1
|
Pawlik MT, Rinneberg G, Koch A, Meyringer H, Loew TH, Kjellberg A. Is there a rationale for hyperbaric oxygen therapy in the patients with Post COVID syndrome? : A critical review. Eur Arch Psychiatry Clin Neurosci 2024; 274:1797-1817. [PMID: 39545965 PMCID: PMC11579208 DOI: 10.1007/s00406-024-01911-y] [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: 05/20/2023] [Accepted: 09/16/2024] [Indexed: 11/17/2024]
Abstract
The SARS-CoV-2 pandemic has resulted in 762 million infections worldwide from 2020 to date, of which approximately ten percent are suffering from the effects after infection in 2019 (COVID-19) [1, 40]. In Germany, it is now assumed that at least one million people suffer from post-COVID condition with long-term consequences. These have been previously reported in diseases like Myalgic Encephalomyelitis (ME) and Chronic Fatigue Syndrome (CFS). Symptoms show a changing variability and recent surveys in the COVID context indicate that 10-30 % of outpatients, 50 to 70% of hospitalised patients suffer from sequelae. Recent data suggest that only 13% of all ill people were completely free of symptoms after recovery [3, 9]. Current hypotheses consider chronic inflammation, mitochondrial dysfunction, latent viral persistence, autoimmunity, changes of the human microbiome or multilocular sequelae in various organ system after infection. Hyperbaric oxygen therapy (HBOT) is applied since 1957 for heart surgery, scuba dive accidents, CO intoxication, air embolisms and infections with anaerobic pathogens. Under hyperbaric pressure, oxygen is physically dissolved in the blood in higher concentrations and reaches levels four times higher than under normobaric oxygen application. Moreover, the alternation of hyperoxia and normoxia induces a variety of processes at the cellular level, which improves oxygen supply in areas of locoregional hypoxia. Numerous target gene effects on new vessel formation, anti-inflammatory and anti-oedematous effects have been demonstrated [74]. The provision of intermittently high, local oxygen concentrations increases repair and regeneration processes and normalises the predominance of hyperinflammation. At present time only one prospective, randomized and placebo-controlled study exists with positive effects on global cognitive function, attention and executive function, psychiatric symptoms and pain interference. In conclusion, up to this date HBO is the only scientifically proven treatment in a prospective randomized controlled trial to be effective for cognitive improvement, regeneration of brain network and improvement of cardiac function. HBOT may have not only theoretical but also potential impact on targets of current pathophysiology of Post COVID condition, which warrants further scientific studies in patients.
Collapse
Affiliation(s)
- M T Pawlik
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany.
- Institute of Experimental Medicine, Christian-Albrechts-University of Kiel c/o German Naval Medical Institute, Kronshagen, Germany.
| | - G Rinneberg
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany
| | - A Koch
- Institute of Experimental Medicine, Christian-Albrechts-University of Kiel c/o German Naval Medical Institute, Kronshagen, Germany
| | - H Meyringer
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany
| | - T H Loew
- Department of Psychosomatic Medicine, University Hospital Regensburg, Regensburg, Germany
| | - A Kjellberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
- Perioperative Medicine and Intensive Care, Medical Unit Intensive Care and Thoracic surgery, Karolinska University Hospital, Stockholm, Sweden
| |
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 2024; 91:102076. [PMID: 39603502 DOI: 10.1016/j.molmet.2024.102076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [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
|
Abdel-Rahman M, Hussein AA, Ahmed-Farid OA, Sawi AA, Abdel Moneim AE. Intermittent fasting alerts neurotransmitters and oxidant/antioxidant status in the brain of rats. Metab Brain Dis 2024; 39:1291-1305. [PMID: 39292431 PMCID: PMC11513736 DOI: 10.1007/s11011-024-01415-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 08/14/2024] [Indexed: 09/19/2024]
Abstract
Several recent studies have attempted to understand how fasting has benefits for body health, especially the nervous system. To evaluate the impact of intermittent fasting on body weight, brain neurotransmitters, brain oxidative stress, and brain-derived neurotrophic factor (BDNF) in several areas of the brain, this study was conducted in rats. Thirty male Wistar rats were randomly divided into two groups. Group 1 (15 rats) served as the control and group 2 (15 rats) underwent intermittent fasting (IF; 24 h) for 1, 7, or 15 days. The findings demonstrated that intermittent fasting significantly reduced body weight. In this sense, brain monoamines and amino acids, namely dopamine, glutamate, aspartate, and oxidative stress markers (malondialdehyde and nitric oxide), decreased significantly after 1 day of IF. However, norepinephrine, serotonin, gamma-amino butyric acid, and glycine increased significantly. Additionally, glutathione levels were markedly elevated in IF. Surprisingly, the neuromodulatory effect of intermittent fasting fluctuates depending on the IF period. To support this fluctuation, BDNF levels increased after 1 day in the hippocampus and decreased after 15 days of intermittent fasting in all areas of the brain tested. In conclusion, our results show that intermittent fasting has beneficial influences on the brain; however, prolonged intermittent fasting can also induce some unfavorable physiological outcomes that prevent optimal neurological function.
Collapse
Affiliation(s)
- Mona Abdel-Rahman
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Aida A Hussein
- Zoology and Entomology Department, Faculty of Science, Suez University, Suez, Egypt
| | - Omar A Ahmed-Farid
- Department of Physiology, National Organization for Drug Control and Research (NODCAR), Giza, Giza Governorate, Egypt
| | - Abdullah A Sawi
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | | |
Collapse
|
4
|
Tansey MG, Boles J, Holt J, Cole C, Neighbarger N, Urs N, Uriarte-Huarte O. Locus coeruleus injury modulates ventral midbrain neuroinflammation during DSS-induced colitis. RESEARCH SQUARE 2024:rs.3.rs-3952442. [PMID: 38559083 PMCID: PMC10980147 DOI: 10.21203/rs.3.rs-3952442/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Parkinson's disease (PD) is characterized by a decades-long prodrome, consisting of a collection of non-motor symptoms that emerges prior to the motor manifestation of the disease. Of these non-motor symptoms, gastrointestinal dysfunction and deficits attributed to central norepinephrine (NE) loss, including mood changes and sleep disturbances, are frequent in the PD population and emerge early in the disease. Evidence is mounting that injury and inflammation in the gut and locus coeruleus (LC), respectively, underlie these symptoms, and the injury of these systems is central to the progression of PD. In this study, we generate a novel two-hit mouse model that captures both features, using dextran sulfate sodium (DSS) to induce gut inflammation and N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) to lesion the LC. We first confirmed the specificity of DSP-4 for central NE using neurochemical methods and fluorescence light-sheet microscopy of cleared tissue, and established that DSS-induced outcomes in the periphery, including weight loss, gross indices of gut injury and systemic inflammation, the loss of tight junction proteins in the colonic epithelium, and markers of colonic inflammation, were unaffected with DSP-4 pre-administration. We then measured alterations in neuroimmune gene expression in the ventral midbrain in response to DSS treatment alone as well as the extent to which prior LC injury modified this response. In this two-hit model we observed that DSS-induced colitis activates the expression of key cytokines and chemokines in the ventral midbrain only in the presence of LC injury and the typical DSS-associated neuroimmune is blunted by pre-LC lesioning with DSP-4. In all, this study supports the growing appreciation for the LC as neuroprotective against inflammation-induced brain injury and draws attention to the potential for NEergic interventions to exert disease-modifying effects under conditions where peripheral inflammation may compromise ventral midbrain dopaminergic neurons and increase the risk for development of PD.
Collapse
|
5
|
Keller BN, Snyder AE, Coker CR, Aguilar EA, O’Brien MK, Bingaman SS, Arnold AC, Hajnal A, Silberman Y. Vagus nerve damage increases alcohol intake and preference in a nonpreferring rat line: Relationship to vagal regulation of the hypothalamic-pituitary-adrenal axis. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:488-498. [PMID: 38311347 PMCID: PMC10939901 DOI: 10.1111/acer.15264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/12/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024]
Abstract
BACKGROUND Clinical and preclinical research indicates that gastric weight loss surgeries, such as Roux-en-Y gastric bypass surgery, can induce alcohol use disorder (AUD). While numerous mechanisms have been proposed for these effects, one relatively unexplored potential mechanism is physical damage to the gastric branch of the vagus nerve, which can occur during bypass surgery. Therefore, we hypothesized that direct damage to the gastric branch of the vagus nerve, without altering other aspects of gastric anatomy, could result in increased alcohol intake. METHODS To test this hypothesis, we compared alcohol intake and preference in multiple models in male Sprague-Dawley rats that received selective gastric branch vagotomy (VX) with rats who underwent sham surgery. Because the vagus nerve regulates hypothalamic-pituitary-adrenal (HPA) axis function, and alterations to HPA function are critical to the escalation of non-dependent alcohol intake, we also tested the hypothesis that gastric VX increases HPA function. RESULTS We found that VX increases alcohol intake and preference in the every-other-day, two-bottle choice test and increases preference for 1 g/kg alcohol in the conditioned place preference test. The effects were selective for alcohol, as sucrose intake and preference were not altered by VX. We also found that VX increases corticotropin releasing factor (CRF) mRNA in the paraventricular nucleus of the hypothalamus (PVN), increases putative PVN CRF neuronal action potential firing, and increases corticosterone levels. CONCLUSIONS Overall, these findings suggest that the vagus nerve may play a critical role in regulating HPA axis function via modulation of PVN CRF mRNA expression and putative PVN CRF neuronal activity. Furthermore, disruptions to vagal regulation of HPA axis function may increase alcohol intake and preference.
Collapse
Affiliation(s)
- Bailey N. Keller
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Angela E. Snyder
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Caitlin R. Coker
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Elizabeth A. Aguilar
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Mary K. O’Brien
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Sarah S. Bingaman
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Amy C. Arnold
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Andras Hajnal
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Yuval Silberman
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| |
Collapse
|
6
|
Boles JS, Holt J, Cole CL, Neighbarger NK, Urs NM, Huarte OU, Tansey MG. Locus coeruleus injury modulates ventral midbrain neuroinflammation during DSS-induced colitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.580010. [PMID: 38405709 PMCID: PMC10888767 DOI: 10.1101/2024.02.12.580010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Parkinson's disease (PD) is characterized by a decades-long prodrome, consisting of a collection of non-motor symptoms that emerges prior to the motor manifestation of the disease. Of these non-motor symptoms, gastrointestinal dysfunction and deficits attributed to central norepinephrine (NE) loss, including mood changes and sleep disturbances, are frequent in the PD population and emerge early in the disease. Evidence is mounting that injury and inflammation in the gut and locus coeruleus (LC), respectively, underlie these symptoms, and the injury of these systems is central to the progression of PD. In this study, we generate a novel two-hit mouse model that captures both features, using dextran sulfate sodium (DSS) to induce gut inflammation and N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) to lesion the LC. We first confirmed the specificity of DSP-4 for central NE using neurochemical methods and fluorescence light-sheet microscopy of cleared tissue, and established that DSS-induced outcomes in the periphery, including weight loss, gross indices of gut injury and systemic inflammation, the loss of tight junction proteins in the colonic epithelium, and markers of colonic inflammation, were unaffected with DSP-4 pre-administration. We then measured alterations in neuroimmune gene expression in the ventral midbrain in response to DSS treatment alone as well as the extent to which prior LC injury modified this response. In this two-hit model we observed that DSS-induced colitis activates the expression of key cytokines and chemokines in the ventral midbrain only in the presence of LC injury and the typical DSS-associated neuroimmune is blunted by pre-LC lesioning with DSP-4. In all, this study supports the growing appreciation for the LC as neuroprotective against inflammation-induced brain injury and draws attention to the potential for NEergic interventions to exert disease-modifying effects under conditions where peripheral inflammation may compromise ventral midbrain dopaminergic neurons and increase the risk for development of PD.
Collapse
Affiliation(s)
- Jake Sondag Boles
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jenny Holt
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Cassandra L. Cole
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Noelle K. Neighbarger
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Nikhil M. Urs
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Oihane Uriarte Huarte
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
7
|
Milicic M, Gaszner B, Berta G, Pintér E, Kormos V. The Lack of TRPA1 Ion Channel Does Not Affect the Chronic Stress-Induced Activation of the Locus Ceruleus. Int J Mol Sci 2024; 25:1765. [PMID: 38339042 PMCID: PMC10855130 DOI: 10.3390/ijms25031765] [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: 12/22/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
We have previously proven the involvement of transient receptor potential ankyrin 1 (TRPA1) in stress adaptation. A lack of TRPA1 affects both urocortin 1 (member of the corticotropin-releasing hormone (CRH) family) content of the Edinger-Westphal nucleus. The noradrenergic locus ceruleus (LC) is also an important player in mood control. We aimed at investigating whether the TRPA1 is expressed in the LC, and to test if the response to chronic variable mild stress (CVMS) is affected by a lack of TRPA1. The TRPA1 expression was examined via RNAscope in situ hybridization. We investigated TRPA1 knockout and wildtype mice using the CVMS model of depression. Tyrosine hydroxylase (TH) and FOSB double immunofluorescence were used to test the functional neuromorphological changes in the LC. No TRPA1 expression was detected in the LC. The TH content was not affected by CVMS exposure. The CVMS-induced FOSB immunosignal did not co-localize with the TH neurons. TRPA1 is not expressed in the LC. A lack of functional TRPA1 receptor neither directly nor indirectly affects the TH content of LC neurons under CVMS.
