1
|
Kartalci Ş, Özgen N, Kartal F, Arslan AK, Koç A, Düzova H. The Effect of Electroconvulsive Therapy on Hippocampal Endoplasmic Reticulum Stress in a Rat Model of Depression. J ECT 2025:00124509-990000000-00288. [PMID: 40245333 DOI: 10.1097/yct.0000000000001144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
OBJECTIVE The mechanisms underlying the effectiveness of electroconvulsive therapy (ECT), one of the most effective treatments for depression, are still unknown. While the increasing endoplasmic reticulum (ER) stress in depression can retrieve with pharmacological agents, ECT's effectiveness has not been examined yet. In this study, we tested how hippocampal ER stress parameters change after repeated ECT in rats in the chronic unpredictable mild stress (CUMS) model. METHODS Rats were divided into 4 groups. Two groups were included in the experimental group, where the CUMS model was applied for 21 days. Then, the experimental ECT model was applied to 2 groups, one experimental and 1 control group, for 21 days. Weight changes results were evaluated at the end of the 21st and 42nd day. In the final step, 3 prominent ER stress-related proteins (Grp-78, Xbp1, and Atf-4) were measured by western blot in the removed hippocampus. RESULTS We found that rats administered CUMS exhibited depression-like behavioral responses such as weight loss. In CUMS-treated rats (group 3), Grp-78 levels increased, ATF4 levels did not change, and Xbp1 levels decreased. While ECT increased Grp-78 levels in normal rats, it did not change Atf-4 levels and reduced Xbp1 levels (group 2). When ECT was applied to rats undergoing the CUMS model, Grp-78 and Xbp1 levels decreased, while Atf-4 levels did not change (group 4). CONCLUSIONS These findings show that increased ER stress may play a role in the pathophysiology of depression and that this increase can be reversed with ECT treatment. These findings need to be confirmed by clinical studies in humans.
Collapse
Affiliation(s)
- Şükrü Kartalci
- From the Department of Pschiatry, The Private Medicabil Hospital, Bursa, Turkey
| | - Nazmi Özgen
- Department of Biostatististics and Medical İnformatics, İnönü University, Faculty of Medicine, Malatya, Turkey
| | - Fatma Kartal
- Kırıkkale University, Faculty of Medicine, Department of psychiatry, Kırıkkale, Turkey
| | - Ahmet Kadir Arslan
- Department of Biostatististics and Medical İnformatics, İnönü University, Faculty of Medicine, Malatya, Turkey
| | - Ahmet Koç
- Department of Medical Genetics, İnönü University, Faculty of Medicine, Malatya, Turkey
| | - Halil Düzova
- Department Of Physiology, Inonu University, Faculty Of Medicine, Malatya, Turkey
| |
Collapse
|
2
|
Hu L, Wu N, Wang J, Cai D. Network pharmacology combined with experimental verification for exploring the potential mechanism of phellodendrine against depression. Sci Rep 2025; 15:1958. [PMID: 39809809 PMCID: PMC11733132 DOI: 10.1038/s41598-024-84771-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025] Open
Abstract
The anti-inflammatory effect of phellodendrine (PHE), derived from Phellodendri Chinensis Cortex, has been verified in previous studies. Major depressive disorder (MDD) is associated with immune dysregulation and inflammatory processes. This study aimed to explore the therapeutic effects of PHE on MDD through network pharmacology and experimental validation. Multiple databases were used to predict the targets of PHE and MDD. The intersection targets between PHE and MDD were obtained to identify as targets for PHE against MDD, followed by protein-protein interaction network, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses. Molecular docking was applied to further confirm the anti-MDD effects of PHE. The mitochondrial DNA (mtDNA) copy number, inflammatory cytokines and pathway-related mRNA expressions in PC12 cell were determined via quantitative PCR (qPCR) and enzyme-linked immunosorbent assay to verify our finding. Thirty-eight intersection targets were obtained between PHE and MDD. PHE exerted an anti-MDD effect by regulating SLC6A4, SLC6A3, SLC6A2, MAOA and other targets through serotonergic synapse, salivary secretion, dopaminergic synapse, and cAMP signalling pathway. In vitro, PHE induced an increment in mtDNA copy number compared with the CORT group. PHE affected the levels of IL6 and IL1β with different concentrations. The mRNA levels of CHRM1, HTR1A and key targets of the PI3K/Akt signalling pathway were also influenced. Our research reveals novel mechanisms underlying the anti-MDD effects of PHE through network pharmacology and experiments, which provides a new direction for the development of antidepressants.
