1
|
Kwon D, Kim Y, Cho SH. Antidepressant Effects of Ginsenoside Rc on L-Alpha-Aminoadipic Acid-Induced Astrocytic Ablation and Neuroinflammation in Mice. Int J Mol Sci 2024; 25:9673. [PMID: 39273621 PMCID: PMC11396248 DOI: 10.3390/ijms25179673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Depression is a prevalent and debilitating mental disorder that affects millions worldwide. Current treatments, such as antidepressants targeting the serotonergic system, have limitations, including delayed onset of action and high rates of treatment resistance, necessitating novel therapeutic strategies. Ginsenoside Rc (G-Rc) has shown potential anti-inflammatory and neuroprotective effects, but its antidepressant properties remain unexplored. This study investigated the antidepressant effects of G-Rc in an L-alpha-aminoadipic acid (L-AAA)-induced mouse model of depression, which mimics the astrocytic pathology and neuroinflammation observed in major depressive disorder. Mice were administered G-Rc, vehicle, or imipramine orally after L-AAA injection into the prefrontal cortex. G-Rc significantly reduced the immobility time in forced swimming and tail suspension tests compared to vehicle treatment, with more pronounced effects than imipramine. It also attenuated the expression of pro-inflammatory cytokines (TNF-α, IL-6, TGF-β, lipocalin-2) and alleviated astrocytic degeneration, as indicated by increased GFAP and decreased IBA-1 levels. Additionally, G-Rc modulated apoptosis-related proteins, decreasing caspase-3 and increasing Bcl-2 levels compared to the L-AAA-treated group. These findings suggest that G-Rc exerts antidepressant effects by regulating neuroinflammation, astrocyte-microglia crosstalk, and apoptotic pathways in the prefrontal cortex, highlighting its potential as a novel therapeutic agent for depression.
Collapse
Affiliation(s)
- Dohyung Kwon
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yunna Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Neuropsychiatry of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung-Hun Cho
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Neuropsychiatry of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
2
|
Li Y, Luo Y, Zhu P, Liang X, Li J, Dou X, Liu L, Qin L, Zhou M, Deng Y, Jiang L, Wang S, Yang W, Tang J, Tang Y. Running exercise improves astrocyte loss, morphological complexity and astrocyte-contacted synapses in the hippocampus of CUS-induced depression model mice. Pharmacol Biochem Behav 2024; 239:173750. [PMID: 38494007 DOI: 10.1016/j.pbb.2024.173750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Although the antidepressant effects of running exercise have been widely reported, further research is still needed to determine the structural bases for these effects. Astrocyte processes physically contact many synapses and directly regulate the numbers of synapses, but it remains unclear whether running exercise can modulate astrocyte morphological complexity and astrocyte-contacted synapses in the hippocampus of the mice with depressive-like behavior. Male C57BL/6 J mice underwent four weeks of running exercise after four weeks of chronic unpredictable stress (CUS). The sucrose preference test (SPT), tail suspension test (TST) and forced swim test (FST) were used to assess anhedonia in mice. Western blotting was used to measure the expression of astrocyte- and synapse-related proteins. Immunofluorescence and 3D reconstruction were used to quantify the density and morphology of astrocytes, and astrocyte-contacted synapses in each hippocampal subregion. Four weeks of running exercise alleviated depressive-like symptoms in mice. The expression of astrocyte- and synapse-related proteins in the hippocampus; astrocyte process lengths, process numbers, and dendritic arborization; and the number of astrocyte-contacted PSD95 positive synapses in the CA2-3 and DG regions were significantly decreased in the mice with depressive-like behavior, and running exercise successfully reserved these changes. Running exercise improved the decreases in astrocyte morphological complexity and astrocyte-contacted PSD95 positive synapses in the CA2-3 and DG regions of the mice with depressive-like behavior, suggesting that the physical interactions between astrocytes and synapses can be increased by running exercise, which might be an important structural basis for the antidepressant effects of running exercise.
Collapse
Affiliation(s)
- Yue Li
- Department of Histology and Embryology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Yanmin Luo
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Peilin Zhu
- Department of Histology and Embryology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Xin Liang
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Department of Pathology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Li
- Department of Histology and Embryology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Xiaoyun Dou
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Li Liu
- Department of Histology and Embryology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Lu Qin
- Department of Histology and Embryology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Mei Zhou
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Yuhui Deng
- Department of Histology and Embryology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Jiang
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Shun Wang
- Department of Histology and Embryology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Wenyu Yang
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Tang
- Department of Histology and Embryology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China.