Collapse
Affiliation(s)
- Milica Milicic
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary; (M.M.); (E.P.)
| | - Balázs Gaszner
- Department of Anatomy, Medical School and Research Group for Mood Disorders, University of Pécs, H-7624 Pécs, Hungary;
| | - Gergely Berta
- Department of Medical Biology, Medical School, University of Pécs, H-7624 Pécs, Hungary;
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary; (M.M.); (E.P.)
| | - Viktória Kormos
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary; (M.M.); (E.P.)
| |
Collapse
|
8
|
Hou Z, Jiang W, Li F, Liu X, Hou Z, Yin Y, Zhang H, Zhang H, Xie C, Zhang Z, Kong Y, Yuan Y. Linking individual variability in functional brain connectivity to polygenic risk in major depressive disorder. J Affect Disord 2023; 329:55-63. [PMID: 36842648 DOI: 10.1016/j.jad.2023.02.104] [Citation(s) in RCA: 2] [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: 12/20/2022] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 02/28/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) is a highly heterogeneous disease, which brings great difficulties to clinical diagnosis and therapy. Its mechanism is still unknown. Prior neuroimaging studies mainly focused on mean differences between patients and healthy controls (HC), largely ignoring individual differences between patients. METHODS This study included 112 MDD patients and 93 HC subjects. Resting-state functional MRI data were obtained to examine the patterns of individual variability of brain functional connectivity (IVFC). The genetic risk of pathways including dopamine, 5-hydroxytryptamine (5-HT), norepinephrine (NE), hypothalamic-pituitary-adrenal (HPA) axis, and synaptic plasticity was assessed by multilocus genetic profile scores (MGPS), respectively. RESULTS The IVFC pattern of the MDD group was similar but higher than that in HCs. The inter-network functional connectivity in the default mode network contributed to altered IVFC in MDD. 5-HT, NE, and HPA pathway genes affected IVFC in MDD patients. The age of onset, duration, severity, and treatment response, were correlated with IVFC. IVFC in the left ventromedial prefrontal cortex had a mediating effect between MGPS of the 5-HT pathway and baseline depression severity. LIMITATIONS Environmental factors and differences in locations of functional areas across individuals were not taken into account. CONCLUSIONS This study found MDD patients had significantly different inter-individual functional connectivity variations than healthy people, and genetic risk might affect clinical manifestations through brain function heterogeneity.
Collapse
Affiliation(s)
- Zhuoliang Hou
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medical, Southeast University, Nanjing 210009, China
| | - Wenhao Jiang
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medical, Southeast University, Nanjing 210009, China
| | - Fan Li
- Jiangsu Provincial Joint International Research Laboratory of Medical Information Processing, School of Computer Science and Engineering, Southeast University, Nanjing 210096, China
| | - Xiaoyun Liu
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medical, Southeast University, Nanjing 210009, China
| | - Zhenghua Hou
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medical, Southeast University, Nanjing 210009, China
| | - Yingying Yin
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medical, Southeast University, Nanjing 210009, China
| | - Haisan Zhang
- Departments of Clinical Magnetic Resonance Imaging, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China
| | - Hongxing Zhang
- Departments of Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453002, China; School of Psychology, Xinxiang Medical University, Xinxiang 453003, China
| | - Chunming Xie
- Department of Neurology, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhijun Zhang
- Department of Neurology, ZhongDa Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Youyong Kong
- Jiangsu Provincial Joint International Research Laboratory of Medical Information Processing, School of Computer Science and Engineering, Southeast University, Nanjing 210096, China.
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, ZhongDa Hospital, School of Medical, Southeast University, Nanjing 210009, China.
| |
Collapse
|
9
|
Landau AM, Jakobsen S, Thomsen MB, Alstrup AKO, Orlowski D, Jacobsen J, Wegener G, Mørk A, Sørensen JCH, Doudet DJ. Combined In Vivo Microdialysis and PET Studies to Validate [ 11C]Yohimbine Binding as a Marker of Noradrenaline Release. Biomolecules 2023; 13:674. [PMID: 37189421 PMCID: PMC10136072 DOI: 10.3390/biom13040674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
The noradrenaline system attracts attention for its role in mood disorders and neurodegenerative diseases but the lack of well-validated methods impairs our understanding when assessing its function and release in vivo. This study combines simultaneous positron emission tomography (PET) and microdialysis to explore if [11C]yohimbine, a selective antagonist radioligand of the α2 adrenoceptors, may be used to assess in vivo changes in synaptic noradrenaline during acute pharmacological challenges. Anesthetised Göttingen minipigs were positioned in a head holder in a PET/CT device. Microdialysis probes were placed in the thalamus, striatum and cortex and dialysis samples were collected every 10 min. Three 90 min [11C]yohimbine scans were acquired: at baseline and at two timepoints after the administration of amphetamine (1-10 mg/kg), a non-specific releaser of dopamine and noradrenaline, or nisoxetine (1 mg/kg), a specific noradrenaline transporter inhibitor. [11C]yohimbine volumes of distribution (VT) were obtained using the Logan kinetic model. Both challenges induced a significant decrease in yohimbine VT, with time courses reflecting their different mechanisms of action. Dialysis samples revealed a significant increase in noradrenaline extracellular concentrations after challenge and an inverse correlation with changes in yohimbine VT. These data suggest that [11C]yohimbine can be used to evaluate acute variations in synaptic noradrenaline concentrations after pharmacological challenges.
Collapse
Affiliation(s)
- Anne Marlene Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, A701, Palle Juul Jensens Boulevard 99, 8200 Aarhus, Denmark
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Steen Jakobsen
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Majken Borup Thomsen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, A701, Palle Juul Jensens Boulevard 99, 8200 Aarhus, Denmark
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, 8200 Aarhus, Denmark
| | | | - Dariusz Orlowski
- Center for Experimental Neuroscience (CENSE), Department of Neurosurgery, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Jan Jacobsen
- Department of Nuclear Medicine & PET-Center, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, A701, Palle Juul Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Arne Mørk
- Synaptic Transmission, H. Lundbeck A/S, Ottiliavej 9, Valby, 2500 Copenhagen, Denmark
| | | | - Doris J. Doudet
- Department of Medicine/Neurology, University of British Columbia, 2221 Wesbrook Mall, Vancouver, BC V6T 2B5, Canada;
| |
Collapse
|
10
|
Fang M, Li Y, Liao Z, Wang G, Cao Q, Li Y, Duan Y, Han Y, Deng X, Wu F, Kamau PM, Lu Q, Lai R. Lipopolysaccharide-binding protein expression is increased by stress and inhibits monoamine synthesis to promote depressive symptoms. Immunity 2023; 56:620-634.e11. [PMID: 36854305 DOI: 10.1016/j.immuni.2023.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 09/11/2022] [Accepted: 02/01/2023] [Indexed: 03/02/2023]
Abstract
Monoamine insufficiency is suggested to be associated with depressive features such as sadness, anhedonia, insomnia, and cognitive dysfunction, but the mechanisms that cause it are unclear. We found that the acute-phase protein lipopolysaccharide-binding protein (LBP) inhibits monoamine biosynthesis by acting as an endogenous inhibitor of dopamine-β-hydroxylase (DBH) and aromatic-L-amino-acid-decarboxylase (DDC). LBP expression was increased in individuals with depression and by diverse stress challenges in mice. LBP antibodies and LBP knockdown inhibited monoamine insufficiency and depression-like features in mice, which worsened with LBP overexpression or administration. Monoamine insufficiency and depression-like symptoms were not induced by stressful stimuli in LBP-deficient mice, further highlighting a role for LBP in stress-induced depression, and a peptide we designed that blocks LBP-DBH and LBP-DDC interactions showed anti-depression effects in mice. This study reveals an important role for LBP in regulating monoamine biosynthesis and suggests that targeting LBP may have potential as a treatment for some individuals with depression.
Collapse
Affiliation(s)
- Mingqian Fang
- Key Laboratory of Animal Models and Human Disease Mechanisms and Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and Sino-African Joint Research Center, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Yu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms and Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and Sino-African Joint Research Center, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; College of Life Science, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhiyi Liao
- Key Laboratory of Animal Models and Human Disease Mechanisms and Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and Sino-African Joint Research Center, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gan Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms and Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and Sino-African Joint Research Center, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Qiqi Cao
- Key Laboratory of Animal Models and Human Disease Mechanisms and Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and Sino-African Joint Research Center, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Ya Li
- First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yong Duan
- First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yanbing Han
- First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Xinyi Deng
- Key Laboratory of Animal Models and Human Disease Mechanisms and Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and Sino-African Joint Research Center, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Feilong Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms and Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and Sino-African Joint Research Center, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peter Muiruri Kamau
- Key Laboratory of Animal Models and Human Disease Mechanisms and Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and Sino-African Joint Research Center, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiumin Lu
- Key Laboratory of Animal Models and Human Disease Mechanisms and Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and Sino-African Joint Research Center, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms and Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), and Sino-African Joint Research Center, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| |
Collapse
|
11
|
Bassil K, Krontira AC, Leroy T, Escoto AIH, Snijders C, Pernia CD, Pasterkamp RJ, de Nijs L, van den Hove D, Kenis G, Boks MP, Vadodaria K, Daskalakis NP, Binder EB, Rutten BPF. In vitro modeling of the neurobiological effects of glucocorticoids: A review. Neurobiol Stress 2023; 23:100530. [PMID: 36891528 PMCID: PMC9986648 DOI: 10.1016/j.ynstr.2023.100530] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Hypothalamic-pituitary adrenal (HPA)axis dysregulation has long been implicated in stress-related disorders such as major depression and post-traumatic stress disorder. Glucocorticoids (GCs) are released from the adrenal glands as a result of HPA-axis activation. The release of GCs is implicated with several neurobiological changes that are associated with negative consequences of chronic stress and the onset and course of psychiatric disorders. Investigating the underlying neurobiological effects of GCs may help to better understand the pathophysiology of stress-related psychiatric disorders. GCs impact a plethora of neuronal processes at the genetic, epigenetic, cellular, and molecular levels. Given the scarcity and difficulty in accessing human brain samples, 2D and 3D in vitro neuronal cultures are becoming increasingly useful in studying GC effects. In this review, we provide an overview of in vitro studies investigating the effects of GCs on key neuronal processes such as proliferation and survival of progenitor cells, neurogenesis, synaptic plasticity, neuronal activity, inflammation, genetic vulnerability, and epigenetic alterations. Finally, we discuss the challenges in the field and offer suggestions for improving the use of in vitro models to investigate GC effects.
Collapse
Affiliation(s)
- Katherine Bassil
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Anthi C Krontira
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany.,International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Thomas Leroy
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Alana I H Escoto
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Clara Snijders
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Cameron D Pernia
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center (UMC) Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Laurence de Nijs
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Daniel van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Marco P Boks
- Psychiatry, UMC Utrecht Brain Center, Utrecht, the Netherlands
| | - Krishna Vadodaria
- Salk Institute for Biological Studies, La Jolla, San Diego, United States
| | | | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany.,International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
12
|
Colzato LS, Elmers J, Beste C, Hommel B. A Prospect to Ameliorate Affective Symptoms and to Enhance Cognition in Long COVID Using Auricular Transcutaneous Vagus Nerve Stimulation. J Clin Med 2023; 12:jcm12031198. [PMID: 36769845 PMCID: PMC9917620 DOI: 10.3390/jcm12031198] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Long COVID, the postviral disorder caused by COVID-19, is expected to become one of the leading causes of disability in Europe. The cognitive consequences of long COVID have been described as "brain fog" and characterized by anxiety and depression, and by cognitive deficits. Long COVID is assumed to be a complex condition arising from multiple causes, including persistent brainstem dysfunction and disrupted vagal signaling. We recommend the potential application of auricular transcutaneous vagus nerve stimulation (atVNS) as an ADD-ON instrument to compensate for the cognitive decline and to ameliorate affective symptoms caused by long COVID. This technique enhances vagal signaling by directly activating the nuclei in the brainstem, which are hypoactive in long COVID to enhance mood and to promote attention, memory, and cognitive control-factors affected by long COVID. Considering that atVNS is a non-pharmacological intervention, its ADD-ON to standard pharmaceutical agents will be useful for non-responders, making of this method a suitable tool. Given that atVNS can be employed as an ecological momentary intervention (EMI), we outline the translational advantages of atVNS in the context of accelerating the cognitive and affective recovery from long COVID.
Collapse
Affiliation(s)
- Lorenza S. Colzato
- Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan 250014, China
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Dresden University of Technology, 01307 Dresden, Germany
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Julia Elmers
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Dresden University of Technology, 01307 Dresden, Germany
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Christian Beste
- Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan 250014, China
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Dresden University of Technology, 01307 Dresden, Germany
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Bernhard Hommel
- Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan 250014, China
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Dresden University of Technology, 01307 Dresden, Germany
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
- Correspondence:
| |
Collapse
|
13
|
Scuotto C, Ilardi CR, Maggi G, Ilardi A, Gamboz N, Staiano M, Borrelli G, La Marra M, Perrella R. What makes us more susceptible to false memories in the era of COVID-19? A focus on vaccines and Green Pass. Brain Behav 2023; 13:e2815. [PMID: 36448933 PMCID: PMC9847604 DOI: 10.1002/brb3.2815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/15/2022] [Accepted: 10/27/2022] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION The coronavirus disease 2019 (COVID-19) pandemic was accompanied by an overabundance of fake news increasing the risk of developing false memories (FMs). Previous studies have shown that the relationship between fake news and FMs could be mediated by some individual variables, including attitudinal biases. We explored the role of these variables in true memories (TMs) and FMs formation, with special emphasis on vaccine- and Green Pass (GP)-related topics. METHOD We set up a large online survey exploring several constructs including media usage, attitude toward vaccines and GP, perceived (PK) and objective knowledge (OK) about COVID-19-related information, fear of the disease, depression and anxiety symptoms, coping mechanisms, and reasoning skills. Then, we asked participants whether they remembered certain news (true or fake), providing confidence ratings. RESULTS Data from 289 respondents (198 females) from the general population were analyzed. Participants with positive attitude reported a greater fear that their loved ones contracted the COVID-19, a more frequent use of traditional media, and a higher PK when compared with respondents with negative attitude. On the whole sample, participants reported higher confidence levels when required to judge their memory of true than fake news; however, participants with positive attitude reported a higher confidence for both true and fake news. The relationship between attitude and TM confidence was mediated by the PK, whereas the relationship between attitude and FM confidence was probably affected by OK. CONCLUSION Attitude can modulate individual behaviors in the context of health issues. The PK and OK may interact with attitude in the memory formation.
