1
|
Xu L, Zhao D, Wang M, Xu L, Cao Y, Meng F, Liu J, Di Z, Wang W, Zhang M, Li C, Jiang S. Control of Methamphetamine-Induced Place Preference Behavior by the Glutamatergic Neural Projections from IC to BLA in Mice. Brain Res Bull 2025:111395. [PMID: 40409603 DOI: 10.1016/j.brainresbull.2025.111395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 05/07/2025] [Accepted: 05/18/2025] [Indexed: 05/25/2025]
Abstract
Drug addiction is closely related to the dysregulation of complex neural circuits. However, the neural pathways underlying methamphetamine (METH)-induced addiction remain unclear. Firstly, we performed ∆FosB (Delta FBJ murine osteosarcoma viral oncogene homolog B) immunofluorescence and fiber-optic recording experiments to assess the neural activity of glutamatergic neurons in the anterior insular cortex (IC) in METH-treated mice. Then, we evaluated the effect of activation or inhibition of glutaminergic neurons in IC on METH-induced conditioned place preference (CPP) behavior through chemogenetic manipulation. Finally, we used adeno-associated virus (AAV)-mediated neural tracing to verify the projections connectivity from IC to the basolateral amygdala (BLA) and investigated their blocking role in METH-mediated CPP behavior using chemogenetic and neural ablation strategies. We found that glutamatergic neurons in the IC were activated by METH. Activation of these neurons enhanced METH-mediated CPP behavior, whereas inhibition of their activity attenuated the CPP expression. Furthermore, we observed robust projections from IC neurons to the BLA. Activation of IC neurons projecting to the BLA enhanced METH-mediated CPP behavior, whereas ablation of BLA neurons receiving projections from IC significantly impaired METH-mediated CPP performance. These results highlight the IC glutaminergic neurons are a major target of METH addiction, with the IC-BLA glutaminergic neural projection playing an important role in regulating METH-mediated CPP behavior. This pathway may provide new insights into the pathophysiology of METH addiction and serve as a potential target for therapeutic strategies.
Collapse
Affiliation(s)
- Lei Xu
- Department of Physiology, Binzhou Medical University, Shandong 264003, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Di Zhao
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Meiqin Wang
- Department of Physiology, Binzhou Medical University, Shandong 264003, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Lihong Xu
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Yifan Cao
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Fantao Meng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Jing Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Zhao Di
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Wentao Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Mengdi Zhang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China.
| | - Chen Li
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China.
| | - Shujun Jiang
- Department of Physiology, Binzhou Medical University, Shandong 264003, China.
| |
Collapse
|
2
|
Ke Y, Wang Y, Hu Y. The leptin/BDNF/TrkB signaling pathway improves corticosteroid combined with chronic restraint stress-induced depressive-like behavior in mice. Neurosci Lett 2025; 859-861:138268. [PMID: 40389053 DOI: 10.1016/j.neulet.2025.138268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 05/06/2025] [Accepted: 05/15/2025] [Indexed: 05/21/2025]
Abstract
This study explored the effect and mechanism of leptin on depressive-like behavior induced by chronic corticosterone injections combined with chronic restraint stress (CORT-CRS) in mice. Differentially expressed genes (DEGs) were extracted using the Gene Expression Omnibus database and Sangerbox tool. Construct protein-protein interaction networks of target DEGs using Cytoscape software, and hub genes brain-derived neurotrophic factor (BDNF) were identified. A mouse model of depression was established using CORT-CRS. Behavioral changes were detected in the mice using the tail suspension, forced swimming, sugar water preference (SPT), and open field tests (OFT). Serum inflammatory factors were measured by Enzyme-linked immunosorbent assay (ELISA). Western blotting was used to detect the protein expression levels of BDNF and tyrosine kinase receptor B (TrkB). Immunofluorescence was used to detect hippocampal neurogenesis in each mouse group. Compared with the control group, mice in the CORT-CRS group presented with marked depression-like behavior and a higher interleukin (IL) -6, IL-1 β, tumor necrosis factor alpha (TNF - α) concentration. Different doses of leptin reversed depressive-like behavior in the CORT-CRS model mice, with significantly increased levels of BDNF, TrkB protein expression (P < 0.01). Furthermore, leptin promoted hippocampal neurogenesis in CORT-CRS-treated mice in vivo. Consequently, leptin alleviates CORT-CRS-induced depression-like behavior in mice by stimulating hippocampal neurogenesis. One possible mechanism could be related to the activation of the BDNF/TrkB signaling pathway, which could pave way for novel therapeutic targets that can be explored to prevent and treat depression.
Collapse
Affiliation(s)
- Yunqian Ke
- Department of Psychiatry, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China.
| | - Yiming Wang
- Department of Psychiatry, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Yongxue Hu
- Department of Psychiatry, The Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| |
Collapse
|
3
|
Wang H, Zhang L, Yang WY, Ji XY, Gao AQ, Wei YH, Ding X, Kang Y, Ding JH, Fan Y, Lu M, Hu G. Visceral adipose tissue-derived extracellular vesicles promote stress susceptibility in obese mice via miR-140-5p. Acta Pharmacol Sin 2025; 46:1221-1235. [PMID: 39930136 PMCID: PMC12032276 DOI: 10.1038/s41401-025-01484-z] [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: 09/06/2024] [Accepted: 01/14/2025] [Indexed: 03/17/2025]
Abstract
Obesity increases the risk of depression. Evidence shows that peripheral inflammation, glycemic dysregulation, and hyperactivity within the hypothalamic-pituitary-adrenal axis are implicated in both obesity and depression. In this study we investigated the impact of visceral adipose tissue (VAT), a crucial characteristic of obesity, on stress susceptibility in obese mice. Age-matched mice were fed with chow diet (CD) or high-fat diet (HFD), respectively, for 12 weeks. CD mice were deprived of VAT and received transplantation of VAT from HFD mice (TransHFD) or CD mice (TransCD). Extracellular vesicles (EVs) were prepared from VAT of CD or HFD mice, and intravenously injected (100 μg, 4 times in 2 weeks) in naïve mice or injected into hippocampus (5 μg, 4 times in 2 weeks) through implanted bilateral cannula. Depression-like behaviors were assessed 14 days after transplantation. We showed that HFD mice exhibited significantly higher body weight gain and impaired insulin and glucose tolerance, accompanied by increased stress susceptibility. Transplantation of VAT or VAT-derived EVs from HFD mice caused synaptic damage and promoted stress susceptibility in recipient mice. Through inhibiting miRNA biogenesis in the VAT and miRNA sequencing analysis, we demonstrated that miR-140-5p was significantly upregulated in both VAT-EVs and hippocampus of HFD mice. Overexpression of hippocampal miR-140-5p in naïve mice not only facilitated acute stress-induced depression-like behaviors, but also decreased hippocampal CREB-BDNF signaling cascade and synaptic plasticity. Conversely, knockdown of miR-140-5p in the VAT, VAT-EVs or hippocampus of HFD mice protected against acute stress, reducing stress susceptibility that were mediated via CREB-BDNF pathway. In summary, VAT-EVs or the cargo miRNAs in obese mice promote synaptic damage and stress susceptibility, providing potential therapeutic targets for metabolism-related affective disorders.
Collapse
Affiliation(s)
- Hao Wang
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Li Zhang
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wan-Yue Yang
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiao-Yi Ji
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - An-Qi Gao
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi-Hong Wei
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xin Ding
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yue Kang
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jian-Hua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Yi Fan
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Gang Hu
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
4
|
Liu C, Zhao D, Yu G, Du H, Xu L, Cao Y, Cui M, Wang W, Wang D, Liu J, Meng F, Hu F, Li W, Du J, Li C. Alleviation of Microglia Mediating Hippocampal Neuron Impairments and Depression-Related Behaviors by Urolithin B via the SIRT1-FOXO1 Pathway. CNS Neurosci Ther 2025; 31:e70379. [PMID: 40237232 PMCID: PMC12000931 DOI: 10.1111/cns.70379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/17/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025] Open
Abstract
AIMS Conventional antidepressants exhibit limited efficacy and delayed onset. This study aimed to elucidate the antidepressant effects of urolithin B (UB) and its regulatory role in microglia-mediated hippocampal neuronal dysfunction. METHODS The mouse model of depression was established using both chronic unpredicted stress (CUS) and lipopolysaccharide (LPS) injection. The therapeutic efficacy of UB was assessed through behavioral paradigms. The microglia activation, cellular cytotoxicity and apoptosis levels, and underlying molecular mechanisms were delineated utilizing proteomics analysis, immunofluorescence staining, real-time PCR and Western blotting. RESULTS UB efficiently alleviated depression-related behaviors, accompanied by suppressed microglia activation, neuroinflammation, changes of classic activation (M1)/alternative activation (M2) polarization and recovered sirtuin-1 (SIRT1) and forkhead box protein O1 (FOXO1) expression in the hippocampus. Additionally, UB reduced the cytotoxicity and apoptosis of HT22 cells and depression-related phenotypes treated by the cellular supernatant from LPS-incubated BV2 cells, which was mediated by the SIRT1-FOXO1 pathway. The proteomics analysis of the cellular supernatant content revealed abundant secreting proteins among the LPS/UB application. CONCLUSION This study confirmed that microglial SIRT1 mediates UB's antidepressant effects, positioning UB as a promising therapeutic candidate for depression by targeting neuroinflammatory pathways.
Collapse
Affiliation(s)
- Cuilan Liu
- Department of Rehabilitation MedicineBinzhou Medical University HospitalBinzhouShandongChina
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Di Zhao
- Department of Rehabilitation MedicineBinzhou Medical University HospitalBinzhouShandongChina
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Guoxing Yu
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
| | - HengWei Du
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
| | - Lihong Xu
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Yifan Cao
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Minghu Cui
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Wentao Wang
- Department of Rehabilitation MedicineBinzhou Medical University HospitalBinzhouShandongChina
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Dan Wang
- Department of Rehabilitation MedicineBinzhou Medical University HospitalBinzhouShandongChina
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Jing Liu
- Department of Rehabilitation MedicineBinzhou Medical University HospitalBinzhouShandongChina
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Fantao Meng
- Department of Rehabilitation MedicineBinzhou Medical University HospitalBinzhouShandongChina
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Fengai Hu
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Wei Li
- Department of Rehabilitation MedicineBinzhou Medical University HospitalBinzhouShandongChina
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
| | - Jing Du
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
| | - Chen Li
- Department of Rehabilitation MedicineBinzhou Medical University HospitalBinzhouShandongChina
- Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
- Department of PsychologyBinzhou Medical University HospitalBinzhouShandongChina
| |
Collapse
|
5
|
Wang M, Xu L, Zhao D, Wang W, Xu L, Cao Y, Meng F, Liu J, Li C, Jiang S. The glutamatergic projections from the PVT to mPFC govern methamphetamine-induced conditional place preference behaviors in mice. J Affect Disord 2025; 371:289-304. [PMID: 39579874 DOI: 10.1016/j.jad.2024.11.053] [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: 02/22/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Drug addiction is closely related to the dysregulation of complex neural circuits. However, the neural connections underlying the symptoms of methamphetamine (METH)-induced addiction have yet to be elucidated. METHODS We conducted ΔFosB (Delta FBJ murine osteosarcoma viral oncogene homolog B) associated immunofluorescence and electrophysiological recording experiments to measure the neural activity of paraventricular thalamus (PVT) neurons in METH treated mice. Then, the METH-mediated conditional place preference (CPP) behaviors were evaluated after chemogenetic manipulation of PVT neurons. Additionally, the neural projection from PVT to medial prefrontal cortex (mPFC) was verified through Adeno-associated virus (AAV) mediating neural tracing method, and its role on METH-mediated CPP behaviors was determined using chemogenetic and neural ablation strategies. RESULTS We found that glutamatergic neurons in PVT were activated by METH. Activating the glutamatergic neurons in PVT promoted the METH-mediated CPP behaviors, while inhibiting these neurons attenuated the CPP behaviors. Moreover, we observed PVT neurons showed robust neuronal projections to mPFC, activation of the mPFC→projecting neurons in PVT or the afferent terminals in mPFC derived from PVT enhanced METH-mediated CPP performance, and ablating the mPFC neurons receipting neural projection from PVT impeded these increased METH-mediated CPP phenotypes. LIMITATIONS The underlying molecular mechanism of the dysfunctional PVT neurons after METH treatment and the PVT neurons regulating the activity of mPFC target neurons remains unclear. CONCLUSIONS These results shed light on that PVT is a key METH addiction-controlling nucleus, and PVT → mPFC projection regulated METH-mediated CPP behaviors, which could serve as a vital pathway for morbidity and treatment for METH-mediated addiction.
