1
|
Hanchate NK. Single-cell genomics meets systems neuroscience: Insights from mapping the brain circuitry of stress. J Neuroendocrinol 2025; 37:e70005. [PMID: 39956535 PMCID: PMC12045673 DOI: 10.1111/jne.70005] [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: 05/08/2024] [Revised: 12/26/2024] [Accepted: 02/03/2025] [Indexed: 02/18/2025]
Abstract
Responses to external and internal dangers is essential for survival and homeostatic regulation. Hypothalamic corticotropin-releasing hormone neurons (CRHNs) play a pivotal role in regulating neuroendocrine responses to fear and stress. In recent years, the application of neurogenetic tools, such as fiber photometry, chemogenetics and optogenetics, have provided new insights into the dynamic neuronal responses of CRHNs during stressful events, offering new perspectives into their functional significance in mediating neurobehavioural responses to stress. Transsynaptic viral tracers have facilitated the comprehensive mapping of neuronal inputs to CRHNs. Furthermore, the development and application of innovative single-cell genomic tools combined with viral tracing have begun to pave the way for a deeper understanding of the transcriptional profiles of neural circuit components, enabling molecular-anatomical circuit mapping. Here, I will discuss how these systems neuroscience approaches and novel single-cell genomic methods are advancing the molecular and functional mapping of stress neurocircuits, their associated challenges and future directions.
Collapse
Affiliation(s)
- Naresh K. Hanchate
- Genetics & Genomic Medicine DepartmentUCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
| |
Collapse
|
2
|
Holt MK, Valderrama N, Polanco MJ, Hayter I, Badenoch EG, Trapp S, Rinaman L. Modulation of stress-related behaviour by preproglucagon neurons and hypothalamic projections to the nucleus of the solitary tract. Mol Metab 2025; 91:102076. [PMID: 39603502 PMCID: PMC11667184 DOI: 10.1016/j.molmet.2024.102076] [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: 09/26/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
Stress-induced behaviours are driven by complex neural circuits and some neuronal populations concurrently modulate diverse behavioural and physiological responses to stress. Glucagon-like peptide-1 (GLP-1)-producing preproglucagon (PPG) neurons within the lower brainstem caudal nucleus of the solitary tract (cNTS) are particularly sensitive to stressful stimuli and are implicated in multiple physiological and behavioural responses to interoceptive and psychogenic threats. However, the afferent inputs driving stress-induced activation of PPG neurons are largely unknown, and the role of PPG neurons in anxiety-like behaviour is controversial. Through chemogenetic manipulations we reveal that cNTS PPG neurons have the ability to moderately increase anxiety-like behaviours in mice in a sex-dependent manner. Using an intersectional approach, we show that input from the paraventricular nucleus of the hypothalamus (PVN) drives activation of both the cNTS as a whole and PPG neurons in particular in response to acute restraint stress, but that while this input is rich in corticotropin-releasing hormone (CRH), PPG neurons do not express significant levels of receptors for CRH and are not activated following lateral ventricle delivery of CRH. Finally, we demonstrate that cNTS-projecting PVN neurons are necessary for the ability of restraint stress to suppress food intake in male mice. Our findings reveal sex differences in behavioural responses to PPG neural activation and highlight a hypothalamic-brainstem pathway in stress-induced hypophagia.
Collapse
Affiliation(s)
- Marie K Holt
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA; Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, UCL, London, UK; University of Warwick, School of Life Sciences, Coventry, UK.
| | - Natalia Valderrama
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA
| | - Maria J Polanco
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA
| | - Imogen Hayter
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, UCL, London, UK; GlaxoSmithKline Pharmaceuticals, London, UK
| | | | - Stefan Trapp
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, UCL, London, UK
| | - Linda Rinaman
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA
| |
Collapse
|
3
|
Burke MR, Sotiropoulos I, Waites CL. The multiple roles of chronic stress and glucocorticoids in Alzheimer's disease pathogenesis. Trends Neurosci 2024; 47:933-948. [PMID: 39307629 PMCID: PMC11563862 DOI: 10.1016/j.tins.2024.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 11/15/2024]
Abstract
Chronic stress and the accompanying long-term elevation of glucocorticoids (GCs), the stress hormones of the body, increase the risk and accelerate the progression of Alzheimer's disease (AD). Signatures of AD include intracellular tau (MAPT) tangles, extracellular amyloid β (Aβ) plaques, and neuroinflammation. A growing body of work indicates that stress and GCs initiate cellular processes underlying these pathologies through dysregulation of protein homeostasis and trafficking, mitochondrial bioenergetics, and response to damage-associated stimuli. In this review, we integrate findings from mechanistic studies in rodent and cellular models, wherein defined chronic stress protocols or GC administration have been shown to elicit AD-related pathology. We specifically discuss the effects of chronic stress and GCs on tau pathogenesis, including hyperphosphorylation, aggregation, and spreading, amyloid precursor protein (APP) processing and trafficking culminating in Aβ production, immune priming by proinflammatory cytokines and disease-associated molecular patterns, and alterations to glial cell and blood-brain barrier (BBB) function.
Collapse
Affiliation(s)
- Mia R Burke
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Pathobiology and Mechanisms of Disease Graduate Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Ioannis Sotiropoulos
- Institute of Biosciences and Applications, National Centre for Scientific Research (NCSR) Demokritos, Agia Paraskevi, Greece
| | - Clarissa L Waites
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Department of Neuroscience, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
4
|
Laule C, Sayar-Atasoy N, Aklan I, Kim H, Ates T, Davis D, Atasoy D. Stress integration by an ascending adrenergic-melanocortin circuit. Neuropsychopharmacology 2024; 49:1361-1372. [PMID: 38326456 PMCID: PMC11251172 DOI: 10.1038/s41386-024-01810-9] [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: 08/22/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
Stress is thought to be an important contributing factor for eating disorders; however, neural substrates underlying the complex relationship between stress and appetite are not fully understood. Using in vivo recordings from awake behaving mice, we show that various acute stressors activate catecholaminergic nucleus tractus solitarius (NTSTH) projections in the paraventricular hypothalamus (PVH). Remarkably, the resulting adrenergic tone inhibits MC4R-expressing neurons (PVHMC4R), which are known for their role in feeding suppression. We found that PVHMC4R silencing encodes negative valence in sated mice and is required for avoidance induced by visceral malaise. Collectively, these findings establish PVHMC4R neurons as an effector of stress-activated brainstem adrenergic input in addition to the well-established hypothalamic-pituitary-adrenal axis. Convergent modulation of stress and feeding by PVHMC4R neurons implicates NTSTH → PVHMC4R input in stress-associated appetite disorders.
Collapse
Affiliation(s)
- Connor Laule
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nilufer Sayar-Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Iltan Aklan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hyojin Kim
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Tayfun Ates
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Debbie Davis
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
5
|
Parks BJ, Salazar P, Morrison L, McGraw MK, Gunnell M, Tobacyk J, Brents LK, Berquist MD. Limited bedding and nesting increases ethanol drinking in female rats. Pharmacol Biochem Behav 2024; 239:173756. [PMID: 38555037 PMCID: PMC11088506 DOI: 10.1016/j.pbb.2024.173756] [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: 12/20/2023] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
Prenatal opioid exposure (POE) and postnatal adverse experiences are early life adversities (ELA) that often co-occur and increase problematic alcohol (EtOH) drinking during adolescence. We investigated the relationship between POE, postnatal adversity, and adolescent EtOH drinking in rats. We also sought to determine whether ELAs affect alpha-adrenoceptor density in the brain because the noradrenergic system is involved in problematic alcohol drinking and its treatment. We hypothesized that the combination of POE and postnatal adversity will increase alcohol drinking in rats compared to rats with exposure to either adversity alone or to control. We also predicted that POE and postnatal adversity would increase α1-adrenoceptor density and decrease α2-adrenoceptor density in brain to confer a stress-responsive phenotype. Pregnant rats received morphine (15 mg/kg/day) or saline via subcutaneous minipumps from gestational day 9 until birth. Limited bedding and nesting (LBN) procedures were introduced from postnatal day (PD) 3-11 to mimic early life adversity-scarcity. Offspring rats (PD 31-33) were given opportunities to drink EtOH (20 %, v/v) using intermittent-access, two-bottle choice (with water) procedures. Rats given access to EtOH were assigned into sub-groups that were injected with either yohimbine (1 mg/kg, ip) or vehicle (2 % DMSO, ip) 30 min prior to each EtOH access session to determine the effects of α2-adrenoceptor inhibition on alcohol drinking. We harvested cortices, brainstems, and hypothalami from EtOH-naïve littermates on either PD 30 or PD 70 and conducted radioligand receptor binding assays to quantify α1- and α2-adrenoceptor densities. Contrary to our hypothesis, only LBN alone increased EtOH intake in female adolescent rats compared to female rats with POE. Neither POE nor LBN affected α1- or α2-adrenoceptor densities in the cortex, brainstem, or hypothalamus of early- or late-aged adolescent rats. These results suggest a complex interaction between ELA type and sex on alcohol drinking.
Collapse
Affiliation(s)
- B J Parks
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - P Salazar
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - L Morrison
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - M K McGraw
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - M Gunnell
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - J Tobacyk
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - L K Brents
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - M D Berquist
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America.
| |
Collapse
|
6
|
Meier JK, Schwabe L. Consistently increased dorsolateral prefrontal cortex activity during the exposure to acute stressors. Cereb Cortex 2024; 34:bhae159. [PMID: 38642105 PMCID: PMC11031141 DOI: 10.1093/cercor/bhae159] [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: 12/01/2023] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 04/22/2024] Open
Abstract
Stress has a major impact on our mental health. Nonetheless, it is still not fully understood how the human brain responds to ongoing stressful events. Here, we aimed to determine the cortical dynamics during the exposure to ecologically valid, standardized stressors. To this end, we conducted 3 experiments in which healthy participants underwent the Trier Social Stress Test (experiments 1 and 2) and the Socially Evaluated Cold Pressor Test (experiment 3) or a respective control manipulation, while we measured their cortical activity using functional near-infrared spectroscopy. Increases in salivary cortisol and subjective stress levels confirmed the successful stress induction in all experiments. Results of experiment 1 showed significantly increased cortical activity, in particular in the dorsolateral prefrontal cortex, during the exposure to the Trier Social Stress Test. Experiment 2 replicated this finding and showed further that this stress-related increase in dorsolateral prefrontal cortex activity was transient and limited to the period of the Trier Social Stress Test. Experiment 3 demonstrated the increased dorsolateral prefrontal cortex activity during the Socially Evaluated Cold Pressor Test, suggesting that this increase is generalizable and not specific to the Trier Social Stress Test. Together, these data show consistently that dorsolateral prefrontal cortex activity is not reduced, as commonly assumed, but increased under stress, which may promote coping with the ongoing stressor.
Collapse
Affiliation(s)
| | - Lars Schwabe
- Department of Cognitive Psychology, Universität Hamburg, Von-Melle-Park 5, 20146 Hamburg, Germany
| |
Collapse
|
7
|
Grantham EK, Tiwari GR, Ponomareva O, Harris RA, Lopez MF, Becker HC, Mayfield RD. Transcriptome changes in the nucleus of the solitary tract induced by repeated stress, alcohol dependence, or stress-induced drinking in dependent mice. Neuropharmacology 2024; 242:109768. [PMID: 37865137 PMCID: PMC10688594 DOI: 10.1016/j.neuropharm.2023.109768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/23/2023]
Abstract
Stress increases alcohol consumption in dependent animals and contributes to the development of alcohol use disorder. The nucleus of the solitary tract (NTS) is a critical brainstem region for integrating and relaying central and peripheral signals to regulate stress responses, but it is not known if it plays a role in alcohol dependence- or in stress-induced escalations in alcohol drinking in dependent mice. Here, we used RNA-sequencing and bioinformatics analyses to study molecular adaptations in the NTS of C57BL/6J male mice that underwent an ethanol drinking procedure that uses exposure to chronic intermittent ethanol (CIE) vapor, forced swim stress (FSS), or both conditions (CIE + FSS). Transcriptome profiling was performed at three different times after the last vapor cycle (0-hr, 72-hr, and 168-hr) to identify changes in gene expression associated with different stages of ethanol intoxication and withdrawal. In the CIE and CIE + FSS groups at 0-hr, there was upregulation of genes enriched for cellular response to type I interferon (IFN) and type I IFN- and cytokine-mediated signaling pathways, while the FSS group showed upregulation of neuronal genes. IFN signaling was the top gene network positively correlated with ethanol consumption levels in the CIE and CIE + FSS groups. Results from different analyses (differential gene expression, weighted gene coexpression network analysis, and rank-rank hypergeometric overlap) indicated that activation of type I IFN signaling would be expected to increase ethanol consumption. The CIE and CIE + FSS groups also shared an immune signature in the NTS as has been demonstrated in other brain regions after chronic ethanol exposure. A temporal-based clustering analysis revealed a unique expression pattern in the CIE + FSS group that suggests the interaction of these two stressors produces adaptations in synaptic and glial functions that may drive stress-induced drinking.