Collapse
Affiliation(s)
- Chiara Scuotto
- Department of Psychology, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Ciro Rosario Ilardi
- Department of Psychology, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Gianpaolo Maggi
- Department of Psychology, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Alfonso Ilardi
- Inmates Ward, Department of Internal Medicine, Antonio Cardarelli Hospital, Naples, Italy
| | - Nadia Gamboz
- Laboratory of Experimental Psychology, Suor Orsola Benincasa University, Naples, Italy
| | - Maria Staiano
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giovanni Borrelli
- Department of Psychology, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Marco La Marra
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Raffaella Perrella
- Department of Psychology, University of Campania Luigi Vanvitelli, Caserta, Italy
| |
Collapse
|
14
|
Locus Coeruleus-Noradrenergic Neurons Regulate Stress Coping During Subchronic Exposure to Social Threats: A Characteristic Feature in Postpartum Female Mice. Cell Mol Neurobiol 2022:10.1007/s10571-022-01314-4. [PMID: 36577871 DOI: 10.1007/s10571-022-01314-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 12/17/2022] [Indexed: 12/29/2022]
Abstract
Stress-coping strategies have been implicated in depression. The control of stress coping may improve the symptom and higher prevalence of depression during the postpartum period in women. However, the neuronal mechanisms underlying stress coping remain to be fully elucidated in postpartum women. In this study, we examined how locus coeruleus-noradrenergic (LC-NA) neurons, which have been associated with both stress coping and depression, regulate changes in coping style induced by subchronic exposure to unfamiliar male mice as a social threat in postpartum female mice. In contrast to virgin females, dams exposed to unfamiliar males daily for four consecutive days showed reduced immobility duration in the forced swim test, indicating that exposure to unfamiliar males decreased passive stress coping in dams. Exposure to unfamiliar males also decreased sucrose palatability in the sucrose preference test and suppressed the crouching behavior in the maternal care test but did not affect anxiety-like behavior in the hole-board test in dams. In fiber photometry analyses, LC-NA neurons showed differential activity between dams and virgin females in response to unfamiliar males. Chemogenetic inhibition of LC-NA neurons during exposure to unfamiliar males prevented the social threat-induced decrease in immobility duration in the forced swim test in dams. Furthermore, inhibition or activation of LC-NA neurons exacerbated crouching behavior in dams. These results indicate that LC-NA neurons regulate the social threat-induced decrease in passive stress coping and relieve social threat-induced inhibition of maternal care in postpartum female mice.
Collapse
|
15
|
Pace-Schott EF, Seo J, Bottary R. The influence of sleep on fear extinction in trauma-related disorders. Neurobiol Stress 2022; 22:100500. [PMID: 36545012 PMCID: PMC9761387 DOI: 10.1016/j.ynstr.2022.100500] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/21/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
In Posttraumatic Stress Disorder (PTSD), fear and anxiety become dysregulated following psychologically traumatic events. Regulation of fear and anxiety involves both high-level cognitive processes such as cognitive reattribution and low-level, partially automatic memory processes such as fear extinction, safety learning and habituation. These latter processes are believed to be deficient in PTSD. While insomnia and nightmares are characteristic symptoms of existing PTSD, abundant recent evidence suggests that sleep disruption prior to and acute sleep disturbance following traumatic events both can predispose an individual to develop PTSD. Sleep promotes consolidation in multiple memory systems and is believed to also do so for low-level emotion-regulatory memory processes. Consequently sleep disruption may contribute to the etiology of PTSD by interfering with consolidation in low-level emotion-regulatory memory systems. During the first weeks following a traumatic event, when in the course of everyday life resilient individuals begin to acquire and consolidate these low-level emotion-regulatory memories, those who will develop PTSD symptoms may fail to do so. This deficit may, in part, result from alterations of sleep that interfere with their consolidation, such as REM fragmentation, that have also been found to presage later PTSD symptoms. Here, sleep disruption in PTSD as well as fear extinction, safety learning and habituation and their known alterations in PTSD are first briefly reviewed. Then neural processes that occur during the early post-trauma period that might impede low-level emotion regulatory processes through alterations of sleep quality and physiology will be considered. Lastly, recent neuroimaging evidence from a fear conditioning and extinction paradigm in patient groups and their controls will be considered along with one possible neural process that may contribute to a vulnerability to PTSD following trauma.
Collapse
Affiliation(s)
- Edward F. Pace-Schott
- Massachusetts General Hospital, Department of Psychiatry, Charlestown, MA, USA
- Harvard Medical School, Department of Psychiatry, Charlestown, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Corresponding author. Harvard Medical School, Massachusetts General Hospital - East, CNY 149 13th Street, Charlestown, MA, 02129, USA.
| | - Jeehye Seo
- Massachusetts General Hospital, Department of Psychiatry, Charlestown, MA, USA
- Harvard Medical School, Department of Psychiatry, Charlestown, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Korea University, Department of Brain & Cognitive Engineering, Seongbuk-gu, Seoul, South Korea
| | - Ryan Bottary
- Massachusetts General Hospital, Department of Psychiatry, Charlestown, MA, USA
- Harvard Medical School, Department of Psychiatry, Charlestown, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Psychology and Neuroscience, Boston College, Chestnut Hill, MA, USA
| |
Collapse
|
16
|
Hinckley JD, Danielson CK. Elucidating the Neurobiologic Etiology of Comorbid PTSD and Substance Use Disorders. Brain Sci 2022; 12:brainsci12091166. [PMID: 36138902 PMCID: PMC9496654 DOI: 10.3390/brainsci12091166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 11/26/2022] Open
Abstract
Early childhood maltreatment and other traumatic event experiences ("trauma") are common among youth, including those with substance use problems including substance use disorders (SUD). Particularly, interpersonal violence is associated with high rates of comorbidity between posttraumatic stress disorder (PTSD) and SUD, and these comorbid disorders exhibit high levels of overlapping symptomatology. Theoretical models proposed to explain the bidirectional relationship between PTSD and SUD include the self-medication hypothesis and susceptibility hypothesis. In this article, we explore neurobiologic changes associated with trauma, PTSD, and SUD that underly dysregulated stress response. Examining lessons learned from recent translational and clinical research, we propose that further elucidating the neurobiologic etiology of comorbid PTSD and SUD will require a collaborative, interdisciplinary approach, including the integration of preclinical and clinical studies, exploration of biologic markers in clinical studies, and accumulation of larger studies and longitudinal studies with the power to study PTSD and SUD. Such research can transform the field and ultimately reduce high rates and costly impairment of co-occurring PTSD and SUD across the lifespan.
Collapse
Affiliation(s)
- Jesse D. Hinckley
- Division of Addiction Science, Treatment & Prevention, Department of Psychiatry, University of Colorado School of Medicine, 1890 N Revere Court, MS-F570, Aurora, CO 80045, USA
- Correspondence:
| | - Carla Kmett Danielson
- National Crime Victims Research & Treatment Center, Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, 67 President Street, MSC 861, Charleston, SC 29425, USA
| |
Collapse
|
17
|
Wang H, Zhang J, Yu Z, Hu N, Du Y, He X, Pan D, Pu L, Zhang X, Li J. Association between changes in social capital and mental well-being among older people in China. PeerJ 2022; 10:e13938. [PMID: 36042863 PMCID: PMC9420402 DOI: 10.7717/peerj.13938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/02/2022] [Indexed: 01/19/2023] Open
Abstract
Background The mental well-being of older people has become social concern under aging times in China. Social capital has been linked to mental well-being. Our aims were to explore how social capital and the state of mental well-being of older people were changing and what the relationship between them was. Methods Data were from six waves of the China Family Panel Studies that were conducted between 2010 and 2020, and a total of 1,055 participants aged 60 and over were included in the analysis. The Generalized Estimated Equation model (GEE) was used to clarify the long-term relationship, and to use GEE we first defined how time points were related, in other words, an appropriate working correlation structure was supposed to choose. Therefore, correlation coefficient between measurements at two time points was calculated to choose the exchange structure. All the analyses were performed in the statistical software Stata 15.0. Results The mental well-being of older people has deteriorated over time, especially we found that between 2014 and 2016, the mental well-being of older people plummeted. In addition, cognitive social capital was positively correlated with mental well-being, while structural social capital was inverse. Conclusions Policymakers are supposed to take into account the long-term impact of cognitive and structural social capital on the mental well-being of older people and to provide them with projects aimed at increasing cognitive social capital and turning the pressure of structural social capital into a source of happiness in life.
Collapse
Affiliation(s)
- Huihui Wang
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Jingni Zhang
- Department of Science and Education, Qinghai Provincial People’s Hospital, Xining, Qinghai Province, China
| | - Zhenfan Yu
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Naifan Hu
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yurun Du
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaoxue He
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Degong Pan
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Lining Pu
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xue Zhang
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Jiangping Li
- Department of Epidemiology and Health Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China,Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical College, Yinchuan, Ningxia Hui Autonomous Region, China
| |
Collapse
|
18
|
Wang Y, Liu Z, Sun D, Sun L, Cao G, Dai J. The Connectome and Chemo-Connectome Databases for Mice Brain Connection Analysis. Front Neuroanat 2022; 16:886925. [PMID: 35756500 PMCID: PMC9218099 DOI: 10.3389/fnana.2022.886925] [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: 03/01/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
The various brain functions rely on the intricate connection networks and certain molecular characteristics of neurons in the brain. However, the databases for the mouse brain connectome and chemo-connectome are still inadequate, hindering the brain circuital and functional analysis. Here, we created mice brain connectome and chemo-connectome databases based on mouse brain projection data of 295 non-overlapping brain areas and in situ hybridization (ISH) data of 50 representative neurotransmission-related genes from the Allen Brain Institute. Based on this connectome and chemo-connectome databases, functional connection patterns and detailed chemo-connectome for monoaminergic nuclei were analyzed and visualized. These databases will aid in the comprehensive research of the mouse connectome and chemo-connectome in the whole brain and serve as a convenient resource for systematic analysis of the brain connection and function.
Collapse
Affiliation(s)
- Yang Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhixiang Liu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
| | - Da Sun
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Leqiang Sun
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Biomedical Center, Huazhong Agricultural University, Wuhan, China
| | - Jinxia Dai
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
19
|
Keller BN, Hajnal A, Browning KN, Arnold AC, Silberman Y. Involvement of the Dorsal Vagal Complex in Alcohol-Related Behaviors. Front Behav Neurosci 2022; 16:801825. [PMID: 35330845 PMCID: PMC8940294 DOI: 10.3389/fnbeh.2022.801825] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/19/2022] [Indexed: 12/20/2022] Open
Abstract
The neurobiological mechanisms that regulate the development and maintenance of alcohol use disorder (AUD) are complex and involve a wide variety of within and between systems neuroadaptations. While classic reward, preoccupation, and withdrawal neurocircuits have been heavily studied in terms of AUD, viable treatment targets from this established literature have not proven clinically effective as of yet. Therefore, examination of additional neurocircuitries not classically studied in the context of AUD may provide novel therapeutic targets. Recent studies demonstrate that various neuropeptides systems are important modulators of alcohol reward, seeking, and intake behaviors. This includes neurocircuitry within the dorsal vagal complex (DVC), which is involved in the control of the autonomic nervous system, control of intake of natural rewards like food, and acts as a relay of interoceptive sensory information via interactions of numerous gut-brain peptides and neurotransmitter systems with DVC projections to central and peripheral targets. DVC neuron subtypes produce a variety of neuropeptides and transmitters and project to target brain regions critical for reward such as the mesolimbic dopamine system as well as other limbic areas important for the negative reinforcing and aversive properties of alcohol withdrawal such as the extended amygdala. This suggests the DVC may play a role in the modulation of various aspects of AUD. This review summarizes the current literature on neurotransmitters and neuropeptides systems in the DVC (e.g., norepinephrine, glucagon-like peptide 1, neurotensin, cholecystokinin, thyrotropin-releasing hormone), and their potential relevance to alcohol-related behaviors in humans and rodent models for AUD research. A better understanding of the role of the DVC in modulating alcohol related behaviors may lead to the elucidation of novel therapeutic targets for drug development in AUD.