Collapse
Affiliation(s)
- Meiqin Wang
- Department of Physiology, Binzhou Medical University, Shandong 264003, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Lei Xu
- Department of Physiology, Binzhou Medical University, Shandong 264003, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Di Zhao
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Wentao Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Lihong Xu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Yifan Cao
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Fantao Meng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Jing Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Chen Li
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China.
| | - Shujun Jiang
- Department of Physiology, Binzhou Medical University, Shandong 264003, China.
| |
Collapse
|
6
|
Yang M, Tian S, Han X, Xu L, You J, Wu M, Cao Y, Jiang Y, Zheng Z, Liu J, Meng F, Li C, Wang X. Interleukin-11Rα2 in the hypothalamic arcuate nucleus affects depression-related behaviors and the AKT-BDNF pathway. Gene 2025; 933:148966. [PMID: 39341516 DOI: 10.1016/j.gene.2024.148966] [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: 01/26/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Depression is a widespread emotional disorder with complex pathogenesis. An essential function of the hypothalamus is to regulate emotional disorders. However, further investigation is required to identify the pathogenic genes and molecular mechanisms that contribute to the onset of depression within the hypothalamus. Through RNA-sequencing analysis, this study identified the upregulated expression of interleukin-11 receptor alpha 2 (IL-11Rα2) in the hypothalamus of mice with chronic unpredictable stress (CUS)-induced depression. This substantial increase in IL-11Rα2, not IL-11Rα1 expression levels in the hypothalamus under the influence of CUS was found to be associated with depression-related behaviors. We further showed that IL-11Rα2 is expressed in the arcuate nucleus (ARC) proopiomelanocortin (POMC) neurons of the hypothalamus. Male and female mice exhibited behaviors association with depression, when IL-11Rα2 or its ligand IL-11 was overexpressed in the ARC POMC neurons through the action of an adeno-associated virus. In addition, reductions in the expression levels of proteins involved in the protein kinase B signaling pathways and brain-derived neurotrophic factor were observed upon overexpression of IL-11Rα2 in the hypothalamic ARC. This study emphasizes the importance of IL-11Rα2 in the hypothalamus ARC in the development of depression, and presents it as a potential novel target for depression treatment.
Collapse
Affiliation(s)
- Mengyu Yang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shulei Tian
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiaofeng Han
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lihong Xu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jingjing You
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Min Wu
- Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yifan Cao
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yuting Jiang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ziteng Zheng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Chen Li
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Xuezhen Wang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
7
|
Zhang Z, He Z, Pan J, Yuan M, Lang Y, Wei X, Zhang C. The interaction of BDNF with estrogen in the development of hypertension and obesity, particularly during menopause. Front Endocrinol (Lausanne) 2024; 15:1384159. [PMID: 39655343 PMCID: PMC11625588 DOI: 10.3389/fendo.2024.1384159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
The expression of BDNF in both neuronal and non-neuronal cells is influenced by various stimuli, including prenatal developmental factors and postnatal conditions such as estrogens, dietary habits, and lifestyle factors like obesity, blood pressure, and aging. Central BDNF plays a crucial role in modulating how target tissues respond to these stimuli, influencing the pathogenesis of hypertension, mitigating obesity, and protecting neurons from aging. Thus, BDNF serves as a dynamic mediator of environmental influences, reflecting an individual's unique history of exposure. Estrogens, on the other hand, regulate various processes to maintain overall physiological well-being. Through nuclear estrogen receptors (ERα, ERβ) and the membrane estrogen receptor (GPER1), estrogens modulate transcriptional processes and signaling events that regulate the expression of target genes, such as ERα, components of the renin-angiotensin system (RAS), and hormone-sensitive lipase. Estrogens are instrumental in maintaining the set point for blood pressure and energy balance. BDNF and estrogens work cooperatively to prevent obesity by favoring lipolysis, and counteractively regulate blood pressure to adapt to the environment. Estrogen deficiency leads to menopause in women with low central BDNF level. This review delves into the complex mechanisms involving BDNF and estrogen, especially in the context of hypertension and obesity, particularly among postmenopausal women. The insights gained aim to inform the development of comprehensive therapeutic strategies for these prevalent syndromes affecting approximately 68% of adults.
Collapse
Affiliation(s)
- Zhongming Zhang
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
- School of Medicine, Zhengzhou University of Industrial Technology, Xinzheng, Henan, China
| | - Ziyi He
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Jing Pan
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Minghui Yuan
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Yini Lang
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Xiaomeng Wei
- School of Medicine, Zhengzhou University of Industrial Technology, Xinzheng, Henan, China
| | - Chaoyun Zhang
- Zhang Zhongjing College of Chinese Medicine, Henan Key Laboratory of Zhang Zhongjing’s Formulas for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| |
Collapse
|
8
|
Carrillo F, Palomba NP, Ghirimoldi M, Didò C, Fortunato G, Khoso S, Giloni T, Santilli M, Bocci T, Priori A, Pietracupa S, Modugno N, Barberis E, Manfredi M, Signorelli P, Esposito T. Multiomics approach discloses lipids and metabolites profiles associated to Parkinson's disease stages and applied therapies. Neurobiol Dis 2024; 202:106698. [PMID: 39427845 DOI: 10.1016/j.nbd.2024.106698] [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: 08/07/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024] Open
Abstract
Profiling circulating lipids and metabolites in Parkinson's disease (PD) patients could be useful not only to highlight new pathways affected in PD condition but also to identify sensitive and effective biomarkers for early disease detection and potentially effective therapeutic interventions. In this study we adopted an untargeted omics approach in three groups of patients (No L-Dopa, L-Dopa and DBS) to disclose whether long-term levodopa treatment with or without deep brain stimulation (DBS) could reflect a characteristic lipidomic and metabolomic signature at circulating level. Our findings disclosed a wide up regulation of the majority of differentially regulated lipid species that increase with disease progression and severity. We found a relevant modulation of triacylglycerols and acyl-carnitines, together with an altered profile in adiponectin and leptin, that can differentiate the DBS treated group from the others PD patients. We found a highly significant increase of exosyl ceramides (Hex2Cer) and sphingoid bases (SPB) in PD patients mainly in DBS group (p < 0.0001), which also resulted in a highly accurate diagnostic performance. At metabolomic level, we found a wide dysregulation of pathways involved in the biosynthesis and metabolism of several amino acids. The most interesting finding was the identification of a specific modulation of L-glutamic acid in the three groups of patients. L-glutamate levels increased slightly in No L-Dopa and highly in L-Dopa patients while decreased in DBS, suggesting that DBS therapy might have a beneficial effect on the glutamatergic cascade. All together, these data provide novel insights into the molecular and metabolic alterations underlying PD therapy and might be relevant for PD prediction, diagnosis and treatment.
Collapse
Affiliation(s)
- Federica Carrillo
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council, Naples, Italy
| | | | - Marco Ghirimoldi
- Biological Mass Spectrometry Lab, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Camilla Didò
- Biological Mass Spectrometry Lab, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Giorgio Fortunato
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council, Naples, Italy
| | - Shahzaib Khoso
- Biological Mass Spectrometry Lab, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | | | | | - Tommaso Bocci
- "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| | - Alberto Priori
- "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; Clinical Neurology Unit, "Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo", Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| | - Sara Pietracupa
- IRCCS INM Neuromed, Pozzilli, IS, Italy; Department of Human Neuroscience, Sapienza University of Rome, Italy
| | | | - Elettra Barberis
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy; Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | - Marcello Manfredi
- Biological Mass Spectrometry Lab, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Paola Signorelli
- "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; Biochemistry Laboratory, IRCCS Policlinico San Donato, Milano Italy
| | - Teresa Esposito
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council, Naples, Italy; IRCCS INM Neuromed, Pozzilli, IS, Italy.
| |
Collapse
|
9
|
Valladolid-Acebes I. Hippocampal Leptin Resistance and Cognitive Decline: Mechanisms, Therapeutic Strategies and Clinical Implications. Biomedicines 2024; 12:2422. [PMID: 39594988 PMCID: PMC11591892 DOI: 10.3390/biomedicines12112422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Leptin, an adipokine essential for regulating energy balance, exerts important effects on brain function, notably within the hippocampus, a region integral to learning and memory. Leptin resistance, characterized by diminished responsiveness to elevated leptin levels, disrupts hippocampal function and exacerbates both obesity and cognitive impairments. Scope: This review critically examines how leptin resistance impairs hippocampal synaptic plasticity processes, specifically affecting long-term potentiation (LTP) and long-term depression (LTD), which are crucial for cognitive performance. Findings: Recent research highlights that leptin resistance disrupts N-methyl-D-aspartate (NMDA) receptor dynamics and hippocampal structure, leading to deficits in spatial learning and memory. Additionally, high-fat diets (HFDs), which contribute to leptin resistance, further deteriorate hippocampal function. Potential therapeutic strategies, including leptin sensitizers, show promise in mitigating brain disorders associated with leptin resistance. Complementary interventions such as caloric restriction and physical exercise also enhance leptin sensitivity and offer potential benefits to alleviating cognitive impairments. Aims of the review: This review synthesizes recent findings on the molecular pathways underlying leptin resistance and its impact on synaptic transmission and plasticity in the hippocampus. By identifying potential therapeutic targets, this work aims to provide an integrated approach for addressing cognitive deficits in obesity, ultimately improving the quality of life for affected individuals.
