1
|
Oliveira A, Azevedo M, Seixas R, Silva S, Martinho R, Serrão P, Silva E, Moreira-Rodrigues M. Hippocampus muscarinic M4 receptor mRNA expression may influence central cholinergic activity, causing fear memory strengthening by peripheral adrenaline. Neuropharmacology 2025; 271:110382. [PMID: 39988278 DOI: 10.1016/j.neuropharm.2025.110382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Adrenaline (Ad) strengthens contextual fear memory by increasing blood glucose, possibly enhancing hippocampus acetylcholine synthesis. Nevertheless, it is unclear if peripheral Ad influences the cholinergic system, contributing to contextual fear memory strengthening. We aimed to evaluate whether peripheral Ad alters muscarinic receptor expression and if the cholinergic system is involved in peripheral Ad contextual fear memory strengthening effect. Wild-type (WT) and Ad-deficient male mice (129 × 1/SvJ) underwent a fear conditioning procedure followed by intraperitoneal pre-training and pre-context administration of Ad (0.1 mg/kg), atropine (10 mg/kg), methylatropine (0.5 mg/kg), Ad (0.1 mg/kg) plus atropine (10 mg/kg) or vehicle (NaCl, 0.9%). Shock responsiveness and freezing behaviour were accessed. Hippocampal M1, M2, and M4 mRNA expression were evaluated. Ad-deficient mice presented decreased hippocampal muscarinic M4 subtype receptor mRNA expression compared to WT mice. In Ad-administered Ad-deficient mice, hippocampal muscarinic M4 subtype receptor mRNA expression increased compared with vehicle-administered Ad-deficient mice. On the context day, atropine-administered WT mice presented decreased freezing behaviour compared to vehicle or methylatropine-administered WT mice. Moreover, Ad plus atropine-administered Ad-deficient mice led to decreased freezing behaviour compared to Ad-administered Ad-deficient mice. In conclusion, Ad-deficient mice's contextual fear memory impairment was associated with hippocampal muscarinic M4 subtype receptor down expression, which was reversed by Ad. This may be related to contextual fear memory consolidation or retrieval induced by peripheral Ad. Furthermore, the effect of Ad contextual fear memory might be due to increased hippocampus muscarinic subtype M4 expression, which may contribute to increased cholinergic activity in the central nervous system.
Collapse
Affiliation(s)
- Ana Oliveira
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Márcia Azevedo
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Rafaela Seixas
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Soraia Silva
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Raquel Martinho
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Paula Serrão
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Biomedicine, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal
| | - Elisabete Silva
- Ageing and Stress Group, i3S- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; Faculty of Veterinary Medicine, Lusófona University, Lisbon, Portugal
| | - Mónica Moreira-Rodrigues
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal.
| |
Collapse
|
2
|
Murck H, Karailiev P, Karailievova L, Puhova A, Jezova D. Treatment with Glycyrrhiza glabra Extract Induces Anxiolytic Effects Associated with Reduced Salt Preference and Changes in Barrier Protein Gene Expression. Nutrients 2024; 16:515. [PMID: 38398838 PMCID: PMC10893552 DOI: 10.3390/nu16040515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
We have previously identified that low responsiveness to antidepressive therapy is associated with higher aldosterone/cortisol ratio, lower systolic blood pressure, and higher salt preference. Glycyrrhiza glabra (GG) contains glycyrrhizin, an inhibitor of 11β-hydroxysteroid-dehydrogenase type-2 and antagonist of toll-like receptor 4. The primary hypothesis of this study is that food enrichment with GG extract results in decreased anxiety behavior and reduced salt preference under stress and non-stress conditions. The secondary hypothesis is that the mentioned changes are associated with altered gene expression of barrier proteins in the prefrontal cortex. Male Sprague-Dawley rats were exposed to chronic mild stress for five weeks. Both stressed and unstressed rats were fed a diet with or without an extract of GG roots for the last two weeks. GG induced anxiolytic effects in animals independent of stress exposure, as measured in elevated plus maze test. Salt preference and intake were significantly reduced by GG under control, but not stress conditions. The gene expression of the barrier protein claudin-11 in the prefrontal cortex was increased in control rats exposed to GG, whereas stress-induced rise was prevented. Exposure to GG-enriched diet resulted in reduced ZO-1 expression irrespective of stress conditions. In conclusion, the observed effects of GG are in line with a reduction in the activity of central mineralocorticoid receptors. The treatment with GG extract or its active components may, therefore, be a useful adjunct therapy for patients with subtypes of depression and anxiety disorders with heightened renin-angiotensin-aldosterone system and/or inflammatory activity.
Collapse
Affiliation(s)
- Harald Murck
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, 35039 Marburg, Germany
| | - Peter Karailiev
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Lucia Karailievova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Agnesa Puhova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| |
Collapse
|
3
|
Feng L, Sharma A, Wang Z, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Li C, Zhang Z, Lin C, Huang H, Manzhulo I, Wiklund L, Sharma HS. Nanowired delivery of dl-3-n-butylphthalide with antibodies to alpha synuclein potentiated neuroprotection in Parkinson's disease with emotional stress. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 171:47-82. [PMID: 37783563 DOI: 10.1016/bs.irn.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Stress is one of the most serious consequences of life leading to several chronic diseases and neurodegeneration. Recent studies show that emotional stress and other kinds of anxiety and depression adversely affects Parkinson's disease symptoms. However, the details of how stress affects Parkinson's disease is still not well known. Traumatic brain injury, stroke, diabetes, post-traumatic stress disorders are well known to modify the disease precipitation, progression and persistence. However, show stress could influence Parkinson's disease is still not well known. The present investigation we examine the role of immobilization stress influencing Parkinson's disease brain pathology in model experiments. In ore previous report we found that mild traumatic brain injury exacerbate Parkinson's disease brain pathology and nanodelivery of dl-3-n-butylphthalide either alone or together with mesenchymal stem cells significantly attenuated Parkinson's disease brain pathology. In this chapter we discuss the role of stress in exacerbating Parkinson's disease pathology and nanowired delivery of dl-3-n-butylphthalide together with monoclonal antibodies to alpha synuclein (ASNC) is able to induce significant neuroprotection. The possible mechanisms of dl-3-n-butylphthalide and ASNC induced neuroprotection and suitable clinical therapeutic strategy is discussed.
Collapse
Affiliation(s)
- Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, P.R. China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Zhenguo Wang
- Shijiazhuang Pharma Group NBP Pharmaceutical Co. Ltd., Economic and Technological Development Zone, Shijiazhuang City, Hebei Province, P.R. China
| | - Dafin F Muresanu
- Dept. Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Dept. Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Department of Anesthesiology, Boston University, Albany str, Boston, MA, United States
| | - Cong Li
- Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, Guangdong, P.R. China
| | - Ziquiang Zhang
- Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, Guangdong, P.R. China
| | - Chen Lin
- Department of Neurosurgery, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing, P.R. China
| | - Hongyun Huang
- Beijing Hongtianji Neuroscience Academy, Beijing, P.R. China
| | - Igor Manzhulo
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
4
|
Medina-Rodriguez EM, Beurel E. Blood brain barrier and inflammation in depression. Neurobiol Dis 2022; 175:105926. [PMID: 36375722 PMCID: PMC10035601 DOI: 10.1016/j.nbd.2022.105926] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 10/26/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022] Open
Abstract
The blood brain barrier (BBB) is a vital structure to protect the brain, tightly filtering the passage of nutrients and molecules from the blood to the brain. This is critical for maintaining the proper functioning of the brain, and any disruption in the BBB has detrimental consequences often leading to diseases. It is not clear whether disruption of the BBB occurs first in depression or is the consequence of the disease, however disruption of the BBB has been observed in depressed patients and evidence points to the role of important culprits in depression, stress and inflammation in disrupting the integrity of the BBB. The mechanisms whereby stress, and inflammation affect the BBB remain to be fully understood. Yet, the role of cytokines in regulating tight junction protein expression seems crucial. Altogether, the findings in depression suggest that acting at the BBB level might provide therapeutic benefit in depression.
Collapse
Affiliation(s)
- Eva M Medina-Rodriguez
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, FL 33136, United States of America
| | - Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, FL 33136, United States of America; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States of America.
| |
Collapse
|
5
|
The Blood-Brain Barrier: Much More Than a Selective Access to the Brain. Neurotox Res 2021; 39:2154-2174. [PMID: 34677787 DOI: 10.1007/s12640-021-00431-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/30/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022]
Abstract
The blood-brain barrier is a dynamic structure, collectively referred to as the neurovascular unit. It is responsible for the exchange of blood, oxygen, ions, and other molecules between the peripheral circulation and the brain compartment. It is the main entrance to the central nervous system and as such critical for the maintenance of its homeostasis. Dysfunction of the blood-brain barrier is a characteristic of several neurovascular pathologies. Moreover, physiological changes, environmental factors, nutritional habits, and psychological stress can modulate the tightness of the barrier. In this contribution, we summarize our current understanding of structure and function of this important component of the brain. We also describe the neurological deficits associated with its damage. A special emphasis is placed in the effect of the exposure to xenobiotics and pollutants in the permeability of the barrier. Finally, current protective strategies as well as the culture models to study this fascinating structure are discussed.
Collapse
|
6
|
Taghadosi Z, Zarifkar A, Razban V, Owjfard M, Aligholi H. Effect of chronically electric foot shock stress on spatial memory and hippocampal blood brain barrier permeability. Behav Brain Res 2021; 410:113364. [PMID: 33992668 DOI: 10.1016/j.bbr.2021.113364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
Maintaining blood-brain barrier (BBB) contributes critically to preserving normal brain functions. According to the available evidence, intense or chronic exposure to stress would potentially affect different brain structures, such as the hippocampus, negatively. The purpose of this study was to define the relationship between the BBB permeability of the hippocampus and the performance of spatial learning and memory under chronically electric foot shock stress. Sixteen rats were divided into the control and stress groups equally. Animals in the stress group were exposed to foot shock (1 mA, 1 Hz) for 10-s duration every 60 s (1 h/day) for 10 consecutive days. The anxiety-related behavior, spatial learning, and memory were assessed by an Open Field (OF) and the Morris Water Maze (MWM) respectively. The hippocampal BBB permeability was determined by Evans blue penetration assay. Our results demonstrated that the stress model not only increased locomotor activities in the OF test but reduced spatial learning and memory in MWM. Moreover, these effects coincided with a significant increase in hippocampal BBB permeability. In sum, the stress model can be used in future studies focusing on the relationship between stress and BBB permeability of the hippocampus.