Collapse
Affiliation(s)
- Lili Hu
- College of Basic Medicine, Shanxi University of Chinese Medicine, No. 121 DaXue Street, Jinzhong, 030619, China.
| | - Na Wu
- College of Basic Medicine, Shanxi University of Chinese Medicine, No. 121 DaXue Street, Jinzhong, 030619, China
| | - Jue Wang
- College of Basic Medicine, Shanxi University of Chinese Medicine, No. 121 DaXue Street, Jinzhong, 030619, China
| | - Donghui Cai
- College of Basic Medicine, Shanxi University of Chinese Medicine, No. 121 DaXue Street, Jinzhong, 030619, China
| |
Collapse
|
3
|
Huang H, Ji F, Qi G, Cao Y, He X, Wang H, Jiang Z. Rehmannioside A promotes the osteoblastic differentiation of MC3T3-E1 cells via the PI3K/AKT signaling pathway and inhibits glucocorticoid-induced bone loss in vivo. J Pharmacol Sci 2024; 156:247-257. [PMID: 39608850 DOI: 10.1016/j.jphs.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is a widespread disease characterized by low bone density. There remains a lack of effective means for osteoporosis. Rehmannioside A (ReA), an iridoid glycoside, exhibits various pharmacological activities. This study aimed to explore the role and mechanism of ReA in osteogenic differentiation of osteoblasts. Cell viability, reactive oxygen species (ROS) generation, and cell apoptosis were assessed using corresponding assay kits. Real-time quantitative polymerase chain reaction, Western blotting, and alkaline phosphatase (ALP) staining were performed to evaluate the osteogenic differentiation of MC3T3-E1 cells. Alizarin red S staining was used to assess the mineralization of MC3T3-E1 cells. Protein expression associated with the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling pathway was analyzed using Western blotting. Micro-computed tomography, histopathological, and immunohistochemical analyses were performed to determine the therapeutic effect of ReA on GIOP in vivo.The results showed that ReA promoted the osteogenic differentiation of MC3T3-E1 cells by regulating the PI3K/AKT signaling pathway and protected mice against glucocorticoid-induced bone loss by promoting osteoblast-mediated bone formation in vivo. The findings of the current study revealed that ReA is a potential therapeutic agent for osteoporosis.
Collapse
Affiliation(s)
- Haisheng Huang
- Department of Orthopedic Surgery, SHANGHAI TCM-INTEGRATED Hospital Shanghai University of TCM, Shanghai, China
| | - Fang Ji
- Department of Day Ward, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guobin Qi
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai, China
| | - Yuting Cao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai, China
| | - Xuecheng He
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai, China
| | - Hao Wang
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Zengxin Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
4
|
Zhong G, Wang X, Zhang Q, Zhang X, Fang X, Li S, Pan Y, Ma Y, Wang X, Wan T, Wang Q. Exploring the therapeutic implications of natural compounds modulating apoptosis in vascular dementia. Phytother Res 2024; 38:5270-5289. [PMID: 39223915 DOI: 10.1002/ptr.8316] [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: 02/21/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Vascular dementia (VaD) is a prevalent form of dementia stemming from cerebrovascular disease, manifesting in memory impairment and executive dysfunction, thereby imposing a substantial societal burden. Unfortunately, no drugs have been approved for the treatment of VaD due to its intricate pathogenesis, and the development of innovative and efficacious medications is urgently needed. Apoptosis, a programmed cell death process crucial for eliminating damaged or unwanted cells within an organism, assumes pivotal roles in embryonic development and tissue homeostasis maintenance. An increasing body of evidence indicates that apoptosis may significantly influence the onset and progression of VaD, and numerous natural compounds have demonstrated significant therapeutic potential. Here, we discuss the molecular mechanisms underlying apoptosis and its correlation with VaD. We also provide a crucial reference for developing innovative pharmaceuticals by systematically reviewing the latest research progress concerning the neuroprotective effects of natural compounds on VaD by regulating apoptosis. Further high-quality clinical studies are imperative to firmly ascertain these natural compounds' clinical efficacy and safety profiles in the treatment of VaD.
