1
|
Su C, Huang T, Zhang M, Zhang Y, Zeng Y, Chen X. Glucocorticoid receptor signaling in the brain and its involvement in cognitive function. Neural Regen Res 2025; 20:2520-2537. [PMID: 39248182 PMCID: PMC11801288 DOI: 10.4103/nrr.nrr-d-24-00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/07/2024] [Accepted: 07/06/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-adrenal axis regulates the secretion of glucocorticoids in response to environmental challenges. In the brain, a nuclear receptor transcription factor, the glucocorticoid receptor, is an important component of the hypothalamic-pituitary-adrenal axis's negative feedback loop and plays a key role in regulating cognitive equilibrium and neuroplasticity. The glucocorticoid receptor influences cognitive processes, including glutamate neurotransmission, calcium signaling, and the activation of brain-derived neurotrophic factor-mediated pathways, through a combination of genomic and non-genomic mechanisms. Protein interactions within the central nervous system can alter the expression and activity of the glucocorticoid receptor, thereby affecting the hypothalamic-pituitary-adrenal axis and stress-related cognitive functions. An appropriate level of glucocorticoid receptor expression can improve cognitive function, while excessive glucocorticoid receptors or long-term exposure to glucocorticoids may lead to cognitive impairment. Patients with cognitive impairment-associated diseases, such as Alzheimer's disease, aging, depression, Parkinson's disease, Huntington's disease, stroke, and addiction, often present with dysregulation of the hypothalamic-pituitary-adrenal axis and glucocorticoid receptor expression. This review provides a comprehensive overview of the functions of the glucocorticoid receptor in the hypothalamic-pituitary-adrenal axis and cognitive activities. It emphasizes that appropriate glucocorticoid receptor signaling facilitates learning and memory, while its dysregulation can lead to cognitive impairment. This provides clues about how glucocorticoid receptor signaling can be targeted to overcome cognitive disability-related disorders.
Collapse
Affiliation(s)
- Chonglin Su
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Taiqi Huang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Meiyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Xingxing Chen
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Wu Y, Wang Y, Lu Y, Yan J, Zhao H, Yang R, Pan J. Research advances in huntingtin-associated protein 1 and its application prospects in diseases. Front Neurosci 2024; 18:1402996. [PMID: 38975245 PMCID: PMC11224548 DOI: 10.3389/fnins.2024.1402996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
Huntingtin-associated protein 1 (HAP1) was the first protein discovered to interact with huntingtin. Besides brain, HAP1 is also expressed in the spinal cord, dorsal root ganglion, endocrine, and digestive systems. HAP1 has diverse functions involving in vesicular transport, receptor recycling, gene transcription, and signal transduction. HAP1 is strongly linked to several neurological diseases, including Huntington's disease, Alzheimer's disease, epilepsy, ischemic stroke, and depression. In addition, HAP1 has been proved to participate in cancers and diabetes mellitus. This article provides an overview of HAP1 regarding the tissue distribution, cell localization, functions, and offers fresh perspectives to investigate its role in diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jingying Pan
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| |
Collapse
|
3
|
İş Ö, Wang X, Reddy JS, Min Y, Yilmaz E, Bhattarai P, Patel T, Bergman J, Quicksall Z, Heckman MG, Tutor-New FQ, Can Demirdogen B, White L, Koga S, Krause V, Inoue Y, Kanekiyo T, Cosacak MI, Nelson N, Lee AJ, Vardarajan B, Mayeux R, Kouri N, Deniz K, Carnwath T, Oatman SR, Lewis-Tuffin LJ, Nguyen T, Carrasquillo MM, Graff-Radford J, Petersen RC, Jr Jack CR, Kantarci K, Murray ME, Nho K, Saykin AJ, Dickson DW, Kizil C, Allen M, Ertekin-Taner N. Gliovascular transcriptional perturbations in Alzheimer's disease reveal molecular mechanisms of blood brain barrier dysfunction. Nat Commun 2024; 15:4758. [PMID: 38902234 PMCID: PMC11190273 DOI: 10.1038/s41467-024-48926-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/17/2024] [Indexed: 06/22/2024] Open
Abstract
To uncover molecular changes underlying blood-brain-barrier dysfunction in Alzheimer's disease, we performed single nucleus RNA sequencing in 24 Alzheimer's disease and control brains and focused on vascular and astrocyte clusters as main cell types of blood-brain-barrier gliovascular-unit. The majority of the vascular transcriptional changes were in pericytes. Of the vascular molecular targets predicted to interact with astrocytic ligands, SMAD3, upregulated in Alzheimer's disease pericytes, has the highest number of ligands including VEGFA, downregulated in Alzheimer's disease astrocytes. We validated these findings with external datasets comprising 4,730 pericyte and 150,664 astrocyte nuclei. Blood SMAD3 levels are associated with Alzheimer's disease-related neuroimaging outcomes. We determined inverse relationships between pericytic SMAD3 and astrocytic VEGFA in human iPSC and zebrafish models. Here, we detect vast transcriptome changes in Alzheimer's disease at the gliovascular-unit, prioritize perturbed pericytic SMAD3-astrocytic VEGFA interactions, and validate these in cross-species models to provide a molecular mechanism of blood-brain-barrier disintegrity in Alzheimer's disease.