Collapse
Affiliation(s)
- Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| |
Collapse
|
10
|
Wang D, Zhao D, Wang W, Hu F, Cui M, Liu J, Meng F, Liu C, Qiu C, Liu D, Xu Z, Wang Y, Zhang Y, Li W, Li C. How do lateral septum projections to the ventral CA1 influence sociability? Neural Regen Res 2024; 19:1789-1801. [PMID: 38103246 PMCID: PMC10960288 DOI: 10.4103/1673-5374.389304] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/10/2023] [Accepted: 08/02/2023] [Indexed: 12/18/2023] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202408000-00033/figure1/v/2023-12-16T180322Z/r/image-tiff Social dysfunction is a risk factor for several neuropsychiatric illnesses. Previous studies have shown that the lateral septum (LS)-related pathway plays a critical role in mediating social behaviors. However, the role of the connections between the LS and its downstream brain regions in social behaviors remains unclear. In this study, we conducted a three-chamber test using electrophysiological and chemogenetic approaches in mice to determine how LS projections to ventral CA1 (vCA1) influence sociability. Our results showed that gamma-aminobutyric acid (GABA)-ergic neurons were activated following social experience, and that social behaviors were enhanced by chemogenetic modulation of these neurons. Moreover, LS GABAergic neurons extended their functional neural connections via vCA1 glutamatergic pyramidal neurons, and regulating LSGABA→vCA1Glu neural projections affected social behaviors, which were impeded by suppressing LS-projecting vCA1 neuronal activity or inhibiting GABAA receptors in vCA1. These findings support the hypothesis that LS inputs to the vCA1 can control social preferences and social novelty behaviors. These findings provide new insights regarding the neural circuits that regulate sociability.
Collapse
Affiliation(s)
- Dan Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Di Zhao
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Wentao Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Fengai Hu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Minghu Cui
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Jing Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Fantao Meng
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Cuilan Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Changyun Qiu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Dunjiang Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Zhicheng Xu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Yameng Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Yu Zhang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- College of Nursing, Binzhou Medical University, Binzhou, Shandong Province, China
| | - Wei Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Chen Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| |
Collapse
|
11
|
Jiang Y, Xu L, Cao Y, Meng F, Jiang S, Yang M, Zheng Z, Zhang Y, Yang L, Wang M, Sun G, Liu J, Li C, Cui M. Effects of Interleukin-19 overexpression in the medial prefrontal cortex on anxiety-related behaviors, BDNF expression and p38/JNK/ERK pathways. Brain Res Bull 2024; 212:110952. [PMID: 38636611 DOI: 10.1016/j.brainresbull.2024.110952] [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: 01/05/2024] [Revised: 03/27/2024] [Accepted: 04/14/2024] [Indexed: 04/20/2024]
Abstract
Anxiety is a prevalent mental illness known for its high incidence, comorbidity, and tendency to recur, posing significant societal and individual burdens. Studies have highlighted Interleukin-19 (IL-19) as having potential relevance in neuropsychiatric disorders. Our previous research revealed that IL-19 overexpression in colonies exacerbated anxiety-related behaviors induced by dextran sodium sulfate/stress. However, the precise role and molecular mechanisms of IL-19 in anxiety regulation remain uncertain. In this study, we initiated an acute restraint stress (ARS)-induced anxious mouse model and identified heightened expression of IL-19 and IL-20Rα in the medial prefrontal cortex (mPFC) of ARS mice. Notably, IL-19 and IL-20Rα were predominantly present in the excitatory pyramidal neurons of the mPFC under both basal and ARS conditions. Utilizing the adeno-associated virus (AAV) strategy, we demonstrated that IL-19 overexpression in the mPFC induced anxiety-related behaviors and elevated stress susceptibility. Additionally, we observed decreased protein levels of brain-derived neurotrophic factor (BDNF) and postsynaptic density protein 95 (PSD95) in the mPFC of IL-19 overexpression mice, accompanied by reduced phosphorylation of in the p38, JNK, and Erk signaling pathways. These findings emphasize the role of IL-19 in modulating anxiety-related behaviors within the mPFC and suggest its potential as a pathological gene and therapeutic target for anxiety.
Collapse
Affiliation(s)
- Yuting Jiang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lihong Xu
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yifan Cao
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shujun Jiang
- Department of Physiology, Binzhou Medical University, Shandong, China
| | - Mengyu Yang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ziteng Zheng
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yi Zhang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lu Yang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Meiqin Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Physiology, Binzhou Medical University, Shandong, China
| | - Guizhi Sun
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Chen Li
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Minghu Cui
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
12
|
Chen HS, Wang F, Chen JG. Epigenetic mechanisms in depression: Implications for pathogenesis and treatment. Curr Opin Neurobiol 2024; 85:102854. [PMID: 38401316 DOI: 10.1016/j.conb.2024.102854] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 02/26/2024]
Abstract
The risk of depression is influenced by both genetic and environmental factors. It has been suggested that epigenetic mechanisms may mediate the risk of depression following exposure to adverse life events. Epigenetics encompasses stable alterations in gene expression that are controlled through transcriptional, post-transcriptional, translational, or post-translational processes, including DNA modifications, chromatin remodeling, histone modifications, RNA modifications, and non-coding RNA (ncRNA) regulation, without any changes in the DNA sequence. In this review, we explore recent research advancements in the realm of epigenetics concerning depression. Furthermore, we evaluate the potential of epigenetic changes as diagnostic and therapeutic biomarkers for depression.
Collapse
Affiliation(s)
- Hong-Sheng Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan 430030, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan 430030, China; The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, China
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan 430030, China; The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, China.
| |
Collapse
|
13
|
Ge X, Hu M, Zhou M, Fang X, Chen X, Geng D, Wang L, Yang X, An H, Zhang M, Lin D, Zheng M, Cui X, Wang Q, Wu Y, Zheng K, Huang XF, Yu Y. Overexpression of forebrain PTP1B leads to synaptic and cognitive impairments in obesity. Brain Behav Immun 2024; 117:456-470. [PMID: 38336024 DOI: 10.1016/j.bbi.2024.02.008] [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: 11/23/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024] Open
Abstract
Obesity has reached pandemic proportions and is a risk factor for neurodegenerative diseases, including Alzheimer's disease. Chronic inflammation is common in obese patients, but the mechanism between inflammation and cognitive impairment in obesity remains unclear. Accumulative evidence shows that protein-tyrosine phosphatase 1B (PTP1B), a neuroinflammatory and negative synaptic regulator, is involved in the pathogenesis of neurodegenerative processes. We investigated the causal role of PTP1B in obesity-induced cognitive impairment and the beneficial effect of PTP1B inhibitors in counteracting impairments of cognition, neural morphology, and signaling. We showed that obese individuals had negative relationship between serum PTP1B levels and cognitive function. Furthermore, the PTP1B level in the forebrain increased in patients with neurodegenerative diseases and obese cognitive impairment mice with the expansion of white matter, neuroinflammation and brain atrophy. PTP1B globally or forebrain-specific knockout mice on an obesogenic high-fat diet showed enhanced cognition and improved synaptic ultrastructure and proteins in the forebrain. Specifically, deleting PTP1B in leptin receptor-expressing cells improved leptin synaptic signaling and increased BDNF expression in the forebrain of obese mice. Importantly, we found that various PTP1B allosteric inhibitors (e.g., MSI-1436, well-tolerated in Phase 1 and 1b clinical trials for obesity and type II diabetes) prevented these alterations, including improving cognition, neurite outgrowth, leptin synaptic signaling and BDNF in both obese cognitive impairment mice and a neural cell model of PTP1B overexpression. These findings suggest that increased forebrain PTP1B is associated with cognitive decline in obesity, whereas inhibition of PTP1B could be a promising strategy for preventing neurodegeneration induced by obesity.
Collapse
Affiliation(s)
- Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Menglu Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoli Fang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu 221006, China
| | - Xi Chen
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, NSW 2522, Australia
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu 221006, China
| | - Li Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Huimei An
- HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Peking University, Beijing 10096, China
| | - Meng Zhang
- HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Peking University, Beijing 10096, China
| | - Danhong Lin
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoying Cui
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4113, Australia; Queensland Centre for Mental Health Research, Wacol, QLD, 4076, Australia
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China.
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Xu-Feng Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Illawarra Health and Medical Research Institute (IHMRI) and School of Medical, Indigenous, and Health, University of Wollongong, NSW 2522, Australia.
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
14
|
Liu J, Meng F, Wang W, Wu M, Zhang Y, Cui M, Qiu C, Hu F, Zhao D, Wang D, Liu C, Liu D, Xu Z, Wang Y, Li W, Li C. Medial prefrontal cortical PPM1F alters depression-related behaviors by modifying p300 activity via the AMPK signaling pathway. CNS Neurosci Ther 2023; 29:3624-3643. [PMID: 37309288 PMCID: PMC10580341 DOI: 10.1111/cns.14293] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/14/2023] Open
Abstract
AIMS Protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F) is a serine/threonine phosphatase, and its dysfunction in depression in the hippocampal dentate gyrus has been previously identified. Nevertheless, its role in depression of another critical emotion-controlling brain region, the medial prefrontal cortex (mPFC), remains unclear. We explored the functional relevance of PPM1F in the pathogenesis of depression. METHODS The gene expression levels and colocalization of PPM1F in the mPFC of depressed mice were measured by real-time PCR, western blot and immunohistochemistry. An adeno-associated virus strategy was applied to determine the impact of knockdown or overexpression of PPM1F in the excitatory neurons on depression-related behaviors under basal and stress conditions in both male and female mice. The neuronal excitability, expression of p300 and AMPK phosphorylation levels in the mPFC after knockdown of PPM1F were measured by electrophysiological recordings, real-time PCR and western blot. The depression-related behavior induced by PPM1F knockdown after AMPKα2 knockout or the antidepressant activity of PPM1F overexpression after inhibiting acetylation activity of p300 was evaluated. RESULTS Our results indicate that the expression levels of PPM1F were largely decreased in the mPFC of mice exposed to chronic unpredictable stress (CUS). Behavioral alterations relevant to depression emerged with short hairpin RNA (shRNA)-mediated genetic knockdown of PPM1F in the mPFC, while overexpression of PPM1F produced antidepressant activity and ameliorated behavioral responses to stress in CUS-exposed mice. Molecularly, PPM1F knockdown decreased the excitability of pyramidal neurons in the mPFC, and restoring this low excitability decreased the depression-related behaviors induced by PPM1F knockdown. PPM1F knockdown reduced the expression of CREB-binding protein (CBP)/E1A-associated protein (p300), a histone acetyltransferase (HAT), and induced hyperphosphorylation of AMPK, resulting in microglial activation and upregulation of proinflammatory cytokines. Conditional knockout of AMPK revealed an antidepressant phenotype, which can also block depression-related behaviors induced by PPM1F knockdown. Furthermore, inhibiting the acetylase activity of p300 abolished the beneficial effects of PPM1F elevation on CUS-induced depressive behaviors. CONCLUSION Our findings demonstrate that PPM1F in the mPFC modulates depression-related behavioral responses by regulating the function of p300 via the AMPK signaling pathway.