Collapse
Affiliation(s)
- Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Wang
- Department of Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qian Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xueying Zhang
- The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China
| | - Xiaoling Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuting Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yaru Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yujie Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuejing Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Wan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
5
|
Zhang N, Zhang S, Dong X. Plant-derived bioactive compounds and their novel role in central nervous system disorder treatment via ATF4 targeting: A systematic literature review. Biomed Pharmacother 2024; 176:116811. [PMID: 38795641 DOI: 10.1016/j.biopha.2024.116811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
Central nervous system (CNS) disorders exhibit exceedingly intricate pathogenic mechanisms. Pragmatic and effective solutions remain elusive, significantly compromising human life and health. Activating transcription factor 4 (ATF4) participates in the regulation of multiple pathophysiological processes, including CNS disorders. Considering the widespread involvement of ATF4 in the pathological process of CNS disorders, the targeted regulation of ATF4 by plant-derived bioactive compounds (PDBCs) may become a viable strategy for the treatment of CNS disorders. However, the regulatory relationship between PDBCs and ATF4 remains incompletely understood. Here, we aimed to comprehensively review the studies on PDBCs targeting ATF4 to ameliorate CNS disorders, thereby offering novel directions and insights for the treatment of CNS disorders. A computerized search was conducted on PubMed, Embase, Web of Science, and Google Scholar databases to identify preclinical experiments related to PDBCs targeting ATF4 for the treatment of CNS disorders. The search timeframe was from the inception of the databases to December 2023. Two assessors conducted searches using the keywords "ATF4," "Central Nervous System," "Neurological," "Alzheimer's disease," "Parkinson's Disease," "Stroke," "Spinal Cord Injury," "Glioblastoma," "Traumatic Brain Injury," and "Spinal Cord Injury." Overall, 31 studies were included, encompassing assessments of 27 PDBCs. Combining results from in vivo and in vitro studies, we observed that these PDBCs, via ATF4 modulation, prevent the deposition of amyloid-like fibers such as Aβ, tau, and α-synuclein. They regulate ERS, reduce the release of inflammatory factors, restore mitochondrial membrane integrity to prevent oxidative stress, regulate synaptic plasticity, modulate autophagy, and engage anti-apoptotic mechanisms. Consequently, they exert neuroprotective effects in CNS disorders. Numerous PDBCs targeting ATF4 have shown potential in facilitating the restoration of CNS functionality, thereby presenting expansive prospects for the treatment of such disorders. However, future endeavors necessitate high-quality, large-scale, and comprehensive preclinical and clinical studies to further validate this therapeutic potential.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, the Seventh Clinical College of China Medical University, No. 24 Central Street, Xinfu District, Fushun, Liaoning 113000, China
| | - Shun Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao street, Heping District, Shenyang, Liaoning 110000, China.
| |
Collapse
|
6
|
Wang C, Zhou N, Li M, Chen H. Rehmannioside A inhibits the activity of CYP3A4, 2C9 and 2D6 in vitro. Xenobiotica 2024; 54:195-200. [PMID: 38385556 DOI: 10.1080/00498254.2024.2321969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/19/2024] [Indexed: 02/23/2024]
Abstract
To assess the effect of Rehmannioside A on CYP450s activity and to estimate its inhibitory properties.The effect of Rehmannioside A on the activity of major CYP450s in human liver microsomes (HLMs) was assessed with the corresponding substrates and marker reactions, and compared with a blank control and the respective inhibitors. Suppression of CYP3A4, 2C9 and 2D6 was assessed by the dose-dependent assay and fitted with non-competitive or competitive inhibition models. The inhibition of CYP3A4 was determined in a time-dependent manner.Rehmannioside A suppressed the activity of CYP3A4, 2C9, and 2D6 with IC50 values of 10.08, 12.62, and 16.43 μM, respectively. Suppression of CYP3A4 was fitted to a non-competitive model with Ki value of 5.08 μM, whereas CYP2C9 and 2D6 were fitted to a competitive model with Ki values of 6.25 and 8.14 μM. Additionally, the inhibitory effect on CYP3A4 was time-dependent with KI value of 8.47 μM-1 and a Kinact of 0.048 min-1.In vitro suppression of CYP3A, 2C9 and 2D6 by Rehmannioside A indicated that Rehmannioside A or its source herbs may interact with drugs metabolised by these CYP450s, which could guide the clinical application.