Collapse
Affiliation(s)
- Zohreh Taghadosi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asadollah Zarifkar
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Neuroscience Research Center and Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Science and Technology, Shiraz University of Medical Science, Shiraz, Iran
| | - Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Aligholi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Salmina AB, Komleva YK, Malinovskaya NA, Morgun AV, Teplyashina EA, Lopatina OL, Gorina YV, Kharitonova EV, Khilazheva ED, Shuvaev AN. Blood-Brain Barrier Breakdown in Stress and Neurodegeneration: Biochemical Mechanisms and New Models for Translational Research. BIOCHEMISTRY (MOSCOW) 2021; 86:746-760. [PMID: 34225598 DOI: 10.1134/s0006297921060122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Blood-brain barrier (BBB) is a structural and functional element of the neurovascular unit (NVU), which includes cells of neuronal, glial, and endothelial nature. The main functions of NVU include maintenance of the control of metabolism and chemical homeostasis in the brain tissue, ensuring adequate blood flow in active regions, regulation of neuroplasticity processes, which is realized through intercellular interactions under normal conditions, under stress, in neurodegeneration, neuroinfection, and neurodevelopmental diseases. Current versions of the BBB and NVU models, static and dynamic, have significantly expanded research capabilities, but a number of issues remain unresolved, in particular, personification of the models for a patient. In addition, application of both static and dynamic models has an important problem associated with the difficulty in reproducing pathophysiological mechanisms responsible for the damage of the structural and functional integrity of the barrier in the diseases of the central nervous system. More knowledge on the cellular and molecular mechanisms of BBB and NVU damage in pathology is required to solve this problem. This review discusses current state of the cellular and molecular mechanisms that control BBB permeability, pathobiochemical mechanisms and manifestations of BBB breakdown in stress and neurodegenerative diseases, as well as the problems and prospects of creating in vitro BBB and NVU models for translational studies in neurology and neuropharmacology. Deciphering BBB (patho)physiology will open up new opportunities for further development in the related areas of medicine such as regenerative medicine, neuropharmacology, and neurorehabilitation.
Collapse
Affiliation(s)
- Alla B Salmina
- Division of Brain Sciences, Research Center of Neurology, Moscow, 125367, Russia. .,Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| | - Yuliya K Komleva
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| | - Nataliya A Malinovskaya
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| | - Andrey V Morgun
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| | - Elena A Teplyashina
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| | - Olga L Lopatina
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| | - Yana V Gorina
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| | - Ekaterina V Kharitonova
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| | - Elena D Khilazheva
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| | - Anton N Shuvaev
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| |
Collapse
|
8
|
Sun H, Hu H, Liu C, Sun N, Duan C. Methods used for the measurement of blood-brain barrier integrity. Metab Brain Dis 2021; 36:723-735. [PMID: 33635479 DOI: 10.1007/s11011-021-00694-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/11/2021] [Indexed: 01/12/2023]
Abstract
The blood-brain barrier (BBB) comprises the interface between blood, brain and cerebrospinal fluid. Its primary function, which is mainly carried out by tight junctions, is to stabilize the tightly controlled microenvironment of the brain. To study the development and maintenance of the BBB, as well as various roles their intrinsic mechanisms that play in neurological disorders, suitable measurements are required to demonstrate integrity and functional changes at the interfaces between the blood and brain tissue. Markers and plasma proteins with different molecular weight (MW) are used to measure the permeability of BBB. In addition, the expression changes of tight-junction proteins form the basic structure of BBB, and imaging modalities are available to study the disruption of BBB. In the present review, above mentioned methods are depicted in details, together with the pros and cons as well as the differences between these methods, which maybe benefit research studies focused on the detection of BBB breakdown.
Collapse
Affiliation(s)
- Huixin Sun
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Huiling Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Chuanjie Liu
- Weihai City Key Laboratory of Autoimmunity, Weihai Central Hospital, Weihai, 264400, Shandong Province, China
| | - Nannan Sun
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Chaohui Duan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
9
|
Karailiev P, Hlavacova N, Chmelova M, Homer NZM, Jezova D. Tight junction proteins in the small intestine and prefrontal cortex of female rats exposed to stress of chronic isolation starting early in life. Neurogastroenterol Motil 2021; 33:e14084. [PMID: 33497497 DOI: 10.1111/nmo.14084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/30/2020] [Accepted: 01/04/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Simultaneous evaluation of barrier protein expression in the gut and the brain and their modulation under stress conditions have not been studied before now. As the permeability and function of the gut and blood-brain barrier are different and both express the MRs, we hypothesized that stress of post-weaning social isolation induces changes in tight junction protein expression in the gut which are (1) independent of changes in the brain and (2) are mediated via the mineralocorticoid receptor (MR). METHODS First, using UPLC-MS/MS we have successfully validated and selected a dose (1.2 mg/rat/day) of the MR antagonist spironolactone to treat female rats exposed to stress of chronic isolation or control conditions from postnatal day 21 for 9 weeks. KEY RESULTS Isolation stress caused an enhancement of gene expression of occludin and ZO-1 and a decrease in claudin-5 and MR expression in both the small intestine and prefrontal cortex. Isolation stress failed to decrease claudin-5 (small intestine) and MR (prefrontal cortex) gene expression in spironolactone-treated rats. MR blockade resulted in a decrease in claudin-15 expression in the small intestine. Anxiogenic effect of chronic stress, measured in elevated plus-maze test, was partly prevented by spironolactone treatment. CONCLUSIONS & INFERENCES Claudins, the main regulators of intestinal barrier permeability responded to chronic stress of social isolation and/or simultaneous blockade of MR in female rats by alterations independent of changes in the brain cortex. The results suggest a physiological role of MR in the control of claudin expression in the small intestine, but not in the brain cortex.
Collapse
Affiliation(s)
- Peter Karailiev
- Institute of Experimental Endocrinology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Natasa Hlavacova
- Institute of Experimental Endocrinology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Magdalena Chmelova
- Institute of Experimental Endocrinology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh CRF, Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
10
|
Bargi R, Hosseini M, Asgharzadeh F, Khazaei M, Shafei MN, Beheshti F. Protection Against Blood-Brain Barrier Permeability as a Possible Mechanism for Protective Effects of Thymoquinone Against Sickness Behaviors Induced by Lipopolysaccharide in Rats. Jundishapur J Nat Pharm Prod 2021; 16. [DOI: 10.5812/jjnpp.67765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
Background: Blood-brain barrier (BBB), as well-known protection for the brain, plays an active role in normal homeostasis. It might be changed by a range of inflammatory mediators to have a role in sickness behaviors. Objectives: Regarding the anti-inflammatory effects of thymoquinone (TQ), its protection against BBB permeability, as a possible mechanism for protective effects against sickness behaviors elicited by lipopolysaccharide (LPS), was evaluated in rats. Methods: The animals were grouped as follows and treated (n = 10 in each): (1) control (saline); (2) LPS 1 mg/kg, was injected two hours before behavioral tests for two weeks; (3-5) 2, 5, and 10 mg/kg TQ, respectively was injected 30 min before LPS injection. Open-field (OF), elevated plus-maze (EPM) and Forced Swimming test (FST) were done. Finally, the animals were anesthetized to evaluate for BBB permeability using Evans blue (EB) dye method. Results: Compared with control, LPS decreased the peripheral distance and crossing and also total crossing and distance in OF, (P < 0.01 - P < 0.001). The central crossing and distance and central time in all three treatment groups were more than LPS (P < 0.05 - P < 0.001). LPS also reduced the entries and the time spent in the open arm while increased the time spent in the closed arm in EPM (P < 0.05 - P < 0.001). The effects of LPS were reversed by TQ (P < 0.05 - P < 0.001). In FST, the immobility time and active time were increased and decreased by LPS compared with control (P < 0.001), respectively. In all three TQ-treated groups, the active and climbing times were more while the immobility time was fewer than the LPS (P < 0.05 - P < 0.001). The animals of the LPS group showed more EB dye content in their brain tissue than the control group (P < 0.05 - P < 0.001). TQ significantly reduced EB dye content of the brain tissues (P < 0.05 - P < 0.001). Conclusions: According to this study, protection against BBB permeability as a possible mechanism for the protective effects of TQ against sickness behaviors induced by LPS might be suggested.
Collapse
|
11
|
Segarra M, Aburto MR, Acker-Palmer A. Blood-Brain Barrier Dynamics to Maintain Brain Homeostasis. Trends Neurosci 2021; 44:393-405. [PMID: 33423792 DOI: 10.1016/j.tins.2020.12.002] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 10/03/2020] [Accepted: 12/07/2020] [Indexed: 01/18/2023]
Abstract
The blood-brain barrier (BBB) is a dynamic platform for exchange of substances between the blood and the brain parenchyma, and it is an essential functional gatekeeper for the central nervous system (CNS). While it is widely recognized that BBB disruption is a hallmark of several neurovascular pathologies, an aspect of the BBB that has received somewhat less attention is the dynamic modulation of BBB tightness to maintain brain homeostasis in response to extrinsic environmental factors and physiological changes. In this review, we summarize how BBB integrity adjusts in critical stages along the life span, as well as how BBB permeability can be altered by common stressors derived from nutritional habits, environmental factors and psychological stress.
Collapse
Affiliation(s)
- Marta Segarra
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Max-von-Laue-Strasse 15, D-60438, Frankfurt am Main, Germany; Cardio-Pulmonary Institute (CPI), Max-von-Laue-Strasse 15, D-60438, Frankfurt am Main, Germany.
| | - Maria R Aburto
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Max-von-Laue-Strasse 15, D-60438, Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Neuro and Vascular Guidance, Buchmann Institute for Molecular Life Sciences (BMLS) and Institute of Cell Biology and Neuroscience, Max-von-Laue-Strasse 15, D-60438, Frankfurt am Main, Germany; Cardio-Pulmonary Institute (CPI), Max-von-Laue-Strasse 15, D-60438, Frankfurt am Main, Germany; Max Planck Institute for Brain Research, Max-von-Laue-Strasse 4, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
12
|
Cellular mechanisms and molecular signaling pathways in stress-induced anxiety, depression, and blood-brain barrier inflammation and leakage. Inflammopharmacology 2020; 28:643-665. [PMID: 32333258 DOI: 10.1007/s10787-020-00712-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/15/2020] [Indexed: 12/17/2022]
Abstract
Depression and anxiety are comorbid conditions in many neurological or psychopathological disorders. Stress is an underlying event that triggers development of anxiety and depressive-like behaviors. Recent experimental data indicate that anxiety and depressive-like behaviors occurring as a result of stressful situations can cause blood-brain barrier (BBB) dysfunction, which is characterized by inflammation and leakage. However, the underlying mechanisms are not completely understood. This paper sought to review recent experimental preclinical and clinical data that suggest possible molecular mechanisms involved in development of stress-induced anxiety and depression with associated BBB inflammation and leakage. Critical therapeutic targets and potential pharmacological candidates for treatment of stress-induced anxiety and depression with associated BBB dysfunctions are also discussed.