Collapse
Affiliation(s)
- Emily K Grantham
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Gayatri R Tiwari
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Olga Ponomareva
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Marcello F Lopez
- Department of Psychiatry & Behavioral Sciences and Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Howard C Becker
- Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 28425, USA; Department of Psychiatry & Behavioral Sciences and Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Veterans Affairs Medical Center, Charleston, SC, 20401, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
8
|
Pace SA, Myers B. Hindbrain Adrenergic/Noradrenergic Control of Integrated Endocrine and Autonomic Stress Responses. Endocrinology 2023; 165:bqad178. [PMID: 38015813 DOI: 10.1210/endocr/bqad178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Hindbrain adrenergic/noradrenergic nuclei facilitate endocrine and autonomic responses to physical and psychological challenges. Neurons that synthesize adrenaline and noradrenaline target hypothalamic structures to modulate endocrine responses while descending spinal projections regulate sympathetic function. Furthermore, these neurons respond to diverse stress-related metabolic, autonomic, and psychosocial challenges. Accordingly, adrenergic and noradrenergic nuclei are integrative hubs that promote physiological adaptation to maintain homeostasis. However, the precise mechanisms through which adrenaline- and noradrenaline-synthesizing neurons sense interoceptive and exteroceptive cues to coordinate physiological responses have yet to be fully elucidated. Additionally, the regulatory role of these cells in the context of chronic stress has received limited attention. This mini-review consolidates reports from preclinical rodent studies on the organization and function of brainstem adrenaline and noradrenaline cells to provide a framework for how these nuclei coordinate endocrine and autonomic physiology. This includes identification of hindbrain adrenaline- and noradrenaline-producing cell groups and their role in stress responding through neurosecretory and autonomic engagement. Although temporally and mechanistically distinct, the endocrine and autonomic stress axes are complementary and interconnected. Therefore, the interplay between brainstem adrenergic/noradrenergic nuclei and peripheral physiological systems is necessary for integrated stress responses and organismal survival.
Collapse
Affiliation(s)
- Sebastian A Pace
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Brent Myers
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
9
|
Sayar-Atasoy N, Laule C, Aklan I, Kim H, Yavuz Y, Ates T, Coban I, Koksalar-Alkan F, Rysted J, Davis D, Singh U, Alp MI, Yilmaz B, Cui H, Atasoy D. Adrenergic modulation of melanocortin pathway by hunger signals. Nat Commun 2023; 14:6602. [PMID: 37857606 PMCID: PMC10587058 DOI: 10.1038/s41467-023-42362-8] [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: 01/10/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023] Open
Abstract
Norepinephrine (NE) is a well-known appetite regulator, and the nor/adrenergic system is targeted by several anti-obesity drugs. To better understand the circuitry underlying adrenergic appetite control, here we investigated the paraventricular hypothalamic nucleus (PVN), a key brain region that integrates energy signals and receives dense nor/adrenergic input, using a mouse model. We found that PVN NE level increases with signals of energy deficit and decreases with food access. This pattern is recapitulated by the innervating catecholaminergic axon terminals originating from NTSTH-neurons. Optogenetic activation of rostral-NTSTH → PVN projection elicited strong motivation to eat comparable to overnight fasting whereas its inhibition attenuated both fasting-induced & hypoglycemic feeding. We found that NTSTH-axons functionally targeted PVNMC4R-neurons by predominantly inhibiting them, in part, through α1-AR mediated potentiation of GABA release from ARCAgRP presynaptic terminals. Furthermore, glucoprivation suppressed PVNMC4R activity, which was required for hypoglycemic feeding response. These results define an ascending nor/adrenergic circuit, NTSTH → PVNMC4R, that conveys peripheral hunger signals to melanocortin pathway.
Collapse
Affiliation(s)
- Nilufer Sayar-Atasoy
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Connor Laule
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Iltan Aklan
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hyojin Kim
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yavuz Yavuz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Tayfun Ates
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ilknur Coban
- Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | | | - Jacob Rysted
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Debbie Davis
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Uday Singh
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Muhammed Ikbal Alp
- Department of Physiology, School of Medicine, Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Bayram Yilmaz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Huxing Cui
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Deniz Atasoy
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
10
|
Buo C, Bearss RJ, Novak AG, Anello AE, Dakin JJ, Piet R. Serotonin stimulates female preoptic area kisspeptin neurons via activation of type 2 serotonin receptors in mice. Front Endocrinol (Lausanne) 2023; 14:1212854. [PMID: 37900129 PMCID: PMC10602649 DOI: 10.3389/fendo.2023.1212854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
Background The neuroendocrine control of ovulation is orchestrated by neuronal circuits that ultimately drive the release of gonadotropin-releasing hormone (GnRH) from the hypothalamus to trigger the preovulatory surge in luteinizing hormone (LH) secretion. While estrogen feedback signals are determinant in triggering activation of GnRH neurons, through stimulation of afferent kisspeptin neurons in the rostral periventricular area of the third ventricle (RP3VKISS1 neurons), many neuropeptidergic and classical neurotransmitter systems have been shown to regulate the LH surge. Among these, several lines of evidence indicate that the monoamine neurotransmitter serotonin (5-HT) has an excitatory, permissive, influence over the generation of the surge, via activation of type 2 5-HT (5-HT2) receptors. The mechanisms through which this occurs, however, are not well understood. We hypothesized that 5-HT exerts its influence on the surge by stimulating RP3VKISS1 neurons in a 5-HT2 receptor-dependent manner. Methods We tested this using kisspeptin neuron-specific calcium imaging and electrophysiology in brain slices obtained from male and female mice. Results We show that exogenous 5-HT reversibly increases the activity of the majority of RP3VKISS1 neurons. This effect is more prominent in females than in males, is likely mediated directly at RP3VKISS1 neurons and requires activation of 5-HT2 receptors. The functional impact of 5-HT on RP3VKISS1 neurons, however, does not significantly vary during the estrous cycle. Conclusion Taken together, these data suggest that 5-HT2 receptor-mediated stimulation of RP3VKISS1 neuron activity might be involved in mediating the influence of 5-HT on the preovulatory LH surge.
Collapse
Affiliation(s)
- Carrie Buo
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Robin J. Bearss
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Alyssa G. Novak
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Anna E. Anello
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Jordan J. Dakin
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
11
|
Trifilio E, Shortell D, Olshan S, O’Neal A, Coyne J, Lamb D, Porges E, Williamson J. Impact of transcutaneous vagus nerve stimulation on healthy cognitive and brain aging. Front Neurosci 2023; 17:1184051. [PMID: 37575296 PMCID: PMC10416636 DOI: 10.3389/fnins.2023.1184051] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/02/2023] [Indexed: 08/15/2023] Open
Abstract
Evidence for clinically meaningful benefits of transcutaneous vagus nerve stimulation (VNS) has been rapidly accumulating over the past 15 years. This relatively novel non-invasive brain stimulation technique has been applied to a wide range of neuropsychiatric disorders including schizophrenia, obsessive compulsive disorder, panic disorder, post-traumatic stress disorder, bipolar disorder, and Alzheimer's disease. More recently, non-invasive forms of VNS have allowed for investigations within healthy aging populations. These results offer insight into protocol considerations specific to older adults and how to translate those results into effective clinical trials and, ultimately, effective clinical care. In this review, we characterize the possible mechanisms by which non-invasive VNS may promote healthy aging (e.g., neurotransmitter effects, inflammation regulation, functional connectivity changes), special considerations for applying non-invasive VNS in an older adult population (e.g., vagus nerve changes with age), and how non-invasive VNS may be used in conjunction with existing behavioral interventions (e.g., cognitive behavioral therapy, cognitive training) to promote healthy emotional and cognitive aging.
Collapse
Affiliation(s)
- Erin Trifilio
- Center for OCD and Anxiety Related Disorders, Department of Psychiatry, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, United States
- Brain Rehabilitation and Research Center, Malcom Randall VAMC, Gainesville, FL, United States
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Destin Shortell
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Sarah Olshan
- Brain Rehabilitation and Research Center, Malcom Randall VAMC, Gainesville, FL, United States
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Alexandria O’Neal
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Jozee Coyne
- Center for OCD and Anxiety Related Disorders, Department of Psychiatry, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, United States
- Brain Rehabilitation and Research Center, Malcom Randall VAMC, Gainesville, FL, United States
| | - Damon Lamb
- Center for OCD and Anxiety Related Disorders, Department of Psychiatry, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, United States
- Brain Rehabilitation and Research Center, Malcom Randall VAMC, Gainesville, FL, United States
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Eric Porges
- Brain Rehabilitation and Research Center, Malcom Randall VAMC, Gainesville, FL, United States
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - John Williamson
- Center for OCD and Anxiety Related Disorders, Department of Psychiatry, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, United States
- Brain Rehabilitation and Research Center, Malcom Randall VAMC, Gainesville, FL, United States
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
- Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
12
|
Kopruszinski CM, Watanabe M, Martinez AL, Moreira de Souza LH, Dodick DW, Moutal A, Neugebauer V, Porreca F, Navratilova E. Kappa opioid receptor agonists produce sexually dimorphic and prolactin-dependent hyperalgesic priming. Pain 2023; 164:e263-e273. [PMID: 36625833 PMCID: PMC10285741 DOI: 10.1097/j.pain.0000000000002835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/21/2022] [Indexed: 01/11/2023]
Abstract
ABSTRACT Repeated stress produces hyperalgesic priming in preclinical models, but underlying mechanisms remain uncertain. As stress engages kappa opioid receptors (KORs), we hypothesized that repeated administration of KOR agonists might mimic, in part, stress-induced hyperalgesic priming. The potential contribution of circulating prolactin (PRL) and dysregulation of the expression of PRL receptor (PRLR) isoforms in sensory neurons after KOR agonist administration was also investigated. Mice received 3 daily doses of U-69593 or nalfurafine as a "first-hit" stimulus followed by assessment of periorbital tactile allodynia. Sixteen days after the first KOR agonist administration, animals received a subthreshold dose of inhalational umbellulone, a TRPA1 agonist, as the second-hit stimulus and periorbital allodynia was assessed. Cabergoline, a dopamine D2 receptor agonist, was used to inhibit circulating PRL in additional cohorts. Prolactin receptor isoforms were quantified in the V1 region of the trigeminal ganglion after repeated doses of U-69593. In both sexes, KOR agonists increased circulating PRL and produced allodynia that resolved within 14 days. Hyperalgesic priming, revealed by umbellulone-induced allodynia in animals previously treated with the KOR agonists, also occurred in both sexes. However, repeated U-69593 downregulated the PRLR long isoform in trigeminal neurons only in female mice. Umbellulone-induced allodynia was prevented by cabergoline co-treatment during priming with KOR agonists in female, but not male, mice. Hyperalgesic priming therefore occurs in both sexes after either biased or nonbiased KOR agonists. However, a PRL/PRLR-dependence is observed only in female nociceptors possibly contributing to pain in stress-related pain disorders in females.
Collapse
Affiliation(s)
- Caroline M. Kopruszinski
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States. Moutal is now with the Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Moe Watanabe
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States. Moutal is now with the Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Ashley L. Martinez
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States. Moutal is now with the Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Luiz Henrique Moreira de Souza
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States. Moutal is now with the Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - David W. Dodick
- Department of Neurology, Mayo Clinic, Phoenix, AZ, United States
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States. Moutal is now with the Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience and Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States. Moutal is now with the Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States
- Department of Neurology, Mayo Clinic, Phoenix, AZ, United States
| | - Edita Navratilova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States. Moutal is now with the Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
13
|
Haynes RL, Trachtenberg F, Darnall R, Haas EA, Goldstein RD, Mena OJ, Krous HF, Kinney HC. Altered 5-HT2A/C receptor binding in the medulla oblongata in the sudden infant death syndrome (SIDS): Part I. Tissue-based evidence for serotonin receptor signaling abnormalities in cardiorespiratory- and arousal-related circuits. J Neuropathol Exp Neurol 2023; 82:467-482. [PMID: 37226597 PMCID: PMC10209647 DOI: 10.1093/jnen/nlad030] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023] Open
Abstract
The sudden infant death syndrome (SIDS), the leading cause of postneonatal infant mortality in the United States, is typically associated with a sleep period. Previously, we showed evidence of serotonergic abnormalities in the medulla (e.g. altered serotonin (5-HT)1A receptor binding), in SIDS cases. In rodents, 5-HT2A/C receptor signaling contributes to arousal and autoresuscitation, protecting brain oxygen status during sleep. Nonetheless, the role of 5-HT2A/C receptors in the pathophysiology of SIDS is unclear. We hypothesize that in SIDS, 5-HT2A/C receptor binding is altered in medullary nuclei that are key for arousal and autoresuscitation. Here, we report altered 5-HT2A/C binding in several key medullary nuclei in SIDS cases (n = 58) compared to controls (n = 12). In some nuclei the reduced 5-HT2A/C and 5-HT1A binding overlapped, suggesting abnormal 5-HT receptor interactions. The data presented here (Part 1) suggest that a subset of SIDS is due in part to abnormal 5-HT2A/C and 5-HT1A signaling across multiple medullary nuclei vital for arousal and autoresuscitation. In Part II to follow, we highlight 8 medullary subnetworks with altered 5-HT receptor binding in SIDS. We propose the existence of an integrative brainstem network that fails to facilitate arousal and/or autoresuscitation in SIDS cases.