Collapse
|
20
|
Hess EM, Riggs LM, Michaelides M, Gould TD. Mechanisms of ketamine and its metabolites as antidepressants. Biochem Pharmacol 2022; 197:114892. [PMID: 34968492 PMCID: PMC8883502 DOI: 10.1016/j.bcp.2021.114892] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 02/06/2023]
Abstract
Treating major depression is a medical need that remains unmet by monoaminergic therapeutic strategies that commonly fail to achieve symptom remission. A breakthrough in the treatment of depression was the discovery that the anesthetic (R,S)-ketamine (ketamine), when administered at sub-anesthetic doses, elicits rapid (sometimes within hours) antidepressant effects in humans that are otherwise resistant to monoaminergic-acting therapies. While this finding was revolutionary and led to the FDA approval of (S)-ketamine (esketamine) for use in adults with treatment-resistant depression and suicidal ideation, the mechanisms underlying how ketamine or esketamine elicit their effects are still under active investigation. An emerging view is that metabolism of ketamine may be a crucial step in its mechanism of action, as several metabolites of ketamine have neuroactive effects of their own and may be leveraged as therapeutics. For example, (2R,6R)-hydroxynorketamine (HNK), is readily observed in humans following ketamine treatment and has shown therapeutic potential in preclinical tests of antidepressant efficacy and synaptic potentiation while being devoid of the negative adverse effects of ketamine, including its dissociative properties and abuse potential. We discuss preclinical and clinical studies pertaining to how ketamine and its metabolites produce antidepressant effects. Specifically, we explore effects on glutamate neurotransmission through N-methyl D-aspartate receptors (NMDARs) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), synaptic structural changes via brain derived neurotrophic factor (BDNF) signaling, interactions with opioid receptors, and the enhancement of serotonin, norepinephrine, and dopamine signaling. Strategic targeting of these mechanisms may result in novel rapid-acting antidepressants with fewer undesirable side effects compared to ketamine.
Collapse
Affiliation(s)
- Evan M Hess
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Lace M Riggs
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.,Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD 21224, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Departments of Pharmacology and Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Baltimore Veterans Affairs Medical Center, Veterans Affairs Maryland Health Care System, Baltimore, MD 21201, USA.
| |
Collapse
|
21
|
Functional Coupling of the Locus Coeruleus Is Linked to Successful Cognitive Control. Brain Sci 2022; 12:brainsci12030305. [PMID: 35326262 PMCID: PMC8946131 DOI: 10.3390/brainsci12030305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 11/20/2022] Open
Abstract
The locus coeruleus (LC) is a brainstem structure that sends widespread efferent projections throughout the mammalian brain. The LC constitutes the major source of noradrenaline (NE), a modulatory neurotransmitter that is crucial for fundamental brain functions such as arousal, attention, and cognitive control. This role of the LC-NE is traditionally not believed to reflect functional influences on the frontoparietal network or the striatum, but recent advances in chemogenetic manipulations of the rodent brain have challenged this notion. However, demonstrations of LC-NE functional connectivity with these areas in the human brain are surprisingly sparse. Here, we close this gap. Using an established emotional stroop task, we directly compared trials requiring response conflict control with trials that did not require this, but were matched for visual stimulus properties, response modality, and controlled for pupil dilation differences across both trial types. We found that LC-NE functional coupling with the parietal cortex and regions of the striatum is substantially enhanced during trials requiring response conflict control. Crucially, the strength of this functional coupling was directly related to individual reaction time differences incurred by conflict resolution. Our data concur with recent rodent findings and highlight the importance of converging evidence between human and nonhuman neurophysiology to further understand the neural systems supporting adaptive and maladaptive behavior in health and disease.
Collapse
|
22
|
Jung JY, Cho SE, Kim N, Kang CK, Kang SG. Decreased resting-state functional connectivity of the habenula-cerebellar in a major depressive disorder. Front Psychiatry 2022; 13:925823. [PMID: 36147982 PMCID: PMC9485485 DOI: 10.3389/fpsyt.2022.925823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND In animal experiments, the habenula and septal nuclei are known as the key brain areas of depression. However, there are few magnetic resonance imaging (MRI) studies on the functional connectivity between these areas and the subcortical areas in humans with major depression. We aimed to investigate the difference in resting-state functional connectivity (RSFC) among the major regions of interest (ROI) in the subcortical areas, including both the habenula and septal nuclei. METHODS We performed the seed-to-voxel analysis to investigate the RSFC between both the habenula and septal nucleus, as well as other subcortical regions. Furthermore, ROI-to-ROI analysis was performed among the combinations of ROI pairs in the subcortical areas. RESULTS The seed-to-voxel analysis showed a lower RSFC between the left habenula and the cerebellum in major depressive disorder (MDD) than in healthy controls (HCs). As a result of ROI-to-ROI analysis in subcortical areas, a total of 31 pairs of FCs in the MDD group showed a lower RSFC than in the HCs group. CONCLUSION This study revealed a lower RSFC between the left habenula and cerebellum in patients with MDD and reduced RSFC among numerous subcortical areas. These new findings on the neural circuitry of MDD might contribute to an in-depth understanding of depression.
Collapse
Affiliation(s)
- Ju-Yeon Jung
- Department of Health Science, Gachon University Graduate School, Incheon, South Korea
| | - Seo-Eun Cho
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Nambeom Kim
- Department of Biomedical Engineering Research Center, Gachon University, Incheon, South Korea
| | - Chang-Ki Kang
- Department of Radiological Science, College of Health Science, Gachon University, Incheon, South Korea
| | - Seung-Gul Kang
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| |
Collapse
|
23
|
Vaseghi S, Zarrabian S, Haghparast A. Reviewing the role of the orexinergic system and stressors in modulating mood and reward-related behaviors. Neurosci Biobehav Rev 2021; 133:104516. [PMID: 34973302 DOI: 10.1016/j.neubiorev.2021.104516] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 01/22/2023]
Abstract
In this review study, we aimed to introduce the orexinergic system as an important signaling pathway involved in a variety of cognitive functions such as memory, motivation, and reward-related behaviors. This study focused on the role of orexinergic system in modulating reward-related behavior, with or without the presence of stressors. Cross-talk between the reward system and orexinergic signaling was also investigated, especially orexinergic signaling in the ventral tegmental area (VTA), the nucleus accumbens (NAc), and the hippocampus. Furthermore, we discussed the role of the orexinergic system in modulating mood states and mental illnesses such as depression, anxiety, panic, and posttraumatic stress disorder (PTSD). Here, we narrowed down our focus on the orexinergic signaling in three brain regions: the VTA, NAc, and the hippocampus (CA1 region and dentate gyrus) for their prominent role in reward-related behaviors and memory. It was concluded that the orexinergic system is critically involved in reward-related behavior and significantly alters stress responses and stress-related psychiatric and mood disorders.
Collapse
Affiliation(s)
- Salar Vaseghi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Shahram Zarrabian
- Department of Anatomical Sciences & Cognitive Neuroscience, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box 19615-1178, Tehran, Iran.
| |
Collapse
|
24
|
Negative respiratory sinus arrhythmia (nRSA) in the MRI-scanner - a physiologic phenomenon observed during elevated anxiety in healthy persons. Physiol Behav 2021; 245:113676. [PMID: 34919919 DOI: 10.1016/j.physbeh.2021.113676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022]
Abstract
Recently, we reported on a rare manifestation of respiratory sinus arrhythmia (RSA), namely the "switched-off" RSA (Rassler et al., 2018), also called negative RSA (nRSA). It was found in a minority of healthy persons during elevated fMRI-related anxiety characterized by slow spontaneous breathing and synchronous slow beat-to-beat interval (RRI) oscillations. From 23 healthy scanner naïve participants of an fMRI study consisting of 4 resting states, we selected resting states with highest state anxiety (AS) from 10 participants (AS=24.6±2.5) and compared them to those with lowest AS of the same participants (AS=15.1±3.8, p<0.001). During elevated anxiety, the percentage of nRSA (nRSA%) was more than twice of RSA (p=0.045), while RSA prevailed during low anxiety. This indicates that nRSA might be related to elevated anxiety. Interestingly, nRSA was not only associated with slow RRI and breathing oscillations, but also occurred at "normal" breathing rates in the 0.20-0.35 Hz range. We often observed coupled RRI oscillations at 0.1 or 0.15 Hz and respiration at 0.3 Hz (rate ratio 1:3 or 1:2) with respiration-synchronous 0.3 Hz-wavelets in the RRI rhythm (termed "superposition") indicating a reduced dominance of the respiratory rhythm over the RRI rhythm. This novel finding is supported by the work of Perlitz et al., (2004) on a "0.15 Hz rhythm" in brainstem. The concept behind such a 1:n ratio is a pacemaker-like rhythm in the brainstem that "drives" the cardiac RRI signal and secondarily also respiration as reflected in the 1:n rate ratio.
Collapse
|
25
|
Processing of fMRI-related anxiety and bi-directional information flow between prefrontal cortex and brain stem. Sci Rep 2021; 11:22348. [PMID: 34785719 PMCID: PMC8595881 DOI: 10.1038/s41598-021-01710-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/19/2021] [Indexed: 12/30/2022] Open
Abstract
Brain-heart synchronization is fundamental for emotional-well-being and brain-heart desynchronization is characteristic for anxiety disorders including specific phobias. Recording BOLD signals with functional magnetic resonance imaging (fMRI) is an important noninvasive diagnostic tool; however, 1-2% of fMRI examinations have to be aborted due to claustrophobia. In the present study, we investigated the information flow between regions of interest (ROI's) in the cortex and brain stem by using a frequency band close to 0.1 Hz. Causal coupling between signals important in brain-heart interaction (cardiac intervals, respiration, and BOLD signals) was studied by means of Directed Transfer Function based on the Granger causality principle. Compared were initial resting states with elevated anxiety and final resting states with low or no anxiety in a group of fMRI-naïve young subjects. During initial high anxiety the results showed an increased information flow from the middle frontal gyrus (MFG) to the pre-central gyrus (PCG) and to the brainstem. There also was an increased flow from the brainstem to the PCG. While the top-down flow during increased anxiety was predominant, the weaker ascending flow from brainstem structures may characterize a rhythmic pacemaker-like activity that (at least in part) drives respiration. We assume that these changes in information flow reflect successful anxiety processing.
Collapse
|
26
|
Scuotto C, Ilardi CR, Avallone F, Maggi G, Ilardi A, Borrelli G, Gamboz N, La Marra M, Perrella R. Objective Knowledge Mediates the Relationship between the Use of Social Media and COVID-19-Related False Memories. Brain Sci 2021; 11:1489. [PMID: 34827488 PMCID: PMC8615822 DOI: 10.3390/brainsci11111489] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
The exposure to relevant social and/or historical events can increase the generation of false memories (FMs). The Coronavirus Disease 2019 (COVID-19) pandemic is a calamity challenging health, political, and journalistic bodies, with media generating confusion that has facilitated the spread of fake news. In this respect, our study aims at investigating the relationships between memories (true memories, TMs vs. FMs) for COVID-19-related news and different individual variables (i.e., use of traditional and social media, COVID-19 perceived and objective knowledge, fear of the disease, depression and anxiety symptoms, reasoning skills, and coping mechanisms). One hundred and seventy-one university students (131 females) were surveyed. Overall, our results suggested that depression and anxiety symptoms, reasoning skills, and coping mechanisms did not affect the formation of FMs. Conversely, the fear of loved ones contracting the infection was found to be negatively associated with FMs. This finding might be due to an empathy/prosociality-based positive bias boosting memory abilities, also explained by the young age of participants. Furthermore, objective knowledge (i) predicted an increase in TMs and decrease in FMs and (ii) significantly mediated the relationships between the use of social media and development of both TMs and FMs. In particular, higher levels of objective knowledge strengthened the formation of TMs and decreased the development of FMs following use of social media. These results may lead to reconsidering the idea of social media as the main source of fake news. This claim is further supported by either the lack of substantial differences between the use of traditional and social media among participants reporting FMs or the positive association between use of social media and levels of objective knowledge. The knowledge about the topic rather than the type of source would make a difference in the process of memory formation.
Collapse
Affiliation(s)
- Chiara Scuotto
- Department of Psychology, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy; (C.S.); (C.R.I.); (G.M.); (G.B.); (R.P.)
| | - Ciro Rosario Ilardi
- Department of Psychology, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy; (C.S.); (C.R.I.); (G.M.); (G.B.); (R.P.)
| | - Francesco Avallone
- Department of Family Medicine, McGill University, Montreal, QC H3S1Z1, Canada;
| | - Gianpaolo Maggi
- Department of Psychology, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy; (C.S.); (C.R.I.); (G.M.); (G.B.); (R.P.)
| | - Alfonso Ilardi
- Inmates Ward, Department of Internal Medicine, Antonio Cardarelli Hospital, 80131 Naples, Italy;
| | - Giovanni Borrelli
- Department of Psychology, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy; (C.S.); (C.R.I.); (G.M.); (G.B.); (R.P.)
| | - Nadia Gamboz
- Laboratory of Experimental Psychology, Suor Orsola Benincasa University, 80135 Naples, Italy;
| | - Marco La Marra
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Raffaella Perrella
- Department of Psychology, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy; (C.S.); (C.R.I.); (G.M.); (G.B.); (R.P.)
| |
Collapse
|
27
|
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
|
28
|
Duan KM, Fang C, Yang SQ, Yang ST, Xiao JD, Chang H, Lin GX, Zhang LB, Peng MC, Liu ZQ, Wang SY. Genetic Polymorphism of rs13306146 Affects α2AAR Expression and Associated With Postpartum Depressive Symptoms in Chinese Women Who Received Cesarean Section. Front Genet 2021; 12:675386. [PMID: 34306020 PMCID: PMC8294467 DOI: 10.3389/fgene.2021.675386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/17/2021] [Indexed: 01/20/2023] Open
Abstract
Postpartum depressive symptom (PDS) is a common psychological and mental disorder after giving birth. Our previous studies showing the application of dexmedetomidine, an α2-AR agonist, can significantly improve maternal sleep, as well as relieve and reduce the incidence of PDS. This study investigated the association between α2 A AR gene polymorphisms and PDS. A total of 568 cesarean section patients were enrolled; the incidence of PDS is 18.13% (103 with PDS, 465 with non-PDS). The Edinburgh Postpartum Depression Scale score ≥10 was used to diagnose PDS at 42 days after delivery. The single-nucleotide polymorphisms of α2AR were sequenced by pyrosequencing. The effect of rs13306146 A > G polymorphism on α2AR transcription and the regulation of miR-646 on α2AR expression were assessed by dual luciferase reporter assays or gene transfection. Increased stress during pregnancy, poor relationship between mother-in-law and daughter-in-law, spousal relationship, domestic violence, antenatal depression, self-harm ideation, and stressful life events were all associated with increased PDS incidence (p < 0.05). The logistic regression analysis found that the α2AAR rs13306146 polymorphism was associated with PDS after adjusting confounding variables. The transcriptional function of the α2AAR rs13306146 A allele was decreased compared with the G allele, and the α2AAR expression level was correspondingly decreased (p < 0.05), as the strongest binding ability of miR-646 to the α2AAR rs13306146 AA genotype. The effect of α2AAR rs13306146 A > G polymorphism may change the binding ability of miR-646 at the 3'UTR of the α2AAR gene, affecting the expression of α2AAR. This study supports the involvement of the norepinephrine system in the pathogenesis of PDS. Genotypes of α2AAR may be novel and useful biomarkers for PDS.