| | - Yong Tang
- Department of Histology and Embryology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
3
|
Lv XJ, Lv SS, Wang GH, Chang Y, Cai YQ, Liu HZ, Xu GZ, Xu WD, Zhang YQ. Glia-derived adenosine in the ventral hippocampus drives pain-related anxiodepression in a mouse model resembling trigeminal neuralgia. Brain Behav Immun 2024; 117:224-241. [PMID: 38244946 DOI: 10.1016/j.bbi.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/11/2023] [Accepted: 01/14/2024] [Indexed: 01/22/2024] Open
Abstract
Glial activation and dysregulation of adenosine triphosphate (ATP)/adenosine are involved in the neuropathology of several neuropsychiatric illnesses. The ventral hippocampus (vHPC) has attracted considerable attention in relation to its role in emotional regulation. However, it is not yet clear how vHPC glia and their derived adenosine regulate the anxiodepressive-like consequences of chronic pain. Here, we report that chronic cheek pain elevates vHPC extracellular ATP/adenosine in a mouse model resembling trigeminal neuralgia (rTN), which mediates pain-related anxiodepression, through a mechanism that involves synergistic effects of astrocytes and microglia. We found that rTN resulted in robust activation of astrocytes and microglia in the CA1 area of the vHPC (vCA1). Genetic or pharmacological inhibition of astrocytes and connexin 43, a hemichannel mainly distributed in astrocytes, completely attenuated rTN-induced extracellular ATP/adenosine elevation and anxiodepressive-like behaviors. Moreover, inhibiting microglia and CD39, an enzyme primarily expressed in microglia that degrades ATP into adenosine, significantly suppressed the increase in extracellular adenosine and anxiodepressive-like behaviors. Blockade of the adenosine A2A receptor (A2AR) alleviated rTN-induced anxiodepressive-like behaviors. Furthermore, interleukin (IL)-17A, a pro-inflammatory cytokine probably released by activated microglia, markedly increased intracellular calcium in vCA1 astrocytes and triggered ATP/adenosine release. The astrocytic metabolic inhibitor fluorocitrate and the CD39 inhibitor ARL 67156, attenuated IL-17A-induced increases in extracellular ATP and adenosine, respectively. In addition, astrocytes, microglia, CD39, and A2AR inhibitors all reversed rTN-induced hyperexcitability of pyramidal neurons in the vCA1. Taken together, these findings suggest that activation of astrocytes and microglia in the vCA1 increases extracellular adenosine, which leads to pain-related anxiodepression via A2AR activation. Approaches targeting astrocytes, microglia, and adenosine signaling may serve as novel therapies for pain-related anxiety and depression.
Collapse
Affiliation(s)
- Xue-Jing Lv
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Su-Su Lv
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Guo-Hong Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yue Chang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ya-Qi Cai
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Hui-Zhu Liu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Guang-Zhou Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200023, China.
| | - Wen-Dong Xu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Yu-Qiu Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Lei L, Wang YT, Hu D, Gai C, Zhang Y. Astroglial Connexin 43-Mediated Gap Junctions and Hemichannels: Potential Antidepressant Mechanisms and the Link to Neuroinflammation. Cell Mol Neurobiol 2023; 43:4023-4040. [PMID: 37875763 DOI: 10.1007/s10571-023-01426-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 10/14/2023] [Indexed: 10/26/2023]
Abstract
Major depression disorder (MDD) is a neuropsychiatric disorder associated with a high suicide rate and a higher disability rate than any other disease. Evidence suggests that the pathological mechanism of MDD is related to astrocyte dysfunction. Depression is mainly associated with the expression of connexin 43 (Cx43) and the function of Cx43-mediated gap junctions and hemichannels in astrocytes. Moreover, neuroinflammation has been a hotspot in research on the pathology of depression, and Cx43-mediated functions are thought to be involved in neuroinflammation-related depression. However, the specific mechanism of Cx43-mediated functions in neuroinflammation-related depression pathology remains unclear. Therefore, this review summarizes and discusses Cx43 expression, the role of gap junction intercellular communication, and its relationship with neuroinflammation in depression. This review also focuses on the effects of antidepressant drugs (e.g., monoamine antidepressants, psychotropic drugs, and N-methyl-D-aspartate receptor antagonists) on Cx43-mediated function and provides evidence for Cx43 as a novel target for the treatment of MDD. The pathogenesis of MDD is related to astrocyte dysfunction, with reduced Cx43 expression, GJ dysfunction, decreased GJIC and reduced BDNF expression in the depressed brain. The effect of Cx43 on neuroinflammation-related depression involving inflammatory cytokines, glutamate excitotoxicity, and HPA axis dysregulation. Antidepressant drugs targeting Cx43 can effectively relieve depressive symptoms.