Collapse
Affiliation(s)
- Özkan İş
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Xue Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Joseph S Reddy
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Yuhao Min
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Elanur Yilmaz
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Prabesh Bhattarai
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Tulsi Patel
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Zachary Quicksall
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Michael G Heckman
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | | | - Birsen Can Demirdogen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Biomedical Engineering, TOBB University of Economics and Technology, Ankara, Turkey
| | - Launia White
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Vincent Krause
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Mehmet Ilyas Cosacak
- German Center for Neurodegenerative Diseases (DZNE) within Helmholtz Association, Dresden, Germany
| | - Nastasia Nelson
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Annie J Lee
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Badri Vardarajan
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Richard Mayeux
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Naomi Kouri
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Kaancan Deniz
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Troy Carnwath
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Laura J Lewis-Tuffin
- Mayo Clinic Florida Cytometry and Cell Imaging Laboratory, Mayo Clinic, Jacksonville, FL, USA
| | - Thuy Nguyen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Ronald C Petersen
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Alzheimer's Disease Research Center, Rochester, MN, USA
| | | | - Kejal Kantarci
- Mayo Clinic Alzheimer's Disease Research Center, Rochester, MN, USA
| | | | - Kwangsik Nho
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Caghan Kizil
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
4
|
Theodoridou D, Tsiantis CO, Vlaikou AM, Chondrou V, Zakopoulou V, Christodoulides P, Oikonomou ED, Tzimourta KD, Kostoulas C, Tzallas AT, Tsamis KI, Peschos D, Sgourou A, Filiou MD, Syrrou M. Developmental Dyslexia: Insights from EEG-Based Findings and Molecular Signatures-A Pilot Study. Brain Sci 2024; 14:139. [PMID: 38391714 PMCID: PMC10887023 DOI: 10.3390/brainsci14020139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Developmental dyslexia (DD) is a learning disorder. Although risk genes have been identified, environmental factors, and particularly stress arising from constant difficulties, have been associated with the occurrence of DD by affecting brain plasticity and function, especially during critical neurodevelopmental stages. In this work, electroencephalogram (EEG) findings were coupled with the genetic and epigenetic molecular signatures of individuals with DD and matched controls. Specifically, we investigated the genetic and epigenetic correlates of key stress-associated genes (NR3C1, NR3C2, FKBP5, GILZ, SLC6A4) with psychological characteristics (depression, anxiety, and stress) often included in DD diagnostic criteria, as well as with brain EEG findings. We paired the observed brain rhythms with the expression levels of stress-related genes, investigated the epigenetic profile of the stress regulator glucocorticoid receptor (GR) and correlated such indices with demographic findings. This study presents a new interdisciplinary approach and findings that support the idea that stress, attributed to the demands of the school environment, may act as a contributing factor in the occurrence of the DD phenotype.
Collapse
Affiliation(s)
- Daniela Theodoridou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Christos-Orestis Tsiantis
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Angeliki-Maria Vlaikou
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), 45110 Ioannina, Greece
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Vasiliki Chondrou
- Laboratory of Biology, School of Science and Technology, Hellenic Open University, 26335 Patras, Greece
| | - Victoria Zakopoulou
- Department of Speech and Language Therapy, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Pavlos Christodoulides
- Department of Speech and Language Therapy, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
- Laboratory of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Emmanouil D Oikonomou
- Department of Informatics and Telecommunications, School of Informatics & Telecommunications, University of Ioannina, 47100 Arta, Greece
| | - Katerina D Tzimourta
- Department of Electrical and Computer Engineering, University of Western Macedonia, 50100 Kozani, Greece
| | - Charilaos Kostoulas
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Alexandros T Tzallas
- Department of Informatics and Telecommunications, School of Informatics & Telecommunications, University of Ioannina, 47100 Arta, Greece
| | - Konstantinos I Tsamis
- Laboratory of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitrios Peschos
- Laboratory of Physiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Argyro Sgourou
- Laboratory of Biology, School of Science and Technology, Hellenic Open University, 26335 Patras, Greece
| | - Michaela D Filiou
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), 45110 Ioannina, Greece
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Maria Syrrou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
5
|
Gao H, Li Y, Chen X. Interactions between nuclear receptors glucocorticoid receptor α and peroxisome proliferator-activated receptor α form a negative feedback loop. Rev Endocr Metab Disord 2022; 23:893-903. [PMID: 35476174 DOI: 10.1007/s11154-022-09725-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2022] [Indexed: 02/05/2023]
Abstract
Both nuclear receptors glucocorticoid receptor α (GRα) and peroxisome proliferator-activated receptor α (PPARα) are involved in energy and lipid metabolism, and possess anti-inflammation effects. Previous studies indicate that a regulatory loop may exist between them. In vivo and in vitro studies showed that glucocorticoids stimulate hepatic PPARα expression via GRα at the transcriptional level. This stimulation of PPARα by GRα has physiological relevance and PPARα is involved in many glucocorticoid-induced pathophysiological processes, including gluconeogenesis and ketogenesis during fasting, insulin resistance, hypertension and anti-inflammatory effects. PPARα also synergizes with GRα to promote erythroid progenitor self-renewal. As the feedback, PPARα inhibits glucocorticoid actions at pre-receptor and receptor levels. PPARα decreases glucocorticoid production through inhibiting the expression and activity of type-1 11β-hydroxysteroid dehydrogenase, which converts inactive glucocorticoids to active glucocorticoids at local tissues, and also down-regulates hepatic GRα expression, thus forming a complete and negative feedback loop. This negative feedback loop sheds light on prospective multi-drug therapeutic treatments in inflammatory diseases through a combination of glucocorticoids and PPARα agonists. This combination may potentially enhance the anti-inflammatory effects while alleviating side effects on glucose and lipid metabolism due to GRα activation. More investigations are needed to clarify the underlying mechanism and the relevant physiological or pathological significance of this regulatory loop.
Collapse
Affiliation(s)
- Hongjiao Gao
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, 610041, Chengdu, China
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Zunyi Medical University (the First People's Hospital of Zunyi), 563002, Zunyi, China
| | - Yujue Li
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Xiang Chen
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
6
|
Liao HY, Lin YW. Electroacupuncture Attenuates Chronic Inflammatory Pain and Depression Comorbidity through Transient Receptor Potential V1 in the Brain. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1417-1435. [PMID: 34224338 DOI: 10.1142/s0192415x2150066x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Chronic pain is one of the highest costs in clinical therapy, often appearing comorbid with depression. They present with overlapping clinical conditions and common pathological pathways especially in neuroinflammation, both of which can be reversed by electroacupuncture (EA). Transient receptor potential V1 receptor (TRPV1) is a Ca[Formula: see text] permeable ion channel that responds to brain inflammation and has a known role in the development of chronic pain and depression. Here, we investigate the role of TRPV1 and its related molecules in a mouse model of inflammation-induced chronic pain and depression using Complete Freund's adjuvant (CFA). We measured inflammatory mediators in plasma and evaluated the TRPV1 signaling pathway in the medial prefrontal cortex (mPFC), hypothalamus, and periaqueductal gray (PAG) of the mouse brain. Mechanical and thermal hyperalgesia as well as depressive-like behaviors were induced using the open field test and forced swimming test. Therapeutic effects were observed in EA and Trpv1[Formula: see text] mice in measures of chronic pain and depression. Inflammatory mediators induced by CFA injection were attenuated by EA and Trpv1 deletion. TRPV1 and downstream molecules were significantly decreased in the mPFC, hypothalamus, and PAG of mice, effects which were reversed by EA and Trpv1 knockout. We provide novel evidence that these inflammatory mediators modulate the TRPV1 signaling pathway and suggest new potential therapeutic targets for chronic pain and depression.