Collapse
Affiliation(s)
- Congrong Wang
- Department of Pharmacy Center, Shandong Public Health Clinical Center, Jinan, China
| | - Naixiang Zhou
- Department of Office, Jiyang People's Hospital of Jinan, Jinan, China
| | - Mingcui Li
- Department of Pharmacy, Shanghe T.C.M Hospital, Jinan, China
| | - Haixia Chen
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
7
|
Li Y, Li M, Feng S, Xu Q, Zhang X, Xiong X, Gu L. Ferroptosis and endoplasmic reticulum stress in ischemic stroke. Neural Regen Res 2024; 19:611-618. [PMID: 37721292 PMCID: PMC10581588 DOI: 10.4103/1673-5374.380870] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 05/30/2023] [Accepted: 06/14/2023] [Indexed: 09/19/2023] Open
Abstract
Ferroptosis is a form of non-apoptotic programmed cell death, and its mechanisms mainly involve the accumulation of lipid peroxides, imbalance in the amino acid antioxidant system, and disordered iron metabolism. The primary organelle responsible for coordinating external challenges and internal cell demands is the endoplasmic reticulum, and the progression of inflammatory diseases can trigger endoplasmic reticulum stress. Evidence has suggested that ferroptosis may share pathways or interact with endoplasmic reticulum stress in many diseases and plays a role in cell survival. Ferroptosis and endoplasmic reticulum stress may occur after ischemic stroke. However, there are few reports on the interactions of ferroptosis and endoplasmic reticulum stress with ischemic stroke. This review summarized the recent research on the relationships between ferroptosis and endoplasmic reticulum stress and ischemic stroke, aiming to provide a reference for developing treatments for ischemic stroke.
Collapse
Affiliation(s)
- Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Mingyang Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Shi Feng
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Qingxue Xu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xu Zhang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
8
|
Li P, Cao G. PDCD4 silencing alleviates KA‑induced neurotoxicity of HT22 cells by inhibiting endoplasmic reticulum stress via blocking the MAPK/NF‑κB signaling pathway. Exp Ther Med 2024; 27:55. [PMID: 38234627 PMCID: PMC10790171 DOI: 10.3892/etm.2023.12343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/11/2023] [Indexed: 01/19/2024] Open
Abstract
Human programmed cell death 4 (PDCD4) has been reported to participate in multiple neurological diseases. However, the role of PDCD4 in epilepsy, as well as its underlying mechanism, remains unclear. To induce excitotoxicity, 100 µM kainic acid (KA) was applied for the stimulation of HT22 cells for 12 h. Initially, the mRNA and protein expression levels of PDCD4 were evaluated using reverse transcription-quantitative PCR and western blotting. A lactate dehydrogenase assay was performed to detect cell injury. Cell apoptosis was assessed using flow cytometry and western blotting was performed to determine the expression levels of apoptosis-related proteins. Oxidative stress was detected using dichlorodihydrofluorescein diacetate staining, and malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) assay kits. Furthermore, the expression levels of MAPK/NF-κB signaling-related proteins and endoplasmic reticulum (ER) stress-related proteins C/EBP homologous protein, glucose-regulated protein 78, activating transcription factor 4 and phosphorylated-eukaryotic initiation factor-2α were assessed by western blotting. It was revealed that PDCD4 expression was markedly elevated in KA-induced HT22 cells, whereas PDCD4 silencing alleviated KA-induced neurotoxicity of HT22 cells by alleviating cell injury and inhibiting apoptosis. In addition, PDCD4 silencing reduced the levels of reactive oxygen species and MDA, but elevated those of SOD and GSH-Px. PDCD4 silencing also suppressed ER stress by blocking the MAPK/NF-κB signaling pathway. By contrast, the MAPK agonist phorbol myristate acetate reversed the effects of PDCD4 silencing on KA-induced neurotoxicity and oxidative stress in HT22 cells. In conclusion, PDCD4 silencing alleviated KA-induced neurotoxicity and oxidative stress in HT22 cells by suppressing ER stress through the inhibition of the MAPK/NF-κB signaling pathway, which may provide novel insights into the treatment of epilepsy.