Collapse
Affiliation(s)
- Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Ya-Ting Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Die Hu
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Cong Gai
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Sunshine Southern Avenue, Fang-Shan District, Beijing, 102488, China.
| |
Collapse
|
5
|
Huang D, Xiao Q, Tang J, Liang X, Wang J, Hu M, Jiang Y, Liu L, Qin L, Zhou M, Li Y, Zhu P, Deng Y, Li J, Zhou C, Luo Y, Tang Y. Positive effects of running exercise on astrocytes in the medial prefrontal cortex in an animal model of depression. J Comp Neurol 2022; 530:3056-3071. [PMID: 35972906 DOI: 10.1002/cne.25397] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/09/2022] [Accepted: 07/27/2022] [Indexed: 11/10/2022]
Abstract
Depression is one of the most common mental illnesses and seriously affects all aspects of life. Running exercise has been suggested to prevent or alleviate the occurrence and development of depression; however, the underlying mechanisms of these effects remain unclear. Independent studies have indicated that astrocytes play essential roles and that the medial prefrontal cortex (mPFC) is an important brain region involved in the pathology underlying depression. However, it is unknown whether running exercise achieves antidepressant effects by affecting the number of astrocytes and glutamate transport function in the mPFC. Here, animal models of depression were established using chronic unpredictable stress (CUS), and depression-like behavior was assessed by the sucrose preference test. After successfully establishing the depression model, experimental animals performed running exercise. Glial fibrillary acidic protein-positive (GFAP+ ) cell number in the mPFC was precisely quantified using immunohistochemical and stereological methods, and the densities of bromodeoxyuridine-positive (BrdU+ ) and BrdU+ /GFAP+ cells in the mPFC were measured using a semiquantitative immunofluorescence assay. Changes in glutamate transporter gene expression in mPFC astrocytes were detected by mRNA sequencing and qRT-PCR. We found that running exercise reversed CUS-induced decreases in sucrose preference, increased astrocyte number and the density of newborn astrocytes, and reversed decreases in gene expression levels of GFAP, S100b, and the glutamate transporters GLT-1 and GLAST in the mPFC of CUS animals. These results suggested that changes in astrocyte number and glutamate transporter function may be potential meditators of the effects of running exercise in the treatment of depression.
Collapse
Affiliation(s)
- Dujuan Huang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Qian Xiao
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China.,Department of Radioactive Medicine, Chongqing Medical University, Chongqing, P. R. China
| | - Jing Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Xin Liang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China.,Department of Pathophysiology, Chongqing Medical University, Chongqing, P. R. China
| | - Jin Wang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Menglan Hu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yanhong Jiang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Li Liu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Lu Qin
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Mei Zhou
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yue Li
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Peilin Zhu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yuhui Deng
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Jing Li
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Chunni Zhou
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yanmin Luo
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China.,Department of Physiology, Chongqing Medical University, Chongqing, P. R. China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
6
|
Mokhtari T. Targeting autophagy and neuroinflammation pathways with plant-derived natural compounds as potential antidepressant agents. Phytother Res 2022; 36:3470-3489. [PMID: 35794794 DOI: 10.1002/ptr.7551] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 11/06/2022]
Abstract
Major depressive disorder (MDD) is a life-threatening disease that presents several characteristics. The pathogenesis of depression still remains poorly understood. Moreover, the mechanistic interactions of natural components in treating depression to target autophagy and neuroinflammation are yet to be evaluated. This study overviewed the effects of plant-derived natural components in regulating critical pathways, particularly neuroinflammation and autophagy, associated with depression. A list of natural components, including luteolin, apigenin, hyperforin, resveratrol, salvianolic acid b, isoliquiritin, nobiletin, andrographolide, and oridonin, have been investigated. All peer-reviewed journal articles were searched by Scopus, MEDLINE, PubMed, Web of Science, and Google Scholar using the appropriated keywords, including depression, neuroinflammation, autophagy, plant, natural components, etc. The neuroinflammation and autophagy dysfunction are critically associated with the pathophysiology of depression. Natural components with higher efficiency and lower complications can be used for targeting neuroinflammation and autophagy. These components with different doses showed the beneficial antidepressant properties in rodents. These can modulate autophagy markers, mainly AMPK, LC3II/LC3I ratio, Beclin-1. Moreover, they can regulate the NLRP3 inflammasome, resulting in the suppression of proinflammatory cytokines (e.g., IL-1β and IL-18). Future in vitro and in vivo studies are required to develop novel therapeutic approaches based on plant-derived active components to treat MDD.
Collapse
Affiliation(s)
- Tahmineh Mokhtari
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
7
|
Ma X, Yang S, Zhang Z, Liu L, Shi W, Yang S, Li S, Cai X, Zhou Q. Rapid and sustained restoration of astrocytic functions by ketamine in depression model mice. Biochem Biophys Res Commun 2022; 616:89-94. [DOI: 10.1016/j.bbrc.2022.03.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022]
|
8
|
Lam YY, Tsai SF, Chen PC, Kuo YM, Chen YW. Pioglitazone rescues high-fat diet-induced depression-like phenotypes and hippocampal astrocytic deficits in mice. Biomed Pharmacother 2021; 140:111734. [PMID: 34022606 DOI: 10.1016/j.biopha.2021.111734] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022] Open
Abstract
The prevalence of diabetes is rapidly increasing worldwide and is highly associated with the incidence of depression. Pioglitazone, a Peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist, is widely used for treating patients with type 2 diabetes. However, whether pioglitazone alleviates metabolic disorder-related depression and astrocytic deficits remains unclear. Here we showed that 12 weeks of high-fat diet (HFD) feeding (from 8- to 20-week-old) induced not only obesity and insulin resistance, but also depression-like behaviors in mice. Astrocytic activation, a sign closely associated with depression, was also evident in the ventral hippocampus. Four weeks of pioglitazone (10 or 20 mg/kg, daily, from 20- to 24-week-old) treatment alleviated the HFD-induced glucose-metabolic dysfunctions, upregulation of ventral hippocampal GFAP, reduction of the total process lengths and the number of branch points of the ventral hippocampal CA1 GFAP-immunoreactive astrocytes and depressive phenotypes but had no effect on anxiety-like behaviors or hippocampus-related learning and memory in mice. These findings suggest that pioglitazone could be a potential therapeutic agent for metabolic disorders and associated depression.