Collapse
|
13
|
Samuels A. Dose dependent toxicity of glutamic acid: a review. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2020. [DOI: 10.1080/10942912.2020.1733016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Adrienne Samuels
- Office of the administrator, Truth in Labeling Campaign, Chicago, IL, USA
| |
Collapse
|
14
|
Abstract
The blood-brain barrier (BBB) protects the vertebrate central nervous system from harmful blood-borne, endogenous and exogenous substances to ensure proper neuronal function. The BBB describes a function that is established by endothelial cells of CNS vessels in conjunction with pericytes, astrocytes, neurons and microglia, together forming the neurovascular unit (NVU). Endothelial barrier function is crucially induced and maintained by the Wnt/β-catenin pathway and requires intact NVU for proper functionality. The BBB and the NVU are characterized by a specialized assortment of molecular specializations, providing the basis for tightening, transport and immune response functionality.The present chapter introduces state-of-the-art knowledge of BBB structure and function and highlights current research topics, aiming to understanding in more depth the cellular and molecular interactions at the NVU, determining functionality of the BBB in health and disease, and providing novel potential targets for therapeutic BBB modulation. Moreover, we highlight recent advances in understanding BBB and NVU heterogeneity within the CNS as well as their contribution to CNS physiology, such as neurovascular coupling, and pathophysiology, is discussed. Finally, we give an outlook onto new avenues of BBB research.
Collapse
Affiliation(s)
- Fabienne Benz
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.
- Excellence Cluster Cardio Pulmonary System (CPI), Partner Site Frankfurt, Frankfurt, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt/Mainz, Frankfurt, Germany.
| |
Collapse
|
15
|
Moreno-Rius J. The cerebellum under stress. Front Neuroendocrinol 2019; 54:100774. [PMID: 31348932 DOI: 10.1016/j.yfrne.2019.100774] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 07/19/2019] [Accepted: 07/20/2019] [Indexed: 12/22/2022]
Abstract
Stress-related psychiatric conditions are one of the main causes of disability in developed countries. They account for a large portion of resource investment in stress-related disorders, become chronic, and remain difficult to treat. Research on the neurobehavioral effects of stress reveals how changes in certain brain areas, mediated by a number of neurochemical messengers, markedly alter behavior. The cerebellum is connected with stress-related brain areas and expresses the machinery required to process stress-related neurochemical mediators. Surprisingly, it is not regarded as a substrate of stress-related behavioral alterations, despite numerous studies that show cerebellar responsivity to stress. Therefore, this review compiles those studies and proposes a hypothesis for cerebellar function in stressful conditions, relating it to stress-induced psychopathologies. It aims to provide a clearer picture of stress-related neural circuitry and stimulate cerebellum-stress research. Consequently, it might contribute to the development of improved treatment strategies for stress-related disorders.
Collapse
|
16
|
Costa-Ferreira W, Morais-Silva G, Gomes-de-Souza L, Marin MT, Crestani CC. The AT1 Receptor Antagonist Losartan Does Not Affect Depressive-Like State and Memory Impairment Evoked by Chronic Stressors in Rats. Front Pharmacol 2019; 10:705. [PMID: 31293424 PMCID: PMC6598205 DOI: 10.3389/fphar.2019.00705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022] Open
Abstract
The present study investigated the effect of the treatment with the angiotensin II type 1 receptor (AT1) antagonist losartan in the depressive-like state and memory impairment evoked by exposure to either homotypic (i.e., repeated exposure to the same type of stressor) or heterotypic (i.e., exposure to different aversive stimuli) chronic stressors in rats. For this, male Wistar rats were subjected to a 10 days regimen of repeated restraint stress (RRS, homotypic stressor) or chronic variable stress (CVS, heterotypic stressor) while being concurrently treated daily with losartan (30 mg/kg/day, p.o.). Depressive-like state was evaluated by analysis of the alterations considered as markers of depression (decreased sucrose preference and body weight and coat state deterioration), whereas cognitive non-emotional performance was tested using the novel object recognition (NOR) test. Locomotor activity was also evaluated in the open field test. Both RRS and CVS impaired sucrose preference and caused coat state deterioration, whereas only CVS impaired body weight gain. Besides, RRS impaired short-term memory (but not long-term memory) in the NOR test. Neither depressive-like state nor memory impairment evoked by the chronic stressors was affected by the treatment with losartan. Nevertheless, CVS increased the locomotion, which was inhibited by losartan. Taken together, these results provide evidence that the chronic treatment with losartan does not affect the depressive-like state and memory impairment evoked by either homotypic or heterotypic chronic stress regimens in rats.
Collapse
Affiliation(s)
- Willian Costa-Ferreira
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| | - Gessynger Morais-Silva
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| | - Lucas Gomes-de-Souza
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| | - Marcelo T Marin
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| | - Carlos C Crestani
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| |
Collapse
|
17
|
Oliveira A, Martinho R, Serrão P, Moreira-Rodrigues M. Epinephrine Released During Traumatic Events May Strengthen Contextual Fear Memory Through Increased Hippocampus mRNA Expression of Nr4a Transcription Factors. Front Mol Neurosci 2018; 11:334. [PMID: 30319349 PMCID: PMC6167477 DOI: 10.3389/fnmol.2018.00334] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/27/2018] [Indexed: 11/13/2022] Open
Abstract
Epinephrine (EPI) strengthens contextual fear memories by acting on peripheral β2-adrenoceptors. Phenylethanolamine-N-methyltransferase-knockout (Pnmt-KO) mice are EPI-deficient mice and have reduced contextual fear learning. Our aim was to evaluate the molecular mechanisms by which peripheral EPI strengthens contextual fear memory and if a β2-adrenoceptor antagonist can erase contextual fear memories. Pnmt-KO and wild-type (WT) mice were submitted to fear conditioning (FC) procedure after treatment with EPI, norepinephrine (NE), EPI plus ICI 118,551 (selective β2-adrenoceptor antagonist), ICI 118,551 or vehicle (NaCl 0.9%). Catecholamines were separated and quantified by high performance liquid chromatography-electrochemical detection (HPLC-ED). Blood glucose was measured by coulometry. Real-time polymerase chain reaction (qPCR) was used to evaluate mRNA expression of nuclear receptor 4a1 (Nr4a1), Nr4a2 and Nr4a3 in hippocampus samples. In WT mice, plasma EPI concentration was significantly higher after fear acquisition (FA) compared with mice without the test. NE did not increase in plasma after FA and did not strengthen contextual fear memory, contrary to EPI. Freezing induced by EPI was blocked by ICI 118,551 in Pnmt-KO mice. In WT mice, ICI 118,551 blocked blood glucose release into the bloodstream after FA and decreased contextual fear memory. Nr4a1, Nr4a2 and Nr4a3 mRNA expression decreased in Pnmt-KO mice compared with WT mice after FC procedure. In Pnmt-KO mice, EPI induced an increase in mRNA expression of Nr4a2 compared to vehicle. In conclusion, EPI increases in plasma after an aversive experience, possibly improving long-term and old memories, by acting on peripheral β2-adrenoceptors. Glucose could be the mediator of peripheral EPI in the central nervous system, inducing the expression of Nr4a transcription factor genes involved in consolidation of contextual fear memories.
Collapse
Affiliation(s)
- Ana Oliveira
- Laboratory of General Physiology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal.,Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal
| | - Raquel Martinho
- Laboratory of General Physiology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal.,Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal
| | - Paula Serrão
- Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal.,Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal
| | - Mónica Moreira-Rodrigues
- Laboratory of General Physiology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal.,Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal
| |
Collapse
|
18
|
Abstract
Patients with cancer are more likely to develop depression than the general population, which negatively impacts their quality of life and prognosis. In order to identify effective antidepressants catered toward cancer patients, the biology of depression in the context of cancer must be well-understood. Many theories have emerged postulating the mechanisms underlying the development of depressive disorder. Here, we review the role inflammation, a hyperactive hypothalamic-pituitary-adrenal (HPA) axis, and glutamate excitotoxicity may play in cancer-induced depression. Hopefully, novel therapeutics targeting these dysregulated pathways may be potent in ameliorating depressive symptoms in the cancer population.