Collapse
Affiliation(s)
- Robin L Haynes
- CJ Murphy Laboratory for SIDS Research, Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Robert’s Program on Sudden Unexpected Death in Pediatrics, Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| | | | - Ryan Darnall
- CJ Murphy Laboratory for SIDS Research, Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Elisabeth A Haas
- Department of Research, Rady Children’s Hospital, San Diego, California, USA
| | - Richard D Goldstein
- Robert’s Program on Sudden Unexpected Death in Pediatrics, Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Othon J Mena
- San Diego County Medical Examiner Office, San Diego, California, USA
| | - Henry F Krous
- University of California, San Diego, San Diego, California, USA
- Rady Children’s Hospital, San Diego, California, USA
| | - Hannah C Kinney
- CJ Murphy Laboratory for SIDS Research, Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Robert’s Program on Sudden Unexpected Death in Pediatrics, Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Moses TE, Gray E, Mischel N, Greenwald MK. Effects of neuromodulation on cognitive and emotional responses to psychosocial stressors in healthy humans. Neurobiol Stress 2023; 22:100515. [PMID: 36691646 PMCID: PMC9860364 DOI: 10.1016/j.ynstr.2023.100515] [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: 08/10/2022] [Revised: 12/19/2022] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
Physiological and psychological stressors can exert wide-ranging effects on the human brain and behavior. Research has improved understanding of how the sympatho-adreno-medullary (SAM) and hypothalamic-pituitary-adrenocortical (HPA) axes respond to stressors and the differential responses that occur depending on stressor type. Although the physiological function of SAM and HPA responses is to promote survival and safety, exaggerated psychobiological reactivity can occur in psychiatric disorders. Exaggerated reactivity may occur more for certain types of stressors, specifically, psychosocial stressors. Understanding stressor effects and how the body regulates these responses can provide insight into ways that psychobiological reactivity can be modulated. Non-invasive neuromodulation is one way that responding to stressors may be altered; research into these interventions may provide further insights into the brain circuits that modulate stress reactivity. This review focuses on the effects of acute psychosocial stressors and how neuromodulation might be effective in altering stress reactivity. Although considerable research into stress interventions focuses on treating pathology, it is imperative to first understand these mechanisms in non-clinical populations; therefore, this review will emphasize populations with no known pathology and consider how these results may translate to those with psychiatric pathologies.
Collapse
Affiliation(s)
| | | | | | - Mark K. Greenwald
- Corresponding author. Department of Psychiatry and Behavioral Neurosciences, Tolan Park Medical Building, 3901 Chrysler Service Drive, Suite 2A, Detroit, MI, 48201, USA.
| |
Collapse
|
15
|
Tan HE. The microbiota-gut-brain axis in stress and depression. Front Neurosci 2023; 17:1151478. [PMID: 37123352 PMCID: PMC10140437 DOI: 10.3389/fnins.2023.1151478] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Humans and animals are evolved to have instinctive physiological responses to threats. The perception of threat by the brain triggers a multitude of changes across the brain and body. A large body of research have demonstrated that our hardwired survival instinct, the stress response, can become maladaptive and promote major depressive disorders and other neuropsychiatric impairments. However, gaps in our understanding of how chronic stress contributes to depression and mental disorders suggest that we also need to consider factors beyond the biology of the host. The unravelling of the structure and function of microorganisms that humans and animals are host to have driven a paradigm shift in understanding the individual as a collective network composed of the host plus microbes. Well over 90% of bacteria in the body reside in the large intestines, and these microbes in the lower gut function almost like an organ in the body in the way it interacts with the host. Importantly, bidirectional interactions between the gut microbiota and the brain (i.e., the two-way microbiota-gut-brain axis) have been implicated in the pathophysiology of mental disorders including depression. Here, in summarizing the emerging literature, we envisage that further research particularly on the efferent brain-gut-microbiota axis will uncover transformative links in the biology of stress and depression.
Collapse
|
16
|
Liu Y, Li S, Zhang X, Wang L, Li Z, Wu W, Qin X, Zhou J, Ma C, Meng W, Kuang X, Yin F, Xia Q, Jiang B, Yang Y. Corticotropin releasing factor neurons in the visual cortex mediate long-term changes in visual function induced by early adversity. Neurobiol Stress 2022; 21:100504. [DOI: 10.1016/j.ynstr.2022.100504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
|
17
|
Gu YQ, Zhu Y. Underlying mechanisms of mindfulness meditation: Genomics, circuits, and networks. World J Psychiatry 2022; 12:1141-1149. [PMID: 36186506 PMCID: PMC9521538 DOI: 10.5498/wjp.v12.i9.1141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/29/2022] [Accepted: 08/18/2022] [Indexed: 02/05/2023] Open
Abstract
Understanding neuropsychological mechanisms of mindfulness meditation (MM) has been a hot topic in recent years. This review was conducted with the goal of synthesizing empirical relationships via the genomics, circuits and networks between MM and mental disorders. We describe progress made in assessing the effects of MM on gene expression in immune cells, with particular focus on stress-related inflammatory markers and associated biological pathways. We then focus on key brain circuits associated with mindfulness practices and effects on symptoms of mental disorders, and expand our discussion to identify three key brain networks associated with mindfulness practices including default mode network, central executive network, and salience network. More research efforts need to be devoted into identifying underlying neuropsychological mechanisms of MM on how it alleviates the symptoms of mental disorders.
Collapse
Affiliation(s)
- Ying-Qi Gu
- Department of Psychology, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Yi Zhu
- School of Psychology, Hainan Medical University, Haikou 571199, Hainan Province, China
- Department of Psychology, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, Hainan Province, China
| |
Collapse
|
18
|
Moeller JS, Bever SR, Finn SL, Phumsatitpong C, Browne MF, Kriegsfeld LJ. Circadian Regulation of Hormonal Timing and the Pathophysiology of Circadian Dysregulation. Compr Physiol 2022; 12:4185-4214. [PMID: 36073751 DOI: 10.1002/cphy.c220018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Circadian rhythms are endogenously generated, daily patterns of behavior and physiology that are essential for optimal health and disease prevention. Disruptions to circadian timing are associated with a host of maladies, including metabolic disease and obesity, diabetes, heart disease, cancer, and mental health disturbances. The circadian timing system is hierarchically organized, with a master circadian clock located in the suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks throughout the CNS and periphery. The SCN receives light information via a direct retinal pathway, synchronizing the master clock to environmental time. At the cellular level, circadian rhythms are ubiquitous, with rhythms generated by interlocking, autoregulatory transcription-translation feedback loops. At the level of the SCN, tight cellular coupling maintains rhythms even in the absence of environmental input. The SCN, in turn, communicates timing information via the autonomic nervous system and hormonal signaling. This signaling couples individual cellular oscillators at the tissue level in extra-SCN brain loci and the periphery and synchronizes subordinate clocks to external time. In the modern world, circadian disruption is widespread due to limited exposure to sunlight during the day, exposure to artificial light at night, and widespread use of light-emitting electronic devices, likely contributing to an increase in the prevalence, and the progression, of a host of disease states. The present overview focuses on the circadian control of endocrine secretions, the significance of rhythms within key endocrine axes for typical, homeostatic functioning, and implications for health and disease when dysregulated. © 2022 American Physiological Society. Compr Physiol 12: 1-30, 2022.
Collapse
Affiliation(s)
- Jacob S Moeller
- Graduate Group in Endocrinology, University of California, Berkeley, California, USA
| | - Savannah R Bever
- Department of Psychology, University of California, Berkeley, California, USA
| | - Samantha L Finn
- Department of Psychology, University of California, Berkeley, California, USA
| | | | - Madison F Browne
- Department of Psychology, University of California, Berkeley, California, USA
| | - Lance J Kriegsfeld
- Graduate Group in Endocrinology, University of California, Berkeley, California, USA.,Department of Psychology, University of California, Berkeley, California, USA.,Department of Integrative Biology, University of California, Berkeley, California, USA.,The Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
| |
Collapse
|
19
|
Glier S, Campbell A, Corr R, Pelletier‐Baldelli A, Yefimov M, Guerra C, Scott K, Murphy L, Bizzell J, Belger A. Coordination of autonomic and endocrine stress responses to the Trier Social Stress Test in adolescence. Psychophysiology 2022; 59:e14056. [PMID: 35353921 PMCID: PMC9339460 DOI: 10.1111/psyp.14056] [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: 05/11/2021] [Revised: 01/19/2022] [Accepted: 02/24/2022] [Indexed: 11/30/2022]
Abstract
Dysregulations in autonomic and endocrine stress responses are linked to the emergence of psychopathology in adolescence. However, most studies fail to consider the interplay between these systems giving rise to conflicting findings and a gap in understanding adolescent stress response regulation. A multisystem framework-investigation of parasympathetic (PNS), sympathetic (SNS), and hypothalamic pituitary adrenal (HPA) axis components and their coordination-is necessary to understand individual differences in stress response coordination which contribute to stress vulnerabilities. As the first investigation to comprehensively evaluate these three systems in adolescence, the current study employed the Trier Social Stress Test in 72 typically developing adolescents (mean age = 13) to address how PNS, SNS, and HPA stress responses are coordinated in adolescence. Hypotheses tested key predictions of the Adaptive Calibration Model (ACM) of stress response coordination. PNS and SNS responses were assessed via heart rate variability (HRV) and salivary alpha amylase (sAA) respectively. HPA responses were indexed by salivary cortisol. Analyses utilized piecewise growth curve modeling to investigate these aims. Supporting the ACM theory, there was significant hierarchical coordination between the systems such that those with low HRV had higher sAA and cortisol reactivity and those with high HRV had low-to-moderate sAA and cortisol responsivity. Our novel results reveal the necessity of studying multisystem dynamics in an integrative fashion to uncover the true mechanisms of stress response and regulation during development. Additionally, our findings support the existence of characteristic stress response profiles as predicted by the ACM model.
Collapse
Affiliation(s)
- Sarah Glier
- School of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Alana Campbell
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Carolina Institute for Developmental DisabilitiesUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Frank Porter Graham Child Development InstituteUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Rachel Corr
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Andrea Pelletier‐Baldelli
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Carolina Institute for Developmental DisabilitiesUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Frank Porter Graham Child Development InstituteUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Mae Yefimov
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Frank Porter Graham Child Development InstituteUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Carina Guerra
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Kathryn Scott
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Louis Murphy
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Joshua Bizzell
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Carolina Institute for Developmental DisabilitiesUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Frank Porter Graham Child Development InstituteUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Aysenil Belger
- Department of PsychiatryUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Carolina Institute for Developmental DisabilitiesUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Frank Porter Graham Child Development InstituteUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
20
|
Pereira Júnior AA, de Amorim GES, Garcia RCT, Ribeiro JM, Silva AO, Almeida CADF, Ceron CS, Ruginsk SG, Antunes-Rodrigues J, Elias LLK, Dias MVS, Marcourakis T, Torres LH. Nicotine exposure through breastfeeding affects BDNF and synaptic proteins levels in the brain of stressed adult female mice. Int J Dev Neurosci 2022; 82:759-771. [PMID: 36018565 DOI: 10.1002/jdn.10227] [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: 05/29/2022] [Revised: 08/11/2022] [Accepted: 08/22/2022] [Indexed: 11/08/2022] Open
Abstract
Nicotine has been used during pregnancy and lactation as a tobacco harm reduction strategy. However, it is unclear whether nicotine exposure during a critical development period negatively impacts stress responses in adulthood. This study investigated how nicotine, administered via breastfeeding, affects the brain-derived neurotrophic factor (BDNF), synaptic proteins levels, and anxiety-like behavior in adult female mice subjected to stress. Female Swiss mice were exposed to saline or nicotine (8 mg/kg/day) through breastfeeding between their fourth and 17th postnatal days (P) via implanted osmotic mini pumps. The unpredictable chronic mild stress (UCMS) protocol was performed during their adulthood (P65) for 10 consecutive days, followed by the elevated plus maze (EPM) test one day after the protocol. Animals were euthanized and their blood, collected for plasma corticosterone measurements and their brain structures, dissected for BDNF and synaptic proteins analyses. We found no significant differences in corticosterone levels between groups (Saline/Non-stress, Nicotine/Non-stress, Saline/Stress, and Nicotine/Stress). The UCMS protocol hindered weight gain. Mice exposed to nicotine through breastfeeding with or without the UCMS protocol in adulthood showed higher grooming and head dipping frequency; decreased BDNF levels in cerebellum and striatum; increased postsynaptic density protein 95 (PSD-95), synapsin I, and synaptophysin levels in cerebellum; and decreased PSD-95 and synapsin I levels in brainstem. Our results indicate that nicotine exposure through breastfeeding leads to long-lasting behavioral effects and synaptic protein changes, most of which were independent of the UCMS protocol, even after a long nicotine-free period, highlighting the importance of further studies on nicotine exposure during development.