Collapse
|
15
|
Musillo C, Giona L, Ristow M, Zarse K, Siems K, Di Francesco A, Collacchi B, Raggi C, Cirulli F, Berry A. Rosmarinic Acid Improves Cognitive Abilities and Glucose Metabolism in Aged C57Bl/6N Mice While Disrupting Lipid Profile in Young Adults in a Sex-Dependent Fashion. Nutrients 2023; 15:3366. [PMID: 37571303 PMCID: PMC10421458 DOI: 10.3390/nu15153366] [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: 07/03/2023] [Revised: 07/18/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
A growing body of evidence suggests that regular consumption of natural products might promote healthy aging; however, their mechanisms of action are still unclear. Rosmarinic acid (RA) is a polyphenol holding anti-inflammatory, antioxidant and neuroprotective properties. The aim of this study was to characterise the efficacy of an oral administration of RA in promoting healthspan in a mouse model of physiological aging. Aged C57Bl/6 male and female (24-month-old) mice were either administered with RA (500 mg/Kg) or a vehicle in drinking bottles for 52 days while 3-month-old mice receiving the same treatment were used as controls. All subjects were assessed for cognitive abilities in the Morris water maze (MWM) and for emotionality in the elevated-plus maze test (EPM). Brain-derived Neurotrophic Factor (BDNF) protein levels were evaluated in the hippocampus. Since the interaction between metabolic signals and cerebral functions plays a pivotal role in the etiopathogenesis of cognitive decline, the glycaemic and lipid profiles of the mice were also assessed. RA enhanced learning and memory in 24-month-old mice, an effect that was associated to improved glucose homeostasis. By contrast, the lipid profile was disrupted in young adults. This effect was associated with worse glycaemic control in males and with reduced BDNF levels in females, suggesting powerful sex-dependent effects and raising a note of caution for RA administration in young healthy adult subjects.
Collapse
Affiliation(s)
- Chiara Musillo
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (C.M.); (L.G.); (A.D.F.); (B.C.)
| | - Letizia Giona
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (C.M.); (L.G.); (A.D.F.); (B.C.)
| | - Michael Ristow
- Institute of Experimental Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (M.R.); (K.Z.)
| | - Kim Zarse
- Institute of Experimental Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany; (M.R.); (K.Z.)
| | | | - Alessia Di Francesco
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (C.M.); (L.G.); (A.D.F.); (B.C.)
| | - Barbara Collacchi
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (C.M.); (L.G.); (A.D.F.); (B.C.)
| | - Carla Raggi
- National Center for the Control and Evaluation of Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Francesca Cirulli
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (C.M.); (L.G.); (A.D.F.); (B.C.)
| | - Alessandra Berry
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (C.M.); (L.G.); (A.D.F.); (B.C.)
| |
Collapse
|
16
|
Yang S, Fan L, Zhang R, Song C, Shi J, Wang J, Zhang P, Wang H, Zhang Y. Smilagenin induces expression and epigenetic remodeling of BDNF in alzheimer's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154956. [PMID: 37499345 DOI: 10.1016/j.phymed.2023.154956] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/22/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Smilagenin (SMI) is a lipid-soluble steroidal sapogenin, extracted from traditional Chinses medicinal herbs Radix Asparagi, which is extracted from the dry root of Asparagus cochinchinensis (Lour.) Merr. We previously found that SMI significantly increased brain-derived neurotrophic factor (BDNF) expression in Aβ-intoxicated SH-SY5Y cells. METHODS In this study, we performed behavioral tests to analyze cognitive function of WT and APP/PS1 mice treated with or without SMI, and found that SMI could significantly improve the learning and memory ability of APP/PS1 mice. Moreover, immunofluorescence and ELISA results showed that SMI pretreatment could effectively reduce the deposition of β-amyloid plaques in the cortex and hippocampus of APP/PS1 mice (26 mg/kg/day for 60 days) and inhibit the secretion of Aβ1-42 in N2a/APPswe cells (10 μM concentration for 24 hours). RESULTS Mechanistically, SMI enhanced BDNF mRNA expression, elevated the global level of H3AC and H4AC, and increased the expression of P300 in AD models. Furthermore, chromatin immunoprecipitation results showed that SMI could increase the levels of H3AC and H4AC at the promoter of BDNF promoter Ⅱ and Ⅳ, indicating that SMI epigenetically regulates BDNF expression through HAT enhancement. To further verify the critical role of P300 by which SMI upregulated histone acetylation in BDNF, AD mice were treated with SMI and C646 simultaneously. Behavioral experiments showed that the improvement effects of SMI on cognitive impairment were abolished after P300 inhibition in APP/PS1 mice. CONCLUSIONS Our research for the first time demonstrated that SMI showed neuroprotective effects by increasing the expression of P300 protein, thus upregulating histone acetylation levels in the promoter region of BDNF and promoting its transcription. Our findings provide an important theoretical basis for the treatment of Alzheimer's disease with SMI extracted from Asparagus cochinchinensis (Lour.) Merr.
Collapse
Affiliation(s)
- Shuangshuang Yang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Central Laboratory, Shanghai Xuhui Central Hospital, Shanghai 200031, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Shanghai Engineering Research Center of Phase I Clinical Research & Quality Consistency Evaluation for Drugs, Shanghai 200031, China; Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lei Fan
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rui Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chenghuan Song
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiyun Shi
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Pingao Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hao Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yongfang Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
17
|
Hajizadeh Tekmeh H, Vanizor Kural B, Kör S, Arıkan Malkoç M, Yuluğ E, Kutlu A, Abidin İ, Orem A. How does l-theanine treatment affect the levels of serum and hippocampal BDNF, insulin and adipocytokines in diabetic rats? Biochem Biophys Res Commun 2023; 667:95-103. [PMID: 37210871 DOI: 10.1016/j.bbrc.2023.05.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND Diabetes Mellitus (DM), a metabolic disease characterized by the increased blood glucose level, insulin deficiency or ineffectiveness, may cause structural and functional disorders in the brain. l-Theanine (LTN) has the relaxing, psychoactive, antidepressant, anti-inflammatory and antinecrotic properties, and regulates the functions of hippocampus (HP) in brain. In the present study, the aim was to identify the effects LTN on the levels of BDNF, insulin and adipocytokines (TNF-α, leptin, adiponectin and resistin) in both HP and serum of diabetic rats. METHODS 32 male Wistar rats were divided into four groups (n = 8/group): Control, LTN, DM and DM + LTN. Diabetes was induced by by nicotinamide/streptozotocin. 200 mg/kg/day LTN treatment was applied for 28 days. The serum and hippocampal levels of the parameters were determined by using commercial ELISA kits. Additionally, HP tissues examined histopathologically. RESULTS LTN treatment significantly decreased leptin and adiponectin levels in HP tissues in diabetic rats (p < 0.05). Although it decreased the insulin level in both serum and HP, this was not statistically significant. No significant effect on other parameters was observed (p > 0.05). In histopathological analysis, although the damage was reduced by LTN in all sections of HP, this change was significant mainly in CA3 region (p < 0.05). CONCLUSION It was concluded that LTN has the ability to reduce hippocampal degeneration and modulates adipocytokines in diabetic rats.
Collapse
Affiliation(s)
- Hamed Hajizadeh Tekmeh
- Department of Medical Biochemistry, Graduate School of Health Sciences, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Birgul Vanizor Kural
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Sevil Kör
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Meltem Arıkan Malkoç
- Vocational School of Health Sciences, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Esin Yuluğ
- Department of Histology and Embryology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Abdulkadir Kutlu
- Department of Histology and Embryology, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - İsmail Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| | - Asım Orem
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080, Trabzon, Turkey.
| |
Collapse
|
18
|
Ma X, Li Q, Chen G, Xie J, Wu M, Meng F, Liu J, Liu Y, Zhao D, Wang W, Wang D, Liu C, Dai J, Li C, Cui M. Role of Hippocampal miR-132-3p in Modifying the Function of Protein Phosphatase Mg2+/Mn2+-dependent 1 F in Depression. Neurochem Res 2023:10.1007/s11064-023-03926-8. [PMID: 37036545 DOI: 10.1007/s11064-023-03926-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/15/2023] [Accepted: 03/28/2023] [Indexed: 04/11/2023]
Abstract
Depression is a common, severe, and debilitating psychiatric disorder of unclear etiology. Our previous study has shown that protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F) in the hippocampal dentate gyrus (DG) displays significant regulatory effects in depression-related behaviors. miR-132-3p plays a potential role in the etiology of depression. This study explored the effect of miR-132-3p on the onset of depression and the possible underlying mechanism for modulating PPM1F expression during the pathology of depression. We found that miR-132-3p levels in the hippocampus of depressed mice subjected to chronic unpredictable stress (CUS) were dramatically reduced, which were correlated with depression-related behaviors. Knockdown of miR-132-3p in hippocampal DG resulted in depression-related phenotypes and increased susceptibility to stress. miR-132-3p overexpression in hippocampal DG alleviated CUS-induced depression-related performance. We then screened out the potential target genes of miR-132-3p, and we found that the expression profiles of sterol regulatory element-binding transcription factor 1 (Srebf1) and forkhead box protein O3a (FOXO3a) were positively correlated with PPM1F under the condition of miR-132-3p knockdown. Finally, as anticipated, we revealed that the activities of Ca2+/calmodulin-dependent protein kinase II (CAMKII) and adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) were reduced, which underlies the target signaling pathway of PPM1F. In conclusion, our study suggests that miR-132-3p was designed to regulate depression-related behaviors by indirectly regulating PPM1F and targeting Srebf1 and FOXO3a, which have been linked to the pathogenesis and treatment of depression.
Collapse
Affiliation(s)
- Xiangxian Ma
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Qiongyu Li
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Guanhong Chen
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- The first clinical medical college, Binzhou Medical University, Yantai, Shandong, China
| | - Junjie Xie
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- The first clinical medical college, Binzhou Medical University, Yantai, Shandong, China
| | - Min Wu
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yong Liu
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Physiology, Binzhou Medical University, Shandong, China
| | - Di Zhao
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wentao Wang
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Dan Wang
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Cuilan Liu
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Juanjuan Dai
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China.
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China.
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Minghu Cui
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China.
- Medical research center, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China.
