glutamatergic neurons in the PB region project to a wide variety of sites in the cerebral cortex, basal forebrain, bed nucleus of the stria terminalis, amygdala, diencephalon, and brainstem. Foxp2 and Pdyn subpopulations project heavily to the hypothalamus, but not to the cortex, basal forebrain, or amygdala. Among the few differences between Foxp2 and Pdyn cases was a notable lack of Pdyn projections to the ventromedial hypothalamic nucleus. Our results indicate that genetic identity determines connectivity (and therefore, function), providing a framework for mapping all PB output projections based on the genetic identity of its neurons. Using genetic markers to systematically classify PB neurons and their efferent projections will enhance the translation of research findings from experimental animals to humans.
Collapse
Affiliation(s)
- Dake Huang
- Department of Neurology, University of Iowa, Iowa City, Iowa, USA
| | - Fillan S Grady
- Department of Neurology, University of Iowa, Iowa City, Iowa, USA
| | - Lila Peltekian
- Department of Neurology, University of Iowa, Iowa City, Iowa, USA
| | - Joel C Geerling
- Department of Neurology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
38
|
Gasparini S, Howland JM, Thatcher AJ, Geerling JC. Central afferents to the nucleus of the solitary tract in rats and mice. J Comp Neurol 2020; 528:2708-2728. [PMID: 32307700 DOI: 10.1002/cne.24927] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/13/2022]
Abstract
The nucleus of the solitary tract (NTS) regulates life-sustaining functions ranging from appetite and digestion to heart rate and breathing. It is also the brain's primary sensory nucleus for visceral sensations relevant to symptoms in medical and psychiatric disorders. To better understand which neurons may exert top-down control over the NTS, here we provide a brain-wide map of all neurons that project axons directly to the caudal, viscerosensory NTS, focusing on a medial subregion with aldosterone-sensitive HSD2 neurons. Injecting an axonal tracer (cholera toxin b) into the NTS produces a similar pattern of retrograde labeling in rats and mice. The paraventricular hypothalamic nucleus (PVH), lateral hypothalamic area, and central nucleus of the amygdala (CeA) contain the densest concentrations of NTS-projecting neurons. PVH afferents are glutamatergic (express Slc17a6/Vglut2) and are distinct from neuroendocrine PVH neurons. CeA afferents are GABAergic (express Slc32a1/Vgat) and are distributed largely in the medial CeA subdivision. Other retrogradely labeled neurons are located in a variety of brain regions, including the cerebral cortex (insular and infralimbic areas), bed nucleus of the stria terminalis, periaqueductal gray, Barrington's nucleus, Kölliker-Fuse nucleus, hindbrain reticular formation, and rostral NTS. Similar patterns of retrograde labeling result from tracer injections into different NTS subdivisions, with dual retrograde tracing revealing that many afferent neurons project axon collaterals to both the lateral and medial NTS subdivisions. This information provides a roadmap for studying descending axonal projections that may influence visceromotor systems and visceral "mind-body" symptoms.
Collapse
Affiliation(s)
- Silvia Gasparini
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa city, Iowa, USA
| | - Jacob M Howland
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa city, Iowa, USA
| | - Andrew J Thatcher
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa city, Iowa, USA
| | - Joel C Geerling
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa city, Iowa, USA
| |
Collapse
|
39
|
Grady F, Peltekian L, Iverson G, Geerling JC. Direct Parabrachial-Cortical Connectivity. Cereb Cortex 2020; 30:4811-4833. [PMID: 32383444 DOI: 10.1093/cercor/bhaa072] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/17/2020] [Accepted: 02/17/2020] [Indexed: 02/06/2023] Open
Abstract
The parabrachial nucleus (PB) in the upper brain stem tegmentum includes several neuronal subpopulations with a wide variety of connections and functions. A subpopulation of PB neurons projects axons directly to the cerebral cortex, and limbic areas of the cerebral cortex send a return projection directly to the PB. We used retrograde and Cre-dependent anterograde tracing to identify genetic markers and characterize this PB-cortical interconnectivity in mice. Cortical projections originate from glutamatergic PB neurons that contain Lmx1b (81%), estrogen receptor alpha (26%), and Satb2 (20%), plus mRNA for the neuropeptides cholecystokinin (Cck, 48%) and calcitonin gene-related peptide (Calca, 13%), with minimal contribution from FoxP2+ PB neurons (2%). Axons from the PB produce an extensive terminal field in an unmyelinated region of the insular cortex, extending caudally into the entorhinal cortex, and arcing rostrally through the dorsolateral prefrontal cortex, with a secondary terminal field in the medial prefrontal cortex. In return, layer 5 neurons in the insular cortex and other prefrontal areas, along with a dense cluster of cells dorsal to the claustrum, send a descending projection to subregions of the PB that contain cortically projecting neurons. This information forms the neuroanatomical basis for testing PB-cortical interconnectivity in arousal and interoception.