Collapse
Affiliation(s)
- Antonio Alves Pereira Júnior
- Department of Food and Drugs, School of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | | | - Raphael Caio Tamborelli Garcia
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo Diadema, São Paulo, Brazil
| | - Jéssyca Milene Ribeiro
- Department of Food and Drugs, School of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Alessandra Oliveira Silva
- Department of Food and Drugs, School of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | | | - Carla Speroni Ceron
- Department of Food and Drugs, School of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Silvia Graciela Ruginsk
- Department of Physiological Sciences, Biomedical Sciences Institute, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - José Antunes-Rodrigues
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Lucila Leico Kagohara Elias
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Tania Marcourakis
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Larissa Helena Torres
- Department of Food and Drugs, School of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| |
Collapse
|
21
|
Smeltzer EA, Stead SM, Li MF, Samson D, Kumpan LT, Teichroeb JA. Social sleepers: The effects of social status on sleep in terrestrial mammals. Horm Behav 2022; 143:105181. [PMID: 35594742 DOI: 10.1016/j.yhbeh.2022.105181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/12/2022] [Accepted: 04/22/2022] [Indexed: 11/18/2022]
Abstract
Social status among group-living mammals can impact access to resources, such as water, food, social support, and mating opportunities, and this differential access to resources can have fitness consequences. Here, we propose that an animal's social status impacts their access to sleep opportunities, as social status may predict when an animal sleeps, where they sleep, who they sleep with, and how well they sleep. Our review of terrestrial mammals examines how sleep architecture and intensity may be impacted by (1) sleeping conditions and (2) the social experience during wakefulness. Sleeping positions vary in thermoregulatory properties, protection from predators, and exposure to parasites. Thus, if dominant individuals have priority of access to sleeping positions, they may benefit from higher quality sleeping conditions and, in turn, better sleep. With respect to waking experiences, we discuss the impacts of stress on sleep, as it has been established that specific social statuses can be characterized by stress-related physiological profiles. While much research has focused on how dominance hierarchies impact access to resources like food and mating opportunities, differential access to sleep opportunities among mammals has been largely ignored despite its potential fitness consequences.
Collapse
Affiliation(s)
- E A Smeltzer
- Department of Anthropology, University of Toronto Scarborough, 1265 Military Trail, Scarborough, Ontario M1C 1A4, Canada
| | - S M Stead
- Department of Anthropology, University of Toronto Scarborough, 1265 Military Trail, Scarborough, Ontario M1C 1A4, Canada.
| | - M F Li
- Department of Anthropology, University of Toronto, 19 Russell St., Toronto, Ontario M5S 2S2, Canada
| | - D Samson
- Department of Anthropology, University of Toronto Mississauga, 3359 Mississauga Rd., Mississauga, Ontario L5L 1C6, Canada
| | - L T Kumpan
- Department of Anthropology, University of Toronto Scarborough, 1265 Military Trail, Scarborough, Ontario M1C 1A4, Canada
| | - J A Teichroeb
- Department of Anthropology, University of Toronto Scarborough, 1265 Military Trail, Scarborough, Ontario M1C 1A4, Canada
| |
Collapse
|
22
|
Glier S, Campbell A, Corr R, Pelletier-Baldelli A, Belger A. Individual differences in frontal alpha asymmetry moderate the relationship between acute stress responsivity and state and trait anxiety in adolescents. Biol Psychol 2022; 172:108357. [PMID: 35662579 PMCID: PMC10091222 DOI: 10.1016/j.biopsycho.2022.108357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 11/25/2022]
Abstract
Stress is a risk factor in the development and maintenance of psychopathology, particularly anxiety. Despite theory suggesting differences in stress responsivity may explain heterogeneity in anxiety, findings remain contradictory. This may be due to failure to account for individuals' neurobiological states and outdated methodologic analyses which confound conceptually and biologically distinct stress response pathways. In 145 adolescents, this study examined whether individual differences in neural activation underlying motivational states, indexed by resting frontal alpha asymmetry (FAA) before and after the Trier Social Stress Test (TSST), moderate the relationship between stress responsivity (measured by cortisol) and anxiety. Adolescents with rightward FAA activation (indexed by changes in resting FAA pre-to-post TSST) and high trait anxiety showed blunted cortisol reactivities while those with leftward FAA activation and high state anxiety showed prolonged cortisol recoveries. Our work reveals individual differences in vulnerability to psychosocial stressors and is the first study to show that FAA activation moderates the relationships between anxiety and distinct phases of the stress response in adolescents.
Collapse
Affiliation(s)
- Sarah Glier
- School of Medicine at the University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Psychiatry Department at University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Alana Campbell
- Psychiatry Department at University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Carolina Institute for Developmental Disabilities University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rachel Corr
- Psychiatry Department at University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Aysenil Belger
- Psychiatry Department at University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Carolina Institute for Developmental Disabilities University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Frank Porter Graham Child Development Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Choi MP, Rubin AM, Wada H. Suboptimal Embryonic Incubation Temperature Has Long-Term, Sex-Specific Consequences on Beak Coloration and the Behavioral Stress Response in Zebra Finches. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.901303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Secondary sex characteristics, like beak color in some avian species, have indirect impacts on reproductive success, as they are considered to be honest indicators of condition, immunocompetence, and developmental history. However, little is known about the long-term effects of environmental perturbations on the production and maintenance of these secondary sex characteristics in avian species. In zebra finches (Taeniopygia guttata), redder beaks indicate increased carotenoid expression and implantation into beak tissue, and female zebra finches prefer males with pronounced bright red beaks as a mate. The present study examines the long-term effects of embryonic incubation temperature on the maturation of beak color in zebra finches. We also investigated the effects of embryonic incubation temperature on sensitivity to a handling and restraint stressor in adulthood. Specifically, the aims of this study were to examine: (1) whether suboptimal incubation temperatures affect the timing of beak color development and color characteristics before and after sexual maturity, (2) if repeated handling causes short-term changes in beak color and whether color changes are related to embryonic thermal environment, and (3) how thermal stress during incubation alters future responses to a repeated handling stressor. Zebra finch eggs were randomly assigned to one of three incubators: “Control,” “Low,” or “Periodic Cooling.” Beak color (hue, saturation, and value) was quantified before [45, 60, 75 days post-hatch (dph)] and after sexual maturity (95 dph), as well as after repeated handling stress later in adulthood (avg of 386 dph). We found that there were age- and sex- specific effects of incubation treatment on beak hue, where females from periodically cooled eggs had decreased hues (redder) in adulthood. Additionally, eggs laid later in a clutch had decreased beak saturation levels throughout life regardless of incubation environment. We found that females had lower beak hue and saturation following a capture and restraint stressor, while males showed increased beak saturation. Lastly, males subjected to the Low incubation treatment had relatively higher activity levels during restraint than those in the Control group. Overall, these findings suggest that fluctuating incubation temperatures combined with repeated, short-term stressors can have significant, sex-specific effects on sexual ornamentation and behavior.
Collapse
|
24
|
Rabey M, Moloney N. "I Don't Know Why I've Got this Pain!" Allostasis as a Possible Explanatory Model. Phys Ther 2022; 102:6535131. [PMID: 35202474 DOI: 10.1093/ptj/pzac017] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 10/05/2021] [Accepted: 02/18/2022] [Indexed: 11/14/2022]
Abstract
UNLABELLED Explaining the onset and maintenance of pain can be challenging in many clinical presentations. Allostasis encompasses the mechanisms through which humans adapt to stressors to maintain physiological stability. Due to related neuro-endocrine-immune system effects, allostasis and allostatic load (the cumulative effects on the brain and body that develop through the maintenance of physiological stability) offer the potential to explain the development and maintenance of musculoskeletal pain in certain cases. This paper outlines the concept of allostatic load, highlights the evidence for allostatic load in musculoskeletal pain conditions to date, and discusses mechanisms through which allostatic load influences pain, with particular focus on hypothalamic-pituitary-adrenal axis and sympathetic nervous system function and central, brain-driven governance of these systems. Finally, through case examples, consideration is given as to how allostatic load can be integrated into clinical reasoning and how it can be used to help explain pain to individuals and guide clinical decision-making. IMPACT Awareness of the concept of allostatic load, and subsequent assessment of physical and psychological stressors potentially contributing to allostatic load, may facilitate a broader understanding of the multidimensional presentations of many people with pain, both acute and persistent. This may facilitate discussion between clinicians and their patients regarding broader influences on their presentations and drive more targeted and inclusive pain management strategies.
Collapse
Affiliation(s)
- Martin Rabey
- THRIVE Physiotherapy, St Martins, Guernsey, UK.,School of Allied Health, Curtin University, Perth, Western Australia, Australia
| | - Niamh Moloney
- THRIVE Physiotherapy, St Martins, Guernsey, UK.,Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, New South Wales, Australia
| |
Collapse
|
25
|
Yamaguchi N, Kakinuma Y, Yakura T, Naito M, Okada S. Glucose infusion suppresses acute restraint stress-induced peripheral and central sympathetic responses in rats. Auton Neurosci 2022; 239:102957. [DOI: 10.1016/j.autneu.2022.102957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 10/19/2022]
|
26
|
Hosseinian S, Ansari S. Prophylactic effects of dietary ascorbic acid on oxidative stress indices, physiological and behavioural responses of domestic pigeons exposed to road transport stress. Vet Med Sci 2021; 7:2389-2398. [PMID: 34797959 PMCID: PMC8604146 DOI: 10.1002/vms3.609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Transport disturbs birds' welfare and health status which lead to oxidative stress and dietary ascorbic acid mitigates the adverse effects of transport stress. The present study was aimed to evaluate the impacts of ascorbic acid administration on oxidative stress indices, cortisol, H/L ratio, tonic immobility reaction and rectal temperature of pigeons exposed to road transport. A total of 80 clinically healthy pigeons were selected and randomly divided to eight equal groups as follow: (1) Ctrl- : fed by basal diet and no subjected to transport stress; (2) Ctrl+: fed by the basal diet and subjected to transport stress; (3, 4) 1DBS10 and 1DBS16: received ascorbic acid from 1 day before transport stress at doses of 10 g/100 L and 16 g/100 L of drinking water, respectively; (5, 6) 3DBS10 and 3DBS16: treated with ascorbic acid from 3 consecutive days before transport stress at doses of 10 g/100 L and 16 g/100 L, respectively and (7, 8) 7DBS10 and 7DBS16: received ascorbic acid from 7 consecutive days before the transport at doses 10 g/100 L and 16 g/100 L, respectively. Birds were transported for 3 h over a distance of about 200 km. The total antioxidant capacity, malondialdehyde and cortisol were measured before transport and at 6, 24 and 72 h post-transportation. The rectal temperature and tunic immobility reactions were recorded. Dietary ascorbic acid led to a decrease in tonic immobility response, hetrophil to lymphocyte ratio, circulating cortisol and total antioxidant capacity, and an increase in circulating malondialdehyde in pigeons exposed to transport stress compare to Ctrl+ group. In conclusion, ascorbic acid administration at dose 16 g/100 L of drinking water from 3 and 7 days before exposure to stress helps attenuate undesirable effects of oxidative stress in pigeons.
Collapse
Affiliation(s)
- S.A. Hosseinian
- Department of Clinical Science, School of Veterinary MedicineShiraz UniversityShirazIran
| | - S. Ansari
- Department of Clinical Science, School of Veterinary MedicineShiraz UniversityShirazIran
| |
Collapse
|
27
|
Fawley JA, Hegarty DM, Aicher SA, Beaumont E, Andresen MC. Dedicated C-fiber vagal sensory afferent pathways to the paraventricular nucleus of the hypothalamus. Brain Res 2021; 1769:147625. [PMID: 34416255 PMCID: PMC8438991 DOI: 10.1016/j.brainres.2021.147625] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/24/2021] [Accepted: 08/12/2021] [Indexed: 12/22/2022]
Abstract
The nucleus of the solitary tract (NTS) receives viscerosensory information from the vagus nerve to regulate diverse homeostatic reflex functions. The NTS projects to a wide network of other brain regions, including the paraventricular nucleus of the hypothalamus (PVN). Here we examined the synaptic characteristics of primary afferent pathways to PVN-projecting NTS neurons in rat brainstem slices.Expression of the Transient Receptor Potential Vanilloid receptor (TRPV1+ ) distinguishes C-fiber afferents within the solitary tract (ST) from A-fibers (TRPV1-). We used resiniferatoxin (RTX), a TRPV1 agonist, to differentiate the two. The variability in the latency (jitter) of evoked excitatory postsynaptic currents (ST-EPSCs) distinguished monosynaptic from polysynaptic ST-EPSCs. Rhodamine injected into PVN was retrogradely transported to identify PVN-projecting NTS neurons within brainstem slices. Graded shocks to the ST elicited all-or-none EPSCs in rhodamine-positive NTS neurons with latencies that had either low jitter (<200 µs - monosynaptic), high jitter (>200 µs - polysynaptic inputs) or both. RTX blocked ST-evoked TRPV1 + EPSCs whether mono- or polysynaptic. Most PVN-projecting NTS neurons (17/21 neurons) had at least one input polysynaptically connected to the ST. Compared to unlabeled NTS neurons, PVN-projecting NTS neurons were more likely to receive indirect inputs and be higher order. Surprisingly, sEPSC rates for PVN-projecting neurons were double that of unlabeled NTS neurons. The ST synaptic responses for PVN-projecting NTS neurons were either all TRPV1+ or all TRPV1-, including neurons that received both direct and indirect inputs. Overall, PVN-projecting NTS neurons received direct and indirect vagal afferent information with strict segregation regarding TRPV1 expression.