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
19
|
Sun Z, Wang M, Xu L, Li Q, Zhao Z, Liu X, Meng F, Liu J, Wang W, Li C, Jiang S. PPARγ/Adiponectin axis attenuates methamphetamine-induced conditional place preference via the hippocampal AdipoR1 signaling pathway. Prog Neuropsychopharmacol Biol Psychiatry 2023; 125:110758. [PMID: 36972780 DOI: 10.1016/j.pnpbp.2023.110758] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
Methamphetamine (METH) is a highly addictive psychostimulant. The adipocyte-derived hormone adiponectin has a broad spectrum of functions in the brain. However, limited research has been conducted on the effect of adiponectin signaling on METH-induced conditioned place preference (CPP) and knowledge of the underlying neural mechanisms is scarce. The METH induced adult male C57/BL6J mice model were used for testing the therapeutic activities of intraperitoneal injection of AdipoRon or Rosiglitazone, and AdipoR1 overexpression in hippocampal dentate gyrus (DG), and chemogenetic inhibiting the neural activity of DG, and the changes of neurotrophic factors, synaptic molecules, and glutamate receptors, and inflammatory cytokines were also measured. We found that adiponectin expression was significantly reduced in METH addicted patients and mice. Our findings also showed that injection of AdipoRon or Rosiglitazone alleviated the METH-induced CPP behavior. Moreover, the expression of AdipoR1 in the hippocampus was also reduced, and AdipoR1 overexpression blocked the development of METH-induced CPP behavior through regulatory effects on neurotrophic factors, synaptic molecules, and glutamate receptors. The observed inhibitory neural activity of the hippocampal dentate gyrus (DG) induced via a chemogenetic approach produced a therapeutic effect on the METH-induced CPP behavior. Finally, we identified an abnormal expression of some key inflammatory cytokines through the PPARγ/Adiponectin/AdipoR1 axis. This study demonstrates that adiponectin signaling is a promising diagnostic and therapeutic target for METH addiction.
Collapse
Affiliation(s)
- Zongyue Sun
- Department of Physiology, Binzhou Medical University, Shandong 264003, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Meiqin Wang
- Department of Physiology, Binzhou Medical University, Shandong 264003, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Lei Xu
- Department of Physiology, Binzhou Medical University, Shandong 264003, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Qiongyu Li
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Zhongyi Zhao
- Department of Physiology, Binzhou Medical University, Shandong 264003, China
| | - Xuehao Liu
- Department of Physiology, Binzhou Medical University, Shandong 264003, China
| | - Fantao Meng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Jing Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Wentao Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Chen Li
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China.
| | - Shujun Jiang
- Department of Physiology, Binzhou Medical University, Shandong 264003, China.
| |
Collapse
|
20
|
Trinh S, Keller L, Herpertz-Dahlmann B, Seitz J. The role of the brain-derived neurotrophic factor (BDNF) in anorexia nervosa. Psychoneuroendocrinology 2023; 151:106069. [PMID: 36878115 DOI: 10.1016/j.psyneuen.2023.106069] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 01/28/2023] [Accepted: 02/20/2023] [Indexed: 03/08/2023]
Abstract
The brain-derived neurotrophic factor (BDNF) is a growth factor belonging to the neurotrophin family which plays a pivotal role in the differentiation, survival, and plasticity of neurons in the central nervous system. Evidence suggests that BDNF is an important signal molecule in the regulation of energy balance and thus implicated in body weight control. The discovery of BDNF-expressing neurons in the paraventricular hypothalamus which is important in the regulation of energy intake, physical activity, and thermogenesis gives more evidence to the suggested participation of BDNF in eating behavior. Until now it remains questionable whether BDNF can be used as a reliable biomarker for eating disorders such as anorexia nervosa (AN) as available findings on BDNF levels in patients with AN are ambiguous. AN is an eating disorder characterized by a pathological low body weight in combination with a body image disturbance typically developing during adolescence. A severe drive for thinness leads to restrictive eating behavior often accompanied by physical hyperactivity. During therapeutic weight restoration an increase of BDNF expression levels seems desirable as it might improve neuronal plasticity and survival which is essential for learning processes and thereby essential for the success of the psychotherapeutic treatment of patients. On the contrary, the well-known anorexigenic effect of BDNF might favor relapse in patients as soon as the BDNF levels significantly increase during weight rehabilitation. The present review summarizes the association between BDNF and general eating behavior and especially focuses on the eating disorder AN. In this regard findings from preclinical AN studies (activity-based anorexia model) are outlined as well.
Collapse
Affiliation(s)
- Stefanie Trinh
- Institute for Neuroanatomy, University Hospital, RWTH University Aachen, Wendlingweg 2, Aachen D-52074, Germany.
| | - Lara Keller
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, RWTH University Aachen, Neuenhofer Weg 21, Aachen D-52074, Germany.
| | - Beate Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, RWTH University Aachen, Neuenhofer Weg 21, Aachen D-52074, Germany.
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, RWTH University Aachen, Neuenhofer Weg 21, Aachen D-52074, Germany.
| |
Collapse
|
21
|
A comparative study of Western, high-carbohydrate, and standard lab diet consumption throughout adolescence on metabolic and anxiety-related outcomes in young adult male and female Long-Evans rats. Behav Brain Res 2023; 438:114184. [PMID: 36336161 DOI: 10.1016/j.bbr.2022.114184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/11/2022] [Accepted: 10/27/2022] [Indexed: 11/05/2022]
Abstract
Anxiety and obesity are prevalent health concerns that are affected by diet in rodents and humans. How diet influences the development and maintenance of anxiety and obesity has been challenging to characterize, in part, due to methodological differences in chosen experimental and control diets. Within the same experiment, anxiety- and obesity-related effects were characterized in rats fed a Western diet (WD) relative to two control diets. Sixty Long-Evans rats split equally by sex were given standard diet (SD), control (i.e., high-carbohydrate) diet (HCD), or WD from weaning until sacrifice in early adulthood. Anxiety-related behavior was characterized in a modified open field test (mOFT) that allowed for the measurement of defensive behaviors (e.g., hiding within a refuge area), in addition to traditional OF measures (e.g., time in center). Both anxiety-related behaviors and hippocampal CA3 BDNF revealed specific sex differences. Neither adolescent weight gain of male and female rats, nor total body weight in early adulthood, were dependent on administration of HCD or WD, although the WD group consumed the most calories. In males only, administration of either WD or HCD resulted in elevated leptin levels relative to administration of the SD. Results indicate that SDs and HCDs are two distinct types of control diets that can affect comparability of studies and that using an SD might reveal more subtle metabolic changes. Control diet choice should be strongly considered during study design and interpretation, depending on specific research goals. Such studies should include both males and females as these effects are sex-specific.
Collapse
|
22
|
Li Q, Meng F, Ma X, Sun Z, Dai J, Liu J, Li D, Cong P, Xu R, Zhao D, Wang W, Wang D, Liu C, Wang F, Li C, Lian H. The colonic interleukin-19 aggravates the dextran sodium sulfate/stress-induced comorbidities due to colitis and anxiety. Front Immunol 2023; 14:1153344. [PMID: 36936941 PMCID: PMC10018752 DOI: 10.3389/fimmu.2023.1153344] [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: 01/29/2023] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
Comorbidities due to inflammatory bowel disease (IBD) and anxiety are commonly acknowledged; however, their underlying basis is unclear. In the current study, we first conducted a clinical retrospective analysis to identify the enhancive incidence rate of IBD before or after the epidemic of Corona Virus Disease 2019 (COVID-19), with higher Generalized Anxiety Disorder-7 (GAD-7), as well as poorer Gastrointestinal Quality of Life Index (GIQLI). Then, the dextran sodium sulfate (DSS) and chronic unpredictable stress (CUS)-induced IBD and anxiety comorbid models were established with the correlational relations between symptoms of IBD and anxiety-related behaviors. We found dysfunctional up-regulation of a new inflammatory factor interleukin (IL)-19 in the colon of DSS/CUS treated mice. Overexpression of IL-19 in colon induced anxious phenotypes, and accelerated the anxious condition and symptoms of colitis in the DSS/CUS model by promoting the expression of inducible nitric oxide synthase (iNOS), IL-1β, and IL-6 pro-inflammatory factors, and activating signal transducer and activator of transcription 3 (STAT3) signaling pathway in the colon. Furthermore, overexpression of IL-19 in the colon also reduced the expression levels of brain-derived neurotrophic factor (BDNF), extracellular signal-regulated kinase (ERK), and cAMP-response element binding protein (CREB) signaling pathways activity in the hippocampus. These results suggest that IL-19 was a pivotal player in DSS/CUS-induced comorbidities of colitis and anxiety with different signaling pathways for the colon and hippocampus, which provides a candidate gene to explore the pathophysiology of comorbidities due to colitis and anxiety.
Collapse
Affiliation(s)
- Qiongyu Li
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Fantao Meng
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Xiangxian Ma
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, China
| | - Zhe Sun
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Juanjuan Dai
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
| | - Jing Liu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Dan Li
- College of Nursing, Binzhou Medical University, Binzhou, China
| | - Peijia Cong
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Ruixue Xu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Di Zhao
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Wentao Wang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Dan Wang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Cuilan Liu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Faxiang Wang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- *Correspondence: Chen Li, ; Haifeng Lian, ; Faxiang Wang,
| | - Chen Li
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
- *Correspondence: Chen Li, ; Haifeng Lian, ; Faxiang Wang,
| | - Haifeng Lian
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
- *Correspondence: Chen Li, ; Haifeng Lian, ; Faxiang Wang,
| |
Collapse
|
23
|
Matsumura S, Miyakita M, Miyamori H, Kyo S, Ishikawa F, Sasaki T, Jinno T, Tanaka J, Fujita K, Yokokawa T, Goto T, Momma K, Takenaka S, Inoue K. CRTC1 deficiency, specifically in melanocortin-4 receptor-expressing cells, induces hyperphagia, obesity, and insulin resistance. FASEB J 2022; 36:e22645. [PMID: 36349991 DOI: 10.1096/fj.202200617r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 10/06/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022]
Abstract
Melanocortin-4 receptor (MC4R) is a critical regulator of appetite and energy expenditure in rodents and humans. MC4R deficiency causes hyperphagia, reduced energy expenditure, and impaired glucose metabolism. Ligand binding to MC4R activates adenylyl cyclase, resulting in increased levels of intracellular cyclic adenosine monophosphate (cAMP), a secondary messenger that regulates several cellular processes. Cyclic adenosine monophosphate responsive element-binding protein-1-regulated transcription coactivator-1 (CRTC1) is a cytoplasmic coactivator that translocates to the nucleus in response to cAMP and is reportedly involved in obesity. However, the precise mechanism through which CRTC1 regulates energy metabolism remains unknown. Additionally, there are no reports linking CRTC1 and MC4R, although both CRTC1 and MC4R are known to be involved in obesity. Here, we demonstrate that mice lacking CRTC1, specifically in MC4R cells, are sensitive to high-fat diet (HFD)-induced obesity and exhibit hyperphagia and increased body weight gain. Moreover, the loss of CRTC1 in MC4R cells impairs glucose metabolism. MC4R-expressing cell-specific CRTC1 knockout mice did not show changes in body weight gain, food intake, or glucose metabolism when fed a normal-chow diet. Thus, CRTC1 expression in MC4R cells is required for metabolic adaptation to HFD with respect to appetite regulation. Our results revealed an important protective role of CRTC1 in MC4R cells against dietary adaptation.