Collapse
Affiliation(s)
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
19
|
Torres-Oliva M, Schneider J, Wiegleb G, Kaufholz F, Posnien N. Dynamic genome wide expression profiling of Drosophila head development reveals a novel role of Hunchback in retinal glia cell development and blood-brain barrier integrity. PLoS Genet 2018; 14:e1007180. [PMID: 29360820 PMCID: PMC5796731 DOI: 10.1371/journal.pgen.1007180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 02/02/2018] [Accepted: 01/01/2018] [Indexed: 01/01/2023] Open
Abstract
Drosophila melanogaster head development represents a valuable process to study the developmental control of various organs, such as the antennae, the dorsal ocelli and the compound eyes from a common precursor, the eye-antennal imaginal disc. While the gene regulatory network underlying compound eye development has been extensively studied, the key transcription factors regulating the formation of other head structures from the same imaginal disc are largely unknown. We obtained the developmental transcriptome of the eye-antennal discs covering late patterning processes at the late 2nd larval instar stage to the onset and progression of differentiation at the end of larval development. We revealed the expression profiles of all genes expressed during eye-antennal disc development and we determined temporally co-expressed genes by hierarchical clustering. Since co-expressed genes may be regulated by common transcriptional regulators, we combined our transcriptome dataset with publicly available ChIP-seq data to identify central transcription factors that co-regulate genes during head development. Besides the identification of already known and well-described transcription factors, we show that the transcription factor Hunchback (Hb) regulates a significant number of genes that are expressed during late differentiation stages. We confirm that hb is expressed in two polyploid subperineurial glia cells (carpet cells) and a thorough functional analysis shows that loss of Hb function results in a loss of carpet cells in the eye-antennal disc. Additionally, we provide for the first time functional data indicating that carpet cells are an integral part of the blood-brain barrier. Eventually, we combined our expression data with a de novo Hb motif search to reveal stage specific putative target genes of which we find a significant number indeed expressed in carpet cells. The development of different cell types must be tightly coordinated, and the eye-antennal imaginal discs of Drosophila melanogaster represent an excellent model to study the molecular mechanisms underlying this coordination. These imaginal discs contain the anlagen of nearly all adult head structures, such as the antennae, the head cuticle, the ocelli and the compound eyes. While large scale screens have been performed to unravel the gene regulatory network underlying compound eye development, a comprehensive understanding of genome wide expression dynamics throughout head development is still missing to date. We studied the genome wide gene expression dynamics during eye-antennal disc development in D. melanogaster to identify new central regulators of the underlying gene regulatory network. Expression based gene clustering and transcription factor motif enrichment analyses revealed a central regulatory role of the transcription factor Hunchback (Hb). We confirmed that hb is expressed in two polyploid retinal subperineurial glia cells (carpet cells). Our functional analysis shows that Hb is necessary for carpet cell development and we show for the first time that the carpet cells are an integral part of the blood-brain barrier.
Collapse
Affiliation(s)
- Montserrat Torres-Oliva
- Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, Abteilung für Entwicklungsbiologie, GZMB Ernst-Caspari-Haus, Göttingen, Germany
| | - Julia Schneider
- Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, Abteilung für Entwicklungsbiologie, GZMB Ernst-Caspari-Haus, Göttingen, Germany
| | - Gordon Wiegleb
- Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, Abteilung für Entwicklungsbiologie, GZMB Ernst-Caspari-Haus, Göttingen, Germany
| | - Felix Kaufholz
- Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, Abteilung für Entwicklungsbiologie, GZMB Ernst-Caspari-Haus, Göttingen, Germany
| | - Nico Posnien
- Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, Abteilung für Entwicklungsbiologie, GZMB Ernst-Caspari-Haus, Göttingen, Germany
- * E-mail:
| |
Collapse
|
20
|
Li Y, Cheng KC, Liu KF, Peng WH, Cheng JT, Niu HS. Telmisartan Activates PPARδ to Improve Symptoms of Unpredictable Chronic Mild Stress-Induced Depression in Mice. Sci Rep 2017; 7:14021. [PMID: 29070884 PMCID: PMC5656622 DOI: 10.1038/s41598-017-14265-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 10/09/2017] [Indexed: 11/09/2022] Open
Abstract
Major depression is a common mental disorder that has been established to be associated with a decrease in serotonin and/or serotonin transporters in the brain. Peroxisome proliferator-activated receptor δ (PPARδ) has been introduced as a potential target for depression treatment. Telmisartan was recently shown to activate PPARδ expression; therefore, the effectiveness of telmisartan in treating depression was investigated. In unpredictable chronic mild stress (UCMS) model, treatment with telmisartan for five weeks notably decrease in the time spent in the central and the reduced frequency of grooming and rearing in open filed test (OFT) and the decreased sucrose consumption in sucrose preference test (SPT) compared with the paradigms. Telmisartan also reversed the decrease in PPARδ and 5-HTT levels in the hippocampus of depression-like mice. Administration of PPARδ antagonist GSK0660 and direct infusion of sh-PPARδ into the brain blocked the effects of telmisartan on the improvement of depression-like behavior in these mice. Moreover, telmisartan enhanced the expression of PPARδ and 5HTT in H19-7 cells. In conclusion, the obtained results suggest that telmisartan improves symptoms of stress-induced depression in animals under chronic stress through activation of PPARδ. Therefore, telmisartan may be developed as a potential anti-depressant in the future.
Collapse
Affiliation(s)
- Yingxiao Li
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890, Japan
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City, 71003, Taiwan
| | - Kai-Chun Cheng
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890, Japan
| | - Keng-Fan Liu
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Pharmacy, China Medical University, Taichung City, 40401, Taiwan
| | - Wen-Huang Peng
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Pharmacy, China Medical University, Taichung City, 40401, Taiwan
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan City, 71003, Taiwan.
- Institute of Medical Sciences, Chang Jung Christian University, Gueiren, Tainan City, 71101, Taiwan.
| | - Ho-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien City, 97005, Taiwan.
| |
Collapse
|
21
|
Costa-Ferreira W, Vieira JO, Almeida J, Gomes-de-Souza L, Crestani CC. Involvement of Type 1 Angiontensin II Receptor (AT1) in Cardiovascular Changes Induced by Chronic Emotional Stress: Comparison between Homotypic and Heterotypic Stressors. Front Pharmacol 2016; 7:262. [PMID: 27588004 PMCID: PMC4988975 DOI: 10.3389/fphar.2016.00262] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/04/2016] [Indexed: 01/26/2023] Open
Abstract
Consistent evidence has shown an important role of emotional stress in pathogenesis of cardiovascular diseases. Additionally, studies in animal models have demonstrated that daily exposure to different stressor (heterotypic stressor) evokes more severe changes than those resulting from repeated exposure to the same aversive stimulus (homotypic stressor), possibly due to the habituation process upon repeated exposure to the same stressor. Despite these pieces of evidence, the mechanisms involved in the stress-evoked cardiovascular dysfunction are poorly understood. Therefore, the present study investigated the involvement of angiotensin II (Ang II) acting on the type 1 Ang II receptor (AT1) in the cardiovascular dysfunctions evoked by both homotypic and heterotypic chronic emotional stresses in rats. For this purpose, we compared the effect of the chronic treatment with the AT1 receptor antagonist losartan (30 mg/kg/day, p.o.) on the cardiovascular and autonomic changes evoked by the heterotypic stressor chronic variable stress (CVS) and the homotypic stressor repeated restraint stress (RRS). RRS increased the sympathetic tone to the heart and decreased the cardiac parasympathetic activity, whereas CVS decreased the cardiac parasympathetic activity. Additionally, both stressors impaired the baroreflex function. Alterations in the autonomic activity and the baroreflex impairment were inhibited by losartan treatment. Additionally, CVS reduced the body weight and increased the circulating corticosterone; however, these effects were not affected by losartan. In conclusion, these findings indicate the involvement of angiotensin II/AT1 receptors in the autonomic changes evoked by both homotypic and heterotypic chronic stressors. Moreover, the present results provide evidence that the increase in the circulating corticosterone and body weight reduction evoked by heterotypic stressors are independent of AT1 receptors.
Collapse
Affiliation(s)
- Willian Costa-Ferreira
- Faculdade de Ciências Farmacêuticas, UNESP-Universidade Estadual PaulistaAraraquara, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, UFSCar-UNESPSão Carlos, Brazil
| | - Jonas O Vieira
- Faculdade de Ciências Farmacêuticas, UNESP-Universidade Estadual PaulistaAraraquara, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, UFSCar-UNESPSão Carlos, Brazil
| | - Jeferson Almeida
- Faculdade de Ciências Farmacêuticas, UNESP-Universidade Estadual PaulistaAraraquara, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, UFSCar-UNESPSão Carlos, Brazil
| | - Lucas Gomes-de-Souza
- Faculdade de Ciências Farmacêuticas, UNESP-Universidade Estadual PaulistaAraraquara, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, UFSCar-UNESPSão Carlos, Brazil
| | - Carlos C Crestani
- Faculdade de Ciências Farmacêuticas, UNESP-Universidade Estadual PaulistaAraraquara, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, UFSCar-UNESPSão Carlos, Brazil
| |
Collapse
|
22
|
Roszkowski M, Bohacek J. Stress does not increase blood-brain barrier permeability in mice. J Cereb Blood Flow Metab 2016; 36:1304-15. [PMID: 27146513 PMCID: PMC4929709 DOI: 10.1177/0271678x16647739] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/08/2016] [Indexed: 12/28/2022]
Abstract
Several studies have reported that exposure to acute psychophysiological stressors can lead to an increase in blood-brain barrier permeability, but these findings remain controversial and disputed. We thoroughly examined this issue by assessing the effect of several well-established paradigms of acute stress and chronic stress on blood-brain barrier permeability in several brain areas of adult mice. Using cerebral extraction ratio for the small molecule tracer sodium fluorescein (NaF, 376 Da) as a sensitive measure of blood-brain barrier permeability, we find that neither acute swim nor restraint stress lead to increased cerebral extraction ratio. Daily 6-h restraint stress for 21 days, a model for the severe detrimental impact of chronic stress on brain function, also does not alter cerebral extraction ratio. In contrast, we find that cold forced swim and cold restraint stress both lead to a transient, pronounced decrease of cerebral extraction ratio in hippocampus and cortex, suggesting that body temperature can be an important confounding factor in studies of blood-brain barrier permeability. To additionally assess if stress could change blood-brain barrier permeability for macromolecules, we measured cerebral extraction ratio for fluorescein isothiocyanate-dextran (70 kDa). We find that neither acute restraint nor cold swim stress affected blood-brain barrier permeability for macromolecules, thus corroborating our findings that various stressors do not increase blood-brain barrier permeability.