Collapse
Affiliation(s)
- Jessica A Fawley
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, United States
| | - Deborah M Hegarty
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, United States
| | - Sue A Aicher
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, United States
| | - Eric Beaumont
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States
| | - Michael C Andresen
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, United States.
| |
Collapse
|
28
|
Bravo-Tobar ID, Fernández P, Sáez JC, Dagnino-Subiabre A. Long-term effects of stress resilience: Hippocampal neuroinflammation and behavioral approach in male rats. J Neurosci Res 2021; 99:2493-2510. [PMID: 34184764 DOI: 10.1002/jnr.24902] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/07/2021] [Accepted: 05/26/2021] [Indexed: 01/28/2023]
Abstract
Resilience to stress is the ability to quickly adapt to adversity. There is evidence that exposure to prolonged stress triggers neuroinflammation what produces individual differences in stress vulnerability. However, the relationship between stress resilience, neuroinflammation, and depressive-like behaviors remains unknown. The aim of this study was to analyze the long-term effects of social defeat stress (SDS) on neuroinflammation in the hippocampus and depressive-like behaviors. Male rats were subjected to the SDS paradigm. Social interaction was analyzed 1 and 2 weeks after ending the SDS to determine which animals were susceptible or resilient to stress. Neuroinflammation markers glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1, and elevated membrane permeability in astrocytes and microglia, as well as depressive-like behaviors in the sucrose preference test and forced swim test were evaluated in all rats. One week after SDS, resilient rats increased their sucrose preference, and time spent in the floating behavior decreased in the forced swim test compared to susceptible rats. Surprisingly, resilient rats became susceptible to stress, and presented neuroinflammation 2 weeks after SDS. These findings suggest that SDS-induced hippocampal neuroinflammation persists in post-stress stages, regardless of whether rats were initially resilient or not. Our study opens a new approach to understanding the neurobiology of stress resilience.
Collapse
Affiliation(s)
- Iván D Bravo-Tobar
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Paola Fernández
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Juan C Sáez
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, Centre for Integrative Neurobiology and Pathophysiology, Institute of Physiology, Faculty of Sciences, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
29
|
Chaves T, Fazekas CL, Horváth K, Correia P, Szabó A, Török B, Bánrévi K, Zelena D. Stress Adaptation and the Brainstem with Focus on Corticotropin-Releasing Hormone. Int J Mol Sci 2021; 22:ijms22169090. [PMID: 34445795 PMCID: PMC8396605 DOI: 10.3390/ijms22169090] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
Stress adaptation is of utmost importance for the maintenance of homeostasis and, therefore, of life itself. The prevalence of stress-related disorders is increasing, emphasizing the importance of exploratory research on stress adaptation. Two major regulatory pathways exist: the hypothalamic–pituitary–adrenocortical axis and the sympathetic adrenomedullary axis. They act in unison, ensured by the enormous bidirectional connection between their centers, the paraventricular nucleus of the hypothalamus (PVN), and the brainstem monoaminergic cell groups, respectively. PVN and especially their corticotropin-releasing hormone (CRH) producing neurons are considered to be the centrum of stress regulation. However, the brainstem seems to be equally important. Therefore, we aimed to summarize the present knowledge on the role of classical neurotransmitters of the brainstem (GABA, glutamate as well as serotonin, noradrenaline, adrenaline, and dopamine) in stress adaptation. Neuropeptides, including CRH, might be co-localized in the brainstem nuclei. Here we focused on CRH as its role in stress regulation is well-known and widely accepted and other CRH neurons scattered along the brain may also complement the function of the PVN. Although CRH-positive cells are present on some parts of the brainstem, sometimes even in comparable amounts as in the PVN, not much is known about their contribution to stress adaptation. Based on the role of the Barrington’s nucleus in micturition and the inferior olivary complex in the regulation of fine motoric—as the main CRH-containing brainstem areas—we might assume that these areas regulate stress-induced urination and locomotion, respectively. Further studies are necessary for the field.
Collapse
Affiliation(s)
- Tiago Chaves
- Laboratory of Behavioural and Stress Studies, Institute of Experimental Medicine, 1083 Budapest, Hungary; (T.C.); (C.L.F.); (K.H.); (P.C.); (A.S.); (B.T.); (K.B.)
- Janos Szentagothai School of Neurosciences, Semmelweis University, 1083 Budapest, Hungary
| | - Csilla Lea Fazekas
- Laboratory of Behavioural and Stress Studies, Institute of Experimental Medicine, 1083 Budapest, Hungary; (T.C.); (C.L.F.); (K.H.); (P.C.); (A.S.); (B.T.); (K.B.)
- Janos Szentagothai School of Neurosciences, Semmelweis University, 1083 Budapest, Hungary
| | - Krisztina Horváth
- Laboratory of Behavioural and Stress Studies, Institute of Experimental Medicine, 1083 Budapest, Hungary; (T.C.); (C.L.F.); (K.H.); (P.C.); (A.S.); (B.T.); (K.B.)
- Janos Szentagothai School of Neurosciences, Semmelweis University, 1083 Budapest, Hungary
| | - Pedro Correia
- Laboratory of Behavioural and Stress Studies, Institute of Experimental Medicine, 1083 Budapest, Hungary; (T.C.); (C.L.F.); (K.H.); (P.C.); (A.S.); (B.T.); (K.B.)
- Janos Szentagothai School of Neurosciences, Semmelweis University, 1083 Budapest, Hungary
| | - Adrienn Szabó
- Laboratory of Behavioural and Stress Studies, Institute of Experimental Medicine, 1083 Budapest, Hungary; (T.C.); (C.L.F.); (K.H.); (P.C.); (A.S.); (B.T.); (K.B.)
- Janos Szentagothai School of Neurosciences, Semmelweis University, 1083 Budapest, Hungary
| | - Bibiána Török
- Laboratory of Behavioural and Stress Studies, Institute of Experimental Medicine, 1083 Budapest, Hungary; (T.C.); (C.L.F.); (K.H.); (P.C.); (A.S.); (B.T.); (K.B.)
- Janos Szentagothai School of Neurosciences, Semmelweis University, 1083 Budapest, Hungary
| | - Krisztina Bánrévi
- Laboratory of Behavioural and Stress Studies, Institute of Experimental Medicine, 1083 Budapest, Hungary; (T.C.); (C.L.F.); (K.H.); (P.C.); (A.S.); (B.T.); (K.B.)
| | - Dóra Zelena
- Laboratory of Behavioural and Stress Studies, Institute of Experimental Medicine, 1083 Budapest, Hungary; (T.C.); (C.L.F.); (K.H.); (P.C.); (A.S.); (B.T.); (K.B.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- Correspondence:
| |
Collapse
|
30
|
Cao B, Zhao Y, Ren Z, McIntyre RS, Teopiz KM, Gao X, Ding L. Are Physical Activities Associated With Perceived Stress? The Evidence From the China Health and Nutrition Survey. Front Public Health 2021; 9:697484. [PMID: 34414158 PMCID: PMC8369204 DOI: 10.3389/fpubh.2021.697484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/07/2021] [Indexed: 11/23/2022] Open
Abstract
Psychological stress is a negative affective state. The association between physical activity and psychological stress was commonly reported in previous researches. Few published studies with large sample sizes have explored such an association in Chinese population. The current research aims to assess the association between perceived stress and physical activity preferences, as well as the association between risk of high perceived stress and physical activity behaviors (e.g., sports-, transport-, occupational-related physical activity, sedentary activities and time in bed). The data were collected from the China Health and Nutrition Survey (CHNS) in 2015. Generalized linear models and logistic regression models were used to explore the association between perceived stress and physical activity. In total, 11,066 participants were included in the current analysis. For physical activity preferences, we found that “like” preference of all six mentioned domains of activities contributed to lower perceived stress (i.e., lower perceived stress scale −14 score, all β < 0 and p < 0.05). For physical activity behaviors, the results indicated that none/low intensity physical activity behaviors (e.g., sports-, occupational- related, sedentary) were associated with risk of high perceived stress, except that low intensity of transport-related activities seemed to be protective from high stress. However, the association between intensity of physical activity behaviors and perceived stress was not simply the higher the better. High intensity of all these physical activity behaviors was also associated with high perceived stress. Our findings suggest that positive preferences and moderate physical activity behaviors were associated with low perceived stress. The findings herein highlight the effect of regulating physical activity on perceived stress, as well as inform potential strategies to reduce psychological stress.
Collapse
Affiliation(s)
- Bing Cao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, China.,National Demonstration Center for Experimental Psychology Education, Southwest University, Chongqing, China
| | - Yuxiao Zhao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, China
| | - Zhongyu Ren
- College of Physical Education, Southwest University, Chongqing, China
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Kayla M Teopiz
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Xiao Gao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, China.,National Demonstration Center for Experimental Psychology Education, Southwest University, Chongqing, China
| | - Ling Ding
- Pharmacy Department, The Central Hospital of Jiangjin, Chongqing, China
| |
Collapse
|
31
|
Sethna V, Siew J, Gudbrandsen M, Pote I, Wang S, Daly E, Deprez M, Pariante CM, Seneviratne G, Murphy DGM, Craig MC, McAlonan G. Maternal depression during pregnancy alters infant subcortical and midbrain volumes. J Affect Disord 2021; 291:163-170. [PMID: 34038833 DOI: 10.1016/j.jad.2021.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Maternal depression in pregnancy increases the risk for adverse neurodevelopmental outcomes in the offspring. The reason for this is unknown, however, one plausible mechanism may include the impact of maternal antenatal depression on infant brain. Nevertheless, relatively few studies have examined the brain anatomy of infants born to clinically diagnosed mothers. METHODS A legacy magnetic resonance imaging (MRI) dataset was used to compare regional brain volumes in 3-to-6-month-old infants born to women with a clinically confirmed diagnosis of major depressive disorder (MDD) during pregnancy (n = 31) and a reference sample of infants born to women without a current or past psychiatric diagnosis (n = 33). A method designed for analysis of low-resolution scans enabled examination of subcortical and midbrain regions previously found to be sensitive to the parent-child environment. RESULTS Compared with infants of non-depressed mothers, infants exposed to maternal antenatal depression had significantly larger subcortical grey matter volumes and smaller midbrain volumes. There was no association between gestational medication exposure and the infant regional brain volumes examined in our sample. LIMITATIONS Our scanning approach did not allow for an examination of fine-grained structural differences, and without repeated measures of brain volume, it is unknown whether the direction of reported associations are dependent on developmental stage. CONCLUSIONS Maternal antenatal depression is associated with an alteration in infant brain anatomy in early postnatal life; and that this is not accounted for by medication exposure. However, our study cannot address whether anatomical differences impact on future outcomes of the offspring.
Collapse
Affiliation(s)
- Vaheshta Sethna
- Sackler Institute for Translational Neurodevelopment, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK.
| | - Jasmine Siew
- Sackler Institute for Translational Neurodevelopment, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; Department of Experimental Clinical and Health Psychology, Research in Developmental Disorders Lab, Ghent University, Belgium
| | - Maria Gudbrandsen
- Sackler Institute for Translational Neurodevelopment, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Inês Pote
- Sackler Institute for Translational Neurodevelopment, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Siying Wang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, UK
| | - Eileen Daly
- Sackler Institute for Translational Neurodevelopment, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Maria Deprez
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, UK
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology & Perinatal Psychiatry Laboratory, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | | | - Declan G M Murphy
- Sackler Institute for Translational Neurodevelopment, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; NIHR Biomedical Research Centre for Mental Health at the South London and Maudsley NHS Foundation Trust and King's College London, UK
| | - Michael C Craig
- Sackler Institute for Translational Neurodevelopment, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Grainne McAlonan
- Sackler Institute for Translational Neurodevelopment, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; NIHR Biomedical Research Centre for Mental Health at the South London and Maudsley NHS Foundation Trust and King's College London, UK
| |
Collapse
|
32
|
Tillage RP, Foster SL, Lustberg D, Liles LC, McCann KE, Weinshenker D. Co-released norepinephrine and galanin act on different timescales to promote stress-induced anxiety-like behavior. Neuropsychopharmacology 2021; 46:1535-1543. [PMID: 33911187 PMCID: PMC8208976 DOI: 10.1038/s41386-021-01011-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 02/02/2023]
Abstract
Both the noradrenergic and galaninergic systems have been implicated in stress-related neuropsychiatric disorders, and these two neuromodulators are co-released from the stress-responsive locus coeruleus (LC); however, the individual contributions of LC-derived norepinephrine (NE) and galanin to behavioral stress responses are unclear. Here we aimed to disentangle the functional roles of co-released NE and galanin in stress-induced behavior. We used foot shock, optogenetics, and behavioral pharmacology in wild-type (WT) mice and mice lacking either NE (Dbh-/-) or galanin (GalcKO-Dbh) specifically in noradrenergic neurons to isolate the roles of these co-transmitters in regulating anxiety-like behavior in the elevated zero maze (EZM) either immediately or 24 h following stress. Foot shock and optogenetic LC stimulation produced immediate anxiety-like behavior in WT mice, and the effects of foot shock persisted for 24 h. NE-deficient mice were resistant to the anxiogenic effects of acute stress and optogenetic LC stimulation, while mice lacking noradrenergic-derived galanin displayed typical increases in anxiety-like behavior. However, when tested 24 h after foot shock, both Dbh-/- and GalcKO-Dbh mice lacked normal expression of anxiety-like behavior. Pharmacological rescue of NE, but not galanin, in knockout mice during EZM testing was anxiogenic. In contrast, restoring galanin, but not NE, signaling during foot shock normalized stress-induced anxiety-like behavior 24 h later. These results indicate that NE and noradrenergic-derived galanin play complementary, but distinguishable roles in behavioral responses to stress. NE is required for the expression of acute stress-induced anxiety, while noradrenergic-derived galanin mediates the development of more persistent responses following a stressor.