Collapse
Affiliation(s)
- Kai Ming Duan
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Chao Fang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China.,Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Si Qi Yang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Shu Ting Yang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Ji Dong Xiao
- Department of Ultrasonography, Third Xiangya Hospital of Central South University, Changsha, China
| | - Huang Chang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Guo Xin Lin
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Liang Bin Zhang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Ming Chao Peng
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhao Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Sai Ying Wang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
29
|
Kelly L, Seifi M, Ma R, Mitchell SJ, Rudolph U, Viola KL, Klein WL, Lambert JJ, Swinny JD. Identification of intraneuronal amyloid beta oligomers in locus coeruleus neurons of Alzheimer's patients and their potential impact on inhibitory neurotransmitter receptors and neuronal excitability. Neuropathol Appl Neurobiol 2021; 47:488-505. [PMID: 33119191 DOI: 10.1111/nan.12674] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/22/2022]
Abstract
AIMS Amyloid β-oligomers (AβO) are potent modulators of Alzheimer's pathology, yet their impact on one of the earliest brain regions to exhibit signs of the condition, the locus coeruleus (LC), remains to be determined. Of particular importance is whether AβO impact the spontaneous excitability of LC neurons. This parameter determines brain-wide noradrenaline (NA) release, and thus NA-mediated brain functions, including cognition, emotion and immune function, which are all compromised in Alzheimer's patients. Therefore, the aim of the study was to determine the expression profile of AβO in the LC of Alzheimer's patients and to probe their potential impact on the molecular and functional correlates of LC excitability, using a mouse model of increased Aβ production (APP-PSEN1). METHODS AND RESULTS Immunohistochemistry and confocal microscopy, using AβO-specific antibodies, confirmed LC AβO expression both intraneuronally and extracellularly in both Alzheimer's and APP-PSEN1 samples. Patch clamp electrophysiology recordings revealed that APP-PSEN1 LC neuronal hyperexcitability accompanied this AβO expression profile, arising from a diminished inhibitory effect of GABA due to impaired expression and function of the GABA-A receptor (GABAA R) α3 subunit. This altered LC α3-GABAA R expression profile overlapped with AβO expression in samples from both APP-PSEN1 mice and Alzheimer's patients. Finally, strychnine-sensitive glycine receptors (GlyRs) remained resilient to Aβ-induced changes and their activation reversed LC hyperexcitability. CONCLUSIONS The data suggest a close association between AβO and α3-GABAA Rs in the LC of Alzheimer's patients, and their potential to dysregulate LC activity, thereby contributing to the spectrum of pathology of the LC-NA system in this condition.
Collapse
Affiliation(s)
- Louise Kelly
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Mohsen Seifi
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, UK
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Ruolin Ma
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Scott J Mitchell
- Neuroscience, Division of Systems Medicine, Ninewells Hospital & Medical School, Dundee University, Dundee, UK
| | - Uwe Rudolph
- Department of Comparative Biosciences, College of Veterinary Medicine, and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Kirsten L Viola
- Department of Neurobiology and Physiology, Northwestern University, Evanston, IL, USA
| | - William L Klein
- Department of Neurobiology and Physiology, Northwestern University, Evanston, IL, USA
| | - Jeremy J Lambert
- Neuroscience, Division of Systems Medicine, Ninewells Hospital & Medical School, Dundee University, Dundee, UK
| | - Jerome D Swinny
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
30
|
Bethea CL, Cameron JL. Neuro-pharmacological reinstatement of ovulation and associated neurobiology in a macaque model of functional hypothalamic amenorrhoea. Hum Reprod 2021; 36:175-188. [PMID: 33319240 DOI: 10.1093/humrep/deaa296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 09/23/2020] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION What is the underlying neuropathology in a cynomolgus macaque model of functional hypothalamic amenorrhoea (FHA) and can it be normalized to restore ovulation? SUMMARY ANSWER Anovulatory monkeys exhibited increased hypothalamic norepinephrine (NE), kisspeptin and gonadotropin-releasing hormone (GnRH) in the early follicular phase, but administration of the NE reuptake inhibitor (NRI), reboxetine (REB), restored ovulation during stress and normalized NE, kisspeptin and GnRH. WHAT IS KNOWN ALREADY Female cynomolgus macaques, like women, show individual reproductive sensitivity to modest psychosocial and metabolic stress. During stress, resilient females ovulate through two menstrual cycles whereas stress-sensitive (SS) macaques immediately cease ovulation. On Day 5 of a non-stressed menstrual cycle, resilient macaques have less NE synthesizing enzyme [dopamine β-hydroxylase (DBH)], kisspeptin and GnRH innervation of the medial basal hypothalamus but more endogenous serotonin than SS macaques. Stress increased DBH/NE, kisspeptin and GnRH but did not alter serotonin. STUDY DESIGN, SIZE, DURATION In a longitudinal design, 27 adult (7-13 years) female cynomolgus macaques (Macaca fascicularis) with three different levels of sensitivity to stress were monitored with daily vaginal swabs and frequent serum progesterone (P) measurements. Three 90-day experimental periods called 'Cycle Sets' were monitored. A Cycle Set consisted of one ovulatory menstrual cycle without stress, and two cycles, or 60 days, with modest stress. Each Cycle Set was followed by a rest period. During a Cycle Set, individuals were either untreated (placebo) or administered escitalopram (CIT) or REB. Ultimately, half of each sensitivity group was euthanized during stress with CIT or REB treatment and the hypothalamus was obtained. Neurobiological endpoints were compared between CIT and REB treatment groups in stress resilient and SS monkeys. PARTICIPANTS/MATERIALS, SETTING, METHODS The monkeys were housed at the University of Pittsburgh primate facility for the duration of the experiments. Upon euthanasia, their brains and serum samples were shipped to the Oregon National Primate Research Center. The hypothalamus was examined with immunohistochemistry for the expression of DBH (a marker for NE axons), kisspeptin and GnRH. P was measured in the serum samples by radioimmunoassay. MAIN RESULTS AND THE ROLE OF CHANCE Daily administration of REB restored ovulation in 9 of 10 SS animals during stress. Of note, REB significantly increased P secretion during stress in the most sensitive group (P = 0.032), which indicates ovulation. CIT lacked efficacy. REB significantly reduced DBH/NE, kisspeptin and GnRH axon density in the hypothalamus relative to CIT treatment (P = 0.003. 0.018 and 0.0001, respectively) on Day 5 of the menstrual cycle in resilient and sensitive groups. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The US FDA has not approved REB for human use, although it is used in Europe for the treatment of depression/anxiety as EdronaxTR. Whether REB could be useful for the treatment of FHA in women has not been determined. WIDER IMPLICATIONS FOR THE FINDINGS The use of an NRI to treat FHA is a novel approach and the potential reinstatement of ovulation could be straightforward compared to current treatment protocols. The underlying neurobiology provides a compelling case for treating the origin of the pathology, i.e. elevated NE, rather than circumventing the hypothalamus altogether with gonadotropins, which have associated risks such as hyperstimulation syndrome or multiple births. STUDY FUNDING/COMPETING INTEREST(S) Portions of this study were supported by NIH grant HD062864 to C.L.B., NIH grant HD62618 to J.L.C. and C.L.B. and 1P51 OD011092 for the operation of the Oregon National Primate Research Center. There were no competing interests.
Collapse
Affiliation(s)
- Cynthia L Bethea
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA.,Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA.,Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97201, USA
| | - Judy L Cameron
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
31
|
Cui K, Yang F, Tufan T, Raza MU, Zhan Y, Fan Y, Zeng F, Brown RW, Price JB, Jones TC, Miller GW, Zhu MY. Restoration of Noradrenergic Function in Parkinson's Disease Model Mice. ASN Neuro 2021; 13:17590914211009730. [PMID: 33940943 PMCID: PMC8114769 DOI: 10.1177/17590914211009730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dysfunction of the central noradrenergic and dopaminergic systems is the primary neurobiological characteristic of Parkinson’s disease (PD). Importantly, neuronal loss in the locus coeruleus (LC) that occurs in early stages of PD may accelerate progressive loss of dopaminergic neurons. Therefore, restoring the activity and function of the deficient noradrenergic system may be an important therapeutic strategy for early PD. In the present study, the lentiviral constructions of transcription factors Phox2a/2b, Hand2 and Gata3, either alone or in combination, were microinjected into the LC region of the PD model VMAT2 Lo mice at 12 and 18 month age. Biochemical analysis showed that microinjection of lentiviral expression cassettes into the LC significantly increased mRNA levels of Phox2a, and Phox2b, which were accompanied by parallel increases of mRNA and proteins of dopamine β-hydroxylase (DBH) and tyrosine hydroxylase (TH) in the LC. Furthermore, there was considerable enhancement of DBH protein levels in the frontal cortex and hippocampus, as well as enhanced TH protein levels in the striatum and substantia nigra. Moreover, these manipulations profoundly increased norepinephrine and dopamine concentrations in the striatum, which was followed by a remarkable improvement of the spatial memory and locomotor behavior. These results reveal that over-expression of these transcription factors in the LC improves noradrenergic and dopaminergic activities and functions in this rodent model of PD. It provides the necessary groundwork for the development of gene therapies of PD, and expands our understanding of the link between the LC-norepinephrine and dopamine systems during the progression of PD.
Collapse
Affiliation(s)
- Kui Cui
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Fan Yang
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Hong Kong Institute, Asia Metropolitan University, Hong Kong, China
| | - Turan Tufan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Muhammad U Raza
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Yanqiang Zhan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Department of Neurology, Renmin Hospital of the Wuhan University, Wuhan, China
| | - Yan Fan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Department of Biochemistry, Nantong University College of Medicine, Nantong, China
| | - Fei Zeng
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Department of Neurology, Renmin Hospital of the Wuhan University, Wuhan, China
| | - Russell W Brown
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Jennifer B Price
- Department of Biological Sciences, College of Arts and Sciences; East Tennessee State University, Johnson City, United States
| | - Thomas C Jones
- Department of Biological Sciences, College of Arts and Sciences; East Tennessee State University, Johnson City, United States
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailmen School of Public Health, Columbia University, New York, New York, United States
| | - Meng-Yang Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| |
Collapse
|
32
|
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
|
33
|
Yong SJ. Persistent Brainstem Dysfunction in Long-COVID: A Hypothesis. ACS Chem Neurosci 2021; 12:573-580. [PMID: 33538586 PMCID: PMC7874499 DOI: 10.1021/acschemneuro.0c00793] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/01/2021] [Indexed: 02/07/2023] Open
Abstract
Long-COVID is a postviral illness that can affect survivors of COVID-19, regardless of initial disease severity or age. Symptoms of long-COVID include fatigue, dyspnea, gastrointestinal and cardiac problems, cognitive impairments, myalgia, and others. While the possible causes of long-COVID include long-term tissue damage, viral persistence, and chronic inflammation, the review proposes, perhaps for the first time, that persistent brainstem dysfunction may also be involved. This hypothesis can be split into two parts. The first is the brainstem tropism and damage in COVID-19. As the brainstem has a relatively high expression of ACE2 receptor compared with other brain regions, SARS-CoV-2 may exhibit tropism therein. Evidence also exists that neuropilin-1, a co-receptor of SARS-CoV-2, may be expressed in the brainstem. Indeed, autopsy studies have found SARS-CoV-2 RNA and proteins in the brainstem. The brainstem is also highly prone to damage from pathological immune or vascular activation, which has also been observed in autopsy of COVID-19 cases. The second part concerns functions of the brainstem that overlap with symptoms of long-COVID. The brainstem contains numerous distinct nuclei and subparts that regulate the respiratory, cardiovascular, gastrointestinal, and neurological processes, which can be linked to long-COVID. As neurons do not readily regenerate, brainstem dysfunction may be long-lasting and, thus, is long-COVID. Indeed, brainstem dysfunction has been implicated in other similar disorders, such as chronic pain and migraine and myalgic encephalomyelitis or chronic fatigue syndrome.