Collapse
Affiliation(s)
- Martin Roszkowski
- Brain Research Institute, Neuroscience Center Zurich, ETH Zurich, University of Zurich, Winterthurerstrasse, Zurich, Switzerland
| | - Johannes Bohacek
- Brain Research Institute, Neuroscience Center Zurich, ETH Zurich, University of Zurich, Winterthurerstrasse, Zurich, Switzerland
| |
Collapse
|
23
|
Malan-Müller S, Fairbairn L, Daniels WMU, Dashti MJS, Oakeley EJ, Altorfer M, Kidd M, Seedat S, Gamieldien J, Hemmings SMJ. Molecular mechanisms of D-cycloserine in facilitating fear extinction: insights from RNAseq. Metab Brain Dis 2016; 31:135-56. [PMID: 26400817 DOI: 10.1007/s11011-015-9727-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/02/2015] [Indexed: 01/24/2023]
Abstract
D-cycloserine (DCS) has been shown to be effective in facilitating fear extinction in animal and human studies, however the precise mechanisms whereby the co-administration of DCS and behavioural fear extinction reduce fear are still unclear. This study investigated the molecular mechanisms of intrahippocampally administered D-cycloserine in facilitating fear extinction in a contextual fear conditioning animal model. Male Sprague Dawley rats (n = 120) were grouped into four experimental groups (n = 30) based on fear conditioning and intrahippocampal administration of either DCS or saline. The light/dark avoidance test was used to differentiate maladapted (MA) (anxious) from well-adapted (WA) (not anxious) subgroups. RNA extracted from the left dorsal hippocampus was used for RNA sequencing and gene expression data was compared between six fear-conditioned + saline MA (FEAR + SALINE MA) and six fear-conditioned + DCS WA (FEAR + DCS WA) animals. Of the 424 significantly downregulated and 25 significantly upregulated genes identified in the FEAR + DCS WA group compared to the FEAR + SALINE MA group, 121 downregulated and nine upregulated genes were predicted to be relevant to fear conditioning and anxiety and stress-related disorders. The majority of downregulated genes transcribed immune, proinflammatory and oxidative stress systems molecules. These molecules mediate neuroinflammation and cause neuronal damage. DCS also regulated genes involved in learning and memory processes, and genes associated with anxiety, stress-related disorders and co-occurring diseases (e.g., cardiovascular diseases, digestive system diseases and nervous system diseases). Identifying the molecular underpinnings of DCS-mediated fear extinction brings us closer to understanding the process of fear extinction.
Collapse
Affiliation(s)
- Stefanie Malan-Müller
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| | - Lorren Fairbairn
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Willie M U Daniels
- Department of Human Physiology, University of KwaZulu-Natal, Durban, South Africa
| | | | - Edward J Oakeley
- Novartis Institutes for BioMedical Research, Biomarker Development - Human Genetics and Genomics, Genome Technologies, Basel, Switzerland
| | - Marc Altorfer
- Novartis Institutes for BioMedical Research, Biomarker Development - Human Genetics and Genomics, Genome Technologies, Basel, Switzerland
| | - Martin Kidd
- Centre for Statistical Consultation, Stellenbosch University, Stellenbosch, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Junaid Gamieldien
- University of the Western Cape, South African National Bioinformatics Institute, Cape Town, South Africa
| | - Sîan Megan Joanna Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
24
|
Sántha P, Veszelka S, Hoyk Z, Mészáros M, Walter FR, Tóth AE, Kiss L, Kincses A, Oláh Z, Seprényi G, Rákhely G, Dér A, Pákáski M, Kálmán J, Kittel Á, Deli MA. Restraint Stress-Induced Morphological Changes at the Blood-Brain Barrier in Adult Rats. Front Mol Neurosci 2016; 8:88. [PMID: 26834555 PMCID: PMC4712270 DOI: 10.3389/fnmol.2015.00088] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/21/2015] [Indexed: 12/16/2022] Open
Abstract
Stress is well-known to contribute to the development of both neurological and psychiatric diseases. While the role of the blood-brain barrier is increasingly recognized in the development of neurodegenerative disorders, such as Alzheimer's disease, dysfunction of the blood-brain barrier has been linked to stress-related psychiatric diseases only recently. In the present study the effects of restraint stress with different duration (1, 3, and 21 days) were investigated on the morphology of the blood-brain barrier in male adult Wistar rats. Frontal cortex and hippocampus sections were immunostained for markers of brain endothelial cells (claudin-5, occluding, and glucose transporter-1) and astroglia (GFAP). Staining pattern and intensity were visualized by confocal microscopy and evaluated by several types of image analysis. The ultrastructure of brain capillaries was investigated by electron microscopy. Morphological changes and intensity alterations in brain endothelial tight junction proteins claudin-5 and occludin were induced by stress. Following restraint stress significant increases in the fluorescence intensity of glucose transporter-1 were detected in brain endothelial cells in the frontal cortex and hippocampus. Significant reductions in GFAP fluorescence intensity were observed in the frontal cortex in all stress groups. As observed by electron microscopy, 1-day acute stress induced morphological changes indicating damage in capillary endothelial cells in both brain regions. After 21 days of stress thicker and irregular capillary basal membranes in the hippocampus and edema in astrocytes in both regions were seen. These findings indicate that stress exerts time-dependent changes in the staining pattern of tight junction proteins occludin, claudin-5, and glucose transporter-1 at the level of brain capillaries and in the ultrastructure of brain endothelial cells and astroglial endfeet, which may contribute to neurodegenerative processes, cognitive and behavioral dysfunctions.
Collapse
Affiliation(s)
- Petra Sántha
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| | - Szilvia Veszelka
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| | - Zsófia Hoyk
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| | - Mária Mészáros
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| | - Fruzsina R Walter
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| | - Andrea E Tóth
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| | - Lóránd Kiss
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| | - András Kincses
- Biomolecular Electronics Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| | - Zita Oláh
- Department of Psychiatry, Alzheimer's Disease Research Centre, University of Szeged Szeged, Hungary
| | - György Seprényi
- Department of Medical Biology, University of Szeged Szeged, Hungary
| | - Gábor Rákhely
- Department of Biotechnology, University of Szeged Szeged, Hungary
| | - András Dér
- Biomolecular Electronics Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| | - Magdolna Pákáski
- Department of Psychiatry, Alzheimer's Disease Research Centre, University of Szeged Szeged, Hungary
| | - János Kálmán
- Department of Psychiatry, Alzheimer's Disease Research Centre, University of Szeged Szeged, Hungary
| | - Ágnes Kittel
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| | - Mária A Deli
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| |
Collapse
|
25
|
Shukla RK, Gupta R, Srivastava P, Dhuriya YK, Singh A, Chandravanshi LP, Kumar A, Siddiqui MH, Parmar D, Pant AB, Khanna VK. Brain cholinergic alterations in rats subjected to repeated immobilization or forced swim stress on lambda-cyhalothrin exposure. Neurochem Int 2015; 93:51-63. [PMID: 26746386 DOI: 10.1016/j.neuint.2015.12.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 12/03/2015] [Accepted: 12/28/2015] [Indexed: 02/03/2023]
Abstract
Role of immobilization stress (IMS), a psychological stressor and forced swim stress (FSS), a physical stressor was investigated on the neurobehavioral toxicity of lambda-cyhalothrin (LCT), a new generation type-II synthetic pyrethroid. Pre-exposure of rats to IMS (15 min/day) or FSS (3 min/day) for 28 days on LCT (3.0 mg/kg body weight, p.o.) treatment for 3 days resulted to decrease spatial learning and memory and muscle strength associated with cholinergic-muscarinic receptors in frontal cortex and hippocampus as compared to those exposed to IMS or FSS or LCT alone. Decrease in acetylcholinesterase activity, protein expression of ChAT and PKC-β1 associated with decreased mRNA expression of CHRM2, AChE and ChAT in frontal cortex and hippocampus was also evident in rats pre-exposed to IMS or FSS on LCT treatment, compared to rats exposed to IMS or FSS or LCT alone. Interestingly, changes both in behavioral and neurochemical endpoints were marginal in rats subjected to IMS or FSS for 28 days or those exposed to LCT for 3 days alone, compared to controls. The results suggest that stress is an important contributor in LCT induced cholinergic deficits.
Collapse
Affiliation(s)
- Rajendra K Shukla
- Developmenatl Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR - Indian Institute of Toxicology Research, Post Box 80, Vishvigyan Bhawan, 31 MG Marg, Lucknow 226 001, India; Department of Biochemistry, Integral Institute of Medical Sciences & Research, Integral University, Lucknow 226 026, India
| | - Richa Gupta
- Developmenatl Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR - Indian Institute of Toxicology Research, Post Box 80, Vishvigyan Bhawan, 31 MG Marg, Lucknow 226 001, India
| | - Pranay Srivastava
- Developmenatl Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR - Indian Institute of Toxicology Research, Post Box 80, Vishvigyan Bhawan, 31 MG Marg, Lucknow 226 001, India
| | - Yogesh K Dhuriya
- Developmenatl Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR - Indian Institute of Toxicology Research, Post Box 80, Vishvigyan Bhawan, 31 MG Marg, Lucknow 226 001, India
| | - Anshuman Singh
- Developmenatl Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR - Indian Institute of Toxicology Research, Post Box 80, Vishvigyan Bhawan, 31 MG Marg, Lucknow 226 001, India
| | - Lalit P Chandravanshi
- Developmenatl Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR - Indian Institute of Toxicology Research, Post Box 80, Vishvigyan Bhawan, 31 MG Marg, Lucknow 226 001, India
| | - Ajay Kumar
- Department of Biochemistry, Integral Institute of Medical Sciences & Research, Integral University, Lucknow 226 026, India
| | - M Haris Siddiqui
- Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow 226 026, India
| | - Devendra Parmar
- Developmenatl Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR - Indian Institute of Toxicology Research, Post Box 80, Vishvigyan Bhawan, 31 MG Marg, Lucknow 226 001, India
| | - Aditya B Pant
- Developmenatl Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR - Indian Institute of Toxicology Research, Post Box 80, Vishvigyan Bhawan, 31 MG Marg, Lucknow 226 001, India
| | - Vinay K Khanna
- Developmenatl Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR - Indian Institute of Toxicology Research, Post Box 80, Vishvigyan Bhawan, 31 MG Marg, Lucknow 226 001, India.
| |
Collapse
|
26
|
Dextran sulfate sodium-induced colitis alters stress-associated behaviour and neuropeptide gene expression in the amygdala-hippocampus network of mice. Sci Rep 2015; 5:9970. [PMID: 26066467 PMCID: PMC4464346 DOI: 10.1038/srep09970] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/26/2015] [Indexed: 01/07/2023] Open
Abstract
Psychological stress causes disease exacerbation and relapses in inflammatory bowel disease (IBD) patients. Since studies on stress processing during visceral inflammation are lacking, we investigated the effects of experimental colitis as well as psychological stress on neurochemical and neuroendocrine changes as well as behaviour in mice. Dextran sulfate sodium (DSS)-induced colitis and water avoidance stress (WAS) were used as mouse models of colitis and mild psychological stress, respectively. We measured WAS-associated behaviour, gene expression and proinflammatory cytokine levels within the amygdala, hippocampus and hypothalamus as well as plasma levels of cytokines and corticosterone in male C57BL/6N mice. Animals with DSS-induced colitis presented with prolonged immobility during the WAS session, which was associated with brain region-dependent alterations of neuropeptide Y (NPY), NPY receptor Y1, corticotropin-releasing hormone (CRH), CRH receptor 1, brain-derived neurotrophic factor and glucocorticoid receptor gene expression. Furthermore, the combination of DSS and WAS increased interleukin-6 and growth regulated oncogene-α levels in the brain. Altered gut-brain signalling in the course of DSS-induced colitis is thought to cause the observed distinct gene expression changes in the limbic system and the aberrant molecular and behavioural stress responses. These findings provide new insights into the effects of stress during IBD.