Collapse
Affiliation(s)
- Fillan Grady
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52246, USA
| | - Lila Peltekian
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52246, USA
| | - Gabrielle Iverson
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52246, USA
| | - Joel C Geerling
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52246, USA
| |
Collapse
|
40
|
Tseilikman V, Dremencov E, Tseilikman O, Pavlovicova M, Lacinova L, Jezova D. Role of glucocorticoid- and monoamine-metabolizing enzymes in stress-related psychopathological processes. Stress 2020; 23:1-12. [PMID: 31322459 DOI: 10.1080/10253890.2019.1641080] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoid signaling is fundamental in healthy stress coping and in the pathophysiology of stress-related diseases, such as post-traumatic stress disorder (PTSD). Glucocorticoids are metabolized by cytochrome P450 (CYP) as well as 11-β-hydroxysteroid dehydrogenase type 1 (11βHSD1) and 2 (11βHSD2). Acute stress-induced increase in glucocorticoid concentrations stimulates the expression of several CYP sub-types. CYP is primarily responsible for glucocorticoid metabolism and its increased activity can result in decreased circulating glucocorticoids in response to repeated stress stimuli. In addition, repeated stress-induced glucocorticoid release can promote 11βHSD1 activation and 11βHSD2 inhibition, and the 11βHSD2 suppression can lead to apparent mineralocorticoid excess. The activation of CYP and 11βHSD1 and the suppression of 11βHSD2 may at least partly contribute to development of the blunted glucocorticoid response to stressors characteristic in high trait anxiety, PTSD, and other stress-related disorders. Glucocorticoids and glucocorticoid-metabolizing enzymes interact closely with other biomolecules such as inflammatory cytokines, monoamines, and some monoamine-metabolizing enzymes, namely the monoamine oxidase type A (MAO-A) and B (MAO-B). Glucocorticoids boost MAO activity and this decreases monoamine levels and induces oxidative tissue damage which then activates inflammatory cytokines. The inflammatory cytokines suppress CYP expression and activity. This dynamic cross-talk between glucocorticoids, monoamines, and their metabolizing enzymes could be a critical factor in the pathophysiology of stress-related disorders.Lay summaryGlucocorticoids, which are produced and released under the control by brain regulatory centers, are fundamental in the stress response. This review emphasizes the importance of glucocorticoid metabolism and particularly the interaction between the brain and the liver as the major metabolic organ in the body. The activity of enzymes involved in glucocorticoid metabolism is proposed to play not only an important role in positive, healthy glucocorticoid effects, but also to contribute to the development and course of stress-related diseases.