Collapse
Affiliation(s)
- Hsien-Yin Liao
- College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, Taiwan
| | - Yi-Wen Lin
- College of Chinese Medicine, Graduate Institute of Acupuncture Science, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
7
|
Liao HY, Lin YW. Electroacupuncture reduces cold stress-induced pain through microglial inactivation and transient receptor potential V1 in mice. Chin Med 2021; 16:43. [PMID: 34082798 PMCID: PMC8173787 DOI: 10.1186/s13020-021-00451-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/24/2021] [Indexed: 11/16/2022] Open
Abstract
Background The treatment, and efficacy thereof, is considered to be inadequate with specificity to alleviation of Fibromyalgia and its associated pain. Fibromyalgia patients suffer from chronic and persistent widespread pain and generalized tenderness. Transient receptor potential V1 (TRPV1), which is reported as a Ca2+ permeable ion channel that can be activated by inflammation, is reported to be involved in the development of fibromyalgia pain. Methods The current study explored the TRPV1 channel functions as a noxious sensory input in mice cold stress model. It remains unknown whether electroacupuncture (EA) attenuates fibromyalgia pain or affects the TRPV1 pathway. Results We show that cold stress increases mechanical and thermal pain (day 7: mechanical: 1.69 ± 0.41 g; thermal: 4.68 ± 0.56 s), and that EA and Trpv1 deletion counter this increase. EA and Trpv1 deletion reduced the cold stress-induced increase in inflammatory mediators and TRPV1-related molecules in the hypothalamus, periaqueductal gray (PAG), and cerebellum of mice. Conclusions Our results imply that EA has an analgesic effect associated with TRPV1 downregulation. We provide novel evidence that these inflammatory mediators can modulate the TRPV1 signaling pathway and suggest new potential therapeutic targets for fibromyalgia pain.
Collapse
Affiliation(s)
- Hsien-Yin Liao
- College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Yi-Wen Lin
- College of Chinese Medicine, Graduate Institute of Acupuncture Science, China Medical University, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan. .,Chinese Medicine Research Center, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
8
|
Wang B, Xin N, Qian X, Zhai L, Miao Z, Yang Y, Li S, Sun M, Xu X, Li XJ. Ahi1 regulates the nuclear translocation of glucocorticoid receptor to modulate stress response. Transl Psychiatry 2021; 11:188. [PMID: 33782379 PMCID: PMC8007735 DOI: 10.1038/s41398-021-01305-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/21/2021] [Accepted: 03/03/2021] [Indexed: 12/21/2022] Open
Abstract
Stress activates the nuclear translocation of glucocorticoid receptors (GR) to trigger gene expression. Abnormal GR levels can alter the stress responses in animals and therapeutic effects of antidepressants. Here, we reported that stress-mediated nuclear translocation of GR reduced Ahi1 in the stressed cells and mouse brains. Ahi1 interacts with GR to stabilize each other in the cytoplasm. Importantly, Ahi1 deficiency promotes the degradation of GR in the cytoplasm and reduced the nuclear translocation of GR in response to stress. Genetic depletion of Ahi1 in mice caused hyposensitivity to antidepressants under the stress condition. These findings suggest that AHI1 is an important regulator of GR level and may serve as a therapeutic target for stress-related disorders.
Collapse
Affiliation(s)
- Bin Wang
- grid.429222.d0000 0004 1798 0228Institute for Fetology, The First Affiliated Hospital of Soochow University, 215006 Suzhou, China ,grid.263761.70000 0001 0198 0694Institute of Neuroscience, Soochow University, 215123 Suzhou, China
| | - Ning Xin
- grid.263761.70000 0001 0198 0694Institute of Neuroscience, Soochow University, 215123 Suzhou, China ,grid.413389.4Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu China
| | - Xuanchen Qian
- grid.263761.70000 0001 0198 0694Institute of Neuroscience, Soochow University, 215123 Suzhou, China
| | - Lijing Zhai
- grid.263761.70000 0001 0198 0694Institute of Neuroscience, Soochow University, 215123 Suzhou, China
| | - Zhigang Miao
- grid.263761.70000 0001 0198 0694Institute of Neuroscience, Soochow University, 215123 Suzhou, China
| | - Yong Yang
- grid.263761.70000 0001 0198 0694Department of Psychiatry, The Affiliated Guangji Hospital of Soochow University, 215008 Suzhou, China
| | - Shihua Li
- grid.258164.c0000 0004 1790 3548Guangdong Key Laboratory of non-human primate models, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632 Guangzhou, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, 215006, Suzhou, China.
| | - Xingshun Xu
- Institute of Neuroscience, Soochow University, 215123, Suzhou, China. .,Department of Neurology, The First Affiliated Hospital of Soochow University, 215006, Suzhou, China. .,Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, 215123, Suzhou, Jiangsu, China.
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of non-human primate models, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China.
| |
Collapse
|
9
|
Chinese Herbal Medicine for the Treatment of Depression: Effects on the Neuroendocrine-Immune Network. Pharmaceuticals (Basel) 2021; 14:ph14010065. [PMID: 33466877 PMCID: PMC7830381 DOI: 10.3390/ph14010065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
The neuroimmune and neuroendocrine systems are two critical biological systems in the pathogenesis of depression. Clinical and preclinical studies have demonstrated that the activation of the neuroinflammatory response of the immune system and hyperactivity of the hypothalamus–pituitary–adrenal (HPA) axis of the neuroendocrine system commonly coexist in patients with depression and that these two systems bidirectionally regulate one another through neural, immunological, and humoral intersystem interactions. The neuroendocrine-immune network poses difficulties associated with the development of antidepressant agents directed toward these biological systems for the effective treatment of depression. On the other hand, multidrug and multitarget Chinese Herbal Medicine (CHM) has great potential to assist in the development of novel medications for the systematic pharmacotherapy of depression. In this narrative essay, we conclusively analyze the mechanisms of action of CHM antidepressant constituents and formulas, specifically through the modulation of the neuroendocrine-immune network, by reviewing recent preclinical studies conducted using depressive animal models. Some CHM herbal constituents and formulas are highlighted as examples, and their mechanisms of action at both the molecular and systems levels are discussed. Furthermore, we discuss the crosstalk of these two biological systems and the systems pharmacology approach for understanding the system-wide mechanism of action of CHM on the neuroendocrine-immune network in depression treatment. The holistic, multidrug, and multitarget nature of CHM represents an excellent example of systems medicine in the effective treatment of depression.