Collapse
Affiliation(s)
- Ying-Yiu Lam
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Feng Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chun Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yun-Wen Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
9
|
Afridi R, Suk K. Neuroinflammatory Basis of Depression: Learning From Experimental Models. Front Cell Neurosci 2021; 15:691067. [PMID: 34276311 PMCID: PMC8283257 DOI: 10.3389/fncel.2021.691067] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/08/2021] [Indexed: 12/17/2022] Open
Abstract
The neuroinflammatory basis of depression encompasses the detrimental role of otherwise supportive non-neuronal cells and neuroinflammation in hampering neuronal function, leading to depressive behavior. Animals subjected to different stress paradigms show glial cell activation and a surge in proinflammatory cytokines in various brain regions. The concept of sterile inflammation observed in animal models of depression has intrigued many researchers to determine the possible triggers of central immune cell activation. Notably, microglial activation and subsequent phenotypic polarization in depression have been strongly advocated by the wealth of recent preclinical studies; however, findings from human studies have shown contradictory results. Despite intensive investigation, many research gaps still exist to elucidate the molecular mechanisms of neuroinflammatory cascades underlying the pathophysiology of depression. In this mini-review, recent progress in understanding neuroinflammatory mechanisms in light of experimental models of depression will be thoroughly discussed. The challenges of mirroring depression in animal and in vitro models will also be highlighted. Furthermore, prospects of targeting neuroinflammation to treat depressive disorder will be covered.
Collapse
Affiliation(s)
- Ruqayya Afridi
- BK21 Plus KNU Biomedical Convergence Program, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Kyoungho Suk
- BK21 Plus KNU Biomedical Convergence Program, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
10
|
The Association Between Antidepressant Effect of SSRIs and Astrocytes: Conceptual Overview and Meta-analysis of the Literature. Neurochem Res 2021; 46:2731-2745. [PMID: 33527219 DOI: 10.1007/s11064-020-03225-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/19/2022]
Abstract
Major depressive disorders (MDD) a worldwide psychiatric disease, is yet to be adequately controlled by therapies; while the mechanisms of action of antidepressants are yet to be fully characterised. In the last two decades, an increasing number of studies have demonstrated the role of astrocytes in the pathophysiology and therapy of MDD. Selective serotonin reuptake inhibitors (SSRIs) are the most widely used antidepressants. It is generally acknowledged that SSRIs increase serotonin levels in the central nervous system by inhibiting serotonin transporters, although the SSRIs action is not ideal. The SSRIs antidepressant effect develops with considerable delay; their efficacy is low and frequent relapses are common. Neither cellular nor molecular pharmacological mechanisms of SSRIs are fully characterised; in particular their action on astrocytes remain underappreciated. In this paper we overview potential therapeutic mechanisms of SSRIs associated with astroglia and report the results of meta-analysis of studies dedicated to MDD, SSRIs and astrocytes. In particular, we argue that fluoxetine, the representative SSRI, improves depressive-like behaviours in animals treated with chronic mild stress and reverses depression-associated decrease in astrocytic glial fibrillary acidic protein (GFAP) expression. In addition, fluoxetine upregulates astrocytic mRNA expression of 5-hydroxytriptamin/serotonin2B receptors (5-HT2BR). In summary, we infer that SSRIs exert their anti-depressant effect by regulating several molecular and signalling pathways in astrocytes.
Collapse
|
11
|
Becker M, Pinhasov A, Ornoy A. Animal Models of Depression: What Can They Teach Us about the Human Disease? Diagnostics (Basel) 2021; 11:123. [PMID: 33466814 PMCID: PMC7830961 DOI: 10.3390/diagnostics11010123] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/28/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Depression is apparently the most common psychiatric disease among the mood disorders affecting about 10% of the adult population. The etiology and pathogenesis of depression are still poorly understood. Hence, as for most human diseases, animal models can help us understand the pathogenesis of depression and, more importantly, may facilitate the search for therapy. In this review we first describe the more common tests used for the evaluation of depressive-like symptoms in rodents. Then we describe different models of depression and discuss their strengths and weaknesses. These models can be divided into several categories: genetic models, models induced by mental acute and chronic stressful situations caused by environmental manipulations (i.e., learned helplessness in rats/mice), models induced by changes in brain neuro-transmitters or by specific brain injuries and models induced by pharmacological tools. In spite of the fact that none of the models completely resembles human depression, most animal models are relevant since they mimic many of the features observed in the human situation and may serve as a powerful tool for the study of the etiology, pathogenesis and treatment of depression, especially since only few patients respond to acute treatment. Relevance increases by the fact that human depression also has different facets and many possible etiologies and therapies.