Collapse
Affiliation(s)
- Shigenobu Matsumura
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan.,Department of Nutrition, Osaka Metropolitan University, Osaka, Japan
| | - Motoki Miyakita
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Haruka Miyamori
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Satomi Kyo
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Fuka Ishikawa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomoki Jinno
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Jin Tanaka
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kotomi Fujita
- Department of Nutrition, Osaka Metropolitan University, Osaka, Japan
| | - Takumi Yokokawa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Keiko Momma
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Shigeo Takenaka
- Department of Nutrition, Osaka Metropolitan University, Osaka, Japan
| | - Kazuo Inoue
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| |
Collapse
|
24
|
The altered sensitivity of acute stress induced anxiety-related behaviors by modulating insular cortex-paraventricular thalamus-bed nucleus of the stria terminalis neural circuit. Neurobiol Dis 2022; 174:105890. [DOI: 10.1016/j.nbd.2022.105890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/20/2022] Open
|
25
|
Kolesnikova IM, Gaponov AM, Roumiantsev SA, Karbyshev MS, Grigoryeva TV, Makarov VV, Yudin SM, Borisenko OV, Shestopalov AV. Relationship between Blood Microbiome and Neurotrophin Levels in Different Metabolic Types of Obesity. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022060229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
26
|
Hebebrand J, Hildebrandt T, Schlögl H, Seitz J, Denecke S, Vieira D, Gradl-Dietsch G, Peters T, Antel J, Lau D, Fulton S. The role of hypoleptinemia in the psychological and behavioral adaptation to starvation: implications for anorexia nervosa. Neurosci Biobehav Rev 2022; 141:104807. [PMID: 35931221 DOI: 10.1016/j.neubiorev.2022.104807] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/11/2022] [Accepted: 07/31/2022] [Indexed: 12/17/2022]
Abstract
This narrative review aims to pinpoint mental and behavioral effects of starvation, which may be triggered by hypoleptinemia and as such may be amenable to treatment with leptin receptor agonists. The reduced leptin secretion results from the continuous loss of fat mass, thus initiating a graded triggering of diverse starvation related adaptive functions. In light of leptin receptors located in several peripheral tissues and many brain regions adaptations may extend beyond those of the hypothalamus-pituitary-end organ-axes. We focus on gastrointestinal tract and reward system as relevant examples of peripheral and central effects of leptin. Despite its association with extreme obesity, congenital leptin deficiency with its many parallels to a state of starvation allows the elucidation of mental symptoms amenable to treatment with exogenous leptin in both ob/ob mice and humans with this autosomal recessive disorder. For starvation induced behavioral changes with an intact leptin signaling we particularly focus on rodent models for which proof of concept has been provided for the causative role of hypoleptinemia. For humans, we highlight the major cognitive, emotional and behavioral findings of the Minnesota Starvation Experiment to contrast them with results obtained upon a lesser degree of caloric restriction. Evidence for hypoleptinemia induced mental changes also stems from findings obtained in lipodystrophies. In light of the recently reported beneficial cognitive, emotional and behavioral effects of metreleptin-administration in anorexia nervosa we discuss potential implications for the treatment of this eating disorder. We postulate that leptin has profound psychopharmacological effects in the state of starvation.
Collapse
Affiliation(s)
- Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Tom Hildebrandt
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Haiko Schlögl
- Department of Endocrinology, Nephrology, Rheumatology, Division of Endocrinology, University Hospital Leipzig, Liebigstr. 20, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Str. 27, 04103 Leipzig, Germany
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH University Hospital Aachen, Germany
| | - Saskia Denecke
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Diana Vieira
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Gertraud Gradl-Dietsch
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Triinu Peters
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Jochen Antel
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - David Lau
- Department of Nutrition, Neuroscience - University of Montreal & CRCHUM, Montréal QC H3T1J4, Canada
| | - Stephanie Fulton
- Department of Nutrition, Neuroscience - University of Montreal & CRCHUM, Montréal QC H3T1J4, Canada
| |
Collapse
|
27
|
Identification of potential therapeutic and diagnostic characteristics of Alzheimer disease by targeting the miR-132-3p/FOXO3a-PPM1F axis in APP/PS1 mice. Brain Res 2022; 1790:147983. [PMID: 35709892 DOI: 10.1016/j.brainres.2022.147983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 11/22/2022]
Abstract
Alzheimer disease (AD) is a neurodegenerative disorder, which is characterized by progressive impairment of memory and cognition. Early diagnosis and treatment of AD has become a leading topic of research. In this study, we explored the effects of the miR-132-3p/FOXO3a-PPM1F axis on the onset of AD for possible early diagnosis and therapy. We found that miR-132-3p levels in the hippocampus and blood were drastically decreased in APP/PS1 mice from 9 months of age, and bi-directional manipulation of miR-132-3p levels induced magnified effects on learning memory behaviors, and manifestation of AD-related pathological characteristics and inflammatory cytokines in APP/PS1 mice of relevant ages. The hippocampal PPM1F expression levels were significantly elevated in APP/PS1 mice from 3 months of age, which was correlated with miR-132-3p levels at different ages. Overexpression of PPM1F remarkably accelerated the progression of learning memory deficits and associated pathological factors in APP/PS1 mice. Further, we showed that miR-132-3p modulated the expression of PPM1F via FOXO3a in HT22 cells. Finally, using peripheral blood samples of human study participants, we found that the miR-132-3p and PPM1F expression levels in patients with AD were also altered with prominent correlations. In conclusion, miR-132-3p indirectly regulates PPM1F expression by targeting FOXO3a, which could play an extensive role in contributing to the establishment of early diagnosis, treatment, and pathogenesis of AD.
Collapse
|
28
|
Chen LJ, Zhang NN, Zhou CX, Yang ZX, Li YR, Zhang T, Li CR, Wang X, Wang Y, Wang ZB, Xia ZR, Wang ZB, Zhang CL, Guan YC, Sun QY, Zhang D. Gm364 coordinates MIB2/DLL3/Notch2 to regulate female fertility through AKT activation. Cell Death Differ 2022; 29:366-380. [PMID: 34635817 PMCID: PMC8816931 DOI: 10.1038/s41418-021-00861-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 08/21/2021] [Accepted: 08/22/2021] [Indexed: 11/09/2022] Open
Abstract
Many integral membrane proteins might act as indispensable coordinators in specific functional microdomains to maintain the normal operation of known receptors, such as Notch. Gm364 is a multi-pass transmembrane protein that has been screened as a potential female fertility factor. However, there have been no reports to date about its function in female fertility. Here, we found that global knockout of Gm364 decreased the numbers of primordial follicles and growing follicles, impaired oocyte quality as indicated by increased ROS and γ-H2AX, decreased mitochondrial membrane potential, decreased oocyte maturation, and increased aneuploidy. Mechanistically, Gm364 directly binds and anchors MIB2, a ubiquitin ligase, on the membrane. Subsequently, membrane MIB2 ubiquitinates and activates DLL3. Next, the activated DLL3 binds and activates Notch2, which is subsequently cleaved within the cytoplasm to produce NICD2, the intracellular active domain of Notch2. Finally, NICD2 can directly activate AKT within the cytoplasm to regulate oocyte meiosis and quality.
Collapse
Affiliation(s)
- Liang-Jian Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Na-Na Zhang
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
- Center for Reproductive Medicine, the Third Affiliated Hospital of Zhengzhou University, 7 Rehabilitation Front Street, Zhengzhou, 450000, Henan, China
| | - Chun-Xiang Zhou
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
- Drum Tower Hospital Affiliated to Medical College of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Zhi-Xia Yang
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Yan-Ru Li
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
- Reproductive Medical Center, Henan Provincial People's Hospital & Reproductive Medical Center, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Teng Zhang
- State Key Lab of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Beijing, 100101, China
| | - Cong-Rong Li
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Xin Wang
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Yang Wang
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Zi-Bin Wang
- Analysis and Test Center, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Zheng-Rong Xia
- Analysis and Test Center, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Zhen-Bo Wang
- State Key Lab of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Beijing, 100101, China
| | - Cui-Lian Zhang
- Reproductive Medical Center, Henan Provincial People's Hospital & Reproductive Medical Center, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, Henan, China.
| | - Yi-Chun Guan
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China.
- Center for Reproductive Medicine, the Third Affiliated Hospital of Zhengzhou University, 7 Rehabilitation Front Street, Zhengzhou, 450000, Henan, China.
| | - Qing-Yuan Sun
- Fertility Preservation Lab and Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, 466 Xin-Gang-Zhong Road, Guangzhou, 510317, Guangdong, China.
| | - Dong Zhang
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
29
|
Leptin enhances social motivation and reverses chronic unpredictable stress-induced social anhedonia during adolescence. Mol Psychiatry 2022; 27:4948-4958. [PMID: 36138127 PMCID: PMC9763124 DOI: 10.1038/s41380-022-01778-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/12/2022] [Accepted: 09/02/2022] [Indexed: 01/19/2023]
Abstract
Social anhedonia, a loss of interest and pleasure in social interactions, is a common symptom of major depression as well as other psychiatric disorders. Depression can occur at any age, but typically emerges in adolescence or early adulthood, which represents a sensitive period for social interaction that is vulnerable to stress. In this study, we evaluated social interaction reward using a conditioned place preference (CPP) paradigm in adolescent male and female mice. Adolescent mice of both sexes exhibited a preference for the social interaction-associated context. Chronic unpredictable stress (CUS) impaired the development of CPP for social interaction, mimicking social anhedonia in depressed adolescents. Conversely, administration of leptin, an adipocyte-derived hormone, enhanced social interaction-induced CPP in non-stressed control mice and reversed social anhedonia in CUS mice. By dissecting the motivational processes of social CPP into social approach and isolation avoidance components, we demonstrated that leptin treatment increased isolation aversion without overt social reward effect. Further mechanistic exploration revealed that leptin stimulated oxytocin gene transcription in the paraventricular nucleus of the hypothalamus, while oxytocin receptor blockade abolished the leptin-induced enhancement of socially-induced CPP. These results establish that chronic unpredictable stress can be used to study social anhedonia in adolescent mice and provide evidence that leptin modulates social motivation possibly via increasing oxytocin synthesis and oxytocin receptor activation.
Collapse
|
30
|
Hebebrand J, Zorn S, Antel J, von Schnurbein J, Wabitsch M, Gradl-Dietsch G. First Account of Psychological Changes Perceived by a Female with Congenital Leptin Deficiency upon Treatment with Metreleptin. Obes Facts 2022; 15:730-735. [PMID: 35952649 PMCID: PMC9669943 DOI: 10.1159/000526169] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/22/2022] [Indexed: 11/19/2022] Open
Abstract
Two psychiatric interviews of a 39-year old female with congenital leptin deficiency were conducted to define psychological changes 14 and 165 days after initiation of treatment with human recombinant leptin (metreleptin). The most pronounced initial experience related to the reduced preoccupation with food. An improved mood was reported by the patient, which she associated with this reduced preoccupation. Her mood remained elevated upon recontact, whereas she was no longer preoccupied with food. Overall, the interviews provide a vivid account of the subjective experiences upon the initiation of treatment. Some of the findings bear a resemblance to those reported recently in patients with anorexia nervosa who were treated with metreleptin for 1-3 weeks. This case report provides further evidence that metreleptin has strong psychopharmacological effects in patients with absolute or relative leptin deficiency. We strongly recommend profound psychological examinations of patients with congenital leptin deficiency at baseline and after intitiation of treatment with human recombinant leptin to gain further insight into the functions affected by this hormone.