Collapse
|
10
|
Lottering B, Lin YW. Functional characterization of nociceptive mechanisms involved in fibromyalgia and electroacupuncture. Brain Res 2021; 1755:147260. [PMID: 33422528 DOI: 10.1016/j.brainres.2020.147260] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 11/04/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022]
Abstract
The diagnosis and treatment of chronic pain in diseases such as fibromyalgia (FM) are lacking effective standardised protocols that can be widely accessed and implemented by healthcare professionals across the globe. Persistent hyperalgesia and allodynia are characteristic symptoms of FM. This disease has indicated a refractory tendency to conventional treatment ventures, largely resultant from a lack of etiological and pathogenic understanding of the disease development. Emerging evidence indicates that the central nervous system (CNS) plays a critical role in the amplification of pain signals and the neurotransmitters associated therewith. We examined the contribution of the transient receptor potential vanilloid 1 (TRPV1) channel and the major nociceptive components in response to fibromyalgia-like pain in an intermittent cold-stress (ICS) model, in the prefrontal cortex, somatosensory cortex, hippocampus and thalamus areas of the brain. The use of TRPV1 gene deletion mice served to elucidate the role of the TRPV1 receptor in the development and expression of FM-like pain. The results suggest that TRPV1 upregulation is central to the sustained sensation of FM related hyperalgesia. Furthermore, the potential therapeutic benefits of electroacupuncture (EA) at bilateral ST36 acupoint were analysed in order to identify the analgesic effects and mechanism associated with this therapy. The findings indicate that EA treatment successfully attenuated both mechanical and thermal hyperalgesia and suggests that a definitive underlying mechanism of neuromodulation through EA is responsible for providing analgesic benefits to patients suffering from FM.
Collapse
Affiliation(s)
- Bernice Lottering
- College of Chinese Medicine, Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | - Yi-Wen Lin
- College of Chinese Medicine, Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan; Chinese Medicine Research Centre, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
11
|
Lin YW, Chou AIW, Su H, Su KP. Transient receptor potential V1 (TRPV1) modulates the therapeutic effects for comorbidity of pain and depression: The common molecular implication for electroacupuncture and omega-3 polyunsaturated fatty acids. Brain Behav Immun 2020; 89:604-614. [PMID: 32640285 DOI: 10.1016/j.bbi.2020.06.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 01/02/2023] Open
Abstract
Chronic pain and depression are conditions that are highly comorbid and present with overlapping clinical presentations and common pathological biological pathways in neuroinflammation, both of which can be reversed by the use of electroacupuncture (EA) and omega-3 polyunsaturated fatty acids (PUFAs). Transient receptor potential V1 (TRPV1), a Ca2+ permeable ion channel that can be activated by inflammation, is reported to be involved in the development of chronic pain and depression. Here, we investigated the role of TRPV1 and its related pathways in the murine models of cold stress-induced nociception and depression. Female C57BL/6 wild type and TRPV1 knockout mice were subjected to intermittent cold-stress (ICS) to initiate depressive-like and chronic pain behaviors, respectively. The Bio-Plex ELISA technique was utilized to analyze inflammatory mediators in mice plasma. The western blot and immunostaining techniques were used to analyze the presence of TRPV1 and related molecules in the medial prefrontal cortex (mPFC), hippocampus, periaqueductal gray (PAG), and amygdala. The ICS model significantly induced chronic pain (mechanical: 2.55 ± 0.31 g; thermal: 8.12 ± 0.87 s) and depressive-like behaviors (10.95 ± 0.95% in the center zone; 53.14 ± 4.01% in immobility). The treatment efficacy of EA, docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA) were observed in both nociceptive and depression test results. Inflammatory mediators were increased after ICS induction and further reversed by the use of EA, EPA and DHA. A majority of TRPV1 proteins and related molecules were significantly decreased in the mPFC, hippocampus and PAG of mice. This decrease can be reversed by the use of EA, EPA and DHA. In contrast, these molecules were increased in the mice's amygdala, and were attenuated by the use of EA, EPA and DHA. Our findings indicate that these inflammatory mediators can regulate the TRPV1 signaling pathway and initiate new potential therapeutic targets for chronic pain and depression treatment.
Collapse
Affiliation(s)
- Yi-Wen Lin
- College of Chinese Medicine, Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
| | - Ana Isabel Wu Chou
- Department of Psychiatry, Taipei Medical University-Wan Fang Medical Center, Taipei, Taiwan
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Kuan-Pin Su
- College of Medicine, China Medical University, Taichung, Taiwan; Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; An-Nan Hospital, China Medical University, Tainan, Taiwan.
| |
Collapse
|
12
|
Ronchetti S, Gentili M, Ricci E, Migliorati G, Riccardi C. Glucocorticoid-Induced Leucine Zipper as a Druggable Target in Inflammatory Bowel Diseases. Inflamm Bowel Dis 2020; 26:1017-1025. [PMID: 31961437 DOI: 10.1093/ibd/izz331] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Indexed: 12/11/2022]
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders with a complex pathogenesis, affecting people of all ages. They are characterized by alternating phases of clinical relapse and remission, depending on the fine balance between immune cells and the gut microbiota. The cross talk between cells of the immune system and the gut microbiota can result in either tolerance or inflammation, according to multifactorial triggers, ranging from environmental factors to genetic susceptibility. Glucocorticoid (GC) administration remains the first-line treatment for IBDs, although long-term use is limited by development of serious adverse effects. Recently, new alternative pharmacological therapies have been developed, although these are not always effective in IBD patients. There is a constant demand for effective new drug targets to guarantee total remission and improve the quality of life for IBD patients. The glucocorticoid-induced leucine zipper (GILZ) has been implicated as a promising candidate for this purpose, in view of its powerful anti-inflammatory effects that mimic those of GCs while avoiding their unwanted adverse reactions. Here we present and discuss the latest findings about the involvement of GILZ in IBDs.