Collapse
|
27
|
Perinatal Positive and Negative Influences on the Early Neurobehavioral Reflex and Motor Development. PERINATAL PROGRAMMING OF NEURODEVELOPMENT 2015; 10:149-67. [DOI: 10.1007/978-1-4939-1372-5_8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
28
|
Gudiño-Cabrera G, Ureña-Guerrero ME, Rivera-Cervantes MC, Feria-Velasco AI, Beas-Zárate C. Excitotoxicity triggered by neonatal monosodium glutamate treatment and blood-brain barrier function. Arch Med Res 2014; 45:653-9. [PMID: 25431840 DOI: 10.1016/j.arcmed.2014.11.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 11/13/2014] [Indexed: 12/21/2022]
Abstract
It is likely that monosodium glutamate (MSG) is the excitotoxin that has been most commonly employed to characterize the process of excitotoxicity and to improve understanding of the ways that this process is related to several pathological conditions of the central nervous system. Excitotoxicity triggered by neonatal MSG treatment produces a significant pathophysiological impact on adulthood, which could be due to modifications in the blood-brain barrier (BBB) permeability and vice versa. This mini-review analyzes this topic through brief descriptions about excitotoxicity, BBB structure and function, role of the BBB in the regulation of Glu extracellular levels, conditions that promote breakdown of the BBB, and modifications induced by neonatal MSG treatment that could alter the behavior of the BBB. In conclusion, additional studies to better characterize the effects of neonatal MSG treatment on excitatory amino acids transporters, ionic exchangers, and efflux transporters, as well as the role of the signaling pathways mediated by erythropoietin and vascular endothelial growth factor in the cellular elements of the BBB, should be performed to identify the mechanisms underlying the increase in neurovascular permeability associated with excitotoxicity observed in several diseases and studied using neonatal MSG treatment.
Collapse
Affiliation(s)
- Graciela Gudiño-Cabrera
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, México
| | - Monica E Ureña-Guerrero
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, México
| | - Martha C Rivera-Cervantes
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, México
| | - Alfredo I Feria-Velasco
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, México
| | - Carlos Beas-Zárate
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Zapopan, Jalisco, México; División de Neurociencias, CIBO, IMSS, Guadalajara, Jalisco, México.
| |
Collapse
|
29
|
Telmisartan attenuates cognitive impairment caused by chronic stress in rats. Pharmacol Rep 2014; 66:436-41. [DOI: 10.1016/j.pharep.2013.11.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 11/08/2013] [Accepted: 11/26/2013] [Indexed: 01/05/2023]
|
30
|
Wincewicz D, Braszko JJ. Angiotensin II AT1 receptor blockade by telmisartan reduces impairment of spatial maze performance induced by both acute and chronic stress. J Renin Angiotensin Aldosterone Syst 2014; 16:495-505. [DOI: 10.1177/1470320314526269] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 01/26/2014] [Indexed: 01/02/2023] Open
Affiliation(s)
- Dominik Wincewicz
- Department of Clinical Pharmacology, Medical University of Bialystok, Poland
| | - Jan J Braszko
- Department of Clinical Pharmacology, Medical University of Bialystok, Poland
| |
Collapse
|
31
|
Yen LF, Wei VC, Kuo EY, Lai TW. Distinct patterns of cerebral extravasation by Evans blue and sodium fluorescein in rats. PLoS One 2013; 8:e68595. [PMID: 23861924 PMCID: PMC3702610 DOI: 10.1371/journal.pone.0068595] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/31/2013] [Indexed: 11/18/2022] Open
Abstract
The Evans blue dye (EBD; 961 Da) and the sodium fluorescein dye (NaF; 376 Da) are commonly used inert tracers in blood-brain barrier (BBB) research. They are both highly charged low molecular weight (LMW) tracers with similar lipophobic profiles. Nevertheless, the EBD binds to serum albumin (69,000 Da) to become a high molecular weight (HMW) protein tracer when injected into the circulation, whereas the NaF remains an unbound small molecule in the circulation. In this study, rats were injected with equal doses of either EBD or NaF to monitor their blood and tissue distribution. The EBD was largely confined to the circulation with little accumulation in the peripheral organ and even less accumulation in the central tissue, whereas the NaF distributed more evenly between the blood and the peripheral organ but was also largely excluded from the central tissue. Importantly, the EBD crossed the BBB most effectively at the prefrontal cortex and the cerebellum, and most poorly at the striatum. In marked contrast, the NaF was evenly distributed throughout the brain. Finally, the EBD exhibited this same peculiar tissue distribution profile when administered by either bolus injection or slow infusion. Our study suggests that different regions of the brain are equally permeable to LMW inert dyes like the NaF, but are markedly different in permeability to HMW proteins such as EBD-labelled serum albumin.
Collapse
Affiliation(s)
- Lola Fenghuei Yen
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Vivi Chiali Wei
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Eva Yuhua Kuo
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Ted Weita Lai
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
32
|
Xanthos DN, Püngel I, Wunderbaldinger G, Sandkühler J. Effects of peripheral inflammation on the blood-spinal cord barrier. Mol Pain 2012; 8:44. [PMID: 22713725 PMCID: PMC3407004 DOI: 10.1186/1744-8069-8-44] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 06/18/2012] [Indexed: 11/10/2022] Open
Abstract
Background Changes in the blood-central nervous system barriers occur under pathological conditions including inflammation and contribute to central manifestations of various diseases. After short-lasting peripheral and neurogenic inflammation, the evidence is mixed whether there are consistent blood-spinal cord changes. In the current study, we examine changes in the blood-spinal cord barrier after intraplantar capsaicin and λ-carrageenan using several methods: changes in occludin protein, immunoglobulin G accumulation, and fluorescent dye penetration. We also examine potential sex differences in male and female adult rats. Results After peripheral carrageenan inflammation, but not capsaicin inflammation, immunohistochemistry shows occludin protein in lumbar spinal cord to be significantly altered at 72 hours post-injection. In addition, there is also significant immunoglobulin G detected in lumbar and thoracic spinal cord at this timepoint in both male and female rats. However, acute administration of sodium fluorescein or Evans Blue dyes is not detected in the parenchyma at this timepoint. Conclusions Our results show that carrageenan inflammation induces changes in tight junction protein and immunoglobulin G accumulation, but these may not be indicative of a blood-spinal cord barrier breakdown. These changes appear transiently after peak nociception and may be indicative of reversible pathology that resolves together with inflammation.
Collapse
Affiliation(s)
- Dimitris N Xanthos
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | | | | | | |
Collapse
|
33
|
Sirav B, Seyhan N. Effects of radiofrequency radiation exposure on blood-brain barrier permeability in male and female rats. Electromagn Biol Med 2011; 30:253-60. [DOI: 10.3109/15368378.2011.600167] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
Liu J, Hu P, Qi XR, Meng FT, Kalsbeek A, Zhou JN. Acute restraint stress increases intrahypothalamic oestradiol concentrations in conjunction with increased hypothalamic oestrogen receptor β and aromatase mRNA expression in female rats. J Neuroendocrinol 2011; 23:435-443. [PMID: 21392135 DOI: 10.1111/j.1365-2826.2011.02123.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Activation of the hypothalamic-pituitary-adrenal axis is considered to be one of the key physiological responses to stress and, interestingly, shows a marked sex difference. Oestradiol plays an important role in this sex difference. The present study investigated the systemic and intrahypothalamic oestradiol response to physical restraint stress in female rats. We used jugular catheterisation and intrahypothalamic microdialysis to simultaneously measure plasma oestradiol and local oestradiol concentrations in the paraventricular nucleus (PVN) of the hypothalamus. We also assessed corticotrophin-releasing hormone (CRH), aromatase, and oestrogen receptor (ER) α and β mRNA expression in the PVN by quantitative polymerase chain reaction immediately after the acute stress period. As expected, PVN CRH mRNA and plasma corticosterone were significantly increased after acute stress. Interestingly, the local oestradiol concentration in the PVN also increased during the 1-h stress period in pro-oestrus and in ovariectomised (OVX) animals. Aromatase mRNA expression in the PVN was increased markedly in pro-oestrus but only modestly in oestrus. PVN ERβ but not ERα mRNA expression was significantly elevated in pro-oestrous animals. In addition, plasma oestradiol levels increased 10 min after stress, both during pro-oestrus and oestrus but not in OVX animals. To conclude, we report an intra-hypothalamic oestradiol response to restraint stress. The rising hypothalamic oestradiol concentration together with increased ERβ gene expression indicates a positive feedback of hypothalamic oestradiol signalling during acute stress in rats.
Collapse
Affiliation(s)
- J Liu
- Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Chinese Academy of Sciences, Hefei, Anhui, China
| | | | | | | | | | | |
Collapse
|
35
|
Effects of stressors and immune activating agents on peripheral and central cytokines in mouse strains that differ in stressor responsivity. Brain Behav Immun 2011; 25:468-82. [PMID: 21093579 DOI: 10.1016/j.bbi.2010.11.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 11/11/2010] [Accepted: 11/12/2010] [Indexed: 12/29/2022] Open
Abstract
The impact of inflammatory immune activation on behavioral and physiological processes varies with antecedent stressor experiences. We assessed whether immune activation would differentially influence such outcomes as a function of stressor reactivity related to genetic differences. To this end, we assessed the influence of a social stressor (exposure to a dominant mouse) in combination with an acute immune challenge on behavior and on peripheral and central cytokines in stressor-reactive BALB/cByJ mice and the less reactive C57BL/6ByJ strain. As C57BL/6ByJ and BALB/cByJ mice are highly T helper type-1 (Th1) and Th2 responsive, respectively, the stressor effects were assessed in response to different challenges, namely the viral analogue poly I:C and the bacterial endotoxin lipopolysaccharide (LPS). The stressor enhanced the effects of LPS on sickness behaviors and plasma corticosterone particularly in BALB/cByJ mice, whereas the effects of poly I:C, which primarily affects Th1 processes, were not augmented by the stressor. As well, the stressor increased circulating cytokines in LPS treated C57BL/6ByJ mice, whereas the effects of poly I:C were diminished. Finally, like circulating cytokines, mRNA expression of pro-inflammatory cytokines within the prefrontal cortex and hippocampus varied with the mouse strain and with the stressor experience, and with the specific cytokine considered. Together, the experiments indicated that the impact of stressors vary with the nature of the immune challenge to which animals had been exposed. Moreover, given the diversity of the stressor effects on central and peripheral processes, it seems likely that the cytokine changes, HPA activity and sickness operate through independent mechanisms.