Collapse
Affiliation(s)
- Maria Becker
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | - Albert Pinhasov
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | - Asher Ornoy
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel
| |
Collapse
|
12
|
Protective effects of dihydromyricetin on primary hippocampal astrocytes from cytotoxicity induced by comorbid diabetic neuropathic pain and depression. Purinergic Signal 2020; 16:585-599. [PMID: 33155081 DOI: 10.1007/s11302-020-09752-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/26/2020] [Indexed: 01/28/2023] Open
Abstract
Activated astrocytes play a key role in diabetic neuropathic pain and depression. We aimed to assess the protective effects of dihydromyricetin (DHM) on primary hippocampal astrocytes cultured with high glucose (HG), substance P (SP), and corticosterone (CORT). Culturing with HG + SP + CORT resulted in damage to primary hippocampal astrocytes, which simulates the clinical damage caused by comorbidity of diabetic neuropathic pain and depression. Western blot, qPCR, and immunofluorescence analyses revealed that HG + SP + CORT increased P2X7 receptor expression in primary hippocampal astrocytes, which was reversed by DHM treatment. Further, HG + SP + CORT elevated TNF-α, IL-1β, free Ca2+, and ERK1/2 phosphorylation levels, which was inhibited by DHM or P2X7 shRNA treatment. Moreover, DHM significantly reduced the P2X7 agonist-activated currents in HEK293 cells transfected with the P2X7 receptor. These findings suggest that DHM can protect primary hippocampal astrocytes cultured with HG + SP + CORT from P2X7 receptor-mediated damage. Culturing cells with HG + SP + CORT might be a viable cell model for cellular injury exploration of diabetic comorbid pain and depression.
Collapse
|
13
|
Guerrero-Vargas NN, Zárate-Mozo C, Guzmán-Ruiz MA, Cárdenas-Rivera A, Escobar C. Time-restricted feeding prevents depressive-like and anxiety-like behaviors in male rats exposed to an experimental model of shift-work. J Neurosci Res 2020; 99:604-620. [PMID: 33078850 DOI: 10.1002/jnr.24741] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/13/2020] [Accepted: 10/01/2020] [Indexed: 12/21/2022]
Abstract
Individuals who regularly shift their sleep timing, like night and/or shift-workers suffer from circadian desynchrony and are at risk of developing cardiometabolic diseases and cancer. Also, shift-work is are suggested to be a risk factor for the development of mood disorders such as the burn out syndrome, anxiety, and depression. Experimental and clinical studies provide evidence that food intake restricted to the normal activity phase is a potent synchronizer for the circadian system and can prevent the detrimental health effects associated with circadian disruption. Here, we explored whether adult male Wistar rats exposed to an experimental model of shift-work (W-AL) developed depressive and/or anxiety-like behaviors and whether this was associated with neuroinflammation in brain areas involved with mood regulation. We also tested whether time-restricted feeding (TRF) to the active phase could ameliorate circadian disruption and therefore would prevent depressive and anxiety-like behaviors as well as neuroinflammation. In male Wistar rats, W-AL induced depressive-like behavior characterized by hypoactivity and anhedonia and induced increased anxiety-like behavior in the open field test. This was associated with increased number of glial fibrillary acidic protein and IBA-1-positive cells in the prefrontal cortex and basolateral amygdala. Moreover W-AL caused morphological changes in the microglia in the CA3 area of the hippocampus indicating microglial activation. Importantly, TRF prevented behavioral changes and decreased neuroinflammation markers in the brain. Present results add up evidence about the importance that TRF in synchrony with the light-dark cycle can prevent neuroinflammation leading to healthy mood states in spite of circadian disruptive conditions.