Collapse
Affiliation(s)
- Shin Jie Yong
- Department of Biological
Sciences, Sunway University, Petaling Jaya, Selangor 47500, Malaysia
| |
Collapse
|
34
|
Tsan L, Décarie-Spain L, Noble EE, Kanoski SE. Western Diet Consumption During Development: Setting the Stage for Neurocognitive Dysfunction. Front Neurosci 2021; 15:632312. [PMID: 33642988 PMCID: PMC7902933 DOI: 10.3389/fnins.2021.632312] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/19/2021] [Indexed: 01/18/2023] Open
Abstract
The dietary pattern in industrialized countries has changed substantially over the past century due to technological advances in agriculture, food processing, storage, marketing, and distribution practices. The availability of highly palatable, calorically dense foods that are shelf-stable has facilitated a food environment where overconsumption of foods that have a high percentage of calories derived from fat (particularly saturated fat) and sugar is extremely common in modern Westernized societies. In addition to being a predictor of obesity and metabolic dysfunction, consumption of a Western diet (WD) is related to poorer cognitive performance across the lifespan. In particular, WD consumption during critical early life stages of development has negative consequences on various cognitive abilities later in adulthood. This review highlights rodent model research identifying dietary, metabolic, and neurobiological mechanisms linking consumption of a WD during early life periods of development (gestation, lactation, juvenile and adolescence) with behavioral impairments in multiple cognitive domains, including anxiety-like behavior, learning and memory function, reward-motivated behavior, and social behavior. The literature supports a model in which early life WD consumption leads to long-lasting neurocognitive impairments that are largely dissociable from WD effects on obesity and metabolic dysfunction.
Collapse
Affiliation(s)
- Linda Tsan
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States.,Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States
| | - Léa Décarie-Spain
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States
| | - Emily E Noble
- Department of Foods and Nutrition, University of Georgia, Athens, GA, United States
| | - Scott E Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States.,Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
35
|
Sugama S, Kakinuma Y. Noradrenaline as a key neurotransmitter in modulating microglial activation in stress response. Neurochem Int 2020; 143:104943. [PMID: 33340593 DOI: 10.1016/j.neuint.2020.104943] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/09/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022]
Abstract
State of mind can influence susceptibility and progression of diseases and disorders not only in peripheral organs, but also in the central nervous system (CNS). However, the underlying mechanism how state of mind can affect susceptibility to various illnesses in the CNS is not fully understood. Among a number of candidates responsible for stress-induced neuroimmunomodulation, noradrenaline has recently been shown to play crucial roles in the major immune cells of the brain, microglia. In particular, recent studies have demonstrated that noradrenaline may be a key neurotransmitter in modulating microglial cells, thereby determining different cell conditions and responses ranging from resting to activation state depending on host stress level or whether the host is awake or asleep. For instance, microglia under resting conditions may have constructive roles in surveillance, such as debris clearance, synaptic monitoring, pruning, and remodeling. In contrast, once activated, microglia may become less efficient in surveillance activities, and instead implicated in detrimental roles such as cytokine or superoxide release. It is also likely that glial activation, both astrocytes and microglia, are negatively associated with the clearance of brain waste via the glymphatic system. In this review, we discuss the possible underlying mechanism as well as the roles of stress-induced microglial activation.
Collapse
Affiliation(s)
- Shuei Sugama
- Department of Physiology, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo, 113-8602, Japan.
| | - Yoshihiko Kakinuma
- Department of Physiology, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo, 113-8602, Japan
| |
Collapse
|
36
|
Fukuwada N, Kanno M, Yoshida S, Seki K. Gαq protein signaling in the bed nucleus of the stria terminalis regulate the lipopolysaccharide-induced despair-like behavior in mice. AIMS Neurosci 2020; 7:438-458. [PMID: 33263080 PMCID: PMC7701371 DOI: 10.3934/neuroscience.2020027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/05/2020] [Indexed: 11/18/2022] Open
Abstract
Major depressive disorder (MDD) is highly comorbid with anxiety disorders. It has been reported that the bed nucleus of the stria terminalis (BNST) is important for the induction of anxiety and MDD. Recently, the Gαq protein signaling within the BNST is involved in the induction of anxiety through Gαq protein signaling-mediated RNA-editing of GluR2 subunit, which produces the calcium (Ca2+)-impermeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor. On the other hand, the role of Gαq protein signaling within the BNST on the induction of MDD has never been reported yet. Therefore, we investigated whether Gαq protein signaling-producing the Ca2+-impermeable AMPA receptors in the BNST is involved in the lipopolysaccharide (LPS)-induced depressive-like behavior, particularly, despair-like behavior. When mice were systemically challenged with a single dose of LPS (1.2 mg/kg, i.p.), the immobility time during tail suspension test (TST) was increased 24 h after LPS injection. However, pretreatment with bilateral intra-BNST injection of neomycin (6.5 mM, 0.125 µL/side), an inhibitor of phospholipase C that is activated by Gαq protein-coupled receptor stimulation, extended the LPS-induced increase in the immobility time of TST. Furthermore, the co-pretreatment with bilateral intra-BNST injection of neomycin with 1-naphthylacetyl spermine (3 mM, 0.125 µL/side), an antagonist of Ca2+-permeable AMPA receptor, to mimic one of the final forms of Gαq protein activation, abolished the aggravated effect of neomycin and significantly shortened the immobility time compared with the control mice with an intra-BNST injection of artificial cerebrospinal fluid before LPS injection. However, pretreatment with bilateral intra-BNST injection of MDL-12,330A (10 µM, 0.125 µL/side), an inhibitor of adenylyl cyclase that is activated by Gαs protein-coupled receptor stimulation, did not affect the LPS-induced increase in the immobility time of TST. These results indicate that the Gαq protein signaling-mediated RNA-editing of GluR2, which produces the Ca2+-impermeable AMPA receptors within the BNST, regulates the LPS-induced despair-like behavior.
Collapse
Affiliation(s)
- Nao Fukuwada
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Miki Kanno
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Satomi Yoshida
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| | - Kenjiro Seki
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima 963-8611, Japan
| |
Collapse
|
37
|
Grueschow M, Kleim B, Ruff CC. Role of the locus coeruleus arousal system in cognitive control. J Neuroendocrinol 2020; 32:e12890. [PMID: 32820571 DOI: 10.1111/jne.12890] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/18/2020] [Accepted: 07/08/2020] [Indexed: 12/13/2022]
Abstract
Cognitive control lies at the core of human adaptive behaviour. Humans vary substantially in their ability to execute cognitive control with respect to optimally facing environmental challenges, although the neural origins of this heterogeneity are currently not well understood. Recent theoretical frameworks implicate the locus coeruleus noradrenergic arousal system (LC-NE) in that process. Invasive neurophysiological work in rodents has shown that the LC-NE is an important homeostatic control centre of the body. LC-NE innervates the entire neocortex and has particularly strong connections with the cingulate gyrus. In the present study, using a response conflict task, functional magnetic resonance imaging and concurrent pupil dilation measures (a proxy for LC-NE firing), we provide empirical evidence for a decisive role of the LC-NE in cognitive control in humans. We show that the level of individual behavioural adjustment in cognitive control relates to the level of functional coupling between LC-NE and the dorsomedial prefrontal cortex, as well as dorsolateral prefrontal cortex. Moreover, we show that the pupil is substantially more dilated during conflict trials requiring behavioural adjustment than during no conflict trials. In addition, we explore a potential relationship between pupil dilation and neural activity during choice conflict adjustments. Our data provide novel insight into arousal-related influences on cognitive control and suggest pupil dilation as a potential external marker for endogenous neural processes involved in optimising behavioural control. Our results may also be clinically relevant for a variety of pathologies where cognitive control is compromised, such as anxiety, depression, addiction and post-traumatic stress disorder.
Collapse
Affiliation(s)
- Marcus Grueschow
- Department of Economics, Zurich Center for Neuroeconomics (ZNE), University of Zurich, Zurich, Switzerland
| | - Birgit Kleim
- Department of Experimental Psychopathology and Psychotherapy, University of Zurich, Zurich, Switzerland
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Zurich, Switzerland
| | - Christian C Ruff
- Department of Economics, Zurich Center for Neuroeconomics (ZNE), University of Zurich, Zurich, Switzerland
| |
Collapse
|
38
|
Trigeminal input, pupil size and cognitive performance: From oral to brain matter. Brain Res 2020; 1751:147194. [PMID: 33159973 DOI: 10.1016/j.brainres.2020.147194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/14/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022]
Abstract
It has been observed that, in patients affected by temporomandibular disorders (TMDs) and edentulism, a left-right asymmetry in electromyographic (EMG) activity of masseter muscles during clenching and in pupil size at rest (anisocoria) is present. Both are greatly reduced by an orthotic-prosthetic correction. In parallel, the correction significantly improves cognitive performance. These effects are possibly due to the recovery of a cortical balance, via Locus Coeruleus (LC) modulation, whose activity is powerfully affected by the sensorimotor trigeminal input. The role of this functional axis was further investigated in subjects without overt occlusal or dental problems. In these individuals, the EMG asymmetry was significantly correlated to anisocoria at rest, with the dental arches open or in contact. Also in normal subjects, both the EMG and the pupil asymmetry during clenching could be significantly reduced by an orthotic (bite) correction. Closing the arches without bite increased anisocoria and reduced performance in the Spinnler-Tognoni matrices test, as well as the mydriasis induced by a haptic task. When the bite was interposed, anisocoria was reduced, while both performance and task-related mydriasis were enhanced. Since pupil size is considered a proxy of the LC activity, these results suggest that asymmetric occlusion biases the LC discharge and the hemispheric excitability, possibly via a sensorimotor trigeminal imbalance. Removing the anisocoria through bite correction re-establishes a symmetric LC discharge, improving performance and enhancing task-related mydriasis. Therefore, occlusal balancing may represent a tool for improving subjective performance and may be exploited for training and rehabilitative purposes.
Collapse
|
39
|
Ye R, Rua C, O'Callaghan C, Jones PS, Hezemans FH, Kaalund SS, Tsvetanov KA, Rodgers CT, Williams G, Passamonti L, Rowe JB. An in vivo probabilistic atlas of the human locus coeruleus at ultra-high field. Neuroimage 2020; 225:117487. [PMID: 33164875 PMCID: PMC7779564 DOI: 10.1016/j.neuroimage.2020.117487] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/22/2020] [Accepted: 10/17/2020] [Indexed: 12/16/2022] Open
Abstract
Early and profound pathological changes are evident in the locus coeruleus (LC) in dementia and Parkinson's disease, with effects on arousal, attention, cognitive and motor control. The LC can be identified in vivo using non-invasive magnetic resonance imaging techniques which have potential as biomarkers for detecting and monitoring disease progression. Technical limitations of existing imaging protocols have impaired the sensitivity to regional contrast variance or the spatial variability on the rostrocaudal extent of the LC, with spatial mapping consistent with post mortem findings. The current study employs a sensitive magnetisation transfer sequence using ultrahigh field 7T MRI to investigate the LC structure in vivo at high-resolution (0.4 × 0.4 × 0.5 mm). Magnetisation transfer images from 53 healthy older volunteers (52 - 84 years) clearly revealed the spatial features of the LC and were used to create a probabilistic LC atlas for older adults. This atlas may be especially relevant for studying disorders associated with older age. To use the atlas does not require use of the same MT sequence of 7T MRI, provided good co-registration and normalisation is achieved. Consistent rostrocaudal gradients of slice-wise volume, contrast and variance along the LC were observed, mirroring distinctive ex vivo spatial distributions of LC cells in its subregions. The contrast-to-noise ratios were calculated for the peak voxels, and for the averaged signals within the atlas, to accommodate the volumetric differences in estimated contrast. The probabilistic atlas is freely available, and the MRI dataset will be made available for non-commercial research, for replication or to facilitate accurate LC localisation and unbiased contrast extraction in future studies.
Collapse
Affiliation(s)
- Rong Ye
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK.
| | - Catarina Rua
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK; Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Claire O'Callaghan
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine, University of Sydney, Sydney, Australia; Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - P Simon Jones
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Frank H Hezemans
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK; MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Sanne S Kaalund
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Kamen A Tsvetanov
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK; Department of Psychology, University of Cambridge, Cambridge, UK
| | | | - Guy Williams
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Luca Passamonti
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK; Istituto di Bioimmagini e Fisiologia Molecolare (IBFM), Consiglio Nazionale delle Ricerche (CNR), Milano, Italy
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK; MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
40
|
Coutens B, Rekik K, Harster A, Etienne P, Noirot V, Frances B, Moulédous L, Guiard BP. A Citrus Based Sensory Functional Food Ingredient Induces Antidepressant-like Effects: Possible Involvement of an Interplay between the Olfactory and the Serotonergic Systems. Neuroscience 2020; 451:149-163. [PMID: 33039523 DOI: 10.1016/j.neuroscience.2020.09.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/17/2020] [Accepted: 09/20/2020] [Indexed: 12/28/2022]
Abstract
In the present study, we examined the neurobehavioral effects of a sensory functional food ingredient mainly based on Citrus sinensis extracts (D11399) using a battery of tests recapitulating various endophenotypes of depression such as anxiety in the open field (OF), the elevated plus-maze (EPM), and the novelty suppressed feeding (NSF), self-care in the splash test (ST), despair in the forced swimming task (FST) but also anhedonia in the sucrose preference test (SPT) in mice. A one-week oral administration of D11399 promoted anxiolytic- and antidepressant-like responses in naïve mice subjected to the NSF and FST. In a marked contrast, the administration of D11399 by oral gavage or the inhibition of olfaction by methimazole prevented such beneficial effects. We further investigated the neurobehavioral properties of a ten-week oral administration of D11399 in the corticosterone (CORT) mouse model of depression. Interestingly, D11399 also elicited anxiolytic- and antidepressant-like effects in various paradigms. To characterize the putative underpinning neurobiological mechanisms in CORT mice, we investigated whether cellular and molecular processes commonly associated with antidepressant responses such as monoaminergic neurotransmission and neuronal maturation in the hippocampus were impacted. Although D11399 did not modify the hippocampal extracellular levels of monoamines (i.e. serotonin and norepinephrine), it reversed the ability of CORT to decrease serotonin neurons firing rate in the dorsal raphe and neuronal maturation in the hippocampus. These findings suggest that the anxiolytic- and antidepressant-like effects of this sensory functional food ingredient are closely related with olfaction and likely a concomitant change in the activity of the central serotonergic system. Further experiments are warranted to precise the neuronal circuits linking sensorial and emotional modalities and identify innovative therapeutic strategies aimed to relieve depressive endophenotypes.