Collapse
Affiliation(s)
- Rachel P. Tillage
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University, Atlanta, GA USA
| | - Stephanie L. Foster
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University, Atlanta, GA USA
| | - Daniel Lustberg
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University, Atlanta, GA USA
| | - L. Cameron Liles
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University, Atlanta, GA USA
| | - Katharine E. McCann
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University, Atlanta, GA USA
| | | |
Collapse
|
33
|
Constantin S, Pizano K, Matson K, Shan Y, Reynolds D, Wray S. An Inhibitory Circuit From Brainstem to GnRH Neurons in Male Mice: A New Role for the RFRP Receptor. Endocrinology 2021; 162:6132086. [PMID: 33564881 PMCID: PMC8016070 DOI: 10.1210/endocr/bqab030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 12/31/2022]
Abstract
RFamide-related peptides (RFRPs, mammalian orthologs of gonadotropin-inhibitory hormone) convey circadian, seasonal, and social cues to the reproductive system. They regulate gonadotropin secretion by modulating gonadotropin-releasing hormone (GnRH) neurons via the RFRP receptor. Mice lacking this receptor are fertile but exhibit abnormal gonadotropin responses during metabolic challenges, such as acute fasting, when the normal drop in gonadotropin levels is delayed. Although it is known that these food intake signals to the reproductive circuit originate in the nucleus tractus solitarius (NTS) in the brainstem, the phenotype of the neurons conveying the signal remains unknown. Given that neuropeptide FF (NPFF), another RFamide peptide, resides in the NTS and can bind to the RFRP receptor, we hypothesized that NPFF may regulate GnRH neurons. To address this question, we used a combination of techniques: cell-attached electrophysiology on GnRH-driven green fluorescent protein-tagged neurons in acute brain slices; calcium imaging on cultured GnRH neurons; and immunostaining on adult brain tissue. We found (1) NPFF inhibits GnRH neuron excitability via the RFRP receptor and its canonical signaling pathway (Gi/o protein and G protein-coupled inwardly rectifying potassium channels), (2) NPFF-like fibers in the vicinity of GnRH neurons coexpress neuropeptide Y, (3) the majority of NPFF-like cell bodies in the NTS also coexpress neuropeptide Y, and (4) acute fasting increased NPFF-like immunoreactivity in the NTS. Together these data indicate that NPFF neurons within the NTS inhibit GnRH neurons, and thus reproduction, during fasting but prior to the energy deficit.
Collapse
Affiliation(s)
- Stephanie Constantin
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Katherine Pizano
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Kaya Matson
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Yufei Shan
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Daniel Reynolds
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
- Correspondence: Dr. Susan Wray, Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive MSC 3703, Building 35, Room 3A1012, Bethesda, MD 20892, USA.
| |
Collapse
|
34
|
Grueschow M, Stenz N, Thörn H, Ehlert U, Breckwoldt J, Brodmann Maeder M, Exadaktylos AK, Bingisser R, Ruff CC, Kleim B. Real-world stress resilience is associated with the responsivity of the locus coeruleus. Nat Commun 2021; 12:2275. [PMID: 33859187 PMCID: PMC8050280 DOI: 10.1038/s41467-021-22509-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 03/04/2021] [Indexed: 02/02/2023] Open
Abstract
Individuals may show different responses to stressful events. Here, we investigate the neurobiological basis of stress resilience, by showing that neural responsitivity of the noradrenergic locus coeruleus (LC-NE) and associated pupil responses are related to the subsequent change in measures of anxiety and depression in response to prolonged real-life stress. We acquired fMRI and pupillometry data during an emotional-conflict task in medical residents before they underwent stressful emergency-room internships known to be a risk factor for anxiety and depression. The LC-NE conflict response and its functional coupling with the amygdala was associated with stress-related symptom changes in response to the internship. A similar relationship was found for pupil-dilation, a potential marker of LC-NE firing. Our results provide insights into the noradrenergic basis of conflict generation, adaptation and stress resilience.
Collapse
Affiliation(s)
- Marcus Grueschow
- Zurich Center for Neuroeconomics (ZNE), Department of Economics, University of Zurich, Zurich, Switzerland.
| | - Nico Stenz
- Division of Experimental Psychopathology and Psychotherapy, Dept of Psychology, University of Zurich, Zurich, Switzerland
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Zurich, Switzerland
| | - Hanna Thörn
- Division of Experimental Psychopathology and Psychotherapy, Dept of Psychology, University of Zurich, Zurich, Switzerland
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Zurich, Switzerland
- Division of Clinical Psychology and Psychotherapy, Dept of Psychology, University of Zurich, Zurich, Switzerland
| | - Ulrike Ehlert
- Division of Clinical Psychology and Psychotherapy, Dept of Psychology, University of Zurich, Zurich, Switzerland
| | - Jan Breckwoldt
- Medical School, Deanery, University of Zurich, Zurich, Switzerland
| | | | | | - Roland Bingisser
- Department of Emergency Medicine, University Hospital Basel, Basel, Switzerland
| | - Christian C Ruff
- Zurich Center for Neuroeconomics (ZNE), Department of Economics, University of Zurich, Zurich, Switzerland
| | - Birgit Kleim
- Division of Experimental Psychopathology and Psychotherapy, Dept of Psychology, University of Zurich, Zurich, Switzerland.
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
35
|
Dygalo NN, Kalinina TS, Shishkina GT. Stress-induced expression pattern of glutamate signaling genes associated with anhedonia. Stress 2020; 23:700-707. [PMID: 32814471 DOI: 10.1080/10253890.2020.1812574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chronic stress can predispose vulnerable individuals to mood disorders, including depression. Glutamate, one of the key participants in this process, may exert both pathological and therapeutic psycho-emotional effects. However, the role of expression of genes encoding proteins that provide glutamatergic signal is still unclear. In this study, we attempted to distinguish changes in expression of glutamatergic genes associated with stress-induced anhedonia, a core symptom of depression, from those related to other stress-related effects. For this, expression of genes was compared between rats after a short-term stress, which did not yet cause depressive-like symptoms, and animals exposed chronically to different stressors that produce anhedonia-like responses. The changes in gene expression induced by chronic restraint or forced swimming concomitantly with anhedonia development demonstrated similar for both stressors patterns. Main features of the expression patterns include the decrease in mRNA levels for AMPA and NMDA subunits in the midbrain and hippocampus that is consistent with the hypothesis that "monoamine (serotonin)-Glutamate/GABA long neural circuit" involved in mood regulation. The decrease in expression of these subunits in the midbrain may attenuate glutamatergic drive on the serotonergic neurons promoting a shift of excitation/inhibition balance between glutamate and GABA in the forebrain regions resulting in anhedonia. In general, changes in expression of multiple genes involved in glutamatergic neurotransmission in the forebrain and brainstem regions suggest that stress-induced anhedonia may result from the network dysfunction of this neurotransmitter system.
Collapse
Affiliation(s)
- Nikolay N Dygalo
- Laboratory of Functional Neurogenomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Tatyana S Kalinina
- Laboratory of Functional Neurogenomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
| | - Galina T Shishkina
- Laboratory of Functional Neurogenomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
| |
Collapse
|
36
|
Herman JP, Nawreen N, Smail MA, Cotella EM. Brain mechanisms of HPA axis regulation: neurocircuitry and feedback in context Richard Kvetnansky lecture. Stress 2020; 23:617-632. [PMID: 33345670 PMCID: PMC8034599 DOI: 10.1080/10253890.2020.1859475] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
Regulation of stress reactivity is a fundamental priority of all organisms. Stress responses are critical for survival, yet can also cause physical and psychological damage. This review provides a synopsis of brain mechanisms designed to control physiological responses to stress, focusing primarily on glucocorticoid secretion via the hypothalamo-pituitary-adrenocortical (HPA) axis. The literature provides strong support for multi-faceted control of HPA axis responses, involving both direct and indirect actions at paraventricular nucleus (PVN) corticotropin releasing hormone neurons driving the secretory cascade. The PVN is directly excited by afferents from brainstem and hypothalamic circuits, likely relaying information on homeostatic challenge. Amygdala subnuclei drive HPA axis responses indirectly via disinhibition, mediated by GABAergic relays onto PVN-projecting neurons in the hypothalamus and bed nucleus of the stria terminalis (BST). Inhibition of stressor-evoked HPA axis responses is mediated by an elaborate network of glucocorticoid receptor (GR)-containing circuits, providing a distributed negative feedback signal that inhibits PVN neurons. Prefrontal and hippocampal neurons play a major role in HPA axis inhibition, again mediated by hypothalamic and BST GABAergic relays to the PVN. The complexity of the regulatory process suggests that information on stressors is integrated across functional disparate brain circuits prior to accessing the PVN, with regions such as the BST in prime position to relay contextual information provided by these sources into appropriate HPA activation. Dysregulation of the HPA in disease is likely a product of inappropriate checks and balances between excitatory and inhibitory inputs ultimately impacting PVN output.
Collapse
Affiliation(s)
- James P Herman
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
- Cincinnati Veterans Administration Medical Center, Cincinnati, OH, USA
| | - Nawshaba Nawreen
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Marissa A Smail
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Evelin M Cotella
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- Cincinnati Veterans Administration Medical Center, Cincinnati, OH, USA
| |
Collapse
|
37
|
Psilopanagioti A, Makrygianni M, Nikou S, Logotheti S, Papadaki H. Nucleobindin 2/nesfatin-1 expression and colocalisation with neuropeptide Y and cocaine- and amphetamine-regulated transcript in the human brainstem. J Neuroendocrinol 2020; 32:e12899. [PMID: 32902020 DOI: 10.1111/jne.12899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 11/26/2022]
Abstract
Feeding is a complex behaviour entailing elaborate interactions between forebrain, hypothalamic and brainstem neuronal circuits via multiple orexigenic and anorexigenic neuropeptides. Nucleobindin-2 (NUCB2)/nesfatin-1 is a negative regulator of food intake and body weight with a widespread distribution in rodent brainstem nuclei. However, its localisation pattern in the human brainstem is unknown. The present study aimed to explore NUCB2/nesfatin-1 immunoexpression in human brainstem nuclei and its possible correlation with body weight. Sections of human brainstem from 20 autopsy cases (13 males, seven females; eight normal weight, six overweight, six obese) were examined using immunohistochemistry and double immunofluorescence labelling. Strong immunoreactivity for NUCB2/nesfatin-1 was displayed in various brainstem areas, including the locus coeruleus, medial and lateral parabrachial nuclei, pontine nuclei, raphe nuclei, nucleus of the solitary tract, dorsal motor nucleus of vagus (10N), area postrema, hypoglossal nucleus, reticular formation, inferior olive, cuneate nucleus, and spinal trigeminal nucleus. NUCB2/nesfatin-1 was shown to extensively colocalise with neuropeptide Y and cocaine- and amphetamine-regulated transcript in the locus coeruleus, dorsal raphe nucleus and solitary tract. Interestingly, in the examined cases, NUCB2/nesfatin-1 protein expression was lower in obese than normal weight subjects in the solitary tract (P = 0.020). The findings of the present study provide neuroanatomical support for a role for NUCB2/nesfatin-1 in feeding behaviour and energy balance. The widespread distribution of NUCB2/nesfatin-1 in the human brainstem nuclei may be indicative of its pleiotropic effects on autonomic, neuroendocrine and behavioural processes. In the solitary tract, a key integrator of energy status, altered neurochemistry may contribute to obesity. Further research is necessary to decipher human brainstem energy homeostasis circuitry, which, despite its importance, remains inadequately characterised.
Collapse
Affiliation(s)
- Aristea Psilopanagioti
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, Patras, Greece
| | - Maria Makrygianni
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, Patras, Greece
| | - Sofia Nikou
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, Patras, Greece
| | - Souzana Logotheti
- Department of Pathology, School of Medicine, University of Patras, Patras, Greece
| | - Helen Papadaki
- Department of Anatomy-Histology-Embryology, School of Medicine, University of Patras, Patras, Greece
| |
Collapse
|
38
|
Goncharova ND. The HPA Axis under Stress and Aging: Individual Vulnerability is Associated with Behavioral Patterns and Exposure Time. Bioessays 2020; 42:e2000007. [PMID: 32666621 DOI: 10.1002/bies.202000007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/05/2020] [Indexed: 12/16/2022]
Abstract
With aging, incidence of severe stress-related diseases increases. However, mechanisms, underlying individual vulnerability to stress and age-related diseases are not clear. The goal of this review is to analyze finding from the recent literature on age-related characteristics of the hypothalamic-pituitary-adrenal (HPA) axis associated with stress reactivity in animals that show behavioral signs of anxiety and depression under mild stress, and in human patients with anxiety disorders and depression with emphasis on the impact of the circadian rhythm and the negative feedback mechanisms involved in the stress response. One can conclude that HPA axis reaction to psycho-emotional stress, at least acute stress, increases in the aged individuals with anxiety and depression behavior. Elevated stress reactivity is associated with disruption of the circadian rhythm and the mineralocorticoid receptor-mediated glucocorticoid negative feedback. The disordered function of the HPA axis in individuals with anxiety and depression behavior can contribute to aging-related pathology.