Collapse
Affiliation(s)
- Natalí N Guerrero-Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Carmen Zárate-Mozo
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mara A Guzmán-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Alfredo Cárdenas-Rivera
- Centro de Investigación en Bioingeniería, Universidad de Ingeniería y Tecnología, Lima, Perú
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
14
|
Giacobbe J, Pariante CM, Borsini A. The innate immune system and neurogenesis as modulating mechanisms of electroconvulsive therapy in pre-clinical studies. J Psychopharmacol 2020; 34:1086-1097. [PMID: 32648795 PMCID: PMC7672674 DOI: 10.1177/0269881120936538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Electroconvulsive therapy (ECT) is a powerful and fast-acting anti-depressant strategy, often used in treatment-resistant patients. In turn, patients with treatment-resistant depression often present an increased inflammatory response. The impact of ECT on several pathophysiological mechanisms of depression has been investigated, with a focus which has largely been on cellular and synaptic plasticity. Although changes in the immune system are known to influence neurogenesis, these processes have principally been explored independently from each other in the context of ECT. OBJECTIVE The aim of this review was to compare the time-dependent consequences of acute and chronic ECT on concomitant innate immune system and neurogenesis-related outcomes measured in the central nervous system in pre-clinical studies. RESULTS During the few hours following acute electroconvulsive shock (ECS), the expression of the astrocytic reactivity marker glial fibrillary acidic protein (GFAP) and inflammatory genes, such as cyclooxygenase-2 (COX2), were significantly increased together with the neurogenic brain-derived neurotrophic factor (BDNF) and cell proliferation. Similarly, chronic ECS caused an initial upregulation of the same astrocytic marker, immune genes, and neurogenic factors. Interestingly, over time, inflammation appeared to be dampened, while glial activation and neurogenesis were maintained, after either acute or chronic ECS. CONCLUSION Regardless of treatment duration ECS would seemingly trigger a rapid increase in inflammatory molecules, dampened over time, as well as a long-lasting activation of astrocytes and production of growth and neurotrophic factors, leading to cell proliferation. This suggests that both innate immune system response and neurogenesis might contribute to the efficacy of ECT.
Collapse
Affiliation(s)
| | | | - Alessandra Borsini
- Alessandra Borsini, King’s College London, Institute of Psychiatry, Psychology & Neuroscience, Division of Psychological Medicine, Stress, Psychiatry and Immunology Lab & Perinatal Psychiatry, The Maurice Wohl Clinical Neuroscience Institute, Cutcombe Road, London SE5 9RT, UK.
| |
Collapse
|
15
|
Understanding the effects of chronic benzodiazepine use in depression: a focus on neuropharmacology. Int Clin Psychopharmacol 2020; 35:243-253. [PMID: 32459725 DOI: 10.1097/yic.0000000000000316] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Benzodiazepines are frequently prescribed on an ongoing basis to individuals with depression, mainly to alleviate anxiety or insomnia, despite current guideline recommendations that continuous use should not exceed 4 weeks. Currently, there are no efficacy trials published beyond 8 weeks. Several antidepressant trials demonstrate that the concomitant use of a benzodiazepine is associated with poorer depressive outcomes and functional status; however, it is unclear why this is the case. Patients with depression receiving a benzodiazepine may reflect a more ill or high anxiety group, although even within anxiety disorders, the use of a benzodiazepine is associated with poorer outcomes. The neuroadaptive consequences of long-term benzodiazepine use may be a factor underlying these findings. Chronic benzodiazepine use results in decreased gamma-aminobutyric acid and monoaminergic function, as well as interference with neurogenesis, which are all purported to play a role in antidepressant efficacy. This review will discuss the oppositional neuropharmacological interactions between chronic benzodiazepine use and antidepressant mechanism of action, which could result in reduced antidepressant efficacy and function in depression.
Collapse
|
16
|
Connexin 43: A novel ginsenoside Rg1-sensitive target in a rat model of depression. Neuropharmacology 2020; 170:108041. [DOI: 10.1016/j.neuropharm.2020.108041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/31/2022]
|
17
|
Stenovec M, Li B, Verkhratsky A, Zorec R. Astrocytes in rapid ketamine antidepressant action. Neuropharmacology 2020; 173:108158. [PMID: 32464133 DOI: 10.1016/j.neuropharm.2020.108158] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/27/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022]
Abstract
Ketamine, a general anaesthetic and psychotomimetic drug, exerts rapid, potent and long-lasting antidepressant effect, albeit the cellular and molecular mechanisms of this action are yet to be discovered. Besides targeting neuronal NMDARs fundamental for synaptic transmission, ketamine affects the function of astroglia the key homeostatic cells of the central nervous system that contribute to pathophysiology of psychiatric diseases including depression. Here we review studies revealing that (sub)anaesthetic doses of ketamine elevate intracellular cAMP concentration ([cAMP]i) in astrocytes, attenuate stimulus-evoked astrocyte calcium signalling, which regulates exocytotic secretion of gliosignalling molecules, and stabilize the vesicle fusion pore in a narrow configuration possibly hindering cargo discharge or vesicle recycling. Next we discuss how ketamine affects astroglial capacity to control extracellular K+ by reducing cytoplasmic mobility of vesicles delivering the inward rectifying potassium channel (Kir4.1) to the plasmalemma. Modified astroglial K+ buffering impacts upon neuronal excitability as demonstrated in the lateral habenula rat model of depression. Finally, we highlight the recent discovery that ketamine rapidly redistributes cholesterol in the plasmalemma of astrocytes, but not in fibroblasts nor in neuronal cells. This alteration of membrane structure may modulate a host of processes that synergistically contribute to ketamine's rapid and prominent antidepressant action.