Collapse
Affiliation(s)
- Simona Ronchetti
- Department of Medicine, Pharmacology Division, University of Perugia, Italy
| | - Marco Gentili
- Department of Medicine, Pharmacology Division, University of Perugia, Italy
| | - Erika Ricci
- Department of Medicine, Pharmacology Division, University of Perugia, Italy
| | | | - Carlo Riccardi
- Department of Medicine, Pharmacology Division, University of Perugia, Italy
| |
Collapse
|
13
|
Ng HP, Jennings S, Nelson S, Wang G. Short-Chain Alcohols Upregulate GILZ Gene Expression and Attenuate LPS-Induced Septic Immune Response. Front Immunol 2020; 11:53. [PMID: 32117233 PMCID: PMC7008712 DOI: 10.3389/fimmu.2020.00053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/09/2020] [Indexed: 12/16/2022] Open
Abstract
Alcohol differentially affects human health, depending on the pattern of exposure. Moderate intake provides beneficial mood modulation and an anti-inflammatory effect, while excessive consumption leads to immunosuppression and various alcohol use disorders. The mechanism underlying this bi-phasic action mode of alcohol has not been clearly defined. Our previous publication demonstrated that ethanol, in the absence of glucocorticoids (GCs), induces expression of Glucocorticoid-Induced Leucine Zipper (GILZ), a key molecule that transduces GC anti-inflammatory effect through a non-canonical activation of glucocorticoid receptor (1). Here we report that similar short-chain alcohols, such as ethanol, propanol and isopropanol, share the same property of upregulating GILZ gene expression, and blunt cell inflammatory response in vitro. When mice were exposed to these alcohols, GILZ gene expression in immune cells was augmented in a dose-dependent manner. Monocytes and neutrophils were most affected. The short-chain alcohols suppressed host inflammatory response to lipopolysaccharide (LPS) and significantly reduced LPS-induced mortality. Intriguingly, propanol and isopropanol displayed more potent protection than ethanol at the same dose. Inhibition of ethanol metabolism enhanced the ethanol protective effect, suggesting that it is ethanol, not its derivatives or metabolites, that induces immune suppression. Taken together, short-chain alcohols per se upregulate GILZ gene expression and provide immune protection against LPS toxicity, suggesting a potential measure to counter LPS septic shock in a resource limited situation.
Collapse
Affiliation(s)
- Hang Pong Ng
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Scott Jennings
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Steve Nelson
- Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Guoshun Wang
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
14
|
Does Maternal Stress Affect the Early Embryonic Microenvironment? Impact of Long-Term Cortisol Stimulation on the Oviduct Epithelium. Int J Mol Sci 2020; 21:ijms21020443. [PMID: 32284519 PMCID: PMC7014231 DOI: 10.3390/ijms21020443] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 01/08/2020] [Indexed: 12/21/2022] Open
Abstract
Maternal stress before or during the sensitive preimplantation phase is associated with reproduction failure. Upon real or perceived threat, glucocorticoids (classic stress hormones) as cortisol are synthesized. The earliest “microenvironment” of the embryo consists of the oviduct epithelium and the oviductal fluid generated via the epithelial barrier. However, to date, the direct effects of cortisol on the oviduct are largely unknown. In the present study, we used a compartmentalized in vitro system to test the hypothesis that a prolonged stimulation with cortisol modifies the physiology of the oviduct epithelium. Porcine oviduct epithelial cells were differentiated at the air–liquid interface and basolaterally stimulated with physiological levels of cortisol representing moderate and severe stress for 21 days. Epithelium structure, transepithelial bioelectric properties, and gene expression were assessed. Furthermore, the distribution and metabolism of cortisol was examined. The polarized oviduct epithelium converted basolateral cortisol to cortisone and thereby reduced the amount of bioactive cortisol reaching the apical compartment. However, extended cortisol stimulation affected its barrier function and the expression of genes involved in hormone signaling and immune response. We conclude that continuing maternal stress with long-term elevated cortisol levels may alter the early embryonic environment by modification of basic oviductal functions.
Collapse
|
15
|
Cultured hippocampal neurons of dystrophic mdx mice respond differently from those of wild type mice to an acute treatment with corticosterone. Exp Cell Res 2020; 386:111715. [PMID: 31711918 DOI: 10.1016/j.yexcr.2019.111715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 12/27/2022]
Abstract
Duchenne muscular dystrophy is a lethal genetic disease characterised by progressive degeneration of skeletal muscles induced by deficiency of dystrophin, a cytoskeletal protein expressed in myocytes and in certain neuron populations. The severity of the neurological disorder varies in humans and animal models owing to dysfunction in numerous brain areas, including the hippocampus. Cyclic treatments with high-dose glucocorticoids remain a major pharmacological approach for treating the disease; however, elevated systemic levels of either stress-induced or exogenously administered anti-inflammatory molecules dramatically affect hippocampal activity. In this study, we analysed and compared the response of hippocampal neurons isolated from wild-type and dystrophic mdx mice to acute administration of corticosterone in vitro, without the influence of other glucocorticoid-regulated processes. Our results showed that in neurons of mdx mice, both the genomic and intracellular signalling-mediated responses to corticosterone were affected compared to those in wild-type animals, evoking the characteristic response to detrimental chronic glucocorticoid exposure. Responsiveness to glucocorticoids is, therefore, another function of hippocampal neurons possibly affected by deficiency of Dp427 since embryonic development. Knowing the pivotal role of hippocampus in stress hormone signalling, attention should be paid to the effects that prolonged glucocorticoid treatments may have on this and other brain areas of DMD patients.
Collapse
|
16
|
Luo Y, Yang M, Guo M, Zhong X, Hu Y. Huang Qin Hua Shi decoction for high-temperature- and high-humidity-induced cognitive-behavioral disorder in rats is associated with deactivation of the hypothalamic-pituitary-adrenal axis. J Int Med Res 2019; 47:5752-5766. [PMID: 31547741 PMCID: PMC6862911 DOI: 10.1177/0300060519875369] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objective To investigate the effects of Huang Qin Hua Shi (HQ) decoction on the hypothalamic–pituitary–adrenal (HPA) axis in rats under high-temperature (hT)- and high-humidity (hH)-induced stress. Methods Male rats were randomized into four groups: rats without stress; rats induced with hT (35 ± 1°C) and hH (85 ± 5% humidity); rats induced with hT and hH and treated with HQ decoction; and rats induced with hT and hH and treated with mifepristone. After 3 weeks, rats underwent the Morris water maze and open-field test. Rat hypothalami were analyzed pathologically using hematoxylin and eosin staining and glucocorticoid receptor (GR) mRNA expression was evaluated by in situ hybridization. Serum levels of corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), and corticosteroid (CORT) were assessed by enzyme-linked immunosorbent assay. Results The administration of mifepristone or HQ in stressed rats significantly improved their performance in the Morris water maze test and increased the central-to-peripheral ratio and incidence of deep rearing in the open-field test. Mifepristone and HQ also reversed histological changes in the hypothalami of stressed rats. Compared with control rats, GR mRNA expression in the hypothalamus and serum CRH, ACTH, and CORT were significantly elevated in rats stressed with hT and hH, and these changes were attenuated by mifepristone and HQ. Conclusion HQ decoction protects against hT- and hH-induced cognitive-behavioral disorder and its therapeutic effect is associated with decreased HPA axis activity.