Collapse
Affiliation(s)
- Peng Li
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Guiling Cao
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
9
|
Yuan LL, Cao CY. Rehmannioside A Inhibits TRAF6/MAPK Pathway and Improves Psoriasis by Interfering with the Interaction of HaCaT Cells with IL-17A. Clin Cosmet Investig Dermatol 2023; 16:2585-2596. [PMID: 37752969 PMCID: PMC10519428 DOI: 10.2147/ccid.s430621] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023]
Abstract
Objective As a common chronic inflammatory skin disease, psoriasis seriously affects the physical health and psychological well-being of patients. Various clinical treatments for psoriasis have their own drawbacks, so it is important to find effective and safe drugs. Rehmannioside A (ReA) has anti-inflammatory properties and is the main active ingredient in Fuzhengzhiyanghefuzhiyang decoction (FZHFZY), an herbal compound for the treatment of psoriasis. But no studies have been conducted to determine whether ReA alone can treat psoriasis. Therefore, this study was designed to investigate the effect of ReA in the treatment of psoriasis and its potential mechanism of action. Methods HaCaT cells were treated with ReA and IL-17A alone for 24 h and 48 h, and the most effective concentrations of ReA and interleukin (IL)-17A were found at 25 μg/mL and 100 ng/mL, respectively. A psoriasis cell model was constructed by stimulating HaCaT cells with IL-17A, followed by intervention with ReA. Cell viability and cell cycle distribution were measured by MTT assay and flow cytometry. The expression levels of keratin family members and chemokines were detected by real-time quantitative PCR (RT-qPCR), the levels of pro-inflammatory cytokines by enzyme-linked immunosorbent assay (ELISA), and key proteins of TRAF6/MAPK signaling pathway by Western blot. Results ReA weaken cell viability, down-regulate the expression of keratin family members (KRT6 and KRT17), restore cell cycle distribution to normal distribution, inhibit the release of pro-inflammatory cytokines (IL-6, IL-8 and IL-1β) and lower the expression of chemokines (S100A7, S100A9 and CXCL2) by interfering with the interaction between HaCaT cells and IL-17A. Thus, it exerts an anti-psoriatic effect by reducing the inflammatory response and inhibiting abnormal proliferation of HaCaT cells. Mechanistically, ReA inhibited the TRAF6/MAPK signaling pathway activated by IL-17A stimulation in HaCaT cells. Conclusion ReA has in vitro anti-psoriatic effects and may be a new therapeutic agent for psoriasis.
Collapse
Affiliation(s)
- Li-li Yuan
- Department of Dermatology, Taizhou People’s Hospital, Taizhou, Jiangsu, 225300, People’s Republic of China
| | - Chun-yu Cao
- Department of Dermatology, Taizhou People’s Hospital, Taizhou, Jiangsu, 225300, People’s Republic of China
| |
Collapse
|
10
|
Zhu W, Li W, Jiang J, Wang D, Mao X, Zhang J, Zhang X, Chang J, Yao P, Yang X, Da Costa C, Zhang Y, Yu J, Li H, Li S, Chi X, Li N. Chronic salmon calcitonin exerts an antidepressant effect via modulating the p38 MAPK signaling pathway. Front Mol Neurosci 2023; 16:1071327. [PMID: 36969556 PMCID: PMC10036804 DOI: 10.3389/fnmol.2023.1071327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/15/2023] [Indexed: 03/12/2023] Open
Abstract
Depression is a common recurrent psychiatric disorder with a high lifetime prevalence and suicide rate. At present, although several traditional clinical drugs such as fluoxetine and ketamine, are widely used, medications with a high efficiency and reduced side effects are of urgent need. Our group has recently reported that a single administration of salmon calcitonin (sCT) could ameliorate a depressive-like phenotype via the amylin signaling pathway in a mouse model established by chronic restraint stress (CRS). However, the molecular mechanism underlying the antidepressant effect needs to be addressed. In this study, we investigated the antidepressant potential of sCT applied chronically and its underlying mechanism. In addition, using transcriptomics, we found the MAPK signaling pathway was upregulated in the hippocampus of CRS-treated mice. Further phosphorylation levels of ERK/p38/JNK kinases were also enhanced, and sCT treatment was able only to downregulate the phosphorylation level of p38/JNK, with phosphorylated ERK level unaffected. Finally, we found that the antidepressant effect of sCT was blocked by p38 agonists rather than JNK agonists. These results provide a mechanistic explanation of the antidepressant effect of sCT, suggesting its potential for treating the depressive disorder in the clinic.
Collapse
Affiliation(s)
- Wenhui Zhu
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weifen Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jian Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dilong Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xinliang Mao
- Perfect Life and Health Institute, Zhongshan, Guangdong, China
| | - Jin Zhang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xunzhi Zhang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Jinlong Chang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Peijia Yao
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiuyan Yang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | | | - Ying Zhang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiezhong Yu
- The Fourth People’s Hospital of Datong City, Datong, China
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, Division of Medicine, Faculty of Medical Sciences, University College London, London, United Kingdom
- China-UK Institute for Frontier Science, Shenzhen, China
- *Correspondence: Huiliang Li,
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
- Shupeng Li,
| | - Xinjin Chi
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Xinjin Chi,
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- China-UK Institute for Frontier Science, Shenzhen, China
- The Fifth People’s Hospital of Datong City, Datong, China
- Ningning Li,
| |
Collapse
|