Collapse
Affiliation(s)
- Matjaž Stenovec
- Celica BIOMEDICAL, Tehnološki Park 24, 1000, Ljubljana, Slovenia; Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000, Ljubljana, Slovenia.
| | - Baoman Li
- Practical Teaching Centre, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China; Department of Poison Analysis, School of Forensic Medicine, China Medical University, Shenyang, China.
| | - Alexei Verkhratsky
- Celica BIOMEDICAL, Tehnološki Park 24, 1000, Ljubljana, Slovenia; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK; Achucarro Center for Neuroscience, IKERBASQUE, 48011, Bilbao, Spain.
| | - Robert Zorec
- Celica BIOMEDICAL, Tehnološki Park 24, 1000, Ljubljana, Slovenia; Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000, Ljubljana, Slovenia.
| |
Collapse
|
18
|
Gong Q, Yan XJ, Lei F, Wang ML, He LL, Luo YY, Gao HW, Feng YL, Yang SL, Li J, Du LJ. Proteomic profiling of the neurons in mice with depressive-like behavior induced by corticosterone and the regulation of berberine: pivotal sites of oxidative phosphorylation. Mol Brain 2019; 12:118. [PMID: 31888678 PMCID: PMC6937859 DOI: 10.1186/s13041-019-0518-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/28/2019] [Indexed: 12/16/2022] Open
Abstract
Chronic corticosterone (CORT) stress is an anxiety and depression inducing factor that involves the dysfunction of glucocorticoid receptor (GR), brain-derived neurotrophic factor (BDNF), and neuronal plasticity. However, the regulation of proteomic profiles in neurons suffering CORT stress is remaining elusive. Thus, the proteomic profiles of mouse neuronal C17.2 stem cells were comprehensively investigated by TMT (tandem mass tag)-labeling quantitative proteomics. The quantitative proteomics conjugated gene ontology analysis revealed the inhibitory effect of CORT on the expression of mitochondrial oxidative phosphorylation-related proteins, which can be antagonized by berberine (BBR) treatment. In addition, animal studies showed that changes in mitochondria by CORT can affect neuropsychiatric activities and disturb the physiological functions of neurons via disordering mitochondrial oxidative phosphorylation. Thus, the mitochondrial energy metabolism can be considered as one of the major mechanism underlying CORT-mediated depression. Since CORT is important for depression after traumatic stress disorder, our study will shed light on the prevention and treatment of depression as well as posttraumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Qin Gong
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.,State Key Laboratory of Innovative Drugs and Efficient Energy-saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Xiao-Jin Yan
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fan Lei
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Mu-Lan Wang
- State Key Laboratory of Innovative Drugs and Efficient Energy-saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Lu-Ling He
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.,State Key Laboratory of Innovative Drugs and Efficient Energy-saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Ying-Ying Luo
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.,State Key Laboratory of Innovative Drugs and Efficient Energy-saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Hong-Wei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Yu-Lin Feng
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.,State Key Laboratory of Innovative Drugs and Efficient Energy-saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.,College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Shi-Lin Yang
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.,State Key Laboratory of Innovative Drugs and Efficient Energy-saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.,College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Jun Li
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China. .,State Key Laboratory of Innovative Drugs and Efficient Energy-saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.
| | - Li-Jun Du
- Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.,State Key Laboratory of Innovative Drugs and Efficient Energy-saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China.,College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530000, China
| |
Collapse
|
19
|
Verkhratsky A, Rodrigues JJ, Pivoriunas A, Zorec R, Semyanov A. Astroglial atrophy in Alzheimer’s disease. Pflugers Arch 2019; 471:1247-1261. [DOI: 10.1007/s00424-019-02310-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/23/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
|
20
|
Steardo L, de Filippis R, Carbone EA, Segura-Garcia C, Verkhratsky A, De Fazio P. Sleep Disturbance in Bipolar Disorder: Neuroglia and Circadian Rhythms. Front Psychiatry 2019; 10:501. [PMID: 31379620 PMCID: PMC6656854 DOI: 10.3389/fpsyt.2019.00501] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
The worldwide prevalence of sleep disorders is approximately 50%, with an even higher occurrence in a psychiatric population. Bipolar disorder (BD) is a severe mental illness characterized by shifts in mood and activity. The BD syndrome also involves heterogeneous symptomatology, including cognitive dysfunctions and impairments of the autonomic nervous system. Sleep abnormalities are frequently associated with BD and are often a good predictor of a mood swing. Preservation of stable sleep-wake cycles is therefore a key to the maintenance of stability in BD, indicating the crucial role of circadian rhythms in this syndrome. The symptom most widespread in BD is insomnia, followed by excessive daytime sleepiness, nightmares, difficulty falling asleep or maintaining sleep, poor sleep quality, sleep talking, sleep walking, and obstructive sleep apnea. Alterations in the structure or duration of sleep are reported in all phases of BD. Understanding the role of neuroglia in BD and in various aspects of sleep is in nascent state. Contributions of the different types of glial cells to BD and sleep abnormalities are discussed in this paper.