Collapse
Affiliation(s)
- Vadim Tseilikman
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Eliyahu Dremencov
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
- Institute of Molecular Physiology and Genetics, Centre for Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Olga Tseilikman
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Michaela Pavlovicova
- Institute of Molecular Physiology and Genetics, Centre for Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lubica Lacinova
- Institute of Molecular Physiology and Genetics, Centre for Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
- Faculty of Natural Sciences, University of Saints Cyril and Methodius, Trnava, Slovakia
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
41
|
Koning ASCAM, Buurstede JC, van Weert LTCM, Meijer OC. Glucocorticoid and Mineralocorticoid Receptors in the Brain: A Transcriptional Perspective. J Endocr Soc 2019; 3:1917-1930. [PMID: 31598572 PMCID: PMC6777400 DOI: 10.1210/js.2019-00158] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023] Open
Abstract
Adrenal glucocorticoid hormones are crucial for maintenance of homeostasis and adaptation to stress. They act via the mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs)-members of the family of nuclear receptors. MRs and GRs can mediate distinct, sometimes opposite, effects of glucocorticoids. Both receptor types can mediate nongenomic steroid effects, but they are best understood as ligand-activated transcription factors. MR and GR protein structure is similar; the receptors can form heterodimers on the DNA at glucocorticoid response elements (GREs), and they share a number of target genes. The transcriptional basis for opposite effects on cellular physiology remains largely unknown, in particular with respect to MR-selective gene transcription. In this review, we discuss proven and potential mechanisms of transcriptional specificity for MRs and GRs. These include unique GR binding to "negative GREs," direct binding to other transcription factors, and binding to specific DNA sequences in conjunction with other transcription factors, as is the case for MRs and NeuroD proteins in the brain. MR- and GR-specific effects may also depend on specific interactions with transcriptional coregulators, downstream mediators of transcriptional receptor activity. Current data suggest that the relative importance of these mechanisms depends on the tissue and physiological context. Insight into these processes may not only allow a better understanding of homeostatic regulation but also the development of drugs that target specific aspects of disease.
Collapse
Affiliation(s)
- Anne-Sophie C A M Koning
- Einthoven Laboratory and Department of Medicine, Division of Endocrinology, Leiden University Medical Center, RC Leiden, Netherlands
| | - Jacobus C Buurstede
- Einthoven Laboratory and Department of Medicine, Division of Endocrinology, Leiden University Medical Center, RC Leiden, Netherlands
| | - Lisa T C M van Weert
- Einthoven Laboratory and Department of Medicine, Division of Endocrinology, Leiden University Medical Center, RC Leiden, Netherlands
| | - Onno C Meijer
- Einthoven Laboratory and Department of Medicine, Division of Endocrinology, Leiden University Medical Center, RC Leiden, Netherlands
| |
Collapse
|
42
|
Interaction of central angiotensin II and aldosterone on sodium intake and blood pressure. Brain Res 2019; 1720:146299. [DOI: 10.1016/j.brainres.2019.06.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/12/2019] [Accepted: 06/15/2019] [Indexed: 12/25/2022]
|
43
|
Affiliation(s)
- John W Funder
- From the Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| |
Collapse
|
44
|
de Kloet ER, de Kloet SF, de Kloet CS, de Kloet AD. Top-down and bottom-up control of stress-coping. J Neuroendocrinol 2019; 31:e12675. [PMID: 30578574 PMCID: PMC6519262 DOI: 10.1111/jne.12675] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/17/2022]
Abstract
In this 30th anniversary issue review, we focus on the glucocorticoid modulation of limbic-prefrontocortical circuitry during stress-coping. This action of the stress hormone is mediated by mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs) that are co-expressed abundantly in these higher brain regions. Via both receptor types, the glucocorticoids demonstrate, in various contexts, rapid nongenomic and slower genomic actions that coordinate consecutive stages of information processing. MR-mediated action optimises stress-coping, whereas, in a complementary fashion, the memory storage of the selected coping strategy is promoted via GR. We highlight the involvement of adipose tissue in the allocation of energy resources to central regulation of stress reactions, point to still poorly understood neuronal ensembles in the prefrontal cortex that underlie cognitive flexibility critical for effective coping, and evaluate the role of cortisol as a pleiotropic regulator in vulnerability to, and treatment of, trauma-related psychiatric disorders.
Collapse
Affiliation(s)
- Edo R. de Kloet
- Division of EndocrinologyDepartment of MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Sybren F. de Kloet
- Department of Integrative NeurophysiologyCenter for Neurogenomics and Cognitive ResearchVU‐University of AmsterdamAmsterdamThe Netherlands
| | | | - Annette D. de Kloet
- Department of Physiology and Functional GenomicsUniversity of FloridaGainesvilleFlorida
| |
Collapse
|