Collapse
Affiliation(s)
- Yong Luo
- Graduate College, Naval Medical University, Shanghai, China.,Traditional Chinese Medicine (TCM) Department, the General Hospital of Western Theater Command PLA, Chengdu, China
| | - Min Yang
- Traditional Chinese Medicine (TCM) Department, the General Hospital of Western Theater Command PLA, Chengdu, China
| | - Mingyang Guo
- Traditional Chinese Medicine (TCM) Department, the General Hospital of Western Theater Command PLA, Chengdu, China
| | - Xiaolong Zhong
- Traditional Chinese Medicine (TCM) Department, the General Hospital of Western Theater Command PLA, Chengdu, China
| | - Yonghe Hu
- Traditional Chinese Medicine (TCM) Department, the General Hospital of Western Theater Command PLA, Chengdu, China
| |
Collapse
|
17
|
Lebow MA, Schroeder M, Tsoory M, Holzman-Karniel D, Mehta D, Ben-Dor S, Gil S, Bradley B, Smith AK, Jovanovic T, Ressler KJ, Binder EB, Chen A. Glucocorticoid-induced leucine zipper "quantifies" stressors and increases male susceptibility to PTSD. Transl Psychiatry 2019; 9:178. [PMID: 31346158 PMCID: PMC6658561 DOI: 10.1038/s41398-019-0509-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/24/2019] [Indexed: 12/04/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) selectively develops in some individuals exposed to a traumatic event. Genetic and epigenetic changes in glucocorticoid pathway sensitivity may be essential for understanding individual susceptibility to PTSD. This study focuses on PTSD markers in the glucocorticoid pathway, spotlighting glucocorticoid-induced leucine zipper (GILZ), a transcription factor encoded by the gene Tsc22d3 on the X chromosome. We propose that GILZ uniquely "quantifies" exposure to stressors experienced from late gestation to adulthood and that low levels of GILZ predispose individuals to PTSD in males only. GILZ mRNA and methylation were measured in 396 male and female human blood samples from the Grady Trauma Project cohort (exposed to multiple traumatic events). In mice, changes in glucocorticoid pathway genes were assessed following exposure to stressors at distinct time points: (i) CRF-induced prenatal stress (CRF-inducedPNS) with, or without, additional exposure to (ii) PTSD induction protocol in adulthood, which induces PTSD-like behaviors in a subset of mice. In humans, the number of traumatic events correlated negatively with GILZ mRNA levels and positively with % methylation of GILZ in males only. In male mice, we observed a threefold increase in the number of offspring exhibiting PTSD-like behaviors in those exposed to both CRF-inducedPNS and PTSD induction. This susceptibility was associated with reduced GILZ mRNA levels and epigenetic changes, not found in females. Furthermore, virus-mediated shRNA knockdown of amygdalar GILZ increased susceptibility to PTSD. Mouse and human data confirm that dramatic alterations in GILZ occur in those exposed to a stressor in early life, adulthood or both. Therefore, GILZ levels may help identify at-risk populations for PTSD prior to additional traumatic exposures.
Collapse
Affiliation(s)
- Maya A. Lebow
- 0000 0004 0604 7563grid.13992.30Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel ,0000 0000 9497 5095grid.419548.5Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Mariana Schroeder
- 0000 0004 0604 7563grid.13992.30Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel ,0000 0000 9497 5095grid.419548.5Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Michael Tsoory
- 0000 0004 0604 7563grid.13992.30Department of Veterinary Resources, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Dorin Holzman-Karniel
- 0000 0004 0604 7563grid.13992.30Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Divya Mehta
- 0000 0000 9497 5095grid.419548.5Department of Translational Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Shifra Ben-Dor
- 0000 0004 0604 7563grid.13992.30Department of Biological Services, Bioinformatics and Biological Computing Unit, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Shosh Gil
- 0000 0004 0604 7563grid.13992.30Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Bekh Bradley
- 0000 0004 0419 4084grid.414026.5Atlanta Veterans Affairs Medical Center, Decatur, GA USA ,0000 0001 0941 6502grid.189967.8Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Alicia K. Smith
- 0000 0001 0941 6502grid.189967.8Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Tanja Jovanovic
- 0000 0001 0941 6502grid.189967.8Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Kerry J. Ressler
- 0000 0001 0941 6502grid.189967.8Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Elisabeth B. Binder
- 0000 0000 9497 5095grid.419548.5Department of Translational Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany ,0000 0001 0941 6502grid.189967.8Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, 76100, Rehovot, Israel. .,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804, Munich, Germany.
| |
Collapse
|
18
|
Peripheral blood GILZ mRNA levels in depression and following electroconvulsive therapy. Psychoneuroendocrinology 2019; 101:304-310. [PMID: 30602137 DOI: 10.1016/j.psyneuen.2018.12.234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/20/2018] [Accepted: 12/22/2018] [Indexed: 12/12/2022]
Abstract
Dysregulation of the hypothalamic-pituitary-adrenocortical (HPA)-axis is commonly observed in patients with depression. The delayed feedback system that mediates inhibition of HPA-axis activation is regulated by glucocorticoid receptors (GRs) found in stress-responsive areas of the brain. Glucocorticoid-induced leucine zipper (GILZ) is a key molecule in glucocorticoid biology and is thought to mediate the downstream anti-inflammatory effects of GRs. Previous reports suggest that GILZ levels are altered in the blood and brains of patients with, and animal models of, depression. However, no study has yet investigated the effects of antidepressant treatment on GILZ. Therefore, our aim was to examine peripheral blood GILZ mRNA levels in patients with depression (n = 88) compared to age- and sex-matched healthy controls (n = 63), and in patients with depression following treatment with a course of electroconvulsive therapy (ECT). We also assessed the relationship between GILZ and mood and clinical outcomes following ECT. GILZ mRNA levels were assessed using qRT-PCR. GILZ levels were found to be significantly lower in patients with depression compared to controls (p < 0.002), and ECT further decreased GILZ levels (p = 0.05). Both of these results survived adjustment for potential covariates. However, we found no association between GILZ and mood scores. Overall, these results suggest that GILZ is involved in the pathophysiology of depression and the peripheral molecular response to ECT.