Collapse
|
36
|
Brain metastases of mouse mammary adenocarcinoma is increased by acute stress. Brain Res 2010; 1366:204-10. [DOI: 10.1016/j.brainres.2010.09.085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 09/21/2010] [Accepted: 09/23/2010] [Indexed: 12/22/2022]
|
37
|
Altered functional development of the blood–brain barrier after early life stress in the rat. Brain Res Bull 2009; 79:376-87. [DOI: 10.1016/j.brainresbull.2009.05.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 05/09/2009] [Accepted: 05/13/2009] [Indexed: 11/22/2022]
|
38
|
Gibb J, Hayley S, Gandhi R, Poulter MO, Anisman H. Synergistic and additive actions of a psychosocial stressor and endotoxin challenge: Circulating and brain cytokines, plasma corticosterone and behavioral changes in mice. Brain Behav Immun 2008; 22:573-89. [PMID: 18191534 DOI: 10.1016/j.bbi.2007.12.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 11/28/2007] [Accepted: 12/01/2007] [Indexed: 01/08/2023] Open
Abstract
Activation of the inflammatory immune response may provoke neuroendocrine and central neurochemical effects that are reminiscent of those elicited by traditional stressors, and when administered concurrently may have synergistic effects. The present investigation assessed whether a psychosocial stressor, comprising social disruption, would augment the effects of lipopolysaccharide in mice. It was indeed observed that the social disruption engendered by a period of 2-4 weeks of social isolation (but not 1-7 days of this treatment) followed by regrouping, enhanced the effects of lipopolysaccharide (LPS: 10mug) in the provocation of sickness behavior, as well as plasma corticosterone, IL-6, TNF-alpha and IL-10 levels. Similar effects were not apparent with respect to IL-1beta, IL-4, or IFN-gamma. Synergy between LPS and other stressors (restraint, tail pinch, and loud noise) was not apparent with respect to sickness or plasma corticosterone, provisionally suggesting that social stressors, such as regrouping, may be more powerful or may engage unique neural or neuroendocrine circuits that favour synergistic outcomes. Within the CNS, the LPS and the regrouping stressor synergistically enhanced NE utilization within the prefrontal cortex, and additively influenced hippocampal NE utilization. In contrast to the effects on circulating cytokines, the LPS-induced elevation of IL-1beta, IL-6 and TNF-alpha mRNA expression in the hippocampus, PFC and nucleus tractus solitarius was diminished in animals that had experienced the regrouping stressor. In view of the combined actions of LPS challenge and a social stressor, these data are interpreted as suggesting that models of depression based on immune activation ought to consider the stressor backdrop upon which immune challenges are imposed.
Collapse
Affiliation(s)
- Julie Gibb
- Institute of Neuroscience, Carleton University, LSRB/1125 Colonial By Drive, Ottawa, Ont., Canada K1S 5B6
| | | | | | | | | |
Collapse
|
39
|
Gos T, Bock J, Poeggel G, Braun K. Stress-induced synaptic changes in the rat anterior cingulate cortex are dependent on endocrine developmental time windows. Synapse 2008; 62:229-32. [PMID: 18088062 DOI: 10.1002/syn.20477] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Fetal and neonatal brain development is characterized by developmental time windows during which brain regions or neuron types are specifically sensitive to environmental influences. Previous studies on cortical development have revealed evidence for the hypothesis that the extent and the direction of experience-induced neuronal and synaptic changes correlate with time windows of endocrine development. To further test this hypothesis we exposed rats to neonatal separation stress during different phases of endocrine maturation, i.e. prior, during and after the stress hyporesponsive period (SHRP) of the hypothalamic-pituitary-adrenal (HPA) axis. We show here that only stress during the SHRP resulted in significantly decreased (-29%) spines densities on the basal dendrites of pyramidal cells in layer V of the anterior cingulate cortex (ACd), whereas stress during the other two tested time windows had no effect on these parameters. Dendritic length remained unaffected by stress exposure at any of the tested time windows. These results reveal specific developmental time window for synaptic wiring within the deeper layers of the anterior cingulate cortex, which seem not to be mediated by hormonally induced mechanisms.
Collapse
Affiliation(s)
- Tomasz Gos
- Institute of Forensic Medicine, Medical University of Gdańsk, 80-204 Gdańsk, Poland
| | | | | | | |
Collapse
|
40
|
Burattini C, McGeehan AJ, Griffin WC, Gass JT, Kinder JR, Janak PH, Olive MF. A microdialysis study of extracellular levels of acamprosate and naltrexone in the rat brain following acute and repeated administration. Addict Biol 2008; 13:70-9. [PMID: 18269381 DOI: 10.1111/j.1369-1600.2008.00097.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Acamprosate and naltrexone are widely used in the treatment of alcoholism. However, numerous studies in rodents have shown differential effects of these compounds on alcohol consumption and/or relapse-like behavior following acute versus repeated administration. In order to determine if these differential behavioral effects could be attributable to changes in extracellular levels of these compounds, we used in vivo microdialysis to monitor extracellular levels of acamprosate and naltrexone in the rat medial prefrontal cortex following acute and repeated intraperitoneal administration. For acute treatment, animals received a single administration of acamprosate (100 or 300 mg/kg) or naltrexone (1 or 3 mg/kg). For repeated treatment, animals received once daily treatment with saline, acamprosate (300 mg/kg) or naltrexone (3 mg/kg) for 10 days before a subsequent challenge with the compound according to their respective pretreatment group. Dialysate levels of acamprosate and naltrexone were analyzed by liquid chromatography-tandem mass spectrometry and high performance liquid chromatography, respectively. Following acute administration, peak dialysate concentrations of each compound were dose-dependent, observed within 1 hour of administration, and were found to be in the low micromolar range for acamprosate and in the low to mid-nanomolar range for naltrexone. Pretreatment with acamprosate, but not naltrexone, for 10 days resulted in higher dialysate concentrations of the compound relative to saline-pretreated controls. Thus, repeated administration of acamprosate, but not naltrexone, results in augmented extracellular levels of the compound in the brain relative to saline-pretreated controls, which may explain the need for repeated administration of acamprosate in order to observe effects on alcohol consumption and/or relapse.
Collapse
|
41
|
Dubovicky M, Paton S, Morris M, Mach M, Lucot JB. Effects of combined exposure to pyridostigmine bromide and shaker stress on acoustic startle response, pre-pulse inhibition and open field behavior in mice. J Appl Toxicol 2007; 27:276-83. [PMID: 17265421 DOI: 10.1002/jat.1210] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The present study investigated the effect of combined exposure of pyridostigmine bromide (PB) and chronic shaker stress on acoustic startle responses (ASR), pre-pulse inhibition (PPI) and open field behavior of adult C57BL/6J mice. PB (10 mg kg(-1) day(-1) for 7 days) or saline was administered subcutaneously using osmotic Alzet minipumps implanted under the skin on the back of the mice. At the same time, the mice were exposed to 7 days of intermittent shaker stress. They were tested for ASR (100 dB and 120 dB stimuli) and PPI (70 dB + 100 dB and 70 dB + 120 dB) in the acoustic startle monitor system. The mice were assessed during the shaker stress on days 2 and 7 and 7, 14, 21 and 28 days after discontinuation of treatment. Separate groups of mice were tested in the open field in 15 min sessions on days 1, 3 and 6 during shaker stress and PB treatment. Exposure of mice to PB resulted in an exaggerated ASR, reduced PPI and non-significant decrease in locomotor activity. These behavioral changes were apparent only during exposure to PB. Repeated shaker stress did not have any effect on sensorimotor functions or open field behavior of mice. There was no prolonged or delayed effect of PB and/or stress on individual behavioral variables. The study found C57BL/6J mice to be behaviorally sensitive to PB treatment.
Collapse
Affiliation(s)
- M Dubovicky
- Wright State University, Boonshoft School of Medicine, Department of Pharmacology and Toxicology, Dayton, OH 45435, USA.
| | | | | | | | | |
Collapse
|
42
|
Theoharides TC, Papaliodis D, Tagen M, Konstantinidou A, Kempuraj D, Clemons A. Chronic fatigue syndrome, mast cells, and tricyclic antidepressants. J Clin Psychopharmacol 2005; 25:515-20. [PMID: 16282830 DOI: 10.1097/01.jcp.0000193483.89260.a7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
43
|
Philippens IHCHM, Joosen MJA, Vanwersch RAP. Stress adversely affects efficacy of physostigmine–scopolamine pretreatment against soman in guinea pigs. Pharmacol Biochem Behav 2005; 82:125-32. [PMID: 16126267 DOI: 10.1016/j.pbb.2005.07.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2004] [Revised: 06/13/2005] [Accepted: 07/12/2005] [Indexed: 11/25/2022]
Abstract
During military operations, soldiers may be exposed to mixtures of chemicals and to physical, emotional and psychological stress factors, which all may influence efficacy of any treatment, including the nerve agent pretreatment regimen. The purpose of this study was therefore to investigate the influence of chronic intermittent, variable, unpredictable and uncontrollable stress conditions on the side effects and therapeutic efficacy of the combination of physostigmine (0.025 mg/kg/h) and scopolamine (0.018 mg/kg/h) as a pretreatment against 2 x LD50 soman intoxication in guinea pigs. Stress during pretreatment led to an increase of motor activity and an increase of power in the EEG delta2 frequency band. After chronic stress, exposure of pretreated animals to 2 x LD50 soman resulted in more severe intoxication symptoms, a more persistent effect on the startle response, and considerable more severe and persistent effects on the EEG power-spectrum, indicating irreversible brain damage.