Collapse
Affiliation(s)
- B Coutens
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - K Rekik
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - A Harster
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | | | | | - B Frances
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - L Moulédous
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France
| | - B P Guiard
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, France; Faculté de Pharmacie, Université Paris Sud, Université Paris-Saclay, 92290 Chatenay-Malabry, France.
| |
Collapse
|
41
|
Flasinski T, Dierolf AM, Rost S, Lutz APC, Voderholzer U, Koch S, Bach M, Asenstorfer C, Münch EE, Mertens VC, Vögele C, Schulz A. Altered Interoceptive Awareness in High Habitual Symptom Reporters and Patients With Somatoform Disorders. Front Psychol 2020; 11:1859. [PMID: 32849092 PMCID: PMC7426478 DOI: 10.3389/fpsyg.2020.01859] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/07/2020] [Indexed: 01/07/2023] Open
Abstract
Objective: Altered interoception may play a major role in the etiology of medically unexplained symptoms (MUS). It remains unclear, however, if these alterations concern noticing of signals or if they are limited to the interpretation of signals. We investigated whether individuals with MUS differ in interoceptive awareness as assessed with the Multidimensional Assessment of Interoceptive Awareness (MAIA) questionnaire. Methods: Study 1: A total of 486 individuals completed the Screening for Somatoform Disorders (SOMS-2). Thirty-two individuals each of the upper and lower decile of the SOMS distribution (low symptom reporters/LSR, high symptom reporters/HSR) completed the MAIA. Study 2: MAIA scores of individuals diagnosed with somatoform disorder (SFD; n = 26) were compared to individuals with major depressive disorder (MDD; n = 25) and healthy controls (HC; n = 26). Results: HSR had lower scores than LSR on the MAIA scales Not-Distracting and Not-Worrying. The SFD and MDD groups showed lower scores than HC on the MAIA scales Not-Distracting, Self-Regulation, and Trusting. The MDD group scored lower than the other two groups on the scales Body Listening and Attention Regulation. There were no group differences on the scale Noticing. Conclusion: HSR, SFD, and MDD patients do not differ from HC in the awareness of noticing of interoceptive signal processing, whereas cognitive facets of interoception, such as distraction or self-regulation are differentially affected. This highlights the necessity of including specifically targeted interventions, which improve interoceptive awareness, in the prevention and treatment of SFDs.
Collapse
Affiliation(s)
- Tabea Flasinski
- Clinical Psychophysiology Laboratory, Institute for Health and Behaviour, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Mental Health Research and Treatment Center, Faculty of Psychology, Ruhr-University Bochum, Bochum, Germany
| | - Angelika Margarete Dierolf
- Clinical Psychophysiology Laboratory, Institute for Health and Behaviour, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Silke Rost
- Clinical Psychophysiology Laboratory, Institute for Health and Behaviour, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Annika P C Lutz
- Clinical Psychophysiology Laboratory, Institute for Health and Behaviour, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | | | - Stefan Koch
- Schön Klinik Roseneck, Prien am Chiemsee, Germany
| | | | | | - Eva Elisabeth Münch
- Clinical Psychophysiology Laboratory, Institute for Health and Behaviour, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Vera-Christina Mertens
- Clinical Psychophysiology Laboratory, Institute for Health and Behaviour, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Claus Vögele
- Clinical Psychophysiology Laboratory, Institute for Health and Behaviour, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - André Schulz
- Clinical Psychophysiology Laboratory, Institute for Health and Behaviour, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
42
|
Chang YP, Lee MS, Wu DW, Tsai JH, Ho PS, Lin CHR, Chuang HY. Risk factors for depression in patients with Parkinson's disease: A nationwide nested case-control study. PLoS One 2020; 15:e0236443. [PMID: 32716954 PMCID: PMC7384643 DOI: 10.1371/journal.pone.0236443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/05/2020] [Indexed: 11/30/2022] Open
Abstract
Objectives Patients with Parkinson’s disease (PD) have higher prevalence of depression than the general population; however, the risk factors for depression in PD remain uncertain. Methods/Design Using the 2000–2010 Taiwan National Health Insurance Research Database, we selected 1767 patients aged ≧ 40 years with new-onset PD during 2000–2009. Among them, 324 patients with a new incidence of depression were enrolled as cases and 972 patients without depression were randomly selected as controls. The groups were frequency-matched at a ratio of 1:3 by age, sex, and index year. Thus, this nested case-control study compared differences between the cases and the controls. Logistic regression models were used to identify risk factors for depression in PD. Results Compared with the controls, the odds ratio (OR) of anxiety disorders in the cases was 1.53 (95% confidence interval [95% CI], 1.16–2.02; P = 0.003), after adjusting for the confounding factors of age, sex, index year, geographic region, urban level, monthly income, and other coexisting medical conditions. The OR for sleep disturbances in the cases was 1.49 (95% CI, 1.14–1.96; P = 0.004) compared to the controls, after adjusting these confounding factors. Hence, the risk factors for depression in PD were nonsignificantly associated with physical comorbidities. Conclusions In the present study, depression in PD was significantly associated with anxiety disorders and sleep disturbances. Integrated care for early identification and treatment of neuropsychiatric comorbidities is crucial in patients with new-onset PD so as to prevent further PD degeneration.
Collapse
Affiliation(s)
- Yang-Pei Chang
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Min-Sheng Lee
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Da-Wei Wu
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jui-Hsiu Tsai
- Department of Psychiatry, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chia-Yi, Taiwan
- Program in Environmental and Occupation Medicine, (Taiwan) National Health Research Institutes and Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail: (JHT); (PSH); (CHRL)
| | - Pei-Shan Ho
- Department of Oral Hygiene, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Medical Statistics and Bioinformatics, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail: (JHT); (PSH); (CHRL)
| | - Chun-Hung Richard Lin
- Department of Computer Science and Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
- * E-mail: (JHT); (PSH); (CHRL)
| | - Hung-Yi Chuang
- Program in Environmental and Occupation Medicine, (Taiwan) National Health Research Institutes and Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Occupational Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
43
|
Tona KD, Revers H, Verkuil B, Nieuwenhuis S. Noradrenergic Regulation of Cognitive Flexibility: No Effects of Stress, Transcutaneous Vagus Nerve Stimulation, and Atomoxetine on Task-switching in Humans. J Cogn Neurosci 2020; 32:1881-1895. [PMID: 32644883 DOI: 10.1162/jocn_a_01603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Cognitive flexibility allows us to adaptively switch between different responsibilities in important domains of our daily life. Previous work has elucidated the neurochemical basis underlying the ability to switch responses to a previously nonreinforced exemplar and to switch between attentional sets. However, the role of neuromodulators in task switching, the ability to rapidly switch between two or more cognitive tasks afforded by the same stimuli, is still poorly understood. We attempted to fill this gap by manipulating norepinephrine levels using stress manipulation (Study 1a, n = 48; between-group design), transcutaneous vagus nerve stimulation at two different intensities (Study 1b, n = 48; sham-controlled between-group design), and pharmacological manipulation (Study 2, n = 24; double-blind crossover design), all of which increased salivary cortisol measures. Participants repeatedly switched between two cognitive tasks (classifying a digit as high/low [Task 1] or as odd/even [Task 2]), depending on the preceding cue. On each trial, a cue indicated the task to be performed. The cue-stimulus interval was varied to manipulate the time to prepare for the switch. Participants showed typical switch costs, which decreased with the time available for preparation. None of the manipulations modulated the size of the switch costs or the preparation effect, as supported by frequentist and Bayesian model comparisons. Task-switching performance reflects a complex mix of cognitive control and bottom-up dynamics of task-set representations. Our findings suggest that norepinephrine does not affect either of these aspects of cognitive flexibility.
Collapse
Affiliation(s)
| | | | - Bart Verkuil
- Leiden University.,Leiden Institute for Brain and Cognition
| | | |
Collapse
|
44
|
Lustberg D, Iannitelli AF, Tillage RP, Pruitt M, Liles LC, Weinshenker D. Central norepinephrine transmission is required for stress-induced repetitive behavior in two rodent models of obsessive-compulsive disorder. Psychopharmacology (Berl) 2020; 237:1973-1987. [PMID: 32313981 PMCID: PMC7961804 DOI: 10.1007/s00213-020-05512-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/26/2020] [Indexed: 02/06/2023]
Abstract
RATIONALE Obsessive-compulsive disorder (OCD) is characterized by repetitive behaviors exacerbated by stress. Many OCD patients do not respond to available pharmacotherapies, but neurosurgical ablation of the anterior cingulate cortex (ACC) can provide symptomatic relief. Although the ACC receives noradrenergic innervation and expresses adrenergic receptors (ARs), the involvement of norepinephrine (NE) in OCD has not been investigated. OBJECTIVE To determine the effects of genetic or pharmacological disruption of NE neurotransmission on marble burying (MB) and nestlet shredding (NS), two animal models of OCD. METHODS We assessed NE-deficient (Dbh -/-) mice and NE-competent (Dbh +/-) controls in MB and NS tasks. We also measured the effects of anti-adrenergic drugs on NS and MB in control mice and the effects of pharmacological restoration of central NE in Dbh -/- mice. Finally, we compared c-fos induction in the locus coeruleus (LC) and ACC of Dbh -/- and control mice following both tasks. RESULTS Dbh -/- mice virtually lacked MB and NS behaviors seen in control mice but did not differ in the elevated zero maze (EZM) model of general anxiety-like behavior. Pharmacological restoration of central NE synthesis in Dbh -/- mice completely rescued NS behavior, while NS and MB were suppressed in control mice by anti-adrenergic drugs. Expression of c-fos in the ACC was attenuated in Dbh -/- mice after MB and NS. CONCLUSION These findings support a role for NE transmission to the ACC in the expression of stress-induced compulsive behaviors and suggest further evaluation of anti-adrenergic drugs for OCD is warranted.
Collapse
Affiliation(s)
- Daniel Lustberg
- Department of Human Genetics, Emory University School of Medicine, 615 Michael St., Whitehead 301, Atlanta, GA, 30322, USA
| | - Alexa F Iannitelli
- Department of Human Genetics, Emory University School of Medicine, 615 Michael St., Whitehead 301, Atlanta, GA, 30322, USA
| | - Rachel P Tillage
- Department of Human Genetics, Emory University School of Medicine, 615 Michael St., Whitehead 301, Atlanta, GA, 30322, USA
| | - Molly Pruitt
- University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - L Cameron Liles
- Department of Human Genetics, Emory University School of Medicine, 615 Michael St., Whitehead 301, Atlanta, GA, 30322, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, 615 Michael St., Whitehead 301, Atlanta, GA, 30322, USA.
| |
Collapse
|
45
|
Du X, Yin M, Yuan L, Zhang G, Fan Y, Li Z, Yuan N, Lv X, Zhao X, Zou S, Deng W, Kosten TR, Zhang XY. Reduction of depression-like behavior in rat model induced by ShRNA targeting norepinephrine transporter in locus coeruleus. Transl Psychiatry 2020; 10:130. [PMID: 32366842 PMCID: PMC7198598 DOI: 10.1038/s41398-020-0808-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 04/07/2020] [Accepted: 04/16/2020] [Indexed: 02/05/2023] Open
Abstract
Depression may be associated with reduced monoamine neurotransmission, particularly serotonin and norepinephrine (NE). Reuptake of NE by the norepinephrine transporter (NET) is the primary mechanism by which many of the antidepressants are high-affinity substrates for NET. This study aimed to examine the effect of lentivirus-mediated shRNA targeting NET in locus coeruleus (LC) on depression-like behaviors of rats. We randomly assigned 60 male Wistar rats to 6 experimental groups: (1) Control group: without chronic unpredictable mild stress (CUMS) and without NET-shRNA treatment; (2) shRNA group: without CUMS + NET-shRNA; (3) CUMS group: 3-week CUMS without NET-shRNA; (4) CUMS + nonsense shRNA group; (5) CUMS + amygdala (Amy)-shRNA group; (6) CUMS+ locus coeruleus (LC)-shRNA group. First, recombinant lentiviral vector expressing shRNA (ShRNA-629, ShRNA-330, ShRNA-1222, ShRNA-1146 or ShRNA- negative control) against NET were produced, and their efficiency in knocking down of NET in PC12 cells were assessed by Q-PCR and western blot analysis. Second, shRNA was injected into the rat LC bilaterally to investigate whether it could prevent the depressive-like behavior induced by 3-week CUMS. Third, we tested the depressive-like behavior of the rats in the forced swimming test, the open field test, the sucrose preference test, as well as the body weight gain at the end of the seventh week. Finally, the protein expressions of NET was measured by western blot and the NE levels were measured by high performance liquid chromatography. Q-PCR and western blot showed that the ShRNA-1146 had the best interference efficiency targeting on NET in PC12 cells (p < 0.01). Compared to the depression model group, the immobility time in the forced swimming test was significantly reduced (p < 0.01), but the sucrose preference and the total scores in the open field test were significantly increased (all p < 0.01) in the group treated with shRNA in LC. Furthermore, compared with the depression model group, NET levels were significantly decreased (p < 0.01), but NE levels were significantly increased in the group treated with shRNA in LC (p < 0.05). Our findings suggest that Lentivirus-mediated shRNA targeting NET in LC downregulated NET both in vitro and in vivo, resulting in a significant decrease in depressive-like behavior of rats.