Collapse
|
19
|
Papadopoulou Z, Vlaikou AM, Theodoridou D, Markopoulos GS, Tsoni K, Agakidou E, Drosou-Agakidou V, Turck CW, Filiou MD, Syrrou M. Stressful Newborn Memories: Pre-Conceptual, In Utero, and Postnatal Events. Front Psychiatry 2019; 10:220. [PMID: 31057437 PMCID: PMC6482218 DOI: 10.3389/fpsyt.2019.00220] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 03/26/2019] [Indexed: 12/15/2022] Open
Abstract
Early-life stressful experiences are critical for plasticity and development, shaping adult neuroendocrine response and future health. Stress response is mediated by the autonomous nervous system and the hypothalamic-pituitary-adrenal (HPA) axis while various environmental stimuli are encoded via epigenetic marks. The stress response system maintains homeostasis by regulating adaptation to the environmental changes. Pre-conceptual and in utero stressors form the fetal epigenetic profile together with the individual genetic profile, providing the background for individual stress response, vulnerability, or resilience. Postnatal and adult stressful experiences may act as the definitive switch. This review addresses the issue of how preconceptual in utero and postnatal events, together with individual differences, shape future stress responses. Putative markers of early-life adverse effects such as prematurity and low birth weight are emphasized, and the epigenetic, mitochondrial, and genomic architecture regulation of such events are discussed.
Collapse
Affiliation(s)
- Zoe Papadopoulou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Angeliki-Maria Vlaikou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece.,Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Daniela Theodoridou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Georgios S Markopoulos
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Konstantina Tsoni
- 1st Department of Neonatology and Neonatal Intensive Care Unit, Medical Faculty, Aristotle University School of Health Sciences, Thessaloniki, Greece
| | - Eleni Agakidou
- 1st Department of Neonatology and Neonatal Intensive Care Unit, Medical Faculty, Aristotle University School of Health Sciences, Thessaloniki, Greece
| | - Vasiliki Drosou-Agakidou
- 1st Department of Neonatology and Neonatal Intensive Care Unit, Medical Faculty, Aristotle University School of Health Sciences, Thessaloniki, Greece
| | | | - Michaela D Filiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece.,Max Planck Institute of Psychiatry, Munich, Germany
| | - Maria Syrrou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| |
Collapse
|
20
|
Morris G, Reiche EMV, Murru A, Carvalho AF, Maes M, Berk M, Puri BK. Multiple Immune-Inflammatory and Oxidative and Nitrosative Stress Pathways Explain the Frequent Presence of Depression in Multiple Sclerosis. Mol Neurobiol 2018; 55:6282-6306. [PMID: 29294244 PMCID: PMC6061180 DOI: 10.1007/s12035-017-0843-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022]
Abstract
Patients with a diagnosis of multiple sclerosis (MS) or major depressive disorder (MDD) share a wide array of biological abnormalities which are increasingly considered to play a contributory role in the pathogenesis and pathophysiology of both illnesses. Shared abnormalities include peripheral inflammation, neuroinflammation, chronic oxidative and nitrosative stress, mitochondrial dysfunction, gut dysbiosis, increased intestinal barrier permeability with bacterial translocation into the systemic circulation, neuroendocrine abnormalities and microglial pathology. Patients with MS and MDD also display a wide range of neuroimaging abnormalities and patients with MS who display symptoms of depression present with different neuroimaging profiles compared with MS patients who are depression-free. The precise details of such pathology are markedly different however. The recruitment of activated encephalitogenic Th17 T cells and subsequent bidirectional interaction leading to classically activated microglia is now considered to lie at the core of MS-specific pathology. The presence of activated microglia is common to both illnesses although the pattern of such action throughout the brain appears to be different. Upregulation of miRNAs also appears to be involved in microglial neurotoxicity and indeed T cell pathology in MS but does not appear to play a major role in MDD. It is suggested that the antidepressant lofepramine, and in particular its active metabolite desipramine, may be beneficial not only for depressive symptomatology but also for the neurological symptoms of MS. One clinical trial has been carried out thus far with, in particular, promising MRI findings.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, Geelong, Australia
| | - Edna Maria Vissoci Reiche
- Department of Pathology, Clinical Analysis, and Toxicology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Andrea Murru
- Bipolar Disorders Program, Hospital Clínic Barcelona, IDIBAPS, CIBERSAM, Barcelona, Spain
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, Geelong, Australia
- Department of Psychiatry, Medical University Plovdiv, Plovdiv, Bulgaria
- Department of Psychiatry, Faculty of Medicine, State University of Londrina, Londrina, Brazil
- Revitalis, Waalre, The Netherlands
- Orygen - The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Basant K Puri
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK.
| |
Collapse
|
21
|
Volk N, Pape JC, Engel M, Zannas AS, Cattane N, Cattaneo A, Binder EB, Chen A. Amygdalar MicroRNA-15a Is Essential for Coping with Chronic Stress. Cell Rep 2017; 17:1882-1891. [PMID: 27829158 PMCID: PMC5120368 DOI: 10.1016/j.celrep.2016.10.038] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 04/29/2016] [Accepted: 10/13/2016] [Indexed: 01/08/2023] Open
Abstract
MicroRNAs are important regulators of gene expression and associated with stress-related psychiatric disorders. Here, we report that exposing mice to chronic stress led to a specific increase in microRNA-15a levels in the amygdala-Ago2 complex and a concomitant reduction in the levels of its predicted target, FKBP51, which is implicated in stress-related psychiatric disorders. Reciprocally, mice expressing reduced levels of amygdalar microRNA-15a following exposure to chronic stress exhibited increased anxiety-like behaviors. In humans, pharmacological activation of the glucocorticoid receptor, as well as exposure to childhood trauma, was associated with increased microRNA-15a levels in peripheral blood. Taken together, our results support an important role for microRNA-15a in stress adaptation and the pathogenesis of stress-related psychopathologies. miR-15a levels are elevated in the amygdala-Ago2 complex following chronic stress miR-15a targets FKBP51 and affects behavioral responses to stressful challenges miR-15a is elevated in peripheral human blood following dexamethasone exposure miR-15a is elevated in peripheral human blood of patients exposed to childhood trauma
Collapse
Affiliation(s)
- Naama Volk
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Julius C Pape
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Mareen Engel
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Anthony S Zannas
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Nadia Cattane
- Biological Psychiatry Unit, IRCCS Fatebenefratelli, University of Brescia, 25121 Brescia, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Fatebenefratelli, University of Brescia, 25121 Brescia, Italy; Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London WC2R 2LS, UK
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany.