Collapse
Affiliation(s)
- Ingrid H C H M Philippens
- TNO Prins Maurits Laboratory (TNO-PML), Research Group Medical Countermeasures, P.O. Box 45, 2280 AA Rijswijk ZH, The Netherlands.
| | | | | |
Collapse
|
44
|
Yan E, Castillo-Meléndez M, Nicholls T, Hirst J, Walker D. Cerebrovascular responses in the fetal sheep brain to low-dose endotoxin. Pediatr Res 2004; 55:855-63. [PMID: 14973172 DOI: 10.1203/01.pdr.0000115681.95957.d4] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Clinical and experimental evidence indicate that infection in pregnancy is associated with fetal brain damage. However, the inflammatory processes that compromise the fetal brain are not fully understood. In this study, we used a single, low dose of lipopolysaccharide (LPS, 0.1 microg/kg i.v.) to provoke an acute-phase response in unanesthetized fetal sheep in utero. COX-2 mRNA was increased in the cortex and cerebellum at 24 and 48 h after LPS, and immunoreactive COX-2 protein was increased in perivascular cells throughout gray and white matter at 24 h after LPS administration. Plasma albumin was observed in the parenchyma of the brain in cortex, thalamus, hypothalamus, corpus callosum, fornix, hippocampus, midbrain, subcallosal bundle, and cerebellar Purkinje cells. Large, rounded, lectin-positive cells with the appearance of macrophages were observed around blood vessels in subventricular white matter. These results indicate that blood-brain barrier permeability is increased in the fetal brain after exposure to endotoxin and suggests that cytotoxic and pro-inflammatory substances could pass from the circulation into the brain after peripheral inflammatory stimulation.
Collapse
Affiliation(s)
- Edwin Yan
- Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | | | | | | | | |
Collapse
|
45
|
Theoharides TC, Cochrane DE. Critical role of mast cells in inflammatory diseases and the effect of acute stress. J Neuroimmunol 2004; 146:1-12. [PMID: 14698841 DOI: 10.1016/j.jneuroim.2003.10.041] [Citation(s) in RCA: 315] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Mast cells are not only necessary for allergic reactions, but recent findings indicate that they are also involved in a variety of neuroinflammatory diseases, especially those worsened by stress. In these cases, mast cells appear to be activated through their Fc receptors by immunoglobulins other than IgE, as well as by anaphylatoxins, neuropeptides and cytokines to secrete mediators selectively without overt degranulation. These facts can help us better understand a variety of sterile inflammatory conditions, such as multiple sclerosis (MS), migraines, inflammatory arthritis, atopic dermatitis, coronary inflammation, interstitial cystitis and irritable bowel syndrome, in which mast cells are activated without allergic degranulation.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Department of Pharmacology and Experimental Therapeutics, Tufts-New England Medical Center, Boston, MA, USA.
| | | |
Collapse
|
46
|
Oztaş B, Akgül S, Arslan FB. Influence of surgical pain stress on the blood-brain barrier permeability in rats. Life Sci 2004; 74:1973-9. [PMID: 14967192 DOI: 10.1016/j.lfs.2003.07.054] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2003] [Accepted: 07/31/2003] [Indexed: 11/20/2022]
Abstract
Effect of surgical pain stress on the blood-brain barrier permeability was investigated in rats. The animals were divided into four groups: Group 1: control, Group 2: immobilization stress, Group 3: acute hypertension, Group 4: immobilization stress + surgical pain stress. Bilateral hid paw surgical wounds for cannulations were applied in animals' inguinal regions under diethyl-ether anesthesia, then the animals were awaken from anesthesia to produce surgical pain stress. Evans-blue was used as a blood-brain barrier tracer. There is no significantly blood-brain barrier breakdown after short-time immobilization stress, but after adrenalin hypertension blood-brain barrier permeability was increased especially on frontal and occipital cortices in 50% of the animals. Surgical pain stress increased blood-brain barrier permeabiliy in comparison to acute adrenalin-induced hypertension (p < 0.01). In surgical pain stress-induced animals distinct Evans-blue leakage was observed in the occipital, frontal and parieto-temporal cortices.
Collapse
Affiliation(s)
- Baria Oztaş
- Department of Physiology, Istanbul Faculty of Medicine, Istanbul University, Capa-34390 Istanbul, Turkey.
| | | | | |
Collapse
|
47
|
Esposito P, Basu S, Letourneau R, Jacobson S, Theoharides TC. Corticotropin-releasing factor (CRF) can directly affect brain microvessel endothelial cells. Brain Res 2003; 968:192-8. [PMID: 12663088 DOI: 10.1016/s0006-8993(03)02237-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Stress activates the hypothalamic-pituitary-adrenal (HPA) axis through release of corticotropin releasing factor (CRF), leading to production of glucocorticoids that down regulate immune responses. However, acute stress via CRF also has pro-inflammatory effects. We previously showed that acute stress increases rat blood-brain barrier (BBB) permeability, an effect involving brain mast cells and CRF, as it was absent in W/W(v) mast cell-deficient mice and was blocked by the CRF-receptor antagonist, Antalarmin. We investigated if CRF could also have a direct action on brain microvessel endothelial cells (BMEC) isolated from rat and bovine brain. BMEC were cultured and identified by electron microscopy. Western blot analysis of cultured BMEC identified CRF receptor protein; stimulation with CRF, or it structural analogue urocortin (Ucn) showed that the receptor is functionally coupled to adenylate cyclase as it increased cyclic AMP (cAMP) levels by 2-fold. These findings suggest that CRF could affect BMEC structure or function, as reported for increased cAMP levels by other studies. It is, therefore, possible that CRF may directly regulate BBB permeability, in addition to any effect mediated via brain mast cells.
Collapse
Affiliation(s)
- Pamela Esposito
- Department of Pharmacology, Tufts University School of Medicine, New England Medical Center, 136 Harrison Avenue, Boston, MA 02111, USA
| | | | | | | | | |
Collapse
|
48
|
Chandler N, Jacobson S, Esposito P, Connolly R, Theoharides TC. Acute stress shortens the time to onset of experimental allergic encephalomyelitis in SJL/J mice. Brain Behav Immun 2002; 16:757-63. [PMID: 12776697 DOI: 10.1016/s0889-1591(02)00028-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- N Chandler
- Surgical Research, Tufts University School of Medicine, New England Medical Center, Boston, MA 02111, USA.
| | | | | | | | | |
Collapse
|
49
|
Abdel-Rahman A, Shetty AK, Abou-Donia MB. Disruption of the Blood–Brain Barrier and Neuronal Cell Death in Cingulate Cortex, Dentate Gyrus, Thalamus, and Hypothalamus in a Rat Model of Gulf-War Syndrome. Neurobiol Dis 2002; 10:306-26. [PMID: 12270692 DOI: 10.1006/nbdi.2002.0524] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We investigated the effects of a combined exposure to restraint stress and low doses of chemicals pyridostigmine bromide (PB), N, N-diethyl-m-toluamide (DEET), and permethrin in adult male rats, a model of Gulf-War syndrome. Animals were exposed daily to one of the following for 28 days: (i) a combination of stress and chemicals (PB, 1.3 mg/kg/day; DEET, 40 mg/kg/day; and permethrin, 0.13 mg/kg/day); (ii) stress and vehicle; (iii) chemicals alone; and (iv) vehicle alone. All animals were evaluated for: (i) the disruption of the blood-brain barrier (BBB) using intravenous horseradish peroxidase (HRP) injections and endothelial barrier antigen (EBA) immunostaining; (ii) neuronal cell death using H&E staining, silver staining, and glial fibrillary acidic protein (GFAP) immunostaining; and (iii) acetylcholinesterase (AChE) activity and m2-muscarinic acetylcholine receptors (m2-AChR). Animals subjected to stress and chemicals exhibited both disruption of the BBB and neuronal cell death in the cingulate cortex, the dentate gyrus, the thalamus, and the hypothalamus. Other regions of the brain, although they demonstrated some neuronal cell death, did not exhibit disruption of the BBB. The neuropathological changes in the above four brain regions were highly conspicuous and revealed by a large number of HRP-positive neurons (21-40% of total neurons), a decreased EBA immunostaining (42-51% reduction), a decreased number of surviving neurons (27-40% reduction), the presence of dying neurons (4-10% of total neurons), and an increased GFAP immunostaining (45-51% increase). These changes were also associated with decreased forebrain AChE activity and m2-AchR (19-25% reduction). In contrast, in animals exposed to stress and vehicle or chemicals alone, the above indices were mostly comparable to that of animals exposed to vehicle alone. Thus, a combined exposure to stress and low doses of PB, DEET, and permethrin leads to significant brain injury. The various neurological symptoms reported by Gulf-War veterans could be linked to this kind of brain injury incurred during the war.
Collapse
Affiliation(s)
- Ali Abdel-Rahman
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Caroloina 277109, USA
| | | | | |
Collapse
|
50
|
Madrigal JLM, Moro MA, Lizasoain I, Lorenzo P, Leza JC. Stress-induced increase in extracellular sucrose space in rats is mediated by nitric oxide. Brain Res 2002; 938:87-91. [PMID: 12031539 DOI: 10.1016/s0006-8993(02)02467-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Exposure to physical or psychological stress causes brain damage ranging from minimal behavioural alterations to neurodegeneration. One of the proposed mechanisms for stress-induced neurodegeneration is the overproduction of nitric oxide (NO) and related oxidative-nitrosative compounds via expression of the inducible NO synthase (iNOS). In the present investigation, the effect of acute or chronic immobilisation on blood-brain barrier (BBB) permeability and the possible role of iNOS were studied in adult male Wistar rats. Stress-induced [(14)C]-sucrose uptake by brain tissue correlates with the production of the stable NO metabolites nitrite and nitrate in both peripheral (plasma) and central (brain) compartments. Injection of the specific iNOS inhibitor 1400W (2 mg/kg, i.p.) prevents the stress-induced increase in BBB permeability. Taken together, these findings indicate that iNOS expression mediates stress-induced increase in BBB permeability and support a possible neuroprotective role for specific iNOS inhibitors in this situation.
Collapse
Affiliation(s)
- José L M Madrigal
- Departmento de Farmacología, Facultad de Medicina, Universidad Complutense, Madrid 28040, Spain
| | | | | | | | | |
Collapse
|