Collapse
Affiliation(s)
- Luca Steardo
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| | - Renato de Filippis
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| | - Elvira Anna Carbone
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| | - Cristina Segura-Garcia
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Achucarro Center for Neuroscience, IKERBASQUE, Bilbao, Spain
| | - Pasquale De Fazio
- Psychiatric Unit, Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| |
Collapse
|
21
|
Xu J, Wang R, Liu Y, Wang W, Liu D, Jiang H, Pan F. Short- and long-term alterations of FKBP5-GR and specific microRNAs in the prefrontal cortex and hippocampus of male rats induced by adolescent stress contribute to depression susceptibility. Psychoneuroendocrinology 2019; 101:204-215. [PMID: 30469088 DOI: 10.1016/j.psyneuen.2018.11.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 11/05/2018] [Accepted: 11/06/2018] [Indexed: 02/06/2023]
Abstract
Maladaptation of the hypothalamic-pituitary-adrenal (HPA) axis is involved in susceptibility to depression. Glucocorticoid receptors (GRs) and the co-chaperone protein, FK506 binding protein 51 (FKBP5), play crucial roles in dysfunction of the HPA axis. Further, certain microRNAs (miRNAs), such as miR-124a and miR-18a, which could reduce GR protein expression, contribute to affective disorders, while miR-511 as a regulator of FKBP5 is involved in an increased risk of depression. However, the short-term and persistent impacts of adolescent stress on miR-124a, miR-18a, and miR-511 expressions in the brain are unknown. Using depression models of chronic unpredictable mild stress (CUMS) or dexamethasone administration of adolescent rats, the authors of the present study probed the depressive-like behaviors, GR and FKBP5 expressions, and miR-124a, miR-18a, and miR-511 expressions in the prefrontal cortex and hippocampus. The GR antagonist RU486 was used as intervention. The results revealed that both CUMS and dexamethasone administration in the adolescent period resulted in anhedonia, altered locomotor behaviors, anxiety, and cognitive impairment. A remarkable decrease in GR expression, and increase in FKBP5, miR-124a, and miR-18a expressions were detected in the prefrontal cortex and hippocampus of adolescent rats. Furthermore, the similar long-term changes on behaviors and expressions of GR, FKBP5 and GR-related microRNAs were found in the adult rats following CUMS and dexamethasone treatment in adolescence. However, reduced miR-511 expression was observed only in the prefrontal cortex of adult rats exposed to adolescent CUMS or dexamethasone administration. These data suggested that the downregulation of GR, upregulation of FKBP5, miR-124a, and miR-18a in the prefrontal cortex and hippocampus, and downregulation of miR-511 in the prefrontal cortex were relevant to depressive-like behaviors.
Collapse
Affiliation(s)
- Jingjing Xu
- Department of Medical Psychology and Medical Ethic, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Rui Wang
- Department of Medical Psychology and Medical Ethic, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Yuan Liu
- Department of Medical Psychology and Medical Ethic, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Wei Wang
- Department of Medical Psychology and Medical Ethic, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Dexiang Liu
- Department of Medical Psychology and Medical Ethic, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Hong Jiang
- Department of Medical Psychology and Medical Ethic, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Fang Pan
- Department of Medical Psychology and Medical Ethic, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
22
|
Tactile Stimulation on Adulthood Modifies the HPA Axis, Neurotrophic Factors, and GFAP Signaling Reverting Depression-Like Behavior in Female Rats. Mol Neurobiol 2019; 56:6239-6250. [PMID: 30741369 DOI: 10.1007/s12035-019-1522-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/30/2019] [Indexed: 12/31/2022]
Abstract
Depression is a common psychiatric disease which pharmacological treatment relieves symptoms, but still far from ideal. Tactile stimulation (TS) has shown beneficial influences in neuropsychiatric disorders, but the mechanism of action is not clear. Here, we evaluated the TS influence when applied on adult female rats previously exposed to a reserpine-induced depression-like animal model. Immediately after reserpine model (1 mg/kg/mL, 1×/day, for 3 days), female Wistar rats were submitted to TS (15 min, 3×/day, for 8 days) or not (unhandled). Imipramine (10 mg/kg/mL) was used as positive control. After behavioral assessments, animals were euthanized to collect plasma and prefrontal cortex (PFC). Behavioral observations in the forced swimming test, splash test, and sucrose preference confirmed the reserpine-induced depression-like behavior, which was reversed by TS. Our findings showed that reserpine increased plasma levels of adrenocorticotropic hormone and corticosterone, decreased brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B, and increased proBDNF immunoreactivity in the PFC, which were also reversed by TS. Moreover, TS reestablished glial fibrillary acidic protein and glucocorticoid receptor levels, decreased by reserpine in PFC, while glial cell line-derived neurotrophic factor was increased by TS per se. Our outcomes are showing that TS applied in adulthood exerts a beneficial influence in depression-like behaviors, modulating the HPA axis and regulating neurotrophic factors more effectively than imipramine. Based on this, our proposal is that TS, in the long term, could be considered a new therapeutic strategy for neuropsychiatric disorders improvement in adult life, which may represent an interesting contribution to conventional pharmacological treatment.
Collapse
|