| |
Collapse
|
22
|
Dexamethasone-induced cell death is restricted to specific molecular subgroups of multiple myeloma. Oncotarget 2016; 6:26922-34. [PMID: 26323097 PMCID: PMC4694963 DOI: 10.18632/oncotarget.4616] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/06/2015] [Indexed: 12/12/2022] Open
Abstract
Due to its cytotoxic effect in lymphoid cells, dexamethasone is widely used in the treatment of multiple myeloma (MM). However, only a subset of myeloma patients responds to high-dose dexamethasone. Despite the undeniable anti-myeloma benefits of dexamethasone, significant adverse effects have been reported. We re-evaluate the anti-tumor effect of dexamethasone according to the molecular heterogeneity of MM. We demonstrated that the pro-death effect of dexamethasone is related to the genetic heterogeneity of MM because sensitive cell lines were restricted to MAF and MMSET signature subgroups, whereas all CCND1 cell lines (n = 10) were resistant to dexamethasone. We demonstrated that the glucocorticoid receptor expression was an important limiting factor for dexamethasone-induced cell death and we found a correlation between glucocorticoid receptor levels and the induction of glucocorticoid-induced leucine zipper (GILZ) under dexamethasone treatment. By silencing GILZ, we next demonstrated that GILZ is necessary for Dex induced apoptosis while triggering an imbalance between anti- and pro-apoptotic Bcl-2 proteins. Finally, the heterogeneity of the dexamethasone response was further confirmed in vivo using myeloma xenograft models. Our findings suggested that the effect of dexamethasone should be re-evaluated within molecular subgroups of myeloma patients to improve its efficacy and reduce its adverse effects.
Collapse
|
23
|
Young JJ, Silber T, Bruno D, Galatzer-Levy IR, Pomara N, Marmar CR. Is there Progress? An Overview of Selecting Biomarker Candidates for Major Depressive Disorder. Front Psychiatry 2016; 7:72. [PMID: 27199779 PMCID: PMC4843170 DOI: 10.3389/fpsyt.2016.00072] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/11/2016] [Indexed: 01/01/2023] Open
Abstract
Major depressive disorder (MDD) contributes to a significant worldwide disease burden, expected to be second only to heart disease by 2050. However, accurate diagnosis has been a historical weakness in clinical psychiatry. As a result, there is a demand for diagnostic modalities with greater objectivity that could improve on current psychiatric practice that relies mainly on self-reporting of symptoms and clinical interviews. Over the past two decades, literature on a growing number of putative biomarkers for MDD increasingly suggests that MDD patients have significantly different biological profiles compared to healthy controls. However, difficulty in elucidating their exact relationships within depression pathology renders individual markers inconsistent diagnostic tools. Consequently, further biomarker research could potentially improve our understanding of MDD pathophysiology as well as aid in interpreting response to treatment, narrow differential diagnoses, and help refine current MDD criteria. Representative of this, multiplex assays using multiple sources of biomarkers are reported to be more accurate options in comparison to individual markers that exhibit lower specificity and sensitivity, and are more prone to confounding factors. In the future, more sophisticated multiplex assays may hold promise for use in screening and diagnosing depression and determining clinical severity as an advance over relying solely on current subjective diagnostic criteria. A pervasive limitation in existing research is heterogeneity inherent in MDD studies, which impacts the validity of biomarker data. Additionally, small sample sizes of most studies limit statistical power. Yet, as the RDoC project evolves to decrease these limitations, and stronger studies with more generalizable data are developed, significant advances in the next decade are expected to yield important information in the development of MDD biomarkers for use in clinical settings.
Collapse
Affiliation(s)
- Juan Joseph Young
- Nathan Kline Institute, Orangeburg, NY, USA; Case Western Reserve University, Cleveland, OH, USA; MetroHealth Medical Center, Cleveland, OH, USA
| | - Tim Silber
- Nathan Kline Institute , Orangeburg, NY , USA
| | - Davide Bruno
- Liverpool John Moores University , Liverpool , UK
| | | | - Nunzio Pomara
- Nathan Kline Institute, Orangeburg, NY, USA; New York University School of Medicine, New York, NY, USA; NYU Cohen Veterans Center, New York, NY, USA
| | - Charles Raymond Marmar
- New York University School of Medicine, New York, NY, USA; NYU Cohen Veterans Center, New York, NY, USA
| |
Collapse
|
24
|
Nikolopoulou E, Mytilinaios D, Calogero AE, Kamilaris TC, Troupis T, Chrousos GP, Johnson EO. Modulation of central glucocorticoid receptors in short- and long-term experimental hyperthyroidism. Endocrine 2015; 49:828-41. [PMID: 25722011 DOI: 10.1007/s12020-015-0528-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/10/2015] [Indexed: 01/25/2023]
Abstract
Hyperthyroidism is associated with a significant increase in circulating glucocorticoid levels and hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis. The aim of this study was to examine whether the HPA axis hyperactivity observed in hyperthyroidism may be explained by a disturbed feedback inhibition of endogenous glucocorticoids through two specific intracellular receptors in the brain: the high affinity mineralocorticoid receptor (MR) and the lower affinity glucocorticoid receptor (GR). Cytosolic receptor binding and gene expression was assessed in rats with short (7 days) and long standing (60 days) eu- and hyperthyroidism. Glucocorticoid receptor number and binding affinity (Kd) in the hippocampus were measured using [(3)H2]-dexamethasone radioreceptor assay. In situ hybridization was employed to examine the effects of hyperthyroidism on the GR and MR mRNA levels in the hippocampus and the pituitary. Both short- and long-term hyperthyroid rats showed pronounced reduction in the concentration of cytosolic GR in the hippocampus, without changes in binding affinity or changes in GR expression. In contrast, GR mRNA in the pituitary increased after 7 days and decreased after 60 days of thyroxin treatment. MR mRNA was moderately affected. Hyperthyroidism is associated with significant decreases in hippocampal GR levels supporting the hypothesis that hyperactivity of the HPA axis observed in experimentally induced hyperthyroidism may be attributed, at least in part, to decreased negative feedback at the level of the hippocampus. These findings further support the notion that a central locus is principally responsible for the hyperactivity of the HPA axis observed in hyperthyroidism.
Collapse
Affiliation(s)
- Elena Nikolopoulou
- Department of Anatomy, School of Medicine, University of Athens, 75 Mikras Asias Str., Goudi, 11572, Athens, Greece
| | | | | | | | | | | | | |
Collapse
|