Collapse
Affiliation(s)
- Nadezhda D Goncharova
- Laboratory of Experimental Endocrinology, Research Institute of Medical Primatology, 177 Mira Street, Veseloye, Adler, Sochi, Krasnodar, 354376, Russia
| |
Collapse
|
39
|
PPG neurons in the nucleus of the solitary tract modulate heart rate but do not mediate GLP-1 receptor agonist-induced tachycardia in mice. Mol Metab 2020; 39:101024. [PMID: 32446875 PMCID: PMC7317700 DOI: 10.1016/j.molmet.2020.101024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 01/07/2023] Open
Abstract
Objective Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are used as anti-diabetic drugs and are approved for obesity treatment. However, GLP-1RAs also affect heart rate (HR) and arterial blood pressure (ABP) in rodents and humans. Although the activation of GLP-1 receptors (GLP-1R) is known to increase HR, the circuits recruited are unclear, and in particular, it is unknown whether GLP-1RAs activate preproglucagon (PPG) neurons, the brain source of GLP-1, to elicit these effects. Methods We investigated the effect of GLP-1RAs on heart rate in anaesthetized adult mice. In a separate study, we manipulated the activity of nucleus tractus solitarius (NTS) PPG neurons (PPGNTS) in awake, freely behaving transgenic Glu-Cre mice implanted with biotelemetry probes and injected with AAV-DIO-hM3Dq:mCherry or AAV-mCherry-FLEX-DTA. Results Systemic administration of the GLP-1RA Ex-4 increased resting HR in anaesthetized or conscious mice, but had no effect on ABP in conscious mice. This effect was abolished by β-adrenoceptor blockade with atenolol, but unaffected by the muscarinic antagonist atropine. Furthermore, Ex-4-induced tachycardia persisted when PPGNTS neurons were ablated, and Ex-4 did not induce expression of the neuronal activity marker cFos in PPGNTS neurons. PPGNTS ablation or acute chemogenetic inhibition of these neurons via hM4Di receptors had no effect on resting HR. In contrast, chemogenetic activation of PPGNTS neurons increased resting HR. Furthermore, the application of GLP-1 within the subarachnoid space of the middle thoracic spinal cord, a major projection target of PPG neurons, increased HR. Conclusions These results demonstrate that both systemic application of Ex-4 or GLP-1 and chemogenetic activation of PPGNTS neurons increases HR. Ex-4 increases the activity of cardiac sympathetic preganglionic neurons of the spinal cord without recruitment of PPGNTS neurons, and thus likely recapitulates the physiological effects of PPG neuron activation. These neurons therefore do not play a significant role in controlling resting HR and ABP but are capable of inducing tachycardia and so are likely involved in cardiovascular responses to acute stress. Activation of PPG neurons triggers increases in heart rate in mice. PPG neurons do not provide a tonic sympathetic drive to the heart. The tachycardic effect of systemic Ex-4 is not mediated by PPG neurons. GLP-1 receptor activation has a sympathoexcitatory effect that increases heart rate. Local activation of GLP-1R in the spinal cord is sufficient to elicit tachycardia.
Collapse
|
40
|
Chen P, Lou S, Huang ZH, Wang Z, Shan QH, Wang Y, Yang Y, Li X, Gong H, Jin Y, Zhang Z, Zhou JN. Prefrontal Cortex Corticotropin-Releasing Factor Neurons Control Behavioral Style Selection under Challenging Situations. Neuron 2020; 106:301-315.e7. [DOI: 10.1016/j.neuron.2020.01.033] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 10/31/2019] [Accepted: 01/24/2020] [Indexed: 02/07/2023]
|
41
|
Hanchate NK, Lee EJ, Ellis A, Kondoh K, Kuang D, Basom R, Trapnell C, Buck LB. Connect-seq to superimpose molecular on anatomical neural circuit maps. Proc Natl Acad Sci U S A 2020; 117:4375-4384. [PMID: 32034095 PMCID: PMC7049128 DOI: 10.1073/pnas.1912176117] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The mouse brain contains about 75 million neurons interconnected in a vast array of neural circuits. The identities and functions of individual neuronal components of most circuits are undefined. Here we describe a method, termed "Connect-seq," which combines retrograde viral tracing and single-cell transcriptomics to uncover the molecular identities of upstream neurons in a specific circuit and the signaling molecules they use to communicate. Connect-seq can generate a molecular map that can be superimposed on a neuroanatomical map to permit molecular and genetic interrogation of how the neuronal components of a circuit control its function. Application of this method to hypothalamic neurons controlling physiological responses to fear and stress reveals subsets of upstream neurons that express diverse constellations of signaling molecules and can be distinguished by their anatomical locations.
Collapse
Affiliation(s)
- Naresh K Hanchate
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Eun Jeong Lee
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Andria Ellis
- Department of Genome Sciences, University of Washington, Seattle, WA 98115
| | - Kunio Kondoh
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Donghui Kuang
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Ryan Basom
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA 98115
- The Brotman Baty Institute for Precision Medicine, Seattle, WA 98195
| | - Linda B Buck
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109;
- The Brotman Baty Institute for Precision Medicine, Seattle, WA 98195
| |
Collapse
|
42
|
Singh K, Indovina I, Augustinack JC, Nestor K, García-Gomar MG, Staab JP, Bianciardi M. Probabilistic Template of the Lateral Parabrachial Nucleus, Medial Parabrachial Nucleus, Vestibular Nuclei Complex, and Medullary Viscero-Sensory-Motor Nuclei Complex in Living Humans From 7 Tesla MRI. Front Neurosci 2020; 13:1425. [PMID: 32038134 PMCID: PMC6989551 DOI: 10.3389/fnins.2019.01425] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/17/2019] [Indexed: 11/13/2022] Open
Abstract
The lateral parabrachial nucleus, medial parabrachial nucleus, vestibular nuclei complex, and medullary viscero-sensory-motor (VSM) nuclei complex (the latter including among others the solitary nucleus, vagus nerve nucleus, and hypoglossal nucleus) are anatomically and functionally connected brainstem gray matter structures that convey signals across multiple modalities between the brain and the spinal cord to regulate vital bodily functions. It is remarkably difficult to precisely extrapolate the location of these nuclei from ex vivo atlases to conventional 3 Tesla in vivo images; thus, a probabilistic brainstem template in stereotaxic neuroimaging space in living humans is needed. We delineated these nuclei using single-subject high contrast 1.1 mm isotropic resolution 7 Tesla MRI images. After precise coregistration of nuclei labels to stereotaxic space, we generated a probabilistic template of their anatomical locations. Finally, we validated the nuclei labels in the template by assessing their inter-rater agreement, consistency across subjects and volumes. We also performed a preliminary comparison of their location and microstructural properties to histologic sections of a postmortem human brainstem specimen. In future, the resulting probabilistic template of these brainstem nuclei in stereotaxic space may assist researchers and clinicians in evaluating autonomic, vestibular and VSM nuclei structure, function and connectivity in living humans using conventional 3 Tesla MRI scanners.
Collapse
Affiliation(s)
- Kavita Singh
- Brainstem Imaging Laboratory, Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Iole Indovina
- Department of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences, Rome, Italy.,Laboratory of Neuromotor Physiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Jean C Augustinack
- Laboratory for Computational Neuroimaging, Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Kimberly Nestor
- Laboratory for Computational Neuroimaging, Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - María G García-Gomar
- Brainstem Imaging Laboratory, Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Jeffrey P Staab
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States.,Department of Otorhinolaryngology - Head and Neck Surgery, Mayo Clinic, Rochester, MN, United States
| | - Marta Bianciardi
- Brainstem Imaging Laboratory, Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Rouhiainen A, Kulesskaya N, Mennesson M, Misiewicz Z, Sipilä T, Sokolowska E, Trontti K, Urpa L, McEntegart W, Saarnio S, Hyytiä P, Hovatta I. The bradykinin system in stress and anxiety in humans and mice. Sci Rep 2019; 9:19437. [PMID: 31857655 PMCID: PMC6923437 DOI: 10.1038/s41598-019-55947-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 11/27/2019] [Indexed: 01/06/2023] Open
Abstract
Pharmacological research in mice and human genetic analyses suggest that the kallikrein-kinin system (KKS) may regulate anxiety. We examined the role of the KKS in anxiety and stress in both species. In human genetic association analysis, variants in genes for the bradykinin precursor (KNG1) and the bradykinin receptors (BDKRB1 and BDKRB2) were associated with anxiety disorders (p < 0.05). In mice, however, neither acute nor chronic stress affected B1 receptor gene or protein expression, and B1 receptor antagonists had no effect on anxiety tests measuring approach-avoidance conflict. We thus focused on the B2 receptor and found that mice injected with the B2 antagonist WIN 64338 had lowered levels of a physiological anxiety measure, the stress-induced hyperthermia (SIH), vs controls. In the brown adipose tissue, a major thermoregulator, WIN 64338 increased expression of the mitochondrial regulator Pgc1a and the bradykinin precursor gene Kng2 was upregulated after cold stress. Our data suggests that the bradykinin system modulates a variety of stress responses through B2 receptor-mediated effects, but systemic antagonists of the B2 receptor were not anxiolytic in mice. Genetic variants in the bradykinin receptor genes may predispose to anxiety disorders in humans by affecting their function.
Collapse
Affiliation(s)
- Ari Rouhiainen
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Natalia Kulesskaya
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Marie Mennesson
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland.,Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland.,SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Neuroscience Center, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Zuzanna Misiewicz
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland.,Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland
| | - Tessa Sipilä
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Ewa Sokolowska
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Kalevi Trontti
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland.,Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland.,SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Neuroscience Center, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Lea Urpa
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - William McEntegart
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Suvi Saarnio
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Petri Hyytiä
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland
| | - Iiris Hovatta
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland. .,Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland. .,SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. .,Neuroscience Center, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
44
|
Sharma RK, Yang T, Oliveira AC, Lobaton GO, Aquino V, Kim S, Richards EM, Pepine CJ, Sumners C, Raizada MK. Microglial Cells Impact Gut Microbiota and Gut Pathology in Angiotensin II-Induced Hypertension. Circ Res 2019; 124:727-736. [PMID: 30612527 DOI: 10.1161/circresaha.118.313882] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
RATIONALE Increased microglial activation and neuroinflammation within autonomic brain regions have been implicated in sustained hypertension, and their inhibition by minocycline-an anti-inflammatory antibiotic-produces beneficial effects. These observations led us to propose a dysfunctional brain-gut communication hypothesis for hypertension. However, it has been difficult to reconcile whether an anti-inflammatory or antimicrobial action is the primary beneficial effect of minocycline in hypertension. Accordingly, we utilized chemically modified tetracycline-3 (CMT-3)-a derivative of tetracycline that has potent anti-inflammatory activity-to address this question. OBJECTIVE Test the hypothesis that central administration of CMT-3 would inhibit microglial activation, attenuate neuroinflammation, alter selective gut microbial communities, protect the gut wall from developing hypertension-associated pathology, and attenuate hypertension. METHODS AND RESULTS Rats were implanted with radiotelemetry devices for recording mean arterial pressure. Ang II (angiotensin II) was infused subcutaneously using osmotic mini-pumps to induce hypertension. Another osmotic mini-pump was surgically implanted to infuse CMT-3 intracerebroventricularly. Intracerebroventricular CMT- 3 infusion was also investigated in SHR (spontaneously hypertensive rats). Physiological, pathological, immunohistological parameters, and fecal microbiota were analyzed. Intracerebroventricular CMT-3 significantly inhibited Ang II-induced increases in number of microglia, their activation, and proinflammatory cytokines in the paraventricular nucleus of hypothalamus. Further, intracerebroventricular CMT-3 attenuated increased mean arterial pressure, normalized sympathetic activity, and left ventricular hypertrophy in Ang II rats, as well as in the SHR. Finally, CMT-3 beneficially restored certain gut microbial communities altered by Ang II and attenuated pathological alterations in gut wall. CONCLUSIONS These observations demonstrate that inhibition of microglial activation alone was sufficient to induce significant antihypertensive effects. This was associated with unique changes in gut microbial communities and profound attenuation of gut pathology. They suggest, for the first time, a link between microglia and certain microbial communities that may have implications for treatment of hypertension.