Collapse
Affiliation(s)
- Xiangdong Du
- Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China.
| | - Ming Yin
- grid.263761.70000 0001 0198 0694Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Lian Yuan
- grid.263761.70000 0001 0198 0694Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Guangya Zhang
- grid.263761.70000 0001 0198 0694Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Yan Fan
- grid.263761.70000 0001 0198 0694Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Zhe Li
- grid.263761.70000 0001 0198 0694Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Nian Yuan
- grid.263761.70000 0001 0198 0694Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Xiaoli Lv
- grid.263761.70000 0001 0198 0694Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Xueli Zhao
- grid.263761.70000 0001 0198 0694Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Siyun Zou
- grid.263761.70000 0001 0198 0694Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Wei Deng
- grid.13291.380000 0001 0807 1581Department of Psychiatry and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Thomas R. Kosten
- grid.39382.330000 0001 2160 926XDepartment of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
| | - Xiang Yang Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China. .,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
46
|
Song NN, Ma P, Zhang Q, Zhang L, Wang H, Zhang L, Zhu L, He CH, Mao B, Ding YQ. Rnf220/Zc4h2-mediated monoubiquitylation of Phox2 is required for noradrenergic neuron development. Development 2020; 147:dev185199. [PMID: 32094113 DOI: 10.1242/dev.185199] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/11/2020] [Indexed: 11/20/2022]
Abstract
Noradrenaline belongs to the monoamine system and is involved in cognition and emotional behaviors. Phox2a and Phox2b play essential but non-redundant roles during development of the locus coeruleus (LC), the main noradrenergic (NA) neuron center in the mammalian brain. The ubiquitin E3 ligase Rnf220 and its cofactor Zc4h2 participate in ventral neural tube patterning by modulating Shh/Gli signaling, and ZC4H2 mutation is associated with intellectual disability, although the mechanisms for this remain poorly understood. Here, we report that Zc4h2 and Rnf220 are required for the development of central NA neurons in the mouse brain. Both Zc4h2 and Rnf220 are expressed in developing LC-NA neurons. Although properly initiated at E10.5, the expression of genes associated with LC-NA neurons is not maintained at the later embryonic stages in mice with a deficiency of either Rnf220 or Zc4h2 In addition, we show that the Rnf220/Zc4h2 complex monoubiquitylates Phox2a/Phox2b, a process required for the full transcriptional activity of Phox2a/Phox2b. Our work reveals a role for Rnf220/Zc4h2 in regulating LC-NA neuron development, and this finding may be helpful for understanding the pathogenesis of ZC4H2 mutation-associated intellectual disability.
Collapse
Affiliation(s)
- Ning-Ning Song
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Pengcheng Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Qiong Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Huishan Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650203, China
| | - Longlong Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650203, China
| | - Liang Zhu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650203, China
| | - Chun-Hui He
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Yu-Qiang Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
47
|
Yu HY, Wang SY, Quan CX, Fang C, Luo SC, Li DY, Zhen SS, Ma JH, Duan KM. Dexmedetomidine Alleviates Postpartum Depressive Symptoms following Cesarean Section in Chinese Women: A Randomized Placebo-Controlled Study. Pharmacotherapy 2019; 39:994-1004. [PMID: 31411762 DOI: 10.1002/phar.2320] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVES Few studies have investigated the prophylactic efficacy of dexmedetomidine (DEX) in postpartum depressive symptoms (PDS). A randomized double-blind placebo-controlled trial was conducted to investigate whether the administration of DEX, immediately after delivery and for patient-controlled intravenous analgesia (PCIA), can attenuate PDS. METHODS A total of 600 parturients scheduled for elective cesarean delivery under spinal anesthesia were randomly allocated into the control group (infusion with 0.9% normal saline after delivery and PCIA with sufentanil) and the DEX group (DEX infusion 0.5 μg/kg after delivery and PCIA with DEX plus sufentanil). The prevalence of postpartum depressive disorders was indicated by the Edinburgh Postnatal Depression Scale (EPDS). Postoperative analgesia, sedation, and sleep quality of parturients were also assessed. RESULTS Postpartum blues and PDS prevalence in the DEX, versus control, group were significantly lower (5.0% vs 14.1%, p<0.001; 5.7% vs 16.3%, p<0.001, respectively), especially in parturients with antenatal depression or moderate stress during pregnancy. Compared with the control group, the EPDS score at postpartum days 7 and 42 in the DEX group was significantly lower (4.23 ± 4.37 vs 1.93 ± 3.36, p<0.001; 4.68 ± 4.78 vs 1.99 ± 3.18, p<0.001, respectively), as was the incidence of postpartum self-harm ideation at postpartum days 7 and 42 in the DEX group versus the control group (1.1% vs 4.0%, p=0.03; 0.4% vs 2.9%, p=0.04, respectively). The pain score and the sleep quality in the DEX group were better than that in the control group (p<0.001). CONCLUSION The application of DEX in the early postpartum period can significantly attenuate the incidence of postpartum depressive disorders.
Collapse
Affiliation(s)
- He-Ya Yu
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Sai-Ying Wang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Cheng-Xuan Quan
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Chao Fang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Shi-Chao Luo
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Dan-Yang Li
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Shan-Shan Zhen
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Jia-Hui Ma
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Kai-Ming Duan
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
48
|
Zahola P, Hanics J, Pintér A, Máté Z, Gáspárdy A, Hevesi Z, Echevarria D, Adori C, Barde S, Törőcsik B, Erdélyi F, Szabó G, Wagner L, Kovacs GG, Hökfelt T, Harkany T, Alpár A. Secretagogin expression in the vertebrate brainstem with focus on the noradrenergic system and implications for Alzheimer's disease. Brain Struct Funct 2019; 224:2061-2078. [PMID: 31144035 PMCID: PMC6591208 DOI: 10.1007/s00429-019-01886-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 05/03/2019] [Indexed: 12/04/2022]
Abstract
Calcium-binding proteins are widely used to distinguish neuronal subsets in the brain. This study focuses on secretagogin, an EF-hand calcium sensor, to identify distinct neuronal populations in the brainstem of several vertebrate species. By using neural tube whole mounts of mouse embryos, we show that secretagogin is already expressed during the early ontogeny of brainstem noradrenaline cells. In adults, secretagogin-expressing neurons typically populate relay centres of special senses and vegetative regulatory centres of the medulla oblongata, pons and midbrain. Notably, secretagogin expression overlapped with the brainstem column of noradrenergic cell bodies, including the locus coeruleus (A6) and the A1, A5 and A7 fields. Secretagogin expression in avian, mouse, rat and human samples showed quasi-equivalent patterns, suggesting conservation throughout vertebrate phylogeny. We found reduced secretagogin expression in locus coeruleus from subjects with Alzheimer’s disease, and this reduction paralleled the loss of tyrosine hydroxylase, the enzyme rate limiting noradrenaline synthesis. Residual secretagogin immunoreactivity was confined to small submembrane domains associated with initial aberrant tau phosphorylation. In conclusion, we provide evidence that secretagogin is a useful marker to distinguish neuronal subsets in the brainstem, conserved throughout several species, and its altered expression may reflect cellular dysfunction of locus coeruleus neurons in Alzheimer’s disease.
Collapse
Affiliation(s)
- Péter Zahola
- SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary.,Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - János Hanics
- SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary.,Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - Anna Pintér
- Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - Zoltán Máté
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Anna Gáspárdy
- Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - Zsófia Hevesi
- SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary.,Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Diego Echevarria
- Institute of Neuroscience, University of Miguel Hernandez de Elche, Alicante, Spain
| | - Csaba Adori
- Department of Neuroscience, Karolinska Institutet, Biomedicum 7D, SE-17165, Stockholm, Sweden
| | - Swapnali Barde
- Department of Neuroscience, Karolinska Institutet, Biomedicum 7D, SE-17165, Stockholm, Sweden
| | - Beáta Törőcsik
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Ferenc Erdélyi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gábor Szabó
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Ludwig Wagner
- Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Biomedicum 7D, SE-17165, Stockholm, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, 1090, Vienna, Austria.,Department of Neuroscience, Karolinska Institutet, Biomedicum 7D, SE-17165, Stockholm, Sweden
| | - Alán Alpár
- SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary. .,Department of Anatomy, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
49
|
Tache Y, Larauche M, Yuan PQ, Million M. Brain and Gut CRF Signaling: Biological Actions and Role in the Gastrointestinal Tract. Curr Mol Pharmacol 2018; 11:51-71. [PMID: 28240194 DOI: 10.2174/1874467210666170224095741] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/16/2016] [Accepted: 08/03/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Corticotropin-releasing factor (CRF) pathways coordinate behavioral, endocrine, autonomic and visceral responses to stress. Convergent anatomical, molecular, pharmacological and functional experimental evidence supports a key role of brain CRF receptor (CRF-R) signaling in stress-related alterations of gastrointestinal functions. These include the inhibition of gastric acid secretion and gastric-small intestinal transit, stimulation of colonic enteric nervous system and secretorymotor function, increase intestinal permeability, and visceral hypersensitivity. Brain sites of CRF actions to alter gut motility encompass the paraventricular nucleus of the hypothalamus, locus coeruleus complex and the dorsal motor nucleus while those modulating visceral pain are localized in the hippocampus and central amygdala. Brain CRF actions are mediated through the autonomic nervous system (decreased gastric vagal and increased sacral parasympathetic and sympathetic activities). The activation of brain CRF-R2 subtype inhibits gastric motor function while CRF-R1 stimulates colonic secretomotor function and induces visceral hypersensitivity. CRF signaling is also located within the gut where CRF-R1 activates colonic myenteric neurons, mucosal cells secreting serotonin, mucus, prostaglandin E2, induces mast cell degranulation, enhances mucosal permeability and propulsive motor functions and induces visceral hyperalgesia in animals and humans. CRF-R1 antagonists prevent CRF- and stressrelated gut alterations in rodents while not influencing basal state. DISCUSSION These preclinical studies contrast with the limited clinical positive outcome of CRF-R1 antagonists to alleviate stress-sensitive functional bowel diseases such as irritable bowel syndrome. CONCLUSION The translational potential of CRF-R1 antagonists in gut diseases will require additional studies directed to novel anti-CRF therapies and the neurobiology of brain-gut interactions under chronic stress.
Collapse
Affiliation(s)
- Yvette Tache
- CURE/Digestive Diseases Research Center, G Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine at UCLA and VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073. United States
| | - Muriel Larauche
- CURE/Digestive Diseases Research Center, G Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine at UCLA and VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073. United States
| | - Pu-Qing Yuan
- CURE/Digestive Diseases Research Center, G Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine at UCLA and VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073. United States
| | - Mulugeta Million
- CURE/Digestive Diseases Research Center, G Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine at UCLA and VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073. United States
| |
Collapse
|
50
|
Campos-Lira E, Kelly L, Seifi M, Jackson T, Giesecke T, Mutig K, Koshimizu TAA, Hernandez VS, Zhang L, Swinny JD. Dynamic Modulation of Mouse Locus Coeruleus Neurons by Vasopressin 1a and 1b Receptors. Front Neurosci 2018; 12:919. [PMID: 30618551 PMCID: PMC6295453 DOI: 10.3389/fnins.2018.00919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/22/2018] [Indexed: 11/29/2022] Open
Abstract
The locus coeruleus (LC) is a brainstem nucleus distinguished by its supply of noradrenaline throughout the central nervous system. Apart from modulating a range of brain functions, such as arousal, cognition and the stress response, LC neuronal excitability also corresponds to the activity of various peripheral systems, such as pelvic viscera and the cardiovascular system. Neurochemically diverse inputs set the tone for LC neuronal activity, which in turn modulates these adaptive physiological and behavioral responses essential for survival. One such LC afferent system which is poorly understood contains the neurohormone arginine-vasopressin (AVP). Here we provide the first demonstration of the molecular and functional characteristics of the LC-AVP system, by characterizing its receptor-specific modulation of identified LC neurons and plasticity in response to stress. High resolution confocal microscopy revealed that immunoreactivity for the AVP receptor 1b (V1b) was located on plasma membranes of noradrenergic and non-noradrenergic LC neurons. In contrast, immunoreactivity for the V1a receptor was exclusively located on LC noradrenergic neurons. No specific signal, either at the mRNA or protein level, was detected for the V2 receptor in the LC. Clusters immunoreactive for V1a-b were located in proximity to profiles immunoreactive for GABAergic and glutamatergic synaptic marker proteins. AVP immunopositive varicosities were also located adjacent to labeling for such synaptic markers. Whole-cell patch clamp electrophysiology revealed that the pharmacological activation of V1b receptors significantly increased the spontaneous activity of 45% (9/20) of recorded noradrenergic neurons, with the remaining 55% (11/20) of cells exhibiting a significant decrease in their basal firing patterns. Blockade of V1a and V1b receptors on their own significantly altered LC neuronal excitability in a similar heterogeneous manner, demonstrating that endogenous AVP sets the basal LC neuronal firing rates. Finally, exposing animals to acute stress increased V1b, but not V1a receptor expression, whilst decreasing AVP immunoreactivity. This study reveals the AVP-V1a-b system as a considerable component of the LC molecular architecture and regulator of LC activity. Since AVP primarily functions as a regulator of homeostasis, the data suggest a novel pathway by modulating the functioning of a brain region that is integral to mediating adaptive responses.
Collapse
Affiliation(s)
- Elba Campos-Lira
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Louise Kelly
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Mohsen Seifi
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Torquil Jackson
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Torsten Giesecke
- Department of Anatomy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kerim Mutig
- Department of Anatomy, Charité - Universitätsmedizin Berlin, Berlin, Germany.,I.M. Sechenov First Moscow State Medical University of the Ministry of Healthcare of the Russian Federation (Sechenovskiy University), Moscow, Russia
| | - Taka-Aki A Koshimizu
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke, Japan
| | - Vito S Hernandez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Limei Zhang
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jerome D Swinny
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|