Collapse
Affiliation(s)
- Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stefanie Zorn
- Center for Rare Endocrine Diseases, Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Jochen Antel
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Julia von Schnurbein
- Center for Rare Endocrine Diseases, Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Martin Wabitsch
- Center for Rare Endocrine Diseases, Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
- *Martin Wabitsch,
| | - Gertraud Gradl-Dietsch
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- **Gertraud Gradl-Dietsch,
| |
Collapse
|
31
|
Prenatal Iron Deficiency and Choline Supplementation Interact to Epigenetically Regulate Jarid1b and Bdnf in the Rat Hippocampus into Adulthood. Nutrients 2021; 13:nu13124527. [PMID: 34960080 PMCID: PMC8706459 DOI: 10.3390/nu13124527] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 11/24/2022] Open
Abstract
Early-life iron deficiency (ID) causes long-term neurocognitive impairments and gene dysregulation that can be partially mitigated by prenatal choline supplementation. The long-term gene dysregulation is hypothesized to underlie cognitive dysfunction. However, mechanisms by which iron and choline mediate long-term gene dysregulation remain unknown. In the present study, using a well-established rat model of fetal-neonatal ID, we demonstrated that ID downregulated hippocampal expression of the gene encoding JmjC-ARID domain-containing protein 1B (JARID1B), an iron-dependent histone H3K4 demethylase, associated with a higher histone deacetylase 1 (HDAC1) enrichment and a lower enrichment of acetylated histone H3K9 (H3K9ac) and phosphorylated cAMP response element-binding protein (pCREB). Likewise, ID reduced transcriptional capacity of the gene encoding brain-derived neurotrophic factor (BDNF), a target of JARID1B, associated with repressive histone modifications such as lower H3K9ac and pCREB enrichments at the Bdnf promoters in the adult rat hippocampus. Prenatal choline supplementation did not prevent the ID-induced chromatin modifications at these loci but induced long-lasting repressive chromatin modifications in the iron-sufficient adult rats. Collectively, these findings demonstrated that the iron-dependent epigenetic mechanism mediated by JARID1B accounted for long-term Bdnf dysregulation by early-life ID. Choline supplementation utilized a separate mechanism to rescue the effect of ID on neural gene regulation. The negative epigenetic effects of choline supplementation in the iron-sufficient rat hippocampus necessitate additional investigations prior to its use as an adjunctive therapeutic agent.
Collapse
|
32
|
Liu Y, Wu M, Sun Z, Li Q, Jiang R, Meng F, Liu J, Wang W, Dai J, Li C, Jiang S. Effect of PPM1F in dorsal raphe 5-HT neurons in regulating methamphetamine-induced conditioned place preference performance in mice. Brain Res Bull 2021; 179:36-48. [PMID: 34871711 DOI: 10.1016/j.brainresbull.2021.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 11/02/2022]
Abstract
Methamphetamine (METH), a synthetically produced central nervous system stimulant, is one of the most illicit and addictive drugs worldwide. Protein phosphatase Mg2 + /Mn2 + -dependent 1F F (PPM1F) has been reported to exert multiple biological and cellular functions. Nevertheless, the effects of PPM1F and its neuronal substrates on METH addiction remain unclear. Herein, we first established a METH-induced conditioned place preference (CPP) mouse model. We showed that PPM1F is widely distributed in 5-HT neurons of the dorsal raphe nucleus (DRN), and METH treatment decreased the expression of PPM1F in DRN, which was negatively correlated with METH-induced CPP behaviors. Knockout of PPM1F mediated by adeno-associated virus (AAV) in DRN produced enhanced susceptibility to METH-induced CPP, whereas the overexpression of PPM1F in DRN attenuated METH-induced CPP phenotypes. The expression levels of Tryptophan hydroxylase2 (TPH2) and serotonin transporter (SERT) were down-regulated with a concurrent reduction in 5-hydroxytryptamine (5-HT), tryptophan hydroxylase2 (TPH2)-immunoreactivity neurons and 5-HT levels in DRN of PPM1F knockout mice. In the end, decreased expression levels of PPM1F were found in the blood of METH abusers and METH-taking mice. These results suggest that PPM1F in DRN 5-HT neurons regulates METH-induced CPP behaviors by modulating the key components of the 5-HT neurotransmitter system, which might be an important pathological gene and diagnostic marker for METH-induced addiction.
Collapse
Affiliation(s)
- Yong Liu
- Department of Physiology, Binzhou Medical University, Shandong, China; Medical research center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Min Wu
- Medical research center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Zongyue Sun
- Department of Physiology, Binzhou Medical University, Shandong, China.
| | - Qiongyu Li
- Medical research center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Rong Jiang
- Department of Physiology, Binzhou Medical University, Shandong, China.
| | - Fantao Meng
- Medical research center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Jing Liu
- Medical research center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Wentao Wang
- Medical research center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Juanjuan Dai
- Medical research center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Medical research center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Shujun Jiang
- Department of Physiology, Binzhou Medical University, Shandong, China.
| |
Collapse
|
33
|
Borsdorf B, Dahmen B, Buehren K, Dempfle A, Egberts K, Ehrlich S, Fleischhaker C, Konrad K, Schwarte R, Timmesfeld N, Wewetzer C, Biemann R, Scharke W, Herpertz-Dahlmann B, Seitz J. BDNF levels in adolescent patients with anorexia nervosa increase continuously to supranormal levels 2.5 years after first hospitalization. J Psychiatry Neurosci 2021; 46:E568-E578. [PMID: 34654737 PMCID: PMC8526129 DOI: 10.1503/jpn.210049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/14/2021] [Accepted: 07/01/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) influences brain plasticity and feeding behaviour, and it has been linked to anorexia nervosa in numerous studies. Findings in mostly adult patients point to reduced serum BDNF levels in the acute stage of anorexia nervosa and rising levels with weight recovery. However, it is unclear whether this increase leads to normalization or supranormal levels, a difference that is potentially important for the etiology of anorexia nervosa and relapse. METHODS We measured serum BDNF at admission (n = 149), discharge (n = 130), 1-year follow-up (n = 116) and 2.5-year follow-up (n = 76) in adolescent female patients with anorexia nervosa hospitalized for the first time, and in healthy controls (n = 79). We analyzed associations with body mass index, eating disorder psychopathology and comorbidities. RESULTS Serum BDNF was only nominally lower at admission in patients with anorexia nervosa compared to healthy controls, but it increased continuously and reached supranormal levels at 2.5-year follow-up. BDNF was inversely associated with eating disorder psychopathology at discharge and positively associated with previous weight gain at 1-year follow-up. LIMITATIONS We compensated for attrition and batch effects using statistical measures. CONCLUSION In this largest longitudinal study to date, we found only nonsignificant reductions in BDNF in the acute stage of anorexia nervosa, possibly because of a shorter illness duration in adolescent patients. Supranormal levels of BDNF at 2.5-year follow-up could represent a pre-existing trait or a consequence of the illness. Because of the anorexigenic effect of BDNF, it might play an important predisposing role for relapse and should be explored further in studies that test causality.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jochen Seitz
- From the Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH University Aachen, Germany (Borsdorf, Dahmen, Buehren, Scharke, Herpertz-Dahlmann, Seitz); the kbo-Heckscher Klinikum, Academic Teaching Hospital, Ludwig Maximilian University, Munich, Germany (Buehren); the Institute of Medical Informatics and Statistics, Kiel University, Germany (Dempfle); the Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Wuerzburg, Germany (Egberts); the Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Germany (Ehrlich); the Eating Disorders Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany (Ehrlich); the Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Freiburg, Germany (Fleischhaker); the Child Neuropsychology Section, Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH University Aachen (Konrad); the JARA-Institute Molecular Neuroscience and Neuroimaging (INM-11), Juelich Research Centre, Germany (Konrad); the Oberberg Fachklinik Konraderhof, Cologne-Huerth, Germany (Schwarte); the Institute of Medical Biometry and Epidemiology, Philipps-University Marburg, Germany (Timmesfeld); the Department of Medical Informatics, Biometrics and Epidemiology, Ruhr University Bochum, Germany (Timmesfeld); the Department of Child and Adolescent Psychiatry and Psychotherapy, Cologne City Hospitals, Germany (Wewetzer); the Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Germany (Biemann); the Cognitive and Experimental Psychology, Institute of Psychology, RWTH Aachen University, Germany (Scharke)
| |
Collapse
|
34
|
Meng F, Liu J, Dai J, Lian H, Jiang S, Li Q, Wu M, Wang W, Wang D, Zhao D, Liu C, Qiu C, Li C. PPM1F in Dentate Gyrus Modulates Anxiety-Related Behaviors by Regulating BDNF Expression via AKT/JNK/p-H3S10 Pathway. Mol Neurobiol 2021; 58:3529-3544. [PMID: 33745117 DOI: 10.1007/s12035-021-02340-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/22/2021] [Indexed: 12/15/2022]
Abstract
Anxiety is a serious psychiatric disorder, with a higher incidence rate in women than in men. Protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F), a serine/threonine phosphatase, has been shown to have multiple biological and cellular functions. However, the effects of PPM1F and its neuronal substrates on anxiety remain largely unclear. In this study, we showed that chronic restraint stress (CRS) induced anxiety-related behaviors only in female mice, while acute restraint stress (ARS) produced anxiety-related behaviors in both male and female mice in light-dark and elevated plus maze tests and induced upregulation of PPM1F and downregulation of brain-derived neurotrophic factor (BDNF) expression in the hippocampus. Adeno-associated virus-mediated overexpression of PPM1F or conditional knockout of BDNF in dentate gyrus (DG) led to a more pronounced anxiety-related behavior in female than in male mice as indicated by the behavioral evaluations. Meanwhile, overexpression of PPM1F in the DG decreased total Bdnf exon-specific messenger RNA expression in the hippocampus with the decreased binding activity of phosphorylated H3S10 to its individual promoters in female mice. Furthermore, we identified that overexpression of PPM1F decreased the phosphorylation levels of AKT and JNK in the hippocampus of female mice. These results may suggest that PPM1F regulates anxiety-related behaviors by modulating BDNF expression and H3S10 phosphorylation-mediated epigenetic modification, which may be served as potentially pathological genes associated with anxiety or other mental diseases.