Collapse
Affiliation(s)
- Ravindra K Sharma
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Tao Yang
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Aline C Oliveira
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Gilberto O Lobaton
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Victor Aquino
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Seungbum Kim
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Elaine M Richards
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Carl J Pepine
- Department of Medicine (C.J.P.), College of Medicine, University of Florida, Gainesville
| | - Colin Sumners
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| | - Mohan K Raizada
- From the Department of Physiology and Functional Genomics (R.K.S., T.Y., A.C.O., G.O.L., V.A., S.K., E.M.R., C.S., M.K.R.), College of Medicine, University of Florida, Gainesville
| |
Collapse
|
45
|
Plasencia G, Luedicke JM, Nazarloo HP, Carter CS, Ebner NC. Plasma oxytocin and vasopressin levels in young and older men and women: Functional relationships with attachment and cognition. Psychoneuroendocrinology 2019; 110:104419. [PMID: 31606581 PMCID: PMC6943921 DOI: 10.1016/j.psyneuen.2019.104419] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/19/2019] [Accepted: 08/22/2019] [Indexed: 01/17/2023]
Abstract
A growing literature associates the neuropeptides oxytocin (OT) and arginine vasopressin (AVP) with affiliative and cognitive outcomes. The majority of this work in humans, however, considers these neuropeptides separately. Also, despite evidence that OT and AVP interact with gonadal hormones, still warranted is an examination of sex and age variations in endogenous neuropeptide levels, their interrelations, and their functional relationships with attachment and cognition in humans. This study measured endogenous plasma OT and AVP levels in generally healthy young (18-31 years) and older (63-81 years) men and women to (i) determine levels of and interrelations between OT and AVP; (ii) explore functional relationships with self-reported attachment (attachment anxiety and avoidance) and performance-based cognition (processing speed, verbal memory); and (iii) identify variations in these effects by sex and age. We observed sex- and age-differential patterns of results: Women had higher plasma OT levels than men and older adults had higher plasma AVP levels than young adults. The two neuropeptides were highly negatively intercorrelated across all groups. Functionally, higher AVP levels were associated with greater attachment anxiety and higher OT and lower AVP levels were associated with faster sensorimotor processing speed, with sex and age moderating these effects. This integrated approach identifies variations in endogenous peripheral neuropeptide levels in humans, supporting their sex- and age-specific role as "difference makers" in attachment and cognition.
Collapse
Affiliation(s)
- Gabriela Plasencia
- Department of Psychology, University of Florida, P.O. Box 112250, Gainesville, FL, 32611-2250, USA; Stritch School of Medicine, Loyola University of Chicago, 2160 S 1st Ave, Maywood, IL, 60153, USA.
| | - Joerg M Luedicke
- Department of Psychology, University of Florida, P.O. Box 112250, Gainesville, FL, 32611-2250, USA.
| | | | - C Sue Carter
- Kinsey Institute, Indiana University, Bloomington, IN, 47405, USA.
| | - Natalie C Ebner
- Department of Psychology, University of Florida, P.O. Box 112250, Gainesville, FL, 32611-2250, USA; Department of Aging and Geriatric Research, Institute on Aging, University of Florida, 2004 Mowry Road, Gainesville, FL, 32611, USA.
| |
Collapse
|
46
|
Daviu N, Bruchas MR, Moghaddam B, Sandi C, Beyeler A. Neurobiological links between stress and anxiety. Neurobiol Stress 2019; 11:100191. [PMID: 31467945 PMCID: PMC6712367 DOI: 10.1016/j.ynstr.2019.100191] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/18/2019] [Accepted: 08/02/2019] [Indexed: 11/21/2022] Open
Abstract
Stress and anxiety have intertwined behavioral and neural underpinnings. These commonalities are critical for understanding each state, as well as their mutual interactions. Grasping the mechanisms underlying this bidirectional relationship will have major clinical implications for managing a wide range of psychopathologies. After briefly defining key concepts for the study of stress and anxiety in pre-clinical models, we present circuit, as well as cellular and molecular mechanisms involved in either or both stress and anxiety. First, we review studies on divergent circuits of the basolateral amygdala (BLA) underlying emotional valence processing and anxiety-like behaviors, and how norepinephrine inputs from the locus coeruleus (LC) to the BLA are responsible for acute-stress induced anxiety. We then describe recent studies revealing a new role for mitochondrial function within the nucleus accumbens (NAc), defining individual trait anxiety in rodents, and participating in the link between stress and anxiety. Next, we report findings on the impact of anxiety on reward encoding through alteration of circuit dynamic synchronicity. Finally, we present work unravelling a new role for hypothalamic corticotropin-releasing hormone (CRH) neurons in controlling anxiety-like and stress-induce behaviors. Altogether, the research reviewed here reveals circuits sharing subcortical nodes and underlying the processing of both stress and anxiety. Understanding the neural overlap between these two psychobiological states, might provide alternative strategies to manage disorders such as post-traumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Nuria Daviu
- Hotchkiss Brain Institute. Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Michael R. Bruchas
- Department of Anesthesiology and Pain Medicine. Center for Neurobiology of Addiction, Pain, and Emotion. University of Washington. 1959 NE Pacific Street, J-wing Health Sciences. Seattle, WA 98195, USA
| | - Bita Moghaddam
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, CH, 1015, Lausanne, Switzerland
| | - Anna Beyeler
- Neurocentre Magendie, INSERM 1215, Université de Bordeaux, 146 Rue Léo Saignat, 33000 Bordeaux, France
| |
Collapse
|
47
|
Synaptic Inputs to the Mouse Dorsal Vagal Complex and Its Resident Preproglucagon Neurons. J Neurosci 2019; 39:9767-9781. [PMID: 31666353 PMCID: PMC6891065 DOI: 10.1523/jneurosci.2145-19.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/07/2019] [Accepted: 10/13/2019] [Indexed: 12/17/2022] Open
Abstract
Stress responses are coordinated by widespread neural circuits. Homeostatic and psychogenic stressors activate preproglucagon (PPG) neurons in the caudal nucleus of the solitary tract (cNTS) that produce glucagon-like peptide-1; published work in rodents indicates that these neurons play a crucial role in stress responses. While the axonal targets of PPG neurons are well established, their afferent inputs are unknown. Stress responses are coordinated by widespread neural circuits. Homeostatic and psychogenic stressors activate preproglucagon (PPG) neurons in the caudal nucleus of the solitary tract (cNTS) that produce glucagon-like peptide-1; published work in rodents indicates that these neurons play a crucial role in stress responses. While the axonal targets of PPG neurons are well established, their afferent inputs are unknown. Here we use retrograde tracing with cholera toxin subunit b to show that the cNTS in male and female mice receives axonal inputs similar to those reported in rats. Monosynaptic and polysynaptic inputs specific to cNTS PPG neurons were revealed using Cre-conditional pseudorabies and rabies viruses. The most prominent sources of PPG monosynaptic input include the lateral (LH) and paraventricular (PVN) nuclei of the hypothalamus, parasubthalamic nucleus, lateral division of the central amygdala, and Barrington's nucleus (Bar). Additionally, PPG neurons receive monosynaptic vagal sensory input from the nodose ganglia and spinal sensory input from the dorsal horn. Sources of polysynaptic input to cNTS PPG neurons include the hippocampal formation, paraventricular thalamus, and prefrontal cortex. Finally, cNTS-projecting neurons within PVN, LH, and Bar express the activation marker cFOS in mice after restraint stress, identifying them as potential sources of neurogenic stress-induced recruitment of PPG neurons. In summary, cNTS PPG neurons in mice receive widespread monosynaptic and polysynaptic input from brain regions implicated in coordinating behavioral and physiological stress responses, as well as from vagal and spinal sensory neurons. Thus, PPG neurons are optimally positioned to integrate signals of homeostatic and psychogenic stress. SIGNIFICANCE STATEMENT Recent research has indicated a crucial role for glucagon-like peptide-1-producing preproglucagon (PPG) neurons in regulating both appetite and behavioral and autonomic responses to acute stress. Intriguingly, the central glucagon-like peptide-1 system defined in rodents is conserved in humans, highlighting the translational importance of understanding its anatomical organization. Findings reported here indicate that PPG neurons receive significant monosynaptic and polysynaptic input from brain regions implicated in autonomic and behavioral responses to stress, as well as direct input from vagal and spinal sensory neurons. Improved understanding of the neural pathways underlying the recruitment of PPG neurons may facilitate the development of novel therapies for the treatment of stress-related disorders.
Collapse
|
48
|
O’Mara SM, Aggleton JP. Space and Memory (Far) Beyond the Hippocampus: Many Subcortical Structures Also Support Cognitive Mapping and Mnemonic Processing. Front Neural Circuits 2019; 13:52. [PMID: 31447653 PMCID: PMC6692652 DOI: 10.3389/fncir.2019.00052] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/22/2019] [Indexed: 11/13/2022] Open
Abstract
Memory research remains focused on just a few brain structures-in particular, the hippocampal formation (the hippocampus and entorhinal cortex). Three key discoveries promote this continued focus: the striking demonstrations of enduring anterograde amnesia after bilateral hippocampal damage; the realization that synapses in the hippocampal formation are plastic e.g., when responding to short bursts of patterned stimulation ("long-term potentiation" or LTP); and the discovery of a panoply of spatially-tuned cells, principally surveyed in the hippocampal formation (place cells coding for position; head-direction cells, providing compass-like information; and grid cells, providing a metric for 3D space). Recent anatomical, behavioral, and electrophysiological work extends this picture to a growing network of subcortical brain structures, including the anterior thalamic nuclei, rostral midline thalamic nuclei, and the claustrum. There are, for example, spatially-tuned cells in all of these regions, including cells with properties similar to place cells of the hippocampus proper. These findings add new perspectives to what had been originally been proposed-but often overlooked-half a century ago: that damage to an extended network of structures connected to the hippocampal formation results in diencephalic amnesia. We suggest these new findings extend spatial signaling in the brain far beyond the hippocampal formation, with profound implications for theories of the neural bases of spatial and mnemonic functions.
Collapse
Affiliation(s)
- Shane M. O’Mara
- School of Psychology and Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - John P. Aggleton
- School of Psychology, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
49
|
McNeal N, Watanasriyakul WT, Normann MC, Akinbo OI, Dagner A, Ihm E, Wardwell J, Grippo AJ. The negative effects of social bond disruption are partially ameliorated by sertraline administration in prairie voles. Auton Neurosci 2019; 219:5-18. [PMID: 31122602 PMCID: PMC6540807 DOI: 10.1016/j.autneu.2019.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 12/11/2022]
Abstract
Negative social experiences influence both depression and cardiovascular dysfunction. Many individuals who experience negative mood states or cardiovascular conditions have limited social support. Therefore, investigation of drug treatments that may protect against the consequences of social stress will aid in designing effective treatment strategies. The current study used an animal model to evaluate the protective effect of sertraline administration on behavioral and cardiovascular consequences of social stress. Specifically, male prairie voles (Microtus ochrogaster), which are socially monogamous rodents that share several behavioral and physiological characteristics with humans, were isolated from a socially-bonded female partner, and treated with sertraline (16 mg/kg/day, ip) or vehicle during isolation. Unexpectedly, sertraline did not protect against depression-relevant behaviors, and it was associated with increased short- and long-term heart rate responses. However, sertraline administration improved heart rate variability recovery following a behavioral stressor, including increased parasympathetic regulation, and altered long-term neuronal activity in brain regions that modulate autonomic control and stress reactivity. These results indicate that sertraline may partially protect against the consequences of social stressors, and suggest a mechanism through which sertraline may beneficially influence neurobiological control of cardiac function.
Collapse
Affiliation(s)
- Neal McNeal
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | | | - Marigny C Normann
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Oreoluwa I Akinbo
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Ashley Dagner
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Elliott Ihm
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Joshua Wardwell
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Angela J Grippo
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA.
| |
Collapse
|
50
|
Abstract
Purpose of Review Hypertension is related to impaired metabolic homeostasis and can be regarded as a metabolic disorder. This review presents possible mechanisms by which metabolic disorders increase blood pressure (BP) and discusses the importance of the gut as a novel modulator of BP. Recent Findings Obesity and high salt intake are major risk factors for hypertension. There is a hypothesis of “salt-induced obesity”; i.e., high salt intake may tie to obesity. Heightened sympathetic nervous system (SNS) activity, especially in the kidney and brain, increases BP in obese patients. Adipokines, including adiponectin and leptin, and renin-angiotensin-aldosterone system (RAAS) contribute to hypertension. Adiponectin induced by a high-salt diet may decrease sodium/glucose cotransporter (SGLT) 2 expression in the kidney, which results in reducing BP. High salt can change secretions of adipokines and RAAS-related components. Evidence has been accumulating linking the gastrointestinal tract to BP. Glucagon-like peptide-1 (GLP-1) and ghrelin decrease BP in both rodents and humans. The sweet taste receptor in enteroendocrine cells increases SGLT1 expression and stimulates sodium/glucose absorption. Roux-en-Y gastric bypass improves glycemic and BP control due to reducing the activity of SGLT1. Na/H exchanger isoform 3 (NHE3) increases BP by stimulating the intestinal absorption of sodium. Gastrin functions as an intestinal sodium taste sensor and inhibits NHE3 activity. Intestinal mineralocorticoid receptors also regulate sodium absorption and BP due to changing ENaC activity. Gastric sensing of sodium induces natriuresis, and gastric distension increases BP. Changes in the composition and function of gut microbiota contribute to hypertension. A high-salt/fat diet may disrupt the gut barrier, which results in systemic inflammation, insulin resistance, and increased BP. Gut microbiota regulates BP by secreting vasoactive hormones and short-chain fatty acids. BP-lowering effects of probiotics and antibiotics have been reported. Bariatric surgery improves metabolic disorders and hypertension due to increasing GLP-1 secretion, decreasing leptin secretion and SNS activity, and changing gut microbiome composition. Strategies targeting the gastrointestinal system may be therapeutic options for improving metabolic abnormalities and reducing BP in humans. Summary SNS, brain, adipocytes, RAAS, the kidney, the gastrointestinal tract, and microbiota play important roles in regulating BP. Most notably, the gut could be a novel target for treatment of hypertension as a metabolic disorder.
Collapse
|