Collapse
Affiliation(s)
- Fantao Meng
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256603, Shandong, China
| | - Jing Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256603, Shandong, China
| | - Juanjuan Dai
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Haifeng Lian
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shujun Jiang
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - Qiongyu Li
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Min Wu
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wentao Wang
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256603, Shandong, China
| | - Dan Wang
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256603, Shandong, China
| | - Di Zhao
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256603, Shandong, China
| | - Cuilan Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256603, Shandong, China
| | - Changyun Qiu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256603, Shandong, China
| | - Chen Li
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, 256603, Shandong, China.
| |
Collapse
|
35
|
Zhao D, Liu C, Cui M, Liu J, Meng F, Lian H, Wang D, Hu F, Liu D, Li C. The paraventricular thalamus input to central amygdala controls depression-related behaviors. Exp Neurol 2021; 342:113744. [PMID: 33965409 DOI: 10.1016/j.expneurol.2021.113744] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/20/2021] [Accepted: 05/04/2021] [Indexed: 02/08/2023]
Abstract
The dysregulation of neuronal networks may contribute to the etiology of major depressive disorder (MDD). However, the neural connections underlying the symptoms of MDD have yet to be elucidated. Here, we observed that glutamatergic neurons in the paraventricular thalamus (PVT) were activated by chronic unpredictable stress (CUS) with higher expression numbers of ΔFosB-labeled neurons and protein expression levels, activation of PVT neurons caused depressive-like phenotypes, whereas suppression of PVT neuronal activity induced an antidepressant effect in male, but not female mice, which were achieved by using a chemogenetic approach. Moreover, we found that PVT glutamatergic neurons showed strong neuronal projections to the central amygdala (CeA), activation of the CeA-projecting neurons in PVT or the neuronal terminals of PVT-CeA projection neurons induced depression-related behaviors or showed enhanced stress-induced susceptibility. These results suggest that PVT is a key depression-controlling nucleus, and PVT-CeA projection regulates depression-related behaviors in a sex-dependent manner, which could be served as an essential pathway for morbidity and treatment of depression.
Collapse
Affiliation(s)
- Di Zhao
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Cuilan Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Minghu Cui
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Haifeng Lian
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Dan Wang
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fengai Hu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Dunjiang Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
36
|
Wang D, Wang W, Jiang S, Ma H, Lian H, Meng F, Liu J, Cui M, You J, Liu C, Zhao D, Hu F, Liu D, Li C. Regulation of depression-related behaviors by GABAergic neurons in the lateral septum through periaqueductal gray neuronal projections. J Psychiatr Res 2021; 137:202-214. [PMID: 33691232 DOI: 10.1016/j.jpsychires.2021.02.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 01/28/2021] [Accepted: 02/17/2021] [Indexed: 12/25/2022]
Abstract
Major depressive disorder (MDD) is a serious and widespread mental illness worldwide. The abnormality of neuronal networks may contribute to the etiology of MDD. However, the neural connections underlying the main symptoms of MDD need further elucidation. Here, we found that GABAergic neurons in the lateral septum (LS) were activated by chronic unpredictable stress (CUS), with increased numbers of ΔFosB-labeled neurons. LS neuronal activity was modulated using a chemogenetic approach. Activation of LS neurons caused a depressive phenotype, as shown by increased immobility in the forced swim test, and induced increased susceptibility to subthreshold chronic stress, as indicated by decreased female urine sniffing time and preference for sucrose in depression-related behavior detection, whereas suppression of LS neuronal activity induced an antidepressant effect under basal and stressed conditions. Moreover, we found that the LS showed strong neuronal projections to the dorsal periaqueductal gray (dPAG); activation of dPAG-projecting GABAergic neurons in the LS produced the same depressive behaviors and stress susceptibility as induced by the activation of the majority of LS GABAergic neurons. Finally, we found that activation of neuronal fibers in the dPAG derived from the LS showed depression-related behaviors, as suggested by the decreased female urine sniffing time and sucrose preference in female urine sniffing and sucrose preference tests respectively. Our findings indicate that LS is a key depression-controlling nucleus, and that the LS-PAG projection is an essential effector circuit for morbidity and treatment in depression.
Collapse
Affiliation(s)
- Dan Wang
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wentao Wang
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shujun Jiang
- Department of Physiology, Binzhou Medical University, Yantai, China
| | - He Ma
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Haifeng Lian
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Minghu Cui
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jingjing You
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Cuilan Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Di Zhao
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fengai Hu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Dunjiang Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
37
|
Irving A, Harvey J. Regulation of hippocampal synaptic function by the metabolic hormone leptin: Implications for health and disease. Prog Lipid Res 2021; 82:101098. [PMID: 33895229 DOI: 10.1016/j.plipres.2021.101098] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Significant advances have been made in our understanding of the hormone, leptin and its CNS actions in recent years. It is now evident that leptin has a multitude of brain functions, that extend beyond its established role in the hypothalamic control of energy balance. Additional brain regions including the hippocampus are important targets for leptin, with a high density of leptin receptors (LepRs) expressed in specific hippocampal regions and localised to CA1 synapses. Extensive evidence indicates that leptin has pro-cognitive actions, as it rapidly modifies synaptic efficacy at excitatory Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 synapses and enhances performance in hippocampal-dependent memory tasks. There is a functional decline in hippocampal responsiveness to leptin with age, with significant reductions in the modulatory effects of leptin at SC-CA1 and TA-CA1 synapses in aged, compared to adult hippocampus. As leptin has pro-cognitive effects, this decline in leptin sensitivity is likely to have negative consequences for cognitive function during the aging process. Here we review how evaluation of the hippocampal actions of leptin has improved our knowledge of the regulatory brain functions of leptin in health and provided significant insight into the impact of leptin in age-related neurodegenerative disorders linked to cognitive decline.
Collapse
Affiliation(s)
- Andrew Irving
- School of Biomolecular and Biomedical Science, The Conway Institute, University College Dublin, Dublin, Ireland
| | - Jenni Harvey
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom.
| |
Collapse
|
38
|
PPM1F in hippocampal dentate gyrus regulates the depression-related behaviors by modulating neuronal excitability. Exp Neurol 2021; 340:113657. [PMID: 33639208 DOI: 10.1016/j.expneurol.2021.113657] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/28/2020] [Accepted: 02/21/2021] [Indexed: 01/21/2023]
Abstract
Major depressive disorder (MDD) is a common, serious, debilitating mental illness. Protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F), a serine/threonine phosphatase, has been reported to have multiple biological and cellular functions. However, the effects of PPM1F and its neuronal substrates on depressive behaviors remain largely unknown. Here, we showed that PPM1F is widely distributed in the hippocampus, and chronic unpredictable stress (CUS) can induce increased expression of PPM1F in the hippocampus, which was correlated with depression-associated behaviors. Overexpression of PPM1F mediated by adeno-associated virus (AAV) in the dentate gyrus (DG) produced depression-related behaviors and enhanced susceptibility to subthreshold CUS (SCUS) in both male and female mice, while, knockout of PPM1F in DG produced antidepressant phonotypes under stress conditions. Whole-cell patch-clamp recordings demonstrated that overexpression of PPM1F increased the neuronal excitability of the granule cells in the DG. Consistent with neuronal hyperexcitability, overexpression of PPM1F regulated the expression of certain ion channel genes and induced decreased phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CAMKII) and Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) in hippocampus. These results suggest that PPM1F in the DG regulates depression-related behaviors by modulating neuronal excitability, which might be an important pathological gene for depression or other mental diseases.
Collapse
|
39
|
Li M, Geng R, Li C, Meng F, Zhao H, Liu J, Dai J, Wang X. Dysregulated gene-associated biomarkers for Alzheimer's disease and aging. Transl Neurosci 2021; 12:83-95. [PMID: 33623715 PMCID: PMC7885957 DOI: 10.1515/tnsci-2021-0009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/20/2020] [Accepted: 01/18/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer’s disease (AD), the most common type of dementia, is a neurodegenerative disorder with a hidden onset, including difficult early detection and diagnosis. Nevertheless, the new crucial biomarkers for the diagnosis and pathogenesis of AD need to be explored further. Here, the common differentially expressed genes (DEGs) were identified through a comprehensive analysis of gene expression profiles from the Gene Expression Omnibus (GEO) database. Furthermore, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that these DEGs were mainly associated with biological processes, cellular components, and molecular functions, which are involved in multiple cellular functions. Next, we found that 9 of the 24 genes showed the same regulatory changes in the blood of patients with AD compared to those in the GEO database, and 2 of the 24 genes showed a significant correlation with Montreal Cognitive Assessment scores. Finally, we determined that mice with AD and elderly mice had the same regulatory changes in the identified DEGs in both the blood and hippocampus. Our study identified several potential core biomarkers of AD and aging, which could contribute to the early detection, differential diagnosis, treatment, and pathological analysis of AD.
Collapse
Affiliation(s)
- Min Li
- Department of Neurology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
| | - Rongxin Geng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430000, China
| | - Chen Li
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Fantao Meng
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Hongwei Zhao
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Jing Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Juanjuan Dai
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Xuezhen Wang
- Department of Neurology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
| |
Collapse
|
40
|
Wang K, Zhai Q, Wang S, Li Q, Liu J, Meng F, Wang W, Zhang J, Wang D, Zhao D, Liu C, Dai J, Li C, Cui M, Chen J. Cryptotanshinone ameliorates CUS-induced depressive-like behaviors in mice. Transl Neurosci 2021; 12:469-481. [PMID: 34900345 PMCID: PMC8633587 DOI: 10.1515/tnsci-2020-0198] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/19/2021] [Accepted: 11/02/2021] [Indexed: 12/26/2022] Open
Abstract
Objectives Cryptotanshinone (CPT), a natural quinoid diterpene, isolated from Salvia miltiorrhiza, has shown various pharmacological properties. However, its effect on chronic unpredictable stress (CUS)-induced depression phenotypes and the underlying mechanism remain unclear. Therefore, the aim of this study was to investigate whether CPT could exert an antidepressant effect. Methods We investigated the effects of CPT in a CUS-induced depression model and explored whether these effects were related to the anti-inflammatory and neurogenesis promoting properties by investigating the expression levels of various signaling molecules at the mRNA and protein levels. Results Administration of CPT improved depression-like behaviors in CUS-induced mice. CPT administration increased the levels of doublecortin-positive cells and reversed the decrease in the expression levels of brain-derived neurotrophic factor (BDNF)/tyrosine kinase receptor B (TrkB) signaling transduction, as well as the downstream functional proteins, phosphorylated extracellular regulated protein kinases (p-ERK), and cyclic adenosine monophosphate (cAMP)-response element-binding protein levels (p-CREB) in hippocampus. CPT treatment also inhibited the activation of microglia and suppressed M1 microglial polarization, while promoting M2 microglial polarization by monitoring the expression levels of arginase 1 (Arg-1) and inducible nitric oxide synthase (iNOS), and further inhibited the expression of proinflammatory cytokines, including interleukin (IL)-1, IL-6, and tumor necrosis factor-α (TNF-α), and increased the expression of the anti-inflammatory cytokine IL-10 by regulating nuclear factor-κB (NF-κB) activation. Conclusions CPT relieves the depressive-like state in CUS-induced mice by enhancing neurogenesis and inhibiting inflammation through the BDNF/TrkB and NF-κB pathways and could therefore serve as a promising candidate for the treatment of depression.
Collapse
Affiliation(s)
- Kaixin Wang
- Department of Neurology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China.,Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Department of Internal Medicine, Jinan Hospital, Jinan, Shandong, China
| | - Qingling Zhai
- Department of Neurology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China.,Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Sanwang Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
| | - Qiongyu Li
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wentao Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jinjie Zhang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Dan Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Di Zhao
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Cuilan Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Juanjuan Dai
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.,Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Minghu Cui
- Department of Psychology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
| | - Jinbo Chen
- Department of Neurology, Binzhou Medical University Hospital, No. 661 Huanghe 2nd Road, Binzhou, Shandong, 256603, China
| |